Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 May 31.
Published in final edited form as: Expert Rev Proteomics. 2021 May 31;18(5):379–394. doi: 10.1080/14789450.2021.1931130

Evolution of proteomics technologies for understanding Respiratory Syncytial Virus pathogenesis

Morgan Mann 1, Allan R Brasier 2
PMCID: PMC8277732  NIHMSID: NIHMS1711120  PMID: 34018899

Abstract

Introduction:

Respiratory syncytial virus (RSV) is a major human pathogen associated with long term morbidity. RSV replication occurs primarily in the epithelium, producing a complex cellular response associated with acute inflammation and long-lived changes in pulmonary function and allergic disease. Proteomics approaches provide important insights into post-transcriptional regulatory processes including alterations in cellular complexes regulating the coordinated innate response and epigenome.

Areas covered:

Peer-reviewed proteomics studies of host responses to RSV infections and proteomics techniques were analyzed. Methodologies identified include 1).” bottom-up” discovery proteomics, 2). Organellar proteomics by LC-gel fractionation; 3). Dynamic changes in protein interaction networks by LC-MS; and 4). selective reaction monitoring MS. We introduce recent developments in single-cell proteomics, top-down mass spectrometry, and photo-cleavable surfactant chemistries that will have impact on understanding how RSV induces extracellular matrix (ECM) composition and airway remodeling.

Expert Opinion:

RSV replication induces global changes in the cellular proteome, dynamic shifts in nuclear proteins, and remodeling of epigenetic regulatory complexes linked to the innate response. Pathways discovered by proteomics technologies have led to deeper mechanistic understanding of the roles of heat shock proteins, redox response, transcriptional elongation complex remodeling and ECM secretion remodeling in host responses to RSV infections and pathological sequelae.

Keywords: Affinity purification (AP)-MS, Bromodomain containing protein 4 (BRD4), Extracellular Matrix (ECM), Global bottom up proteomics, Immunoprecipitation-MS, Innate Immune Response, Label-free proteomics, liquid chromatography (LC)-mass spectrometry (MS), photo-cleavable surfactant, Respiratory Syncytial Virus (RSV), Stable Isotopic Dilution (SID), Selective Reaction Monitoring (SRM), 4‐hexylphenylazosulfonate

1. Introduction.

1.1. Respiratory Syncytial Virus.

The goal of this review is to examine the contribution of mass spectrometry-based proteomics technologies to the study of respiratory syncytial virus (RSV) infection, and to describe new methods and technologies in this field which could be applied to improve our understanding of this pathogen and its host response. RSV is an enveloped negative-sense RNA virus that circulates seasonally worldwide. Most often, RSV causes mild, cold-like symptoms, but in the very old and very young, the virus spreads into the lower airways, causing bronchiolitis or pneumonia. Worldwide, RSV was associated with 33.1 million episodes of lower respiratory tract infections (LRTIs), 3.2 million RSV-related hospital admissions, and 118,000 deaths in children less than 5 years of age, predominantly in developing countries [1]. Consequently, RSV infection is responsible for the majority of pediatric hospitalizations of young children [2].

RSV is an Orthopneumovirus belonging to the Pneumoviridae family [3], that forms a filamentous enveloped virion encapsulating a single-stranded, negative sense genome of approximately 15.2 kb in length [4]. Molecular epidemiological studies have shown that epidemic RSV infections occur seasonally, and represent co-circulation of two principal strains of RSV- known as RSV-A and RSV-B viruses [5]. These strains are due to antigenic variations of a hypervariable region of the G glycoprotein. Within each subtype are multiple clades of genetic variants whose abundance and composition are determined by local fitness, transmissability and immune-driven evolution [5]. Recently, rapidly transmissible RSV genotypes have been identified and represent the dominant forms of RSV strains. These include an RSV-A subtype with a 72 nt duplication of G (ON genotype) and an RSV-B subtype with a 60 nt duplication (BA genotype) [6,7]. Although these variants are highly transmissible, it is unclear whether these are more pathogenic than others; however, other genetic RSV variants elicit distinct patterns of immune responses, such as Line 19, a variant that elicits a more robust IL-13 driven T-helper type 2 response than that produced by RSV-A2 and Long strains [8]. The lifecycle of RSV strains are thought to be identical and illustrated in Figure 1.

Figure 1. Life cycle and cellular responses to RSV infection.

Figure 1.

RSV infects epithelial cells in the respiratory tract. Upon binding to extracellular receptors, RSV fuses with the epithelial membrane, forming a fusion pore, permitting the capside entry into the cytoplasm. The RSV replication complex forms on cellular membranes triggering formation of inclusion bodies. After transcription, genome replication results in mature virus that buds from the cell cytoplasm. In the infected cell, pattern recognition receptors trigger an innate immune response that involves the production of reactive oxygen species, as well as the secretion of protective IFNs, cytokines, and exosomes. The application of proteomics technologies has elucidated key cellular responses to RSV infection, including activation of anti-oxidant homeostasis, dynamic regulation of transcriptional complexes in the IIR, and cell type differences in secreted proteins (secretome).

RSV virions are covered in attachment (G) and fusion (F) glycoproteins with a lesser amount of small hydrophobic (SH) proteins within the envelope. RSV F and G mediate viral attachment and entry into cells; RSV primarily infects epithelial cells of the respiratory tract as its initial target [3]. A number of cellular receptors have been identified for RSV including the CX3 chemokine receptor, CXC3CR1[9], Toll like receptor-4 [10], nucleolin [11] and IGF1R [12]. RSV F mediates fusion between the viral envelope and host membrane; because of its relatively invariant structure and is a target of neutralizing antibody formation, RSV F is one of the major targets for anti-viral drug development [13]. Membrane mixing permits the formation of a fusion pore, resulting in delivery of the virion capsid into the host cell, consisting of the nucleocapsid (N), L and P proteins (Figure 1). The RNA genome encodes for the key internal structural proteins (matrix protein [M] and nucleoprotein [N]), proteins required for a functional polymerase complex (phosphoprotein [P] and polymerase [L]), A viral replication complex then forms on internal cellular membranes, consisting of the polymerase (L), P and M2–1 proteins encoded by RSV, along with N protein-encapsulated viral genomic RNA.

The polymerase complex transcribes the nonstructural proteins (NS-1 and NS-2) involved in evasion of the innate immune response, transmembrane glycoproteins (small hydrophobic protein [SH], glycoprotein [G], and fusion protein [F]), and the regulatory M2 proteins (M2–1 antitermination protein and M2–2, involved in transcription/replication regulation [4]. After transcription of the RSV-encoded proteins, the polymerase switches modes and produces genome. The assembled virus then buds from the infected cell, incorporating components of the host plasma membrane.

Productive infection of the respiratory epithelium induces multiple cellular responses, including the formation of inclusion bodies, reactive oxygen stress, and rapid activation of the innate immune response (IIR). IBs appear to be both a site of RSV replication as well a mechanism for modulating the intracellular IIR [14]. A hallmark of the IIR involves resulting in secretion of cytokines [15], interferons [16], exosomes [17], and damage-associated patterns [18] that mediate mucous production and leukocytic inflammation. After viral clearance, immunity is partial and incomplete. Consequently, reinfections occur throughout life, typically causing upper respiratory tract infections. In the elderly, nosocomial RSV infections cause pneumonia. With repetitive infections, innate inflammatory signaling can result in airway remodeling [19]. Airway remodeling involves physical changes to the airways, including enhanced production of extracellular matrix (ECM) and mucous production, producing obstruction, reducing effective airflow and lung function [20]. These changes are significant and irreversible [21].

Presently, no vaccines are available to prevent RSV infection. Both formalin-inactivated and live-attenuated viruses have failed to induce protective responses in clinical trials, and in-fact, have generated exaggerated immune responses [2225]. In the case of a 1969 formalin-inactivated RSV vaccine trial, this resulted in a 80% hospitalization rate in the vaccine-treated infants compared to only 5% in controls [26]. Monoclonal antibodies and pharmaceutical prophylactics are available for high-risk infants, but they are expensive and have questionable efficacy [27,28]. In light of the high prevalence of RSV and severe consequences of repeated infection, it is clear that additional research is required to identify long-term solutions. In this review, we will describe the application of proteomics technologies and how these studies have elucidated key cellular responses to RSV infection, including activation of anti-oxidant homeostasis, dynamic regulation of transcriptional complexes in the IIR, and cell type differences in secreted proteins (secretome).

1.2. Mass Spectrometry & Proteomics.

RSV-infection alters cell physiology through complex and multifaceted mechanisms, including activation of the innate immune reaction through TLR3 Retinoic Acid Inducible Gene I (RIG-I) and other pattern-recognition receptors [16,29], resulting in altered transcription, post-translational modification of regulatory proteins, and epigenetic chromatin regulation [30]. Additionally, perturbations to the extracellular environment and non-infected cells play significant roles in the development of disease pathology [31]. While genomic and transcriptomic approaches can probe some of these changes at the level of gene expression, they are unsuitable for studying the true abundance of regulated proteins and post-translational modifications [32].

In contrast, Mass Spectrometry (MS)-based proteomics methodologies are capable of unambiguously and precisely quantifying changes to both protein expression and the relative abundance of most post-translational modifications (PTMs). In traditional “bottom-up” proteomic analysis, proteins are isolated and digested using trypsin or another protease [33]. The resulting peptides are then sequenced using tandem mass spectrometry, generally coupled to liquid chromatography (LC-MS) to reduce sample complexity. This method is sensitive and highly specific. More importantly, it is unbiased and high-throughput, which makes MS-based proteomics ideal for examining the cellular effects of RSV-infection at a systems biology perspective.

1.3. Survey of Shotgun Proteomics Approaches.

Bottom-up or “Shotgun” proteomics is highly versatile, and when combined with different sample preparation and molecular biology techniques, can be applied to extract an incredible diversity of information from biological systems. Most relevant to the discussion of RSV biology are its applications to protein and PTM quantitation from whole cell lysates, subcellular (e.g. nuclear and cytoplasmic) fractions, tissue matrices, and protein interactomes.

The simplest of these methods involves the direct extraction and digestion of protein from whole cells or tissue, which are then submitted to MS analysis for identification and quantitation. For convenience, we will refer to all such applications as “global bottom-up proteomics” (GBUP). The first RSV-oriented GBUP studies used 2-Dimensional gel electrophoresis (2DE) to separate proteins based on intact mass and isoelectric properties [3436]. Protein stains, (e.g. Coomassie R-250, Sypro-Ruby) were then used to identify differentially abundant proteins, which were subsequently excised, digested in-gel, and identified via Matrix-assisted Laser Desorption Ionization – Time of Flight (MALDI-TOF) mass spectrometry and Peptide Mass Fingerprinting (PMF). Notably, PMF identifies proteins by matching the intact masses of tryptic peptides – not by peptide sequencing; resulting in generally high false-identification rates [37]. With advances in liquid chromatography and mass spectrometers over the last decade, such gel-based methods are no longer the primary tool for quantitative studies [38], nevertheless, they contributed important insights to the field of RSV biology.

More recently, reverse-phase liquid chromatography (RPLC), coupled to high-resolution mass spectrometry has become the standard methodology for bottom-up proteomics. This enables the detection of significantly more proteins and peptides than gel-based methods by improving the front-end separation as compared to 2DE; highly-reproducible LC also facilitates direct, or “label free” quantitation of peptides and proteins via extracted ion profiles [39]. Alternative methods of quantitation have also been applied to RSV research, most notably isobaric quantitation methods such as Stable Isotope Labeling by Amino Acids in Cell Culture (SILAC) [40], which enables direct binary comparisons within a single mass spectrum by altering the mass of otherwise identical peptides from different samples.

GBUP strategies are easily applied to study subcellular and organellar proteomes. Airway tissue is similarly accessible [41,42], but extracellular matrix proteins require additional processing steps for efficient extraction and digestion [4346]. Finally, GBUP can be coupled to common molecular biology affinity purification techniques; Affinity-purification- Mass Spectrometry (AP-MS) allows for deep and unbiased profiling of protein interactomes [47]. We summarize these approaches in Figure 2.

Figure 2. Schematic of Bottom-up Proteomics approaches for studying RSV biology.

Figure 2.

Post-infection with RSV, airway cells, tissue, and extracellular fluids (including Bronchoalveolar Lavage Fluid and Nasopharyngeal Aspirates) are subjected to extracellular matrix (ECM) enrichment, immunoprecipitation, subcellular enrichment, or lysed whole to harvest protein. The extracts are digested using trypsin or another protease, and the resulting peptides are analyzed via LC-MS to identify proteins and post-translational modifications and facilitate quantitation.

One challenge of GBUP is the quantitation of low-abundance proteins. Most commonly, mass spectrometers acquire a limited number of peptides for sequencing, fragmenting only the most abundant precursor ions within a single mass spectrum. In this process (termed “Data-dependent Acquisition”), low abundance peptides and proteins may be missed. To address this, two alternative strategies may be employed: Data-independent acquisition (DIA), which attempts to fragment all precursor ions in a given mass spectrum [48], but greatly increases the complexity of data analysis; and Selected- or Multiple-Reaction Monitoring (SRM/MRM), which targets specific precursor ions for fragmentation and sequencing regardless of their relative abundance in the sample [49], but has relatively low throughput and requires prior knowledge of the target proteins and peptides in the sample. Isotope labeled peptide standards may also be employed to ensure specific identification and accurate quantitation.

2. Applications of MS-based proteomics to cell culture-based RSV systems.

Many RSV-related proteomics studies have been conducted using in-vitro culture systems with malignant, KRAS-mutation transformed cells – most commonly A549 type 2 alveolar epithelial cells. A549 cells are highly differentiated adenocarcinoma cells that maintain characteristics of alveolar type II epithelial cells [50]. These cells are highly permissive for RSV replication [51], express type I and III interferons and cytokines upon RSV infection [15], have endogenous TLR3 and RIG-I pattern recognition receptors [16], and elaborate an integrated interferon response factor (IRF)-nuclear factor kB (NFκB) pathway [5254]. These cells therefore are a valid model system of understanding pathogenesis of RSV-induced lower respiratory tract infections that has provided information on cell stress response validated in animal models and human infections. Other cell types, such as Hep2, have been used. Hep2 are derived from laryngeal carcinoma cells but have been shown to have HeLa contamination [55]. Fewer studies have used primary airway epithelial cultures, despite the benefits of the more human-like model, as historically, their added expense and relatively short lifespan in culture have imposed severe limitations on many typical workflows [56]. In recent years, LC-MS sensitivity has increased to the point where efficient label-free quantitation of primary cells is possible [5759], but these benefits have not been applied to RSV models. For many of the same reasons, multicellular lung organoids – which may recapitulate multicellular interactions seen in pulmonary biology [60,61] - have yet to be applied to RSV proteomics. Nevertheless, some findings from transformed cell lines have been validated in primary cell cultures [17,62] and in-vivo [63,64], lending credence to these models. Another important consideration in the interpretation of these studies is that the cellular models are typically done with high multiplicity of infection (MOIs), so that most cells are actively infected or repetitively infected at the time of analysis.

2.1. Heat Shock Proteins are regulated by RSV infection and are required for viral polymerase activity.

Understanding the protein composition of cellular organelles and how these change in response to RSV infection provides an added dimension of information into the homeostatic response to virus and how the virus may manipulate these changes to enhance its replication. Consequently, substantial effort has been devoted to the characterization of airway epithelial cell nuclear proteome.

In one early approach to address this, subcellular fractionation, high-performance liquid chromatography, 2DE, and MALDI-TOF MS were combined to deeply probe the nuclear proteome of A549 cells and quantify its response to RSV infection [65]. Overall, RSV induced changes to 24 nuclear proteins controlling chromatin remodeling, protein refolding, cytoskeletal structure, membrane function, metabolic processes, mitochondrial function, RNA binding, protein synthesis, signaling, and transcription factor activities. These included large upregulations in the abundance of cytokeratins, heat shock proteins (Hsp60/Hsp70), and components of Nuclear Domain 10 (ND10) structures. These results were used to inform confocal immunofluorescence microscopy experiments which demonstrated that RSV infection disrupted nuclear ND10 structures while simultaneously inducing cytoplasmic aggregation and nuclear accumulation of Hsp70.

In short succession, some of these results were confirmed and given biological significance. McDonald, et. al., also in 2004, demonstrated that the RSV viral polymerase associates with lipid rafts on the cell’s plasma membrane [66]. They accomplished this by isolating lipid rafts via a flotation gradient, and analyzing tryptic peptides via 2-Dimensional nanoflow LC-MS (2D-nLC-MS); this strategy utilized two complementary chromatography methods (Strong Anion Exchange and Reverse-phase Liquid Chromatography) to reduce sample complexity and bypass the low sensitivity of gel-based methods; this maximized the number of proteins that they could identify in their analysis, and enabled them to detect the RSV-L protein, which lacks a suitable antibody for immunoblotting. In 2005, the same group leveraged their 2D-nLC-MS method to show that Hsp70 associated with these raft/polymerase complexes [67]. Furthermore, they demonstrated that anti-Hsp70 antibodies could reduce viral polymerase activity, indicating that the interaction was functional and required for efficient viral replication.

In 2015, this result was further fleshed out by Munday, et. al., who used affinity purification via EGFP-labeled viral proteins, coupled to nanoflow LC-MS, to discover host-cell protein interactors of the viral replication complex [68]. In this AP-MS analysis, they identified numerous heat shock proteins that interacted with the RSV L-protein, including Hsp70 and Hsp90. When they treated their cell culture system with 17-AAG (an Hsp90 inhibitor) they found that the global abundance of cellular RSV proteins and corresponding mRNA plummeted. Finally, they tested the necessity of Hsp70 for viral transcription using an Hsp70 inhibitor and a gel-based cell extract assay, and observed that HSP70 inhibition was required for RSV polymerase activity on an intact nucleocapsid template. These results collectively demonstrated that the RSV infection induces increased expression of Heat Shock Proteins which are in-turn required for viral polymerase activity.

2.2. Elucidation of oxidative stress response mechanisms by subcellular enrichment and MALDI-TOF MS.

RSV replication induces an oxidative stress response in vitro and in vivo, a phenomenon linked to activation of transcription factors important in the innate inflammatory response. In particular, RSV-infection has been shown to inhibit expression of cytoplasmic antioxidants, including Glutathione S-transferase (GST), superoxide dismutase (SOD), and catalase. Despite a deep body of transcriptional regulation, a holistic understanding including post-transcriptional regulation of the viral response has been lacking.

To address this, Jamaluddin, et. al. combined in-solution Isoelectric Focusing (EIF) followed by 2DE and MALDI-TOF peptide mass fingerprinting to examine the A549 nuclear proteome in greater depth and determine its responses to RSV-infection [35]. To enable more accurate quantitation, free cysteine side chains were labeled with fluorescent dyes. In addition to reproducing the results of Brasier, et. al., these authors found that RSV induced shifts to the isoelectric points of several peroxiredoxin (Prdx) isoforms, without changing their apparent abundance. Notably, an siRNA knockdown model demonstrated that cells lacking Prdx-1, Prdx-4, or both, showed increased levels of reactive oxygen species (ROS) and protein carbonylation.

To follow up on this, a novel proteomics labeling technique was developed and applied to covalently link free cysteine side chains with fluorescent dyes to systematically identify proteins sensitive to RSV-induced cysteinyl oxidation in the presence or absence of Prdx-1, Prdx-4, or both. Saturation fluorescence labeling of unoxidized cysteine side chains followed by quantification using 2DE fractionation identified 15 unique proteins, including Lam A, annexin, and centromere E-associated protein that had enhanced oxidative modifications in the Prdx-depleted nuclei. These studies concluded that Prdx-1 and Prdx-4 are essential for preventing oxidative damage in a subset of nuclear intermediate filament and actin binding proteins in RSV-infected epithelial cells.

Identifying the interaction between nuclear oxidant stress response and cytoskeletal changes contributed the following insights to the field: 1). Cytoskeletal reorganization of filamentous actin is required for RSV transcription, assembly and budding. 2). RSV disrupts cellular antioxidant proteins. 3). Subcellular isolation followed by HPLC prefractionation significantly enhanced identification of nuclear processes affected by virus replication.

2.3. The RSV-Nonstructural protein 1 interferes with type I and II interferon expression by suppressing RIG-I activation.

The RSV nonstructural (NS) proteins are small, non-packaged viral proteins that are well-established to inhibit host type I (e.g. IFN-α, IFN-ß) and type III interferon (e.g. IFN-λ) signaling and regulate NF-κB signaling [6976]. However, their mechanisms of action were incompletely understood until recently. To address this knowledge gap, Hastie and colleagues used 2DE and MALDI-TOF/TOF mass spectrometry to identify the effects of a recombinant, NS1-deficient RSV virus on the whole cell proteome of A549 cells [77]. They found that 22 proteins that were differentially abundant between the NS1-deficient and wild-type viruses, including Superoxide Dismutase-2 (SOD2) and several other known type II IFN-inducible proteins. Further investigation using cytokine-treated A549 cells showed that these same proteins were induced by both type I IFNs and type II IFNs (e.g. IFN-γ), but not TNF-α or IL1ß.

Using a type I IFN-deficient Vero cell model, the investigators went on to show that NS1-deficient RSV-induced changes to SOD2 and other markers could be induced in a type I IFN-independent manner, thereby implicating NS1 in blocking type II IFN regulation. The authors additionally showed that this mechanism was independent of the NRF2 oxidative stress response pathway (see Section 3.1), which could also induce expression of SOD2 alongside catalase and Thioredoxin reductase. The authors speculated that this behavior was consistent with targeting of the RIG PRR by NS1 – an observation that has been borne out by later mechanistic studies [78].

2.4. LC-MS analysis indicates mitochondrial disfunction in ROS production and broadens the understanding of anti-viral responses to RSV infection.

In addition to the 2DE/MALDI-TOF studies of RSV infection, several studies have been conducted using online LC-MS. Unfortunately, most such studies omitted biological and/or technical replicates, making their interpretation difficult in relation to other studies. Nevertheless, some specific results were confirmed using additional methods, such as immunoblotting and confocal microscopy, and are worth acknowledging.

In 2010, Munday, et. al. published two papers employing SILAC to quantify RSV-induced changes to both the cytoplasmic and nuclear proteomes of Hep2 cells [79,80]. In these studies, the authors identified concerted reductions to the abundances of mitochondrial proteins such as the Translocase of the Outer Mitochondrial membrane (TOM) complex (e.g. Tom20, Tom22, Tom40, Tom70), as well as Voltage-dependent Anion Channels (VDACs; e.g. VDAC-1, VDAC2, VDAC-3). These changes were induced by both RSV subtypes, although no direct comparison of the two was conducted. Their findings led the investigators to propose that RSV infection altered the permeability of mitochondrial transition pores; a hypothesis that they then confirmed using a live-cell mitochondrial permeability assay. These results suggested that mitochondrial dysfunction may contribute to RSV-induced oxidative damage. A third study from this group applied SILAC to mitochondrial extracts from RSV-infected A549 cells, and confirmed that infection altered the abundances of TOM proteins [81]. The authors demonstrated that knockdown of Tom70 increased viral titer, further indicating a role for these proteins during viral infection.

Somewhat later, Ternette et. al. contributed to these experiments by combining in-solution IEF with LC-MS and Data-independent Analysis (DIA) more deeply probe the whole-cell proteome of RSV-infected A549 cells [82]. Their data quantified over 1000 proteins, reproduced the findings of many earlier studies, and highlighted the cellular accumulation of the Interferon-induced protein with tetratricopeptide repeats 3 (IFIT3) protein, supporting the activation of the interferon response. They also observed differential isoelectric mobility of the 5’-3’-exoribonuclease 2 (XRN2) protein, which they attributed to post-translational modification.

In 2014, Dave et. al. conducted a rigorous examination of A549 whole cell lysates post-RSV infection, utilizing five biological replicates and contrasting the performance of IEF pre-separation with a fractionation-free workflow prior to LC-MS [83]. This study flagged extensive alterations to IFN-induced proteins, and pathway analysis demonstrated that the vast majority (83%) of cellular proteins with differential regulation were controlled by IFNs, Tumor-necrosis Factor Alpha, and NF-κB. Notably, this work indicated that RSV infection induced an IFN-γ response in A549 cells, despite the prevailing belief that A549 epithelial cells should not produce that cytokine. Finally, this study also demonstrated the degree to which proteomics technologies had advanced over the years; using their fractionation-free workflow, the investigators quantified over 5000 proteins from whole cell lysates and five biological replicates; a staggering improvement to what was possible only a few years before.

2.5. Multiplexed quantitative proteomics measurements of the innate immune response.

It is well-established that RSV infection induces type I and III mucosal interferon (IFN) production with paracrine activation of protective, anti-viral interferon stimulated genes (ISGs) [8486]. This results in dynamic nuclear translocation and cytoplasmic shuttling of innate transcription factors and regulatory kinases, as well as expression of pathway inhibitors. However, quantification of this response is extremely challenging. First, the major regulatory components of the IIR, consisting of PRRs, kinases, and transcription factors, are all low-abundance proteins [87,88]. Second, few high affinity detection reagents are available that can reliably detect them in complex cellular samples. Third, classical “shotgun” proteomics is limited by stochastic sampling of a fraction of the proteome in a manner that is usually biased toward higher abundance proteins [33]. Consequently, many important low-abundance proteins are usually not consistently identified across samples. Such fragmentary data sets are not satisfactory for understanding the signaling pathway at the systems level.

These problems have illustrated the need for high-accuracy, high- sensitivity multiplex assays for measuring dynamic changes in the IIR. One such approach is the selective reaction monitoring (SRM)-MS assay. SID-SRM-MS assays for 10 major regulators of IIR (including RelA, IRF3, and RIG-I) have been developed and reported [89]. Application of SID-SRM method reproducibly measured the concentrations of these proteins in cytoplasmic and nuclear compartments of A549 cells over a time course of ds-RNA stimulation, revealing differences in translocation kinetics between associated transcription factors. These studies also elucidated, for the first time, the existence of a negative NF-kB-IRF3 cross-talk pathway, and the consumption of the RIG-I/Tank Binding Kinase (TBK1) “signalsome” in IRF3 signaling.

Coupling the SID-SRM method with affinity enrichment using antibodies or single-stranded DNA aptamers results in dramatic enhancement of the assay sensitivity by enhancing the signal-to-noise ratio [90]. Using an ssDNA aptamer (termed P028F4) that binds to the activated (IκBα-dissociated) form of RelA with a K(D) of 6.4 × 10−10, coupled to SID-SRM MS assay was reported. This assay produced a linear response over 1,000 fold dilution range of input protein and had a 200 amol lower limit of quantification, enabling the amount of nucleoplasmic and chromatin associated RelA could be estimated in response to stimulation. The aptamer-SID-SRM-MS assay quantified the fraction of activated RelA in subcellular extracts, detecting the presence of a cytoplasmic RelA reservoir unresponsive to TNFα stimulation.

In a separate study, Tian, et. al. used these SRM and IP-SRM assays to show that RSV-induced inflammation triggers a dynamic interaction between the pro-inflammatory NF-κB RelA subunit with the epigenetic scaffold Bromodomain-containing Protein 4 [29]. Small molecule BRD4 inhibitors (JQ1+) and siRNA depletion of BRD4 further demonstrated that this interaction was necessary for RSV-induced activation of NF-κB dependent cytokines, but was independent of viral replication. Ultimately these results contributed the investigators’ novel finding that BRD4 coupled NF-κB to the IRF/RIG-I anti-viral amplification loop by contributing to the formation of active transcriptional elongation complexes on NF-κB-dependent pro-inflammatory genes.

2.6. Dynamic Protein Interaction monitoring of RSV regulated epigenetic scaffolds by PASEF-MS

In addition to mediating RSV-induced inflammation, the BRD4 epigenetic scaffold is also associated with the development of airway remodeling and fibrosis, and bromodomain inhibitors prevent downstream airway remodeling in murine models [19,9195]. However, the mechanism of this action is currently unclear.

In a recent study, Morgan Mann et al. examined the dynamic interactome of BRD4 in an attempt to provide context for this phenomenon [96]. They tested their hypothesis that RSV-infection would trigger BRD4 to interact with transcription factors that regulate inflammation and remodeling by examining the makeup of immunopurified BRD4 complexes isolated from A549 cells infected with RSV and treated with a specific BRD4 inhibitor (ZL 0454). To facilitate deep proteomic profiling despite the low cellular abundance of BRD4, the authors employed Parallel Accumulation – Serial Fragmentation MS (PASEF-MS), which utilizes Trapped Ion Mobility separation (TIMS) as an additional mechanism to reduce sample complexity prior to MS-analysis. Additionally, the TIMS dimension increases sensitivity and throughput by focusing ion signals and excluding non-peptide contaminants from fragmentation and sequencing.

In this study, the investigators identified 557 potential BRD4 interactors, and found that a staggering 305 were responsive to RSV-infection, of which 272 demonstrated increased relative abundance on the BRD4 complex post-infection; 95 of these proteins were also disrupted from the RSV-activated complex by the BRD4 inhibitor (Figure 3). Interestingly, these proteins were highly enriched in transcriptional regulators, including members of the AP1 transcription factor complex (i.e. c-JUN, AP1), and the Wnt-signaling pathway (i.e. β-Catenin, γ-Catenin). AP1 is a transcription factor complex involved in cell survival, apoptosis, and inflammation [97,98]; critically, it also contributes to the expression of Interleukin-6 – a key pro-inflammatory cytokine induced by RSV-infection [99,100]. Similarly, the Catenin proteins are transcription factors that are highly correlated with activation of the epithelial-to-mesenchymal transition (EMT) and are linked to asthmatic airway remodeling [101106]. Furthermore, treatment with the BRD4 inhibitor resulted in the partial eviction of c-JUN and the complete eviction of both β-Catenin and γ-Catenin from the BRD4 complex. These results indicated that RSV-infection triggers interactions between BRD4 and pro-inflammation/remodeling transcription factors and suggested the disruption of those interactions was responsible for the observed effects of BRD4 inhibitors in vivo. In a broader sense, this experiment also perfectly demonstrated the unique capability of modern proteomics technologies to analyze entire networks of proteins in a single experiment.

Figure 3. RSV-infection induces dynamic and bromodomain-dependent interactions to the host cellular protein BRD4.

Figure 3.

Cytoscape network visualization of RSV-inducible and bromodomain-dependent BRD4 interactors. Node color represents the RSV-inducible Log2 fold change. Edge color is keyed to the STRING score and represents interaction confidence. Low confidence interactions (STRING score < 0.4) and nodes without interactions are omitted. Reproduced with permission from [96].

3. Proteomics studies conducted in alternative models of RSV-infection.

Owing to the experimental control of the timing and extent of infection, in-vitro cell culture systems – especially A549 alveolar epithelial cells - have dominated the field of RSV-proteomics [34,35,6668,77,82,83,107]. Nevertheless, RSV-infection is a tissue-level event, affecting numerous cell types that can act in concert, and producing marked changes to the extracellular matrix and extracellular fluids [17,30]. Accordingly, investigations focusing on models outside of the petri dish have significant value, and have produced notable data contributing to our understanding of RSV pathogenesis.

3.1. RSV depletion of antioxidant capacity is linked to disease pathogenesis.

In 2011, Hosakote, et. al. continued the work started by Jamaluddin, et. al in 2010 by demonstrating that RSV infection induced a significant decrease in the expression and/or activity of SOD, catalase, GST, and glutathione peroxidase in murine lungs and in the airways of children with severe bronchiolitis [64]. The mechanism of RSV-induced reductions to the expression of these antioxidant genes was determined by mechanistic studies that demonstrated RSV induced degradation of transcription factor Nuclear Factor, Erythroid 2 Like 2 (NRF2), a transcription factor that normally maintains expression of a cluster of antioxidant enzymes including catalase [108]. Catalase is a key enzyme for the dismutation of virus-mediated generation of hydrogen peroxide (H2O2).

To test whether this disruption of this antioxidant pathway was relevant to disease progression, SOD mimetics and stabilized polyethylene glycol-conjugated catalase were administered in an experimental rodent model of RSV infection [109,110]. These manipulations resulted in reduced viral H2O2 production and produced a significant protective effect on RSV-induced inflammation, clinical disease and airway pathology. Collectively, these studies indicated that strategies to augment the host antioxidant pathway may be an important strategy to reduce manifestations of RSV infection.

3.2. Tissue proteomics indicates that RSV Vaccine-related eosinophilia acts through Chil3 and Integrin alpha-M.

In 2015, van Diepen, et. al. turned their attention to the issue of RSV-vaccine-mediated enhanced disease (VMED) [111]. They measured host proteome correlates of VMED by examining the lung proteome of mice challenged with RSV and primed with vaccine preparations based on the RSV F and G-proteins. These vaccine preparations were based on the recombinant vaccinia virus (rVV), and were known to produce phenotypically different host immune responses corresponding to VMED [112,113]. For this model, it had been shown that the rVV-G preparation resulted in the production of neutralizing antibodies and a subsequent reduction to RSV replication, but also produced severe illness and pulmonary eosinophilia. In contrast, the rVV-F preparation resulted in a more effective host defense without pulmonary eosinophilia.

When the lung tissue of the mice was harvested and analyzed, Diepen, et. al. found that rVV-G-treated mice had significantly greater levels of Chitinase-like-protein 3 (Chil3) and Integrin alpha-M (Itgam). Chil3 is a secretory protein most often produced by macrophages and neutrophils, and has chemotactic properties for T lymphocytes, bone marrow polymorphonuclear lymphocytes, and eosinophils [114,115]. Importantly, Chil3 has been demonstrated to trigger extravasation of eosinophils, and mRNA knockdown of Chil3 has been shown to block eosinophilia in a murine model of airway hyperreactiveness [116]. Similarly, Itgam is a component of the Complement Receptor Type 3 (CR3) protein, and facilitates adhesion of neutrophils and eosinophils [117]. This data suggested that Chil3 and Itgam were mechanistically responsible for the high level of eosinophilia observed in the lungs of rVV-G-treated mice.

3.3. GBUP demonstrates the presence of anti-viral and pro-ROS species in BAL fluid and nasopharyngeal aspirates.

In regard to extracellular proteomics, two recent studies have examined the proteome of bovine bronchoalveolar lavage fluid (BALF) and human nasopharyngeal aspirates, respectively. The bovine study, conducted by Häggland, et. al. used nLC-MS to examine the effect of RSV-infection on gauze-filtered BALF from RSV-vaccinated and unvaccinated cows [118]. Between all comparisons, they identified 96 proteins that were associated with the bovine RSV-disease, mostly associated with neutrophil activation and chemotaxis (e.g. Resistin, Elastase, S100-A9) and homeostatic responses to ROS (e.g. SOD, Prdx-6). Markers of lymphocyte and natural killer cell activation/chemotaxis were also enriched by RSV-infection, alongside epithelial cell markers.

The major findings in this study were that lung pathology and histological scoring correlated heavily with increased abundance of neutrophil activation markers and reduced abundance of antioxidants. A secondary finding was that bovine RSV was negatively correlated with Leucine Zipper TFL1, which has been shown to inhibit the epithelial-to-mesenchymal transition (EMT) [119] – potentially establishing a link to the inflammation-associated phenomena of airway remodeling.

Along a different angle, Aljabr, et. al., examined the proteome of nasopharyngeal aspirates from RSV-infected children, and used a combined proteomics and transcriptomics approach to characterize the disease response for potential biomarker development [120]. As has become standard, nLC-MS was applied to profile tryptic peptides derived from the samples, and the group found significant increases to numerous proteins corresponding to interferon-stimulated genes (e.g. ISG15, ICAM1, IFIT1, OAS2), and the Bactericidal/Permeability-Increasing (BPI) proteins, which have been shown to have anti-viral properties in the context of influenza [121].

3.4. Secretome Proteomics reveals cell-type differences in paracrine signaling proteins.

Recent studies have illuminated the findings that airway epithelial cells are derived from multiple lineages and are functionally distinct in their viral responses [122]. Gene-profiling experiments have suggested that lower airway epithelial cells produce greater amounts of T helper type 2 (Th2)-activating CCL-type chemokines than do epithelial cells of the conducting airways [15,123]. Because cellular proteomics undersample secreted proteins and factors Zhao et al. applied a highly sensitive unbiased secretome profiling technique to identify free and membrane-bound exosomes [17]. After developing the workflow and analysis pipeline in immortalized small airway epithelial cells, the profiling was conducted in primary cultures of human airway epithelial cells.

Approximately 1,000 high confidence protein identifications in cell supernatants at a 1% false discovery rate. These common proteins were derived from lysosomal, cytoplasmic, and nuclear compartments. Approximately one third of secretome proteins were exosomal; the remainder were from lysosomal and vacuolar compartments. These investigators also identified 577 differentially expressed proteins (by cell type) from control supernatants and 966 differentially expressed proteins from RSV-infected. Interestingly, many of these secreted proteins do not contain classic signal peptides, suggesting novel and incompletely understood mechanisms controlling innate signaling. For example, the nuclear damage-associated molecular patterns (DAMPs) HMGB1 and histone H3 undergo nuclear export and extracellular secretion in response to RSV; these DAMPs control mononuclear inflammation [18].

Focusing informatics analysis on the 103 proteins secreted by lower airway bronchiolar cells, Th2 activating cytokines (MIP1, TSLP), mucin expressing (CCL20), and fibrogenic cytokines (IL6) were identified (Figure 4). Importantly, all of these factors are dependent on NFκB, further implicating NFκB in immunopathogenesis and remodeling in RSV lower respiratory tract infection. This data underscores the cell type influences on paracrine signaling by the IIR.

Figure 4. Identification of differentially secreted proteins in response to RSV infection.

Figure 4.

(A). Statistical analysis of microarray (SAM) for secretome proteins whose expression differs by cell type in the basal state. X axis, expectation score; Y axis, observation score. The diagonal line shows where false discovery rate = 0.01. Points above (in red) outside the threshold are those with FDR < 0.01, and points below the threshold (in green) are those proteins with FDR > 0.01. Proteins with increased expression in hSAECs are indicated by red points; those decreased are indicated in green. (B). SAM for secretome proteins whose expression di ers due to RSV infection. (C). Expression values of proteins were z-score-normalized data of log2-transformed expression values for biological replicates. Two-dimensional hierarchical clustering was performed, with columns representing cell samples and rows representing individual proteins (green, low expression, red, high expression). Cluster 3 is unique to the bronchiolar-derived human small airway epithelial cells (HSAECs). HSAECs differentially express CCL20, TSLP, IL6, and MIP1. Reproduced with permission from [17].

4.0. Underutilized and unexplored proteomics approaches for RSV-research.

Thus far, we have discussed proteomics technologies and approaches that have been well applied to RSV-research. However, the field of proteomics is fast-moving, and several new technologies and recent advances merit discussion here for their potential applications towards RSV-biology. In this section, we discuss Single-cell proteomics, extracellular matrix proteomics, and Top-down mass spectrometry.

4.1. Single-cell proteomics.

GBUP approaches have provided detailed overviews of RSV-induced changes to cellular protein abundances. However, these measurements represent bulk protein, averaged over millions of cells [124]. Even in cell culture systems with identical genomic backgrounds, stochastic influences can result in significant heterogeneity at the transcript and protein level, and there is a growing consensus that this heterogeneity is an important metric for understanding biological systems.

Historically, proteomics techniques have lagged behind similar genomic and transcriptomic methods [125], and while single-cell sequencing has been available for many years, single-cell proteomics has yet to reach its stride. Nevertheless, rapid advances have been made in the field, aided by improvements in sample preparation, LC-separation, and the application of isobaric labeling and quantitation techniques [126,127]. At present, the state of the art is capable of identifying and quantifying over 1500 proteins from single HeLa cells [128], which already eclipse the number of proteins quantified in early RSV proteomics studies. Notably, Hela cells have similar total protein content to A549 cells, opening the door to analysis of airway biological conditions, such as RSV-infection [129,130]. Further technical advances in the next several years should be anticipated to increase sensitivity even more, potentially allowing analysis of primary cells and stem cells.

While single-cell proteomics is certainly out of reach to most laboratories, the findings so far emphasize the importance of these measurements. A recent preprint from the Matthias Mann group compared single cell transcriptomics to their recently developed single cell proteomics strategy, and found significant disagreement between the two methods [128]; in general, the proteome was far more stable than the transcriptome. Given that viral infection modulates both transcriptional activity and protein abundance, both types of information will likely be required to decipher the actual effects of RSV-infection at single-cell resolution. This will be especially important for investigations into the proteomic landscape of partially infected cells and tissues, as opposed to current cell culture models that use very high MOIs.

4.2. FFPE Proteomics

Formalin-fixed Parrafin-embedded (FFPE) tissue is commonly collected in both clinical and research settings to observe pathological changes to tissue [131]. Owing to the high stability of parrafin-embedded tissue and its ease of storage, significant archives of FFPE tissue have accumulated in the past century. Nevertheless, these samples were historically considered inaccessible to proteomic analysis, owing to the molecular crosslinking induced by formaldehyde. However, this assumption has proven false in the last decade [132,133], and numerous groups since then have developed sophisticated extraction techniques to perform high-throughput characterization of FFPE tissue sections [134136].

In the context of RSV biology, FFPE lung sections have been used in mouse models to characterize the extent of virus-mediated damage and airway remodeling [137], and human samples are sometimes obtained during autopsy [138,139]. These samples represent a rich, but as-of-yet untapped source of data regarding the mechanisms of RSV-mediated remodeling and pathogenesis. Furthermore, in combination with laser-capture microdissection - which has been coupled to FFPE proteomic analysis [132,140] - it is possible to isolate specific cell populations for analysis, and probe differences between tissues with high- and low-grade pathology within a single tissue sample. This development opens the door for sophisticated and high-power experiments to characterize how cellular heterogeneity contributes to RSV pathogenesis.

4.3. Proteomics of extracellular matrices.

Chronic RSV-infection leads to irreversible, fibrotic changes to airway structures and extracellular matrices (ECM), collectively referred to as airway remodeling [21]. However, measuring ECM proteins via proteomics is far more difficult than measuring cellular or secreted proteins, as the various ECM-components form complex macromolecular structures with low solubility in aqueous solution [141143]. In addition, many have proven resistant to typical proteolytic digestion strategies employed [144]. Meanwhile, the workflows developed to alleviate these issues are labor intensive compared to cellular GBUP experiments [4345]. This has resulted in an unfortunate knowledge gap, and to our knowledge, no RSV-oriented ECM proteomics study has been published.

Fortunately, recent advances in surfactant chemistries and sample preparation methodologies show promise in alleviating these issues. In 2020, Knott, et. al. published a streamlined and improved ECM proteomics strategy employing a photocleavable surfactant (4-Hexylphenylazosulfonate) [46]. Using this strategy in combination with 1- and 2-dimensional liquid chromatography, they examined the ECM proteome of mouse mammary tumors, and identified multiple novel PTMs. Importantly, they rigorously established the reproducibility of the method to ensure applicability in biological contexts.

4.4. Post-translational Modifications and Top-down mass spectrometry.

Protein post-translational modifications provide an additional degree of complexity to the discussion of RSV biology, as phosphorylation, ubiquitination, and numerous other PTMs can modulate the function and abundance of key signaling molecules [145]. While the bottom-up proteomics studies described thus far have generally provided useful quantitative data regarding protein abundance, the same cannot be said regarding RSV-relevant post-translational modifications. This is especially glaring given that several studies indirectly detected RSV-induced changes to PTMs via altered protein isoelectric points, and yet the topic has seen no additional attention.

This deficiency most likely reflects the generally low cellular abundance of post-translationally modified proteins compared to their unmodified variants, and the chemical difficulties involved in preserving them throughout long sample-handling and LC-MS workflows. The most commonly applied fragmentation method for peptide sequencing further complicates the matter, as Collision-induced Dissociation (CID) can remove labile PTMs, including phosphorylation, from peptides in a neutral-loss event that is not always obvious in a tandem mass spectrum [146]. In studies where phosphopeptides are expected, the use of Electron-capture or Electon-transfer Fragmentation is recommended (ETD & ETF).

An additional avenue worth exploring is the application of intact protein mass spectrometry to the problem. Intact protein MS, or “Top-down” MS contrasts with the usual “bottom-up” approach in that proteins are not digested with a protease prior to analysis [147,148]. This often requires different extraction techniques, chromatography, and MS-instrumentation, but can simplify sample preparation workflows and preserve sensitive post-translational modifications [149] . In addition, modified protein variants (“proteoforms”) are chemically similar, which enables direct quantitation of PTM stoichiometry. Accordingly, top-down mass spectrometry has been applied to deeply characterize the post-translational modifications of numerous proteins, including muscle tissue proteins [150,151], phosphoproteins [152], and glycoproteins [153155]. For identical reasons, Top-down may also prove useful in examining proteins which are difficult to detect in bottom-up workflows, such as the RSV-G protein.

5.0. Expert Opinion

RSV alters cell biology through complex and multi-faceted mechanisms, with the common phenotypes representing a convergence of numerous factors and signaling pathways. In the face of such complexity, traditional biochemical methods for protein quantitation are ill-suited to make new discoveries, due to their low sensitivity and throughput. MS-based proteomics technologies, in contrast, have proven highly useful in filling in the blanks of RSV-biology. Over the course of nearly twenty years, RSV-oriented proteomics studies have cast a wide net over the cellular protein landscape, resulting in significant findings related to RSV viral replication and transcription. Additional studies have also shed light on host protein correlates of the disease, implicating neutrophils and oxidative damage as key drivers of the disease.

Over this period, MS and related technologies have advanced rapidly, progressing from crude gel-based methods with relatively low sensitivity and throughput to sophisticated unions of mass spectrometry, liquid chromatography, and gas-phase separation, that maximize the proteomic depth and coverage of any given analysis. We expect that this pace will continue, incorporating new technologies and methods such as single-cell and top-down proteomics.

5.1. Strengths

Compared to traditional biochemical approaches for protein quantitation, MS-based proteomics methodologies are incredibly specific, sensitive, and high-throughput. This enables effective quantitation of low-abundance transcription factors and cellular regulators, as well as proteins (e.g. RSV-L) that do not have specific antibodies available for immunodetection. Furthermore, the multiplexed measurements obtained lend themselves well to bioinformatics analysis, enabling recognition of entire biological pathways and protein complexes that are regulated during RSV infection. At present, this combination of advantages is entirely unique to MS-based proteomics.

MS analysis can also serve as a detection endpoint for numerous biological assays; bottom-up proteomics has been robustly applied to global measurement of both cellular and extracellular protein abundances, with interactome analysis also facilitated by affinity purification. Many post-translational modifications can also be preserved and quantified in an unbiased proteomic analysis.

5.2. Weaknesses

Although the mass spectrometer can be coupled to most sample preparation workflows, this often requires additional, labor-intensive steps. While automation and recent developments in sample preparation methodologies have made great progress in overcoming these difficulties, the additional workload may still be intimidating to some researchers.

In addition, the bottom-up proteomics workflows covered by this review are often insufficient to comprehensively study post-translational modifications, owing to their sometimes-uncooperative chemical properties and typically low relative abundance compared to unmodified peptides. While top-down mass spectrometry offers a powerful tool to circumvent the usual limitations, it is technically complicated and low throughput in comparison to bottom-up analysis. Furthermore, it may be overwhelmed by highly modified and diverse proteoforms.

Although many of the cellular models of RSV infection have advanced understanding of host responses to RSV infection used alveolar epithelial-like cells, such as A549, applications of high resolution and sensitive proteomics to primary cells and organoids will lead to additional discoveries from these more differentiated systems.

Finally, the cellular studies reviewed here were performed at high MOIs, an experimental design intentionally to reveal response to viral replication. Little is understood about the paracrine response of uninfected cells in the airways. Single cell proteomic designs using low MOIs will be illuminating studies.

5.3. Potential Opportunities

Immediate research opportunities largely represent applications of new technologies and approaches to older problems. PASEF and similar ion mobility-mass spectrometry workflows facilitates a massive increase to sensitivity and proteome coverage and may enable high-throughput quantitation of RSV-relevant proteins with low sample abundances, or from samples derived from primary cells and other protein sources traditionally avoided due to high cost or low protein content. Meanwhile, laser microdissection and FFPE proteomics workflows may enable nuanced analysis of the effect of tissue and cell-type heterogeneity on RSV-pathogenesis in-vivo.

Additionally, no study has directly compared the effects of the RSV subgroups or different genotypes within a subgroup on the cellular proteome. It is speculated that genomic differences contribute to altered virulence between strains, and proteomic changes may offer insights into the drivers of increased virulence.

5.4. Five-year view.

Over the next five years, we anticipate that single-cell proteomics studies will become sufficiently comprehensive and applicable to tissue to allow unbiased analysis of RSV-induced protein changes with cell-type resolution, and dissection of effects of direct replication vs paracrine signaling. ECM proteomics will be highly complementary to these studies and allow analysis of changes to airway structure and fibrosis. Finally, as top-down proteomics becomes more common and approachable, more comprehensive identification of changes in phosphorylation, oxidation, s-nitrosylation and other pathways will be illuminating.

Collectively, we believe that these developments will enable future studies to deconvolute the complex web of inter-cell signaling present in-vivo RSV-infections. The prospect of more completely deciphering the mechanisms of RSV-induced airway remodeling, for example, is extremely exciting, and appears to be on the horizon. Vaccine development will also benefit, as top-down MS facilitates deeper analysis of RSV-proteins and their relevant post-translational modifications

Article Highlights:

  • Unbiased proteomic profiling studies have discovered viral replication effects on nuclear dot 10 (ND10) structures, heat shock responses and perturbations of anti-oxidant defenses important in disease pathogenesis. These post-translationally regulated homeostatic responses are invisible to gene expression profiling.

  • Affinity purification-MS enables focused studies of dynamic changes in protein-protein interaction networks controlling innate immune response (IIR).

  • Advances selective reaction monitoring to profile of dynamic changes in abundance and subcellular distribution of IIR regulators opens opportunities for understanding how RSV subverts anti-viral responses.

  • Emerging studies implicate RSV replication has substantial effects the formation and remodeling of the extracellular matrix (ECM). New developments in photocleavable Azo dyes are described with the potential to unlock discoveries in ECM remodeling.

  • Advances in proteomics, including top down and single-cell proteomics will advance understanding of post-translational modifications and enable dissection of direct-versus-paracrine effects of viral replication.

Acknowledgments

The authors thank the University of Wisconsin–Madison Human Proteome Program for access to equipment, training and guidance. The authors also thank the NIAID-AI062885-funded IBVC Core. Figure 2 was created with BioRender.com.

Funding

This work was partially supported by NIH grants AI062885 (ARB), NCATS UL1TR002373 (ARB) and R01 AI136994 (ARB). The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript; or in the decision to publish the results.

Footnotes

Declaration of interest

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

References

Papers of special note have been highlighted as:

* of interest

** of considerable interest

  • 1.Shi T, McAllister DA, O’Brien KL et al. Global, regional, and national disease burden estimates of acute lower respiratory infections due to respiratory syncytial virus in young children in 2015: a systematic review and modelling study. Lancet, 390(10098), 946–958 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Group PERfCHPS. Causes of severe pneumonia requiring hospital admission in children without HIV infection from Africa and Asia: the PERCH multi-country case-control study. Lancet, 394(10200), 757–779 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Rima B, Collins P, Easton A et al. ICTV Virus Taxonomy Profile: Pneumoviridae. J Gen Virol, 98(12), 2912–2913 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Collins PL, Fearns R, Graham BS. Respiratory syncytial virus: virology, reverse genetics, and pathogenesis of disease. Current topics in microbiology and immunology, 372, 3–38 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Pandya MC, Callahan SM, Savchenko KG, Stobart CC. A Contemporary View of Respiratory Syncytial Virus (RSV) Biology and Strain-Specific Differences. Pathogens, 8(2) (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Trento A, Galiano M, Videla C et al. Major changes in the G protein of human respiratory syncytial virus isolates introduced by a duplication of 60 nucleotides. J Gen Virol, 84(Pt 11), 3115–3120 (2003). [DOI] [PubMed] [Google Scholar]
  • 7.Agoti CN, Otieno JR, Gitahi CW, Cane PA, Nokes DJ. Rapid spread and diversification of respiratory syncytial virus genotype ON1, Kenya. Emerg Infect Dis, 20(6), 950–959 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lukacs NW, Moore ML, Rudd BD et al. Differential Immune Responses and Pulmonary Pathophysiology Are Induced by Two Different Strains of Respiratory Syncytial Virus. The American Journal of Pathology, 169(3), 977–986 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Tripp RA, Jones LP, Haynes LM, Zheng H, Murphy PM, Anderson LJ. CX3C chemokine mimicry by respiratory syncytial virus G glycoprotein. Nat Immunol, 2(8), 732–738 (2001). [DOI] [PubMed] [Google Scholar]
  • 10.Kurt-Jones EA, Popova L, Kwinn L et al. Pattern recognition receptors TLR4 and CD14 mediate response to respiratory syncytial virus. Nature Immunology, 5, 398–401 (2000). [DOI] [PubMed] [Google Scholar]
  • 11.Tayyari F, Marchant D, Moraes TJ, Duan W, Mastrangelo P, Hegele RG. Identification of nucleolin as a cellular receptor for human respiratory syncytial virus. Nat Med, 17(9), 1132–1135 (2011). [DOI] [PubMed] [Google Scholar]
  • 12.Griffiths CD, Bilawchuk LM, McDonough JE et al. IGF1R is an entry receptor for respiratory syncytial virus. Nature, 583(7817), 615–619 (2020). [DOI] [PubMed] [Google Scholar]
  • 13.McLellan JS, Ray WC, Peeples ME. Structure and function of respiratory syncytial virus surface glycoproteins. Current topics in microbiology and immunology, 372, 83–104 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Lifland AW, Jung J, Alonas E, Zurla C, Crowe JE, Santangelo PJ. Human Respiratory Syncytial Virus Nucleoprotein and Inclusion Bodies Antagonize the Innate Immune Response Mediated by MDA5 and MAVS. Journal of Virology, 86(15), 8245–8258 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Zhang Y, Luxon BA, Casola A, Garofalo RP, Jamaluddin M, Brasier AR. Expression of respiratory syncytial virus-induced chemokine gene networks in lower airway epithelial cells revealed by cDNA microarrays. J Virol, 75(19), 9044–9058 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Liu P, Jamaluddin M, Li K, Garofalo RP, Casola A, Brasier AR. Retinoic Acid-Inducible Gene I Mediates Early Antiviral Response and Toll-Like Receptor 3 Expression in Respiratory Syncytial Virus-Infected Airway Epithelial Cells. The Journal of Virology, 81(3), 1401–1411 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zhao Y, Jamaluddin M, Zhang Y et al. Systematic Analysis of Cell-Type Differences in the Epithelial Secretome Reveals Insights into the Pathogenesis of Respiratory Syncytial Virus-Induced Lower Respiratory Tract Infections. J Immunol, 198(8), 3345–3364 (2017).• Identified cell-type differences in secretome proteins using LC-MS/MS.
  • 18.Hosakote YM, Brasier AR, Casola A, Garofalo RP, Kurosky A. Respiratory Syncytial Virus Infection Triggers Epithelial HMGB1 Release as a Damage-Associated Molecular Pattern Promoting a Monocytic Inflammatory Response. J Virol, 90(21), 9618–9631 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Tian B, Hosoki K, Liu Z et al. Mucosal Bromodomain-Containing Protein 4 (BRD4) Mediates Aeroallergen-induced Inflammation and Remodeling. The Journal of allergy and clinical immunology, 143(4), 1380–1394.e1389 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Bergeron C, Tulic MK, Hamid Q. Airway remodelling in asthma: From benchside to clinical practice. Canadian Respiratory Journal : Journal of the Canadian Thoracic Society, 17(4), e85–e93 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Prakash YS, Halayko AJ, Gosens R, Panettieri RA, Camoretti-Mercado B, Penn RB. An Official American Thoracic Society Research Statement: Current Challenges Facing Research and Therapeutic Advances in Airway Remodeling. American Journal of Respiratory and Critical Care Medicine, 195(2), e4–e19 (2017). [DOI] [PubMed] [Google Scholar]
  • 22.Chin J, Magoffin RL, Shearer LA, Schieble JH, Lennette EH. Field evaluation of a respiratory syncytial virus vaccine and a trivalent parainfluenza virus vaccine in a pediatric population. American Journal of Epidemiology, 89(4), 449–463 (1969). [DOI] [PubMed] [Google Scholar]
  • 23.Fulginiti VA, Eller JJ, Sieber OF, Joyner JW, Minamitani M, Meiklejohn G. Respiratory virus immunization: a field trial of two inactivated respiratory virus vaccines; an aqueous trivalent paratnfluenza virus vaccine and an alum-precipitated respiratory syncytial virus vaccine. American Journal of Epidemiology, 89(4), 435–448 (1969). [DOI] [PubMed] [Google Scholar]
  • 24.Kapikian AZ, Mitchell RH, Chanock RM, Shvedoff RA, Stewart CE. An epidemiologic study of altered clinical reactivity to respiratory syncytial (rs) virus infection in children previously vaccinated with an inactivated rs virus vaccine. American Journal of Epidemiology, 89(4), 405–421 (1969). [DOI] [PubMed] [Google Scholar]
  • 25.Weibel RE, Stokes J, Leagus MB et al. Respiratory virus vaccines. VII. Field evaluation of respiratory syncytial, parainfluenza 1, 2, 3, and Mycoplasma pneumoniae vaccines, 1965 to 1966. The American Review of Respiratory Disease, 96(4), 724–739 (1967). [DOI] [PubMed] [Google Scholar]
  • 26.Kim HW, Canchola JG, Brandt CD et al. Respiratory syncytial virus disease in infants despite prior administration of antigenic inactivated vaccine12. American Journal of Epidemiology, 89(4), 422–434 (1969). [DOI] [PubMed] [Google Scholar]
  • 27.Rezaee F, Linfield D, Harford TJ, Piedimonte G. Ongoing Developments in RSV Prophylaxis: A Clinician’s Analysis Short Title: Ongoing Developments in RSV Prophylaxis. Current opinion in virology, 24, 70–78 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Efficacy of Ribavirin in the Treatment of Respiratory Syncytial Virus Infections in Lung-Transplant Recipients- ClinicalKey.
  • 29.Tian B, Yang J, Zhao Y et al. BRD4 Couples NF-κB/RelA with Airway Inflammation and the IRF-RIG-I Amplification Loop in Respiratory Syncytial Virus Infection. J Virol, 91(6) (2017).• Demonstrated that RSV-induced inflammation is dependent on Bromodomain-containing Protein 4 (BRD4), and that this pathway amplifies the IRF-RIG-I mediated interferon response.
  • 30.Xu X, Qiao D, Mann M, Garofalo RP, Brasier AR. Respiratory Syncytial Virus Infection Induces Chromatin Remodeling to Activate Growth Factor and Extracellular Matrix Secretion Pathways. Viruses, 12(8) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Sun Y, López CB. The innate immune response to RSV: Advances in our understanding of critical viral and host factors. Vaccine, 35(3), 481–488 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Ludvigsen M, Honoré B. Transcriptomics and Proteomics: Integration? In: eLS. (American Cancer Society, 2018) 1–7. [Google Scholar]
  • 33.Zhang Y, Fonslow BR, Shan B, Baek M-C, Yates JR. Protein Analysis by Shotgun/Bottom-up Proteomics. Chemical reviews, 113(4), 2343–2394 (2013).• Describes Bottom-up proteomics in-depth.
  • 34.Brasier AR, Spratt H, Wu Z et al. Nuclear heat shock response and novel nuclear domain 10 reorganization in respiratory syncytial virus-infected a549 cells identified by high-resolution two-dimensional gel electrophoresis. J Virol, 78(21), 11461–11476 (2004).• Identified RSV-induced changes to nuclear heat shock proteins and ND10s using 2DE.
  • 35.Jamaluddin M, Wiktorowicz JE, Soman KV et al. Role of peroxiredoxin 1 and peroxiredoxin 4 in protection of respiratory syncytial virus-induced cysteinyl oxidation of nuclear cytoskeletal proteins. J Virol, 84(18), 9533–9545 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Webster J, Oxley D. Protein identification by MALDI-TOF mass spectrometry. Methods in Molecular Biology (Clifton, N.J.), 800, 227–240 (2012). [DOI] [PubMed] [Google Scholar]
  • 37.Damodaran S, Wood TD, Nagarajan P, Rabin RA. Evaluating Peptide Mass Fingerprinting-based Protein Identification. Genomics, Proteomics & Bioinformatics, 5(3–4), 152–157 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Dupree EJ, Jayathirtha M, Yorkey H, Mihasan M, Petre BA, Darie CC. A Critical Review of Bottom-Up Proteomics: The Good, the Bad, and the Future of This Field. Proteomes, 8(3) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Wong JWH, Cagney G. An overview of label-free quantitation methods in proteomics by mass spectrometry. Methods in Molecular Biology (Clifton, N.J.), 604, 273–283 (2010). [DOI] [PubMed] [Google Scholar]
  • 40.Mann M. Functional and quantitative proteomics using SILAC. Nature Reviews. Molecular Cell Biology, 7(12), 952–958 (2006). [DOI] [PubMed] [Google Scholar]
  • 41.Tian Y, Li H, Gao Y et al. Quantitative proteomic characterization of lung tissue in idiopathic pulmonary fibrosis. Clinical Proteomics, 16(1), 6 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Abdul-Salam Vahitha B, Wharton J, Cupitt J, Berryman M, Edwards Robert J, Wilkins Martin R. Proteomic Analysis of Lung Tissues From Patients With Pulmonary Arterial Hypertension. Circulation, 122(20), 2058–2067 (2010). [DOI] [PubMed] [Google Scholar]
  • 43.Goddard ET, Hill RC, Barrett A et al. Quantitative extracellular matrix proteomics to study mammary and liver tissue microenvironments. The international journal of biochemistry & cell biology, 81(Pt A), 223–232 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Hill RC, Calle EA, Dzieciatkowska M, Niklason LE, Hansen KC. Quantification of Extracellular Matrix Proteins from a Rat Lung Scaffold to Provide a Molecular Readout for Tissue Engineering. Molecular & Cellular Proteomics : MCP, 14(4), 961–973 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Naba A, Pearce OMT, Del Rosario A et al. Characterization of the Extracellular Matrix of Normal and Diseased Tissues Using Proteomics. Journal of Proteome Research, 16(8), 3083–3091 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Brown KA, Tucholski T, Eken C et al. High-Throughput Proteomics Enabled by a Photocleavable Surfactant. Angewandte Chemie International Edition, 59(22), 8406–8410 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Dunham WH, Mullin M, Gingras A-C. Affinity-purification coupled to mass spectrometry: Basic principles and strategies. PROTEOMICS, 12(10), 1576–1590 (2012). [DOI] [PubMed] [Google Scholar]
  • 48.Heaven MR, Funk AJ, Cobbs AL et al. Systematic Evaluation of Data-Independent Acquisition for Sensitive and Reproducible Proteomics – a Prototype Design for a Single Injection Assay. Journal of mass spectrometry : JMS, 51(1), 1–11 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Ebhardt HA. Selected reaction monitoring mass spectrometry: a methodology overview. Methods in Molecular Biology (Clifton, N.J.), 1072, 209–222 (2014). [DOI] [PubMed] [Google Scholar]
  • 50.Lieber M, Smith B, Szakal A, Nelson-Rees W, Todaro G. A continuous tumor-cell line from a human lung carcinoma with properties of type II alveolar epithelial cells. International Journal of Cancer, 17, 62–70 (1976). [DOI] [PubMed] [Google Scholar]
  • 51.Arnold R, Humbert B, Werchaus H, Gallati H, Konig W. Interleukin-8, Interleukin-6, and soluble tumor necrosis factor receptor type I released from a human pulmonary epithelial cell line (A549) exposed to respiratory syncytial virus. Immunology (Oxford), 82, 126–133 (1994). [PMC free article] [PubMed] [Google Scholar]
  • 52.Tian B, Zhao Y, Kalita M et al. CDK9-dependent transcriptional elongation in the innate interferon-stimulated gene response to respiratory syncytial virus infection in airway epithelial cells. J Virol, 87(12), 7075–7092 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Garofalo R, Sabry M, Jamaluddin M et al. 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(12), 8773–8781 (1996). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Bertolusso R, Tian B, Zhao Y et al. Dynamic Cross Talk Model Of The Epithelial Innate Immune Response To Double-Stranded Rna Stimulation: Coordinated Dynamics Emerging From Cell-Level Noise. PLoS ONE, 9(4), e93396 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Gorphe P A comprehensive review of Hep-2 cell line in translational research for laryngeal cancer. American Journal of Cancer Research, 9(4), 644–649 (2019). [PMC free article] [PubMed] [Google Scholar]
  • 56.Howden AJ, Geoghegan V, Katsch K et al. QuaNCAT: quantitating proteome dynamics in primary cells. Nat Methods, 10(4), 343–346 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Foster MW, Gwinn WM, Kelly FL et al. Proteomic Analysis of Primary Human Airway Epithelial Cells Exposed to the Respiratory Toxicant Diacetyl. J Proteome Res, 16(2), 538–549 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Francavilla C, Lupia M, Tsafou K et al. Phosphoproteomics of Primary Cells Reveals Druggable Kinase Signatures in Ovarian Cancer. Cell Rep, 18(13), 3242–3256 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Mathieson T, Franken H, Kosinski J et al. Systematic analysis of protein turnover in primary cells. Nat Commun, 9(1), 689 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Sachs N, Papaspyropoulos A, Zomer-van Ommen DD et al. Long-term expanding human airway organoids for disease modeling. EMBO J, 38(4) (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Miller AJ, Dye BR, Ferrer-Torres D et al. Generation of lung organoids from human pluripotent stem cells in vitro. Nat Protoc, 14(2), 518–540 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Tian B, Widen SG, Yang J et al. The NFκB subunit RELA is a master transcriptional regulator of the committed epithelial-mesenchymal transition in airway epithelial cells. J Biol Chem, 293(42), 16528–16545 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Tian B, Patrikeev I, Ochoa L et al. NF-κB Mediates Mesenchymal Transition, Remodeling, and Pulmonary Fibrosis in Response to Chronic Inflammation by Viral RNA Patterns. Am J Respir Cell Mol Biol, 56(4), 506–520 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Hosakote YM, Jantzi PD, Esham DL et al. Viral-mediated inhibition of antioxidant enzymes contributes to the pathogenesis of severe respiratory syncytial virus bronchiolitis. Am J Respir Crit Care Med, 183(11), 1550–1560 (2011).• Confirmed that RSV-infection inhibits antioxidant proteins in-vivo and implicated NRF2 as the mechanism responsible.
  • 65.Brasier AR, Spratt H, Wu Z et al. Nuclear heat shock response and novel nuclear domain 10 reorganization in respiratory syncytial virus-infected a549 cells identified by high-resolution two-dimensional gel electrophoresis. Journal of Virology, 78(21), 11461–11476 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.McDonald TP, Pitt AR, Brown G, Rixon HWM, Sugrue RJ. Evidence that the respiratory syncytial virus polymerase complex associates with lipid rafts in virus-infected cells: a proteomic analysis. Virology, 330(1), 147–157 (2004). [DOI] [PubMed] [Google Scholar]
  • 67.Brown G, Rixon HWM, Steel J et al. Evidence for an association between heat shock protein 70 and the respiratory syncytial virus polymerase complex within lipid-raft membranes during virus infection. Virology, 338(1), 69–80 (2005). [DOI] [PubMed] [Google Scholar]
  • 68.Munday DC, Wu W, Smith N et al. Interactome Analysis of the Human Respiratory Syncytial Virus RNA Polymerase Complex Identifies Protein Chaperones as Important Cofactors That Promote L-Protein Stability and RNA Synthesis. Journal of Virology, 89(2), 917–930 (2014).• Applied AP-MS to discover that RSV-L functionally interacts with heat shock proteins.
  • 69.Kotelkin A, Belyakov IM, Yang L, Berzofsky JA, Collins PL, Bukreyev A. The NS2 Protein of Human Respiratory Syncytial Virus Suppresses the Cytotoxic T-Cell Response as a Consequence of Suppressing the Type I Interferon Response. Journal of Virology, 80(12), 5958–5967 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Ling Z, Tran KC, Teng MN. Human Respiratory Syncytial Virus Nonstructural Protein NS2 Antagonizes the Activation of Beta Interferon Transcription by Interacting with RIG-I. Journal of Virology, 83(8), 3734–3742 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Lo MS, Brazas RM, Holtzman MJ. Respiratory Syncytial Virus Nonstructural Proteins NS1 and NS2 Mediate Inhibition of Stat2 Expression and Alpha/Beta Interferon Responsiveness. Journal of Virology, 79(14), 9315–9319 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Moore EC, Barber J, Tripp RA. Respiratory syncytial virus (RSV) attachment and nonstructural proteins modify the type I interferon response associated with suppressor of cytokine signaling (SOCS) proteins and IFN-stimulated gene-15 (ISG15). Virology Journal, 5, 116 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Ramaswamy M, Shi L, Varga SM, Barik S, Behlke MA, Look DC. Respiratory syncytial virus nonstructural protein 2 specifically inhibits type I interferon signal transduction. Virology, 344(2), 328–339 (2006). [DOI] [PubMed] [Google Scholar]
  • 74.Spann KM, Tran K-C, Chi B, Rabin RL, Collins PL. Suppression of the Induction of Alpha, Beta, and Gamma Interferons by the NS1 and NS2 Proteins of Human Respiratory Syncytial Virus in Human Epithelial Cells and Macrophages. Journal of Virology, 78(8), 4363–4369 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Spann KM, Tran KC, Collins PL. Effects of Nonstructural Proteins NS1 and NS2 of Human Respiratory Syncytial Virus on Interferon Regulatory Factor 3, NF-κB, and Proinflammatory Cytokines. Journal of Virology, 79(9), 5353–5362 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Swedan S, Musiyenko A, Barik S. Respiratory syncytial virus nonstructural proteins decrease levels of multiple members of the cellular interferon pathways. Journal of Virology, 83(19), 9682–9693 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Hastie ML, Headlam MJ, Patel NB et al. The Human Respiratory Syncytial Virus Nonstructural Protein 1 Regulates Type I and Type II Interferon Pathways. Molecular & Cellular Proteomics : MCP, 11(5), 108–127 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Ban J, Lee N-R, Lee N-J, Lee JK, Quan F-S, Inn K-S. Human Respiratory Syncytial Virus NS 1 Targets TRIM25 to Suppress RIG-I Ubiquitination and Subsequent RIG-I-Mediated Antiviral Signaling. Viruses, 10(12) (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Munday DC, Emmott E, Surtees R et al. Quantitative Proteomic Analysis of A549 Cells Infected with Human Respiratory Syncytial Virus *. Molecular & Cellular Proteomics, 9(11), 2438–2459 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Munday DC, Hiscox JA, Barr JN. Quantitative proteomic analysis of A549 cells infected with human respiratory syncytial virus subgroup B using SILAC coupled to LC-MS/MS. Proteomics, 10(23), 4320–4334 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Munday DC, Howell G, Barr JN, Hiscox JA. Proteomic analysis of mitochondria in respiratory epithelial cells infected with human respiratory syncytial virus and functional implications for virus and cell biology. The Journal of Pharmacy and Pharmacology, 67(3), 300–318 (2015). [DOI] [PubMed] [Google Scholar]
  • 82.Ternette N, Wright C, Kramer HB, Altun M, Kessler BM. Label-free quantitative proteomics reveals regulation of interferon-induced protein with tetratricopeptide repeats 3 (IFIT3) and 5’-3’-exoribonuclease 2 (XRN2) during respiratory syncytial virus infection. Virology Journal, 8(1), 442 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Dave KA, Norris EL, Bukreyev AA et al. A comprehensive proteomic view of responses of A549 type II alveolar epithelial cells to human respiratory syncytial virus infection. Molecular & cellular proteomics: MCP, 13(12), 3250–3269 (2014).• Comprehensive LC-MS analysis of A549 cells infected with RSV.
  • 84.Jamaluddin M, Wang S, Garofalo RP et al. IFN-beta mediates coordinate expression of antigen-processing genes in RSV-infected pulmonary epithelial cells. American journal of physiology. Lung cellular and molecular physiology, 280(2), L248–257 (2001). [DOI] [PubMed] [Google Scholar]
  • 85.Liu P, Lu M, Tian B et al. Expression of an IKKgamma splice variant determines IRF3 and canonical NF-kappaB pathway utilization in ssRNA virus infection. PLoS One, 4(11), e8079 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Smieja J, Jamaluddin M, Brasier AR, Kimmel M. Model-based analysis of interferon-beta induced signaling pathway. Bioinformatics, 24(20), 2363–2369 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Shapira SD, Hacohen N. Systems biology approaches to dissect mammalian innate immunity. Current opinion in immunology, 23(1), 71–77 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Zak DE, Aderem A. Systems biology of innate immunity. Immunological reviews, 227(1), 264–282 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Zhao Y, Tian B, Edeh CB, Brasier AR. Quantitation of the Dynamic Profiles of the Innate Immune Response Using Multiplex Selected Reaction Monitoring–Mass Spectrometry. Molecular & Cellular Proteomics : MCP, 12(6), 1513–1529 (2013).• Development and application of Selected Reaction Monitoring assays for interrogation of the innate immune response.
  • 90.Zhao Y, Widen SG, Jamaluddin M et al. Quantification of Activated NF-κB/RelA Complexes Using ssDNA Aptamer Affinity – Stable Isotope Dilution—Selected Reaction Monitoring—Mass Spectrometry. Molecular & Cellular Proteomics : MCP, 10(6) (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Brasier AR. RSV Reprograms the CDK9•BRD4 Chromatin Remodeling Complex to Couple Innate Inflammation to Airway Remodeling. Viruses, 12(4) (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Liu Z, Tian B, Chen H, Wang P, Brasier AR, Zhou J. Discovery of potent and selective BRD4 inhibitors capable of blocking TLR3-induced acute airway inflammation. European Journal of Medicinal Chemistry, 151, 450–461 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Tian B, Zhao Y, Sun H, Zhang Y, Yang J, Brasier AR. BRD4 mediates NF-κB-dependent epithelial-mesenchymal transition and pulmonary fibrosis via transcriptional elongation. American Journal of Physiology-Lung Cellular and Molecular Physiology, 311(6), L1183–L1201 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Tian B, Liu Z, Yang J et al. Selective Antagonists of the Bronchiolar Epithelial NF-κB-Bromodomain-Containing Protein 4 Pathway in Viral-Induced Airway Inflammation. Cell Rep, 23(4), 1138–1151 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Tian B, Liu Z, Litvinov J et al. Efficacy of Novel Highly Specific Bromodomain-Containing Protein 4 Inhibitors in Innate Inflammation-Driven Airway Remodeling. Am J Respir Cell Mol Biol, 60(1), 68–83 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Mann M, Roberts DS, Zhu Y et al. Discovery of RSV-Induced BRD4 Protein Interactions Using Native Immunoprecipitation and Parallel Accumulation—Serial Fragmentation (PASEF) Mass Spectrometry. Viruses, 13(3), 454 (2021).• Determined that RSV-infection induces dynamic, pro-inflammatory and pro-remodeling changes to the host BRD4 protein interactome.
  • 97.Zenz R, Wagner EF. Jun signalling in the epidermis: From developmental defects to psoriasis and skin tumors. The International Journal of Biochemistry & Cell Biology, 38(7), 1043–1049 (2006). [DOI] [PubMed] [Google Scholar]
  • 98.Wagner EF. Bone development and inflammatory disease is regulated by AP-1 (Fos/Jun). Annals of the Rheumatic Diseases, 69(Suppl 1), i86–i88 (2010). [DOI] [PubMed] [Google Scholar]
  • 99.Xiao W, Hodge DR, Wang L, Yang X, Zhang X, Farrar WL. NF-kappaB activates IL-6 expression through cooperation with c-Jun and IL6-AP1 site, but is independent of its IL6-NFkappaB regulatory site in autocrine human multiple myeloma cells. Cancer Biology & Therapy, 3(10), 1007–1017 (2004). [DOI] [PubMed] [Google Scholar]
  • 100.Khalaf H, Jass J, Olsson P-E. Differential cytokine regulation by NF-κB and AP-1 in Jurkat T-cells. BMC Immunology, 11(1), 26 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Basu S, Cheriyamundath S, Ben-Ze’ev A. Cell–cell adhesion: linking Wnt/β-catenin signaling with partial EMT and stemness traits in tumorigenesis. F1000Research, 7 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Jiang Y-G, Luo Y, He D-l et al. Role of Wnt/β-catenin signaling pathway in epithelial-mesenchymal transition of human prostate cancer induced by hypoxia-inducible factor-1α. International Journal of Urology, 14(11), 1034–1039 (2007). [DOI] [PubMed] [Google Scholar]
  • 103.Tian X, Liu Z, Niu B et al. E-Cadherin/β-Catenin Complex and the Epithelial Barrier. Journal of Biomedicine and Biotechnology, 2011 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Valenta T, Hausmann G, Basler K. The many faces and functions of β-catenin. The EMBO Journal, 31(12), 2714–2736 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Wu Y, Ginther C, Kim J et al. Expression of Wnt3 Activates Wnt/β-Catenin Pathway and Promotes EMT-like Phenotype in Trastuzumab-Resistant HER2-Overexpressing Breast Cancer Cells. Molecular Cancer Research, 10(12), 1597–1606 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Hussain M, Xu C, Lu M, Wu X, Tang L, Wu X. Wnt/β-catenin signaling links embryonic lung development and asthmatic airway remodeling. Biochimica Et Biophysica Acta. Molecular Basis of Disease, 1863(12), 3226–3242 (2017). [DOI] [PubMed] [Google Scholar]
  • 107.Mann M, Roberts DS, Zhu Y et al. Discovery of RSV-Induced BRD4 Protein Interactions Using Native Immunoprecipitation and Parallel Accumulation-Serial Fragmentation (PASEF) Mass Spectrometry. Viruses, 13(3) (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Komaravelli N, Ansar M, Garofalo RP, Casola A. Respiratory syncytial virus induces NRF2 degradation through a promyelocytic leukemia protein ‐ ring finger protein 4 dependent pathway. Free Radical Biology and Medicine, 113, 494–504 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Hosakote YM, Komaravelli N, Mautemps N, Liu T, Garofalo RP, Casola A. Antioxidant mimetics modulate oxidative stress and cellular signaling in airway epithelial cells infected with respiratory syncytial virus. American journal of physiology. Lung cellular and molecular physiology, 303(11), L991–1000 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Ansar M, Ivanciuc T, Garofalo RP, Casola A. Increased Lung Catalase Activity Confers Protection Against Experimental RSV Infection. Scientific reports, 10(1), 3653 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.van Diepen A, Brand HK, de Waal L et al. Host proteome correlates of vaccine-mediated enhanced disease in a mouse model of respiratory syncytial virus infection. Journal of Virology, 89(9), 5022–5031 (2015).• Identified host protein correlates of RSV Vaccine-mediated Enhanced Disease in a murine model.
  • 112.Olmsted RA, Elango N, Prince GA et al. Expression of the F glycoprotein of respiratory syncytial virus by a recombinant vaccinia virus: comparison of the individual contributions of the F and G glycoproteins to host immunity. Proceedings of the National Academy of Sciences of the United States of America, 83(19), 7462–7466 (1986). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Openshaw PJ, Clarke SL, Record FM. Pulmonary eosinophilic response to respiratory syncytial virus infection in mice sensitized to the major surface glycoprotein G. International Immunology, 4(4), 493–500 (1992). [DOI] [PubMed] [Google Scholar]
  • 114.Owhashi M, Arita H, Hayai N. Identification of a novel eosinophil chemotactic cytokine (ECF-L) as a chitinase family protein. The Journal of Biological Chemistry, 275(2), 1279–1286 (2000). [DOI] [PubMed] [Google Scholar]
  • 115.Chang NC, Hung SI, Hwa KY et al. A macrophage protein, Ym1, transiently expressed during inflammation is a novel mammalian lectin. The Journal of Biological Chemistry, 276(20), 17497–17506 (2001). [DOI] [PubMed] [Google Scholar]
  • 116.Iwashita H, Morita S, Sagiya Y, Nakanishi A. Role of eosinophil chemotactic factor by T lymphocytes on airway hyperresponsiveness in a murine model of allergic asthma. American Journal of Respiratory Cell and Molecular Biology, 35(1), 103–109 (2006). [DOI] [PubMed] [Google Scholar]
  • 117.Tan S-M. The leucocyte β2 (CD18) integrins: the structure, functional regulation and signalling properties. Bioscience Reports, 32(3), 241–269 (2012). [DOI] [PubMed] [Google Scholar]
  • 118.Hägglund S, Blodörn K, Näslund K et al. Proteome analysis of bronchoalveolar lavage from calves infected with bovine respiratory syncytial virus—Insights in pathogenesis and perspectives for new treatments. PLoS ONE, 12(10) (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Wei Q, Chen ZH, Wang L et al. LZTFL1 suppresses lung tumorigenesis by maintaining differentiation of lung epithelial cells. Oncogene, 35(20), 2655–2663 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Aljabr W, Armstrong S, Rickett NY et al. High Resolution Analysis of Respiratory Syncytial Virus Infection In Vivo. Viruses, 11(10) (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Akram KM, Moyo NA, Leeming GH et al. An innate defense peptide BPIFA1/SPLUNC1 restricts influenza A virus infection. Mucosal Immunology, 11(1), 71–81 (2018). [DOI] [PubMed] [Google Scholar]
  • 122.Whitsett JA, Alenghat T. Respiratory epithelial cells orchestrate pulmonary innate immunity. Nat Immunol, 16(1), 27–35 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Olzewska B, Casola A, Saito T et al. Cell-specific expression of RANTES, MCP-1, and MIP-1a by lower airway epithelial cells and eosinophils infected with respiratory syncytial virus. Journal of Virology, 72, 4756–4764 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Regev A, Teichmann SA, Lander ES et al. The Human Cell Atlas. eLife, 6, e27041 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Ctortecka C, Mechtler K. The rise of single-cell proteomics. Analytical Science Advances, n/a(n/a)). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Budnik B, Levy E, Harmange G, Slavov N. SCoPE-MS: mass spectrometry of single mammalian cells quantifies proteome heterogeneity during cell differentiation. Genome Biology, 19(1), 161 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Zhu Y, Piehowski PD, Zhao R et al. Nanodroplet processing platform for deep and quantitative proteome profiling of 10–100 mammalian cells. Nature Communications, 9(1), 882 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Brunner A-D, Thielert M, Vasilopoulou CG et al. Ultra-high sensitivity mass spectrometry quantifies single-cell proteome changes upon perturbation. bioRxiv, 2020.2012.2022.423933 (2021). [DOI] [PMC free article] [PubMed]
  • 129.Vuong NQ, Goegan P, Mohottalage S et al. Human lung epithelial cell A549 proteome data after treatment with titanium dioxide and carbon black. Data in Brief, 8, 687–691 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Volpe P, Eremenko‐Volpe T. Quantitative Studies on Cell Proteins in Suspension Cultures. European Journal of Biochemistry, 12(1), 195–200 (1970). [DOI] [PubMed] [Google Scholar]
  • 131.Fox CH, Johnson FB, Whiting J, Roller PP. Formaldehyde fixation. J Histochem Cytochem, 33(8), 845–853 (1985). [DOI] [PubMed] [Google Scholar]
  • 132.Ostasiewicz P, Zielinska DF, Mann M, Wiśniewski JR. Proteome, phosphoproteome, and N-glycoproteome are quantitatively preserved in formalin-fixed paraffin-embedded tissue and analyzable by high-resolution mass spectrometry. J Proteome Res, 9(7), 3688–3700 (2010). [DOI] [PubMed] [Google Scholar]
  • 133.Wakabayashi M, Yoshihara H, Masuda T, Tsukahara M, Sugiyama N, Ishihama Y. Phosphoproteome analysis of formalin-fixed and paraffin-embedded tissue sections mounted on microscope slides. J Proteome Res, 13(2), 915–924 (2014). [DOI] [PubMed] [Google Scholar]
  • 134.Marchione DM, Ilieva I, Devins K et al. HYPERsol: High-Quality Data from Archival FFPE Tissue for Clinical Proteomics. J Proteome Res, 19(2), 973–983 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Uchida Y, Sasaki H, Terasaki T. Establishment and validation of highly accurate formalin-fixed paraffin-embedded quantitative proteomics by heat-compatible pressure cycling technology using phase-transfer surfactant and SWATH-MS. Sci Rep, 10(1), 11271 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Coscia F, Doll S, Bech JM et al. A streamlined mass spectrometry-based proteomics workflow for large-scale FFPE tissue analysis. J Pathol, 251(1), 100–112 (2020). [DOI] [PubMed] [Google Scholar]
  • 137.Becnel D, You D, Erskin J, Dimina DM, Cormier SA. A role for airway remodeling during respiratory syncytial virus infection. Respir Res, 6, 122 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Neilson KA, Yunis EJ. Demonstration of respiratory syncytial virus in an autopsy series. Pediatr Pathol, 10(4), 491–502 (1990). [DOI] [PubMed] [Google Scholar]
  • 139.Wright C, Oliver KC, Fenwick FI, Smith NM, Toms GL. A monoclonal antibody pool for routine immunohistochemical detection of human respiratory syncytial virus antigens in formalin-fixed, paraffin-embedded tissue. J Pathol, 182(2), 238–244 (1997). [DOI] [PubMed] [Google Scholar]
  • 140.Longuespée R, Alberts D, Pottier C et al. A laser microdissection-based workflow for FFPE tissue microproteomics: Important considerations for small sample processing. Methods, 104, 154–162 (2016). [DOI] [PubMed] [Google Scholar]
  • 141.Barrett AS, Wither MJ, Hill RC et al. Hydroxylamine Chemical Digestion for Insoluble Extracellular Matrix Characterization. Journal of Proteome Research, 16(11), 4177–4184 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Frantz C, Stewart KM, Weaver VM. The extracellular matrix at a glance. Journal of Cell Science, 123(24), 4195–4200 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Eyre DR, Paz MA, Gallop PM. Cross-linking in collagen and elastin. Annual Review of Biochemistry, 53, 717–748 (1984). [DOI] [PubMed] [Google Scholar]
  • 144.Lindsey ML, Jung M, Hall ME, DeLeon-Pennell KY. Proteomic analysis of the cardiac extracellular matrix: clinical research applications. Expert review of proteomics, 15(2), 105–112 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Mann M, Jensen ON. Proteomic analysis of post-translational modifications. Nature Biotechnology, 21(3), 255–261 (2003). [DOI] [PubMed] [Google Scholar]
  • 146.Potel CM, Lemeer S, Heck AJR. Phosphopeptide Fragmentation and Site Localization by Mass Spectrometry: An Update. Analytical Chemistry, 91(1), 126–141 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Doerr A Top-down mass spectrometry. Nature Methods, 5(1), 24–24 (2008). [Google Scholar]
  • 148.Toby TK, Fornelli L, Kelleher NL. Progress in Top-Down Proteomics and the Analysis of Proteoforms. Annual review of analytical chemistry (Palo Alto, Calif.), 9(1), 499–519 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Cai W, Tucholski TM, Gregorich ZR, Ge Y. Top-down Proteomics: Technology Advancements and Applications to Heart Diseases. Expert review of proteomics, 13(8), 717–730 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Tiambeng TN, Tucholski T, Wu Z et al. Analysis of Cardiac Troponin Proteoforms by Top-Down Mass Spectrometry. Methods in enzymology, 626, 347–374 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Tiambeng TN, Roberts DS, Brown KA et al. Nanoproteomics enables proteoform-resolved analysis of low-abundance proteins in human serum. Nature Communications, 11(1), 3903 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Roberts DS, Chen B, Tiambeng TN, Wu Z, Ge Y, Jin S. Reproducible Large-Scale Synthesis of Surface Silanized Nanoparticles as an Enabling Nanoproteomics Platform: Enrichment of the Human Heart Phosphoproteome. Nano Res, 12(6), 1473–1481 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Hanisch F-G. Top-down sequencing of O-glycoproteins by in-source decay matrix-assisted laser desorption ionization mass spectrometry for glycosylation site analysis. Analytical Chemistry, 83(12), 4829–4837 (2011). [DOI] [PubMed] [Google Scholar]
  • 154.Nicolardi S, van der Burgt YEM, Dragan I, Hensbergen PJ, Deelder AM. Identification of new apolipoprotein-CIII glycoforms with ultrahigh resolution MALDI-FTICR mass spectrometry of human sera. Journal of Proteome Research, 12(5), 2260–2268 (2013). [DOI] [PubMed] [Google Scholar]
  • 155.Schirm M, Schoenhofen IC, Logan SM, Waldron KC, Thibault P. Identification of unusual bacterial glycosylation by tandem mass spectrometry analyses of intact proteins. Analytical Chemistry, 77(23), 7774–7782 (2005). [DOI] [PubMed] [Google Scholar]

RESOURCES