Next Article in Journal
The Hypersensitive Response to Plant Viruses
Previous Article in Journal
Long-Read High-Throughput Sequencing (HTS) Revealed That the Sf-Rhabdovirus X+ Genome Contains a 3.7 kb Internal Duplication
Previous Article in Special Issue
Allosteric Inhibitors of Zika Virus NS2B-NS3 Protease Targeting Protease in “Super-Open” Conformation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Host Responses to Respiratory Syncytial Virus Infection

Research Center for Emerging Infections and Zoonoses, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
*
Author to whom correspondence should be addressed.
Viruses 2023, 15(10), 1999; https://doi.org/10.3390/v15101999
Submission received: 18 August 2023 / Revised: 22 September 2023 / Accepted: 23 September 2023 / Published: 26 September 2023
(This article belongs to the Special Issue Molecular Biology of RNA Viruses)

Abstract

:
Respiratory syncytial virus (RSV) infections are a constant public health problem, especially in infants and older adults. Virtually all children will have been infected with RSV by the age of two, and reinfections are common throughout life. Since antigenic variation, which is frequently observed among other respiratory viruses such as SARS-CoV-2 or influenza viruses, can only be observed for RSV to a limited extent, reinfections may result from short-term or incomplete immunity. After decades of research, two RSV vaccines were approved to prevent lower respiratory tract infections in older adults. Recently, the FDA approved a vaccine for active vaccination of pregnant women to prevent severe RSV disease in infants during their first RSV season. This review focuses on the host response to RSV infections mediated by epithelial cells as the first physical barrier, followed by responses of the innate and adaptive immune systems. We address possible RSV-mediated immunomodulatory and pathogenic mechanisms during infections and discuss the current vaccine candidates and alternative treatment options.

1. Introduction

Respiratory syncytial virus (RSV) is a widely circulating pathogen in the human population and a major cause of acute lower respiratory tract infections (ALRIs) in infants and older adults. RSV is an enveloped, negative-sense, single-stranded RNA virus belonging to the Orthopneumovirus genus of the family Pneumoviridae [1]. The viral genome consists of ten genes encoding eleven proteins: nonstructural proteins 1 and 2 (NS1/2), nucleocapsid protein (N), matrix protein (M), phosphoprotein (P), small hydrophobic protein (SH), glycoprotein (G), fusion protein (F), large protein (L), and M2 with two overlapping open-reading frames leading to two proteins, M2.1 and M2.2 [1]. Two antigenic subtypes, RSV-A and -B, are distinguished, each with several genotypes [2,3]. Currently, genotypes ON1 (RSV-A) and BA-CC (RSV-B) are circulating and differ from previous genotypes by the presence of sequence duplications of 72 bp and 60 bp, respectively, in the second hypervariable region of the G gene [4]. Both subtypes may co-circulate during an outbreak, with RSV A being the predominant strain in most years, although regional and seasonal differences are common [4].
RSV infections are a major cause of severe ALRI in infants and older adults [5,6,7,8]. In 2019, approx. 33 million RSV cases in infants were reported, of which 3.2 million required hospitalization [9]. Children under six months of age are at higher risk of hospitalization and fatal outcomes, accounting for ~50% of total hospitalized ALRI cases in high-income countries [9]. In older adults, 5.2 million RSV cases of ALRI were reported in the same year, resulting in 470,000 reported hospitalizations and 33,000 in-hospital deaths [10]. During the SARS-CoV-2 pandemic, reduced activity of various respiratory viruses, including RSV, was reported [11,12], probably due to non-pharmaceutical preventive measures to limit the transmission of SARS-CoV-2. Subsequently, increased sizes and numbers of outbreaks, with different times of onset in comparison to previous years, were reported for RSV disease, mainly affecting infants and children [12]. The decrease in RSV infections during the pandemic may have reduced immunity in children, referred to as the ‘immunity dept’, making them more susceptible to severe infections [12,13].
Nearly all infants are infected with RSV by the age of 2 years [14]. Reinfections occur throughout life, while disease severity tends to diminish with subsequent exposures. Older adults, however, represent a population that is also at risk of more severe disease during RSV infection [15,16,17]. Most infections in infants are mild, leading to an upper respiratory tract illness (URTI) or ‘flu-like’ symptoms often accompanied by otitis media [18,19]. Severe ALRI, mainly observed in infants younger than six months of age, can lead to bronchiolitis, pneumonia, and croup [18,19]. ALRIs are characterized by rhinorrhea, dry/wheezy cough, tachypnea, and dyspnea [19]. Infants, the immunocompromised, and older adults have a higher risk of developing severe infections [5,6,7,20,21]. Several studies further indicated a correlation between RSV-associated hospitalization during infancy and the development of asthma and recurrent wheezing later in life [22,23,24,25,26]. However, other factors, like comorbidities, may also account for this [6,27,28].
Virus-neutralizing human monoclonal antibodies directed to the F protein, i.e., Synagis® (Palivizumab) and Beyfortus® (Nirsevimab), have proven effective in lowering the risk of ALRI in high-risk infants when administered prophylactically. However, vaccine development for RSV has been challenging. The first clinical trials in infants in the 1960s with a formalin-inactivated RSV vaccine (FI-RSV) led to enhanced respiratory disease (ERD) in infants following their first natural infection [29]. Since then, vaccine development has been approached more cautiously while research has focused on understanding the immune response and immunopathogenesis during infection. The immaturity of an infant’s immune system as well as immunosenescence and preexisting immunity in older adults represent the most significant challenges for the development of a protective vaccine [30]. Several promising RSV vaccine candidates based on various production platforms have been developed and are currently in clinical trials. Recently, two protein-based vaccines, Arexvy (GlaxoSmithKline Biologicals (GSK), Brentford, UK) and Abrysvo (Pfizer, New York, NY, USA), were approved for use in older adults. Furthermore, a prefusion F-protein-based RSV vaccine candidate was developed for use in pregnant women to protect their infants early in life and was recently approved by the FDA.
Previous work illustrates the importance of understanding the correlation between protective immunity and immunopathogenesis in severe cases of RSV infections for the development of efficient and protective vaccines and therapeutics for those most at risk. This review focuses on the current knowledge of host immune responses to RSV infection, leading to either viral control or more severe disease due to immunopathogenesis, and the current use of this knowledge in vaccine development.

2. Innate Immune Responses to RSV Infection

Although the innate immune response toward infection with different Pneumoviridae members is still far from clear and may differ considerably [31,32,33,34], for RSV infection of humans many different mechanisms have been described. Epithelial cells are the primary target of infection (Figure 1) and secrete proinflammatory mediators upon RSV infection, leading to the recruitment and activation of innate immune cells. Several cell types are involved in the innate immune response, such as polymorphonuclear cells, cytotoxic lymphocytes, i.e., natural killer (NK) cells, or mononuclear phagocytes [35,36]. Tissue-resident macrophages and dendritic cells are among the first innate immune cells activated and mediate the further recruitment of inflammatory leukocytes and lymphocytes, like eosinophils, neutrophils, monocytes, and NK cells, to the site of infection by the secretion of type I IFNs and chemokines [37]. These cells subsequently begin eliminating the virus and simultaneously mediate the activation and recruitment of adaptive immune cells [36]. The initial immune response during epithelial cell infection, the protective role of innate immune cells, and their potential contribution to immunopathogenesis during RSV infections will be discussed in the following sections.

2.1. Infection of Epithelial Cells

The airway epithelium and associated mucus represent the first physiochemical barrier during respiratory infections [38]. The pseudostratified epithelium consists of three major cell types, namely ciliated cells, basal cells, and non-ciliated, secretory goblet cells which are connected by tight junctions [39,40]. The epithelial cells are mainly covered by mucus, contributing to protection from inhaled pathogens. The mucus is comprised of two distinct layers: a periciliary liquid layer, allowing the movement of cilia, and a more viscous upper layer, immobilizing the pathogen. Both layers consist of mucins, defensins, lysozymes, and immunoglobulins [41]. Mucociliary clearance, resulting from the continuous beating of motile cilia and consequently the movement of the outer mucus layer toward the pharynx, forms the first innate defense mechanism against respiratory pathogens. The protective properties of the mucus layer have already been shown for various respiratory viruses, like influenza viruses or coronaviruses [42,43,44,45]. An increased susceptibility of older adults to respiratory infections may be partly explained by a decrease in mucus production, cilia movement, and mucociliary clearance resulting in reduced pathogen removal [42,46].
Once RSV has traversed the mucus layer, it primarily infects airway epithelial cells (AECs), usually via the apical surface [47]. The infection of AECs depends on the cell’s differentiation status as infection increases with ciliogenesis. RSV therefore preferentially infects polarized ciliated AECs resulting in a focal distribution of the infection [48]. Hallmarks of AEC infection are cell sloughing, cilia loss, mucus hypersecretion, and syncytia formation. The loss of cilia and increased mucus production may contribute to airway obstruction during infection [38,49].
Several viral receptors have been proposed, as reviewed in [50]. Among others, nucleolin [51,52], CX3CR1 [53,54,55,56], heparan sulfate proteoglycans (HSPGs) [57,58,59], and intracellular adhesion molecule-1 (ICAM-1) [60] were associated with RSV attachment [50]. The interaction of RSV F and G with the HSPGs on the surface of immortalized cell lines leads to viral attachment and facilitates infection [59]. However, since HSPGs are barely expressed on the apical surface of epithelial cells in vivo, other receptors may be required for RSV infection [59].
CX3CR1 is expressed on various cell types, including innate and adaptive immune cells, and on motile cilia of AECs, which matches the RSV cell tropism during an infection [56,61,62]. Only two ligands are known for CX3CR1: the RSV G protein and CX3CL1 (fractalkine) [61]. CX3CL1 is a transmembrane protein on the surface of epithelial cells and contributes to the cell adhesion of leukocytes during infection [61,63]. Soluble CX3CL1 serves as a chemoattractant for T cells and monocytes via interaction with CX3CR1 on the surface of these cells [64]. RSV G contains a CX3C motif in its conserved central region, implicating a direct interaction between RSV G and CX3CR1 [65]. A study by Ha et al. showed a possible interaction between the receptor and G since a mutation in the CX3C motif (CX3C to CX4C) reduced the infection of epithelial cells in RSV-infected cotton rats [66]. However, a direct biochemical interaction between the RSV G protein and CX3CR1 has not been demonstrated yet. These data show that although G may facilitate the infection of epithelial cells, it is not strictly necessary for it [66]. However, the direct interaction of G and CX3CR1 may not only affect the infection of AECs but also interfere with the CX3CR1-CX3CL1 interaction. Soluble G may thereby act as a chemoattractant resulting in the increased recruitment of CX3CR1+ immune cells to the site of infection.
With the infection of respiratory epithelial cells, antiviral and innate immune responses are initiated. The response of AECs to RSV infection is similar to that against other viral infections like influenza viruses, SARS-CoV-2, or herpesviruses [67,68,69,70,71]. Upon viral attachment and entrance, intra- and extracellular pattern recognition receptors (PRRs) recognize the pathogen and initiate a signaling cascade resulting in the expression of antiviral genes [72]. Among the PRRs, retinoic-acid-inducible gene I (RIG-I)-like receptors (RLRs) and toll-like receptors (TLRs) were shown to play a role in virus recognition [73]. During the early stage of infection, RIG-I senses the viral RNA and activates the transcription factor interferon regulatory factor (IRF)-3, ultimately leading to the upregulation of MHC-I, NLRC5, and IFN-β [73]. During the later stages of an active infection, intracellular TLR3 binds to the viral RNA [73] and activates the transcription factor NF-κB, which induces the expression of type I interferons (IFN) [74,75]. Type I IFN signaling modulates metabolic pathways, thereby setting the cells in an ‘antiviral state’ that restricts viral replication and spreading to uninfected cells. Infection of AECs with RSV consequently induces an altered expression and secretion of chemoattractants and adhesion molecules, e.g., tumor necrosis factor (TNF)-α, CXCL6, CXCL10, RANTES/CCL5, interleukin (IL) 1β, IL-6, IL-8, CCL2, macrophage inflammatory protein 1α (MIP-1α), granulocyte colony-stimulating factor (G-CSF), granulocyte-macrophage colony-stimulating factor (GM-CSF), ICAM-1, vascular cell adhesion protein 1 (VCAM-1), and major histocompatibility complex I/II (MHC-I/II) [76,77,78,79,80,81,82,83]. Subsequently, their secretion triggers the recruitment of innate and adaptive immune cells and their interaction with infected AECs.
The immune modulatory mechanisms of different RSV proteins are well-known and have been reviewed previously [84]. NS1 and NS2 play a significant role in immune modulation by interacting with components of different immune signaling pathways. For example, NS1/2 interferes with the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling by suppressor of cytokine signaling proteins (SOCS)-dependent inhibition of JAK kinases and induction of proteasome-mediated STAT degradation [85,86]. In vitro studies have also shown the capacity of NS1 to translocate into the nucleus, where it interacts with the mediator complex and chromatin, indicating a regulatory effect of NS1 on the host’s gene expression [87]. Further, the respective interactions of NS1 and NS2 with mitochondrial antiviral-signaling protein (MAVS) and RIG-I interfere with the activation of IRF3 and NF-κB [87,88,89,90]. IFN-β synthesis is impaired by RSV infection, and the expression of anti-apoptotic genes is increased [90,91,92]. Following this, previous in vivo and in vitro studies have shown that the modulation of immune responses by NS1/2 inhibited apoptosis but induced necroptosis in RSV-infected AECs, which is considered an inflammatory cell death variant [92,93,94]. The release of HMGB1 into the extracellular space during necroptosis causes the recruitment of proinflammatory and Th2-type cytokines, ultimately resulting in increased airway inflammation and disease severity [94]. Besides NS1/2, an immunomodulatory role was also proposed for RSV nucleoprotein (N). In vitro studies indicate that RSV N induces the formation of inclusion bodies that serve as ‘traps’ for MAVS and melanoma-differentiation-associated gene 5 (MDA-5) during an early stage of infection [95,96]. RSV N interacts with PKR leading to reduced phosphorylation of eIFα and reduction in PKR-mediated signaling [97]. The activity of RSV proteins in infected AECs not only interferes with intrinsic immune pathways but restricts the activation of several genes that are involved in the production of cytokines and chemokines, which are crucial for the recruitment of innate and adaptive immune cells [84].
AECs, thereby, initiate the first antiviral, innate immune response upon RSV infection by the increased expression of adhesion molecules on the surface and the secretion of a variety of cytokines and chemokines that are essential for the recruitment of innate and adaptive immune cells. However, RSV has developed ways to alter and suppress this initial immune response, possibly affecting the activation and recruitment of immune cells during the early phase of infection. As mentioned above, cilia movement and mucus production are important during the first encounter with RSV and other respiratory viruses. However, most in vitro models are not able to represent the airway epithelium in its physiological three-dimensional structure, cellular composition, and motility [98]. Air–liquid interfaces and organoids represent promising systems to investigate virus–host interactions and immune responses during an infection under more physiological conditions, when compared to immortalized cell lines, and are therefore of increasing importance for investigating respiratory viruses [99,100,101].

2.2. Eosinophils

Eosinophils are a subset of granulocytic, polymorphonuclear leukocytes originating in the bone marrow and can be detected in low numbers in the peripheral blood after maturation [102,103]. They can migrate to different tissues, where they contribute to the physiology of the tissue under homeostatic conditions [102,103]. Increased numbers of eosinophils and their recruitment to the infected tissue can be observed during viral infection, mediating the primary antiviral host defense through their phagocytic and antigen-presenting activity. The protective role of eosinophils during infection has been shown for several viruses, like Influenza A virus (IAV), SARS-CoV-2, human parainfluenza virus, and human rhinovirus [104,105,106,107].
Chemoattractants like CCL5 or MIP-1α, secreted by RSV-infected AECs, lead to the recruitment of eosinophils to the site of infection [108,109,110]. Following recruitment, eosinophils sense viral ssRNA in a TLR7-dependent manner [111]. This induces a signaling cascade resulting in the increased expression of the transcription factor IRF7 and, consequently, the increased expression of type I IFNs like IFN-β [111]. Upon interaction with infected AECs and viral RNA, eosinophils are activated, characterized by the increased surface expression of phagocytic surface marker CD11b (Figure 2) [111,112,113]. Activated eosinophils contribute to viral clearance by degranulation, defined by the secretion of RNA-degrading enzymes, such as the eosinophilic cationic protein (ECP) [109,114].
Despite their potential role in virus elimination, in the context of RSV, eosinophils are mostly known for their alleged role in ERD in infants following FI-RSV vaccination. Early studies with the tissue of the two fatal cases (ex vivo) reported an increased infiltration of eosinophils into the lungs compared to control groups [29]. Eosinophilia was consequently considered a hallmark of ERD. In the following years, several studies indicated that eosinophils do not directly contribute to ERD [115]. Vaccination studies with FI-RSV followed by RSV challenge in cotton rats demonstrated that levels of neutrophils and lymphocytes were increased in vaccinated rats compared to unimmunized control rats [116]. In mice depleted of regulatory T cells (Tregs), RSV infection led to increased disease severity and increased levels of eosinophils, CD4+, and CD8+ T cells in the bronchoalveolar lavage (BAL) [117]. CD4+ T cells found in the BALs of these mice had a Th2-type phenotype and secreted IL-13 during infection [117]. Therefore, excessive recruitment of eosinophilia may only result from an unbalanced Th2-biased T-cell response and the increased expression of Th2-type cytokines like IL-4, IL-10, and IL-13 [118].
Apart from their still tentative role in FI-RSV-associated ERD, several in vitro and ex vivo studies suggested a contribution of eosinophils in severe cases of RSV infection (Figure 3). In RSV-infected infants, increased levels of ECP were detected in nasopharyngeal secretions that correlated with increased disease severity compared to uninfected controls [110,119]. The direct interaction or infection of eosinophils by RSV may explain their altered immune response as activated eosinophils secrete proinflammatory cytokines like CCL5, IL-6, and MIP-1α after viral interaction resulting in a proinflammatory milieu and increased influx of proinflammatory cells [79,120,121,122]. In IL-5-deficient mice, eosinophilic infiltration into the lungs was abolished following RSV infection and restored after IL-5 transfer into the mice, indicating a crucial role of IL-5 in eosinophil recruitment [123]. However, eosinophilic recruitment to the site of infection led to airway hyperresponsiveness and increased disease severity compared to sham-infected controls [123]. A recent study showed that infection of neonatal mice with RSV resulted in airway hyperresponsiveness and increased numbers of eosinophils, macrophages, and CD4+ T cells in the lungs compared to mock-infected mice, demonstrating that recruited eosinophils can contribute to disease severity [124].
Together, these studies indicate that excessive eosinophil recruitment to the lungs and subsequent lung pathology may result from altered CD4+ T cell signaling. Eosinophils usually contribute to protection against RSV infections. Th2-biased responses, however, lead to their increased recruitment and activation during infection and result in excessive and proinflammatory immune responses.

2.3. Neutrophils

Neutrophils belong to the polymorphonuclear leukocyte system and constitute the most abundant leukocyte in the blood [125]. Similar to eosinophils, neutrophils can migrate into tissue during a steady state and contribute to tissue homeostasis [125]. Under pathological conditions, like viral infections, neutrophils are involved in virus detection and elimination, cytokine production, recruitment of proinflammatory immune cells, and antigen presentation [126,127]. Neutrophil activity is mainly based on three central mechanisms: phagocytosis, degranulation, and NETosis [126].
During RSV infection, chemoattractants like ICAM-1 and IL-8 produced by epithelial cells lead to the recruitment of neutrophils to the site of infection [75,128]. Following recruitment, neutrophils interact with ICAM-1 on the surface of RSV-infected AECs leading to their activation characterized by increased expression of CD11b, CD18b, and myeloperoxidase (MPO) (Figure 2) [129,130]. Once activated, neutrophils can initiate several mechanisms to inhibit the further spread of infection. The formation of neutrophil extracellular traps (NETs) is a hallmark of neutrophil activation. NETs are extracellular structures of histones, elastase, MPO, and defensins [126]. Viral particles are captured by these structures and neutralized by MPO and defensin secretion [126]. It was shown that the interaction of RSV F protein with neutrophilic bactericidal/permeability-increasing protein (BPI) induces NET formation in a TLR4- and reactive oxygen species (ROS)-dependent manner, thus promoting viral neutralization [131,132]. Neutrophils can further limit viral infection and replication by producing ROS (‘respiratory burst’), virus phagocytosis, and degranulation [129]. Neutrophils can release a subset of chemokines, cytokines, and antimicrobial substances like TNF-α, IL-6, IL-8, IFN, and matrix metalloprotease 9 (MMP9), resulting in the recruitment of monocytes and macrophages [133]. Neutrophils, therefore, can contribute to viral clearance during RSV infections by exhibiting an antiviral response and coordinating the immune signaling of innate and adaptive immune cells.
An immunopathogenic role during RSV infection has also been proposed for neutrophils since their numbers were increased in the airways of infants with severe disease [134,135]. Enhanced interaction of neutrophils with AECs may induce excessive cell damage characterized by lower ciliary activity, cilium loss, and detachment [133,136]. Recent studies further implicated pulmonary neutrophilia as contributing to ERD following FI-RSV vaccination [137]. Direct infection of neutrophils led to the secretion of IL-8, MIP-1α/β, and MPO, resulting in an increased proinflammatory milieu (Figure 3) [138,139]. The increased secretion of IL-8, a potent chemoattractant for neutrophils, by infected neutrophils may lead to further recruitment of neutrophils to the infection site, contributing to immune pathogenesis [139]. Although NETs generally have an antiviral role, their disproportional formation during infections might enhance disease. Increased generation and accumulation of NETs were shown to block the airways of infants with severe infections [132,140]. In a recent experimental human infection study, the inflammatory activity of neutrophils in the respiratory mucosa of volunteers before infection predisposed the latter to symptomatic RSV infection [141]. The presence and activity of neutrophils were linked with impaired antiviral immune responses. Asymptomatic outcomes were characterized by IL-17 expression in the early phase of infection [141]. Preexisting neutrophilic inflammation, therefore, may counteract the protective response during the early stages of infection and instead lead to a higher risk of symptomatic infection [141].
Similar to other immune cells, neutrophils exhibit a bivalent role during RSV infections where time and microenvironmental conditions are critical for neutrophilic activity. Neutrophils may contribute to viral clearance by inhibiting viral spread and coordinating innate and adaptive immune responses. However, excessive neutrophil recruitment and activation of neutrophils can lead to an exaggerated immune response, correlated with disease severity in infants.

2.4. Natural Killer Cells

Bone-marrow-derived NK cells are granular lymphocytes best known for their cytotoxic activity [142]. Since NK cells do not undergo somatic hypermutation like other lymphocytes (i.e., B and T cells), they are considered part of the innate immune response [143]. NK cells are involved in various processes, like maintaining immune homeostasis, clearing tumors or infections, and regulating immune responses by interaction with different immune cells like T cells, macrophages, or dendritic cells [142,144]. NK cells contribute to viral clearance through the secretion of IFN-γ and the lysis of infected cells by perforin- and granzyme-dependent antibody-dependent cellular cytotoxicity (ADCC) [144].
Following RSV infection, NK cells accumulate in the lung and are an important source of IFN-γ, especially during the early phase of infection [145]. Besides their cytotoxic role, NK cells are involved in the IFN-γ-mediated recruitment of CD8+ T cells during infection (Figure 2) [145]. In RSV-infected mice, NK-derived IFN-γ prevented the development of lung eosinophilia in an IL-12-dependent manner [146]. Infection experiments in NK-deficient mice further showed an impaired IFN-γ production resulting in a Th2-skewed immune response and lung disease [147]. In secretions of RSV-infected infants with bronchiolitis, low numbers of NK cells and IFN-γ were detected, indicating that impaired NK activity is associated with more severe disease manifestations [148]. These studies thereby show a cytotoxic and immunoregulatory role of NK cells during RSV infection, mainly by the production of IFN-γ. In TLR4-deficient mice, the recruitment into the lungs and cytotoxic activity of NK cells was diminished upon infection compared to wild-type mice, indicating that RSV F protein activation of TLR4 is important for the recruitment and activity of NK cells [149].
Excessive recruitment of immune cells into the lungs during RSV infection, as seen for eosinophils and neutrophils, can induce immunopathogenic side effects. In RSV-infected mice, NK cells accumulated in the lungs during the early stage of infection and produced large amounts of IFN-γ (Figure 3) [150]. The depletion of NK cells reduced the disease severity and the influx of inflammatory cells like T cells [150]. In studies with nude and wild-type BALB/c mice, increased numbers of NK cells in the lung were associated with airway inflammation and hyperresponsiveness [151]. The depletion of NK cells reduced disease severity and decreased the levels of Th2-type cytokines IL-4, IL-5, and IL-13, indicating that NK cells might contribute to a Th2-skewed immune response at later stages [151]. Direct infection of NK cells might be an explanation for these immune responses. In vitro studies have shown that RSV infects neonatal and adult NK cells efficiently [152]. This infection of NK cells was further increased by sub-neutralizing antibodies, indicating the uptake of viral particles in an Fc-γ-receptor-dependent manner [152]. Infected NK cells were characterized by an increased expression of IFN-γ and a decrease in perforin secretion, suggesting a shift from the cytotoxic to the proinflammatory phenotype of RSV-infected NK cells [152].
The role of NK cells seems to depend on the stage of infection. In mice infected with recombinant RSV expressing IL-18, increased NK cell numbers were detected at early stages that correlated with increased lung inflammation and reduced viral replication [153]. Depleting NK cells during that infection stage reduced the disease severity and increased the viral load. However, the depletion of NK cells at later stages of infection was associated with increased disease severity mediated by the increased influx of CD8+ T cells [153]. This demonstrates the biphasic role of NK cells during RSV infections.
The NK cell response strongly depends on environmental stimuli. At the early stages of infection, the cytotoxic and immune-recruiting activity of NK cells is required to restrict viral spread. However, the cytotoxic activity of NK cells may also affect uninfected cells and thereby contribute to increased lung pathology. At later stages of infection, as immune cells infiltrate the lung, NK activity must shift from a cytotoxic to a regulatory phenotype to limit immune-cell-mediated lung inflammation. An imbalance in this immune response, induced by direct RSV infection or an altered microenvironment, can lead to increased lung damage.

2.5. Monocytes

Monocytes originate in the bone marrow from a common myeloid progenitor and belong to the mononuclear phagocyte system [154]. Monocytes are classified into three groups based on surface markers and function [155]. Classical monocytes represent the largest population (~90%) and are CD14++CD16-CCR2hiCX3CR1low with inflammatory and phagocytic activities [155]. Non-classical monocytes with a CD14+CD16+CCR2lowCX3CR1hi phenotype have antiviral and patrolling activities [155]. Intermediate monocytes represent a transitional population between classic and non-classic monocytes and have a CD14+CD16+CX3CR1hiCCR2low phenotype. Intermediate monocytes are proinflammatory and show increased activity in antigen presentation [155]. Mature monocytes circulate in the blood and infiltrate the tissue in response to chemoattractants. In the steady state, monocytes can differentiate into macrophages and dendritic cells in the presence of granulocyte-macrophage colony-stimulating factor (GM-CSF) and macrophage colony-stimulating factor (M-CSF) [156,157]. During viral infections, monocytes and monocyte-derived phagocytes, i.e., macrophages and dendritic cells, contribute to viral clearance by the secretion of antiviral mediators and phagocytosis of infected cells [135,136], as shown for HIV-1 and the influenza virus [158,159,160].
Monocytes are recruited to the site of infection during the early phase by IL-6, IL-8, CCL2, CCL5, and MIP-1α derived from infected AECs and macrophages [108,161,162]. The kind of recruited monocytes depends strongly on the secreted cytokines. CCL2 mediates classical inflammatory cytokine recruitment while non-classical, anti-inflammatory monocytes are recruited by CX3CL1 [156]. Alterations in the secretion pattern, e.g., mediated by RSV, could lead to a modified and shifted immune response. Following recruitment and activation, monocytes produce CCL2 and MIP-1α/β, resulting in the recruitment of immune cells [108]. They directly contribute to viral clearance by phagocytosis of the virus in a surfactant A-enhanced manner [163]. In response to uptake, monocytes expressed type I IFNs, TNF-α, G-CSF, and IL-6 [109]. A byproduct of monocyte-mediated viral clearance is the decrease in CCL5 expression by AECs and, consequently, the limitation of immune cell infiltration and cell-mediated immunopathology [109]. Growth factors like G-CSF, however, are secreted continuously and mediate the survival and differentiation of recruited cells [109]. Direct interaction of the RSV F protein with TLR4 and CD14 on the surface of monocytes further initiated innate immune pathways that resulted in the secretion of IL-6, IL-8, TNF-α, and IL-1β, which in turn mediated the antiviral immune response [164]. In TLR4-deficient mice, recruitment of CD14+ and NK cells was diminished upon infection while viral clearance was delayed [149,164].
The upregulation of co-stimulatory factors like CD40, CD80, and MHC-I/II on the surface of activated monocytes reflects their role in antigen presentation (Figure 2) [109,165]. Expression of HLA-DR on the surface of monocytes isolated from RSV-infected infants was reduced and correlated with increased disease severity, underlining their importance in antigen presentation and the initiation of adaptive immunity [165]. A previous study analyzed the immune responses in a CD14-deficient patient with recurrent RSV infections and discovered that this deficiency impaired the innate immune response, i.e., a decrease in IL-6 expression, mediated by monocytes to RSV, resulting in inadequate control of infection characterized by recurrent and severe RSV infections [166]. Monocytes, therefore, contribute to viral clearance by phagocytosis of viral particles and by supporting the proliferation, activation, and survival of other immune cells through the secretion of cytokines and upregulation of surface markers.
Several studies also described the involvement of monocytes in immunopathogenesis. In vitro restimulation of monocytes isolated from RSV-infected patients with bronchiolitis (ex vivo) showed an increase in IL-10, associated with recurrent wheezing, and decreased levels of IFN-γ, IL-12, and IL-4, indicating a direct contribution of monocytes to immunosuppression during severe infections (Figure 3) [167]. RSV can productively infect monocytes the same as other immune cells [168,169]. The permissiveness of monocytes to infection depended on their maturation and activation state, as in vitro studies of isolated cord blood monocytes were more permissive to infection than adult monocytes [168]. Infection of neonatal monocytes induced the expression and secretion of proinflammatory cytokine IL-1β [170]. The infection did not affect their cell viability, indicating that RSV inhibits the apoptosis of monocytes and instead alters their cytokine response [170].
The interaction of soluble G with CX3CR1 on the surface of monocytes may lead to altered gene and cytokine expression by inhibiting the nuclear translocation of NF-κB [171]. This change in cytokine response, notably the increased expression of IL-10, may skew for a Th2-typed immune response, a hallmark of severe infections [171]. Following their recruitment to the lungs, monocytes are exposed to different stimuli leading to their differentiation into macrophages with a proinflammatory (M1) or anti-inflammatory phenotype (M2) (described in Section 2.6). Based on this, it can be speculated that RSV alters the lung microenvironment resulting in increased recruitment of non-classical, anti-inflammatory monocytes. Microenvironmental stimuli and possibly the interaction of soluble G with CX3CR1 on their surface may result in alternatively activated, anti-inflammatory monocytes with an M2-like phenotype. This shift in the early immune responses would impair viral clearance.
Recruiting functional monocytes to the lungs in response to stimuli is pivotal for protection and immune regulation. In addition, monocyte recruitment is crucial to replenish the macrophage population during infection. Despite their protective effects, microenvironmental changes, especially due to G protein interactions, can affect their immune response. A modified cytokine expression following infection in the lung may shift monocyte differentiation toward an anti-inflammatory, M2-like phenotype, resulting in delayed viral clearance.

2.6. Macrophages

Macrophages belong to the mononuclear phagocyte system, like monocytes and dendritic cells, and are characterized by their phagocytic activity toward cell debris in different situations, like tissue repair or infections [154,172]. Macrophages circulate in the bloodstream but can migrate and reside in various tissues, like the lung, liver, and brain, in response to microenvironmental stimuli [172]. Two subsets of macrophages are present in the lung: alveolar macrophages, which are present on the surface of the alveolar cavity, and interstitial macrophages, which remain in the interstitium [173,174]. Lung-tissue-resident, alveolar macrophages show two distinct polarization states, M1 and M2, with different functions [156,173]. M1-like macrophages display a proinflammatory phenotype due to the secretion of proinflammatory cytokines, decreased expression of CX3CR1, and the increased expression of CD86, a co-stimulatory surface marker involved in activating T cells, demonstrating their role in antigen presentation [156,173]. M2-like macrophages, on the other hand, are characterized by the increased expression of CX3CR1, and mediate anti-inflammatory responses and contribute to tissue repair [156,173]. As mentioned above, monocytes are recruited to the lungs during viral infection to replenish the macrophage population. It should be noted that monocyte-derived macrophages are a distinct population formed during infection and do not have the role and function of tissue-resident, long-lived macrophages [156]. During the early stages of infection, monocytes differentiate into M1-like macrophages to mediate viral clearance and initiate adaptive immunity [156]. At later stages, monocyte differentiation switches from M1 to M2 polarization to limit the inflammatory response and promote tissue repair [156]. However, macrophage polarization is not irreversible and can shift between the different M-like subsets depending on endogenous stimuli [156].
Macrophages can detect RSV through MAVS-coupled RLRs and IFNAR signaling, producing type I IFNs, like IFN-α/β, IL-6, MIP-1α, and CXCL10 (Figure 2) [161,175,176]. It was shown that the macrophage-mediated release of IFNs is crucial for controlling RSV infection as they recruit immune cells to the infection site [161]. Several studies emphasized the role of macrophages in controlling and clearing viral infection. Depletion or impaired activity of macrophages led to increased viral replication, disease severity, inflammation, and excessive neutrophil/dendritic cell infiltration [177]. It is thought that the depletion of alveolar macrophages resulted in a proinflammatory environment characterized by increased expression of G-CSF, IL-17, TGF-β, MIP-1α, and IL-α/β, and decreased levels of TNF-α, IL-6, and type I IFNs [177]. It could thus be speculated that the functional immaturity of neonatal alveolar macrophages leads to an inefficient uptake of infected cells, recruitment of immune cells, and clearance of damaged cells [178]. The activity of different subsets of macrophages appears to be dependent on the stage of infection, similar to the activation pattern of NK cells. During the early stage of infection, M1-like, proinflammatory macrophages are essential for the restriction of viral spread, removal of infected cells, expression of type I IFNs for immune cell recruitment, and antigen presentation to initiate adaptive immune responses. Later during an infection, an M1 to M2 phenotypic switch can be observed, where M2 macrophages produce type III IFNs in a PPAR-γ-dependent manner and secrete IL-10 to control the immune-cell-mediated inflammatory response and mediate tissue repair [179]. M2 differentiation is thought to be mediated by TLR4 and IFN-β signaling [180]. This phenotypic switch is crucial for suppressing exaggerated, unspecific antiviral responses and preventing unnecessary tissue damage.
A possible contribution of macrophages to immunopathology remains inconclusive and requires further research. Several in vitro and in vivo studies show the infection of macrophages by RSV [181,182,183]. This infection is TLR4- and CX3CR1-dependent and polarizes the differentiation of neonatal macrophages toward an M2-like phenotype (Figure 3) [180,184,185]. It is unclear whether it is an abortive or productive infection, as various studies yielded different conclusions. While one study shows no loss of cell viability upon infection [181], another indicated that RSV induces necroptosis and suggests an enhancement of viral replication by M2-like macrophages, thus contributing to disease severity and lung pathology [183]. Regardless of that, in vitro infection of macrophages, isolated from adult or neonatal humans, has been shown to alter the immune response, characterized by the expression of proinflammatory cytokines like TNF-α, IL-6, and IL-8 [186,187], while the expression of IFN-γ and IL-12, a regulatory cytokine, was impaired [187]. Reduced expression of IFN-γ was associated with impaired activation of macrophages resulting in reduced phagocytosis of the virus and recruitment of immune cells like T cells [188]. These data suggest that the direct interaction with RSV impairs the activation and function of neonatal macrophages by skewing their differentiation toward an M2-like phenotype, thereby contributing to disease severity.
Macrophage responses during RSV infections follow a biphasic course similar to NK cell response. During the early stage of infection, proinflammatory M1-like macrophages are activated to restrict viral spread. At later stages, probably by the arrival of T cells, a phenotype switch towards an anti-inflammatory, immune regulatory M2-like type is necessary to reduce immunopathogenesis. It can be speculated that in infants with severe infections, the change in the macrophage response is impaired, leading to exaggerated T-cell responses and increased lung damage.

2.7. Dendritic Cells

Dendritic cells complete the mononuclear phagocyte system and are considered professional antigen-presenting cells as they bridge innate and adaptive immune responses during infections [189]. Compared to the other two members of the phagocyte system, DCs only display a low phagocytic activity [190]. Two DC subsets have been described based on the expression of surface markers, function, and origin. Conventional or myeloid DCs (cDCs) are involved in tissue damage sensing and the capture and presentation of antigens [191,192]. cDCs are further divided into cDC1s, located directly underneath the airway epithelium and responsible for MHC-I-mediated antigen presentation to CD8+ T cells, and cDC2s, located in the lung parenchyma and presenting antigen to CD4+ T cells [191,192]. Plasmacytoid DCs (pDCs) are found in blood and lymphoid tissues and are an essential source of IFN-α/β [191,192]. While DCs show reduced phagocytic activity and low antigen presentation under homeostatic conditions, their T cell-priming activity increases during viral infections as co-stimulatory markers, like CD40, CD80, and CD86, are upregulated on the surface [191].
Upon infection with RSV, DCs are recruited to the site of infection and sense the virus either through direct infection or by binding viral antigens through PRRs, like TLRs or RLRs [193,194]. As a result, co-stimulatory molecules CD40, CD80, CD83, CD86, and MHC-I/II were found to be upregulated on the surface of infected DCs [195], indicating their maturation (Figure 2). Ex vivo studies implied a ‘two-step process’ for maximal maturation of DCs since only the direct infection of pDCs led to increased expression of CD40 [196]. Once DCs are activated and matured, TLR7 and myeloid differentiation factor 88 (MyD88) mediate the production of antiviral and Th1-type cytokines (CCL5, IL-12, IFN-α, and IFN-β) [176,193,197,198]. Matured DCs then migrate to lung-draining lymph nodes to initiate the virus-specific T-cell response and have an essential role in initiating the virus-specific adaptive immune response and, consequently, in controlling RSV infection.
Despite their protective and crucial role in immunity, DC functions are heavily affected by RSV-induced immune modulatory mechanisms. Several studies showed that the direct infection of DCs impacts subsequent DC-mediated T-cell activation (Figure 3) [199,200]. Upon infection, the RSV N protein is expressed on the surface of infected DCs, which is thought to interfere with DC–T cell synapse formation [201]. This interference impairs or delays the formation of RSV-specific T-cell immunity [201]. Others found a decreased expression of co-stimulatory molecules on infected neonatal DCs that impaired their recruitment to the lymph nodes, ultimately reducing virus-specific T-cell responses compared to the control group [202,203]. The increased expression of IL-4R-α on the surface of neonatal cDC2s, in comparison to cDCs derived from adult mice, may also account for the altered DC responses and impaired recruitment of the cells into the lymph nodes as the deletion of the receptor resulted in increased recruitment to the lymph nodes, reduced Th2 polarization, and upregulation of IL-12 expression [204]. Interaction of the RSV G protein with DC-/L-SIGN inhibits the activation of DCs and interferes with the DC-induced production of cytokines, illustrating an immunomodulatory mechanism by RSV for interfering with the host’s immune response [205].
DCs, as the bridge between innate and adaptive immunity, are of major importance for the induction of virus-specific immune responses. The direct infection of DCs and the associated functional changes may lead to severe consequences in initiating a functional adaptive immune response.

3. Adaptive Immune Responses to RSV Infection

The adaptive immune response represents the second line of defense against infections. Although innate immunity reacts to infection rapidly, the recognition of pathogen patterns is limited since the responsible receptors are germline-encoded [206]. Adaptive immune responses, on the other hand, are characterized by highly specific receptors on the surface of the lymphocytes allowing targeted, pathogen-specific responses. Activation of adaptive immune cells leads to their proliferation and clonal expansion, exerting their effector function, and ultimately to the generation of an ‘immunological memory’ that persists in the host and can be rapidly reactivated during reinfection [207,208]. The adaptive immune system comprises two cell types: T lymphocytes and antibody-secreting B cells [207]. Mature adaptive immune cells reside in secondary lymph nodes and can infiltrate the site of infection after stimulation with antigens presented by innate immune cells, like monocytes, macrophages, or DCs [207]. In the following part, the protective roles of B and T cells and the potential impairment of their function by RSV during infections will be discussed in more detail.

3.1. B Cells

B cells originate in the bone marrow from hemopoietic stem cells [209]. In an immature state, immunoglobulin (Ig)-expressing B cells leave the bone marrow and migrate to secondary lymphoid organs for their final development and maturation [209]. Final B cell activation is mediated by two distinct processes: T-cell-independent and -dependent activation, both of which require the binding of an antigen to the B cell receptor (BCR) [209]. During T-cell-independent activation, antigens with multiple, repeating epitopes are necessary for the cross-linking of BCRs, leading to the proliferation and differentiation of B cells into antibody-secreting plasma cells [209]. For the majority of antigens, however, B cell activation requires the help of CD4+ T helper (Th) cells. During this process, B cells internalize the antigen and present it on the cell surface through MHC-II. The antigen is then recognized by antigen-specific Th cells that initiate B cell proliferation and differentiation by transmitting activating signals [209]. Besides the generation of antibody-secreting plasma cells, the T cell-dependent activation of B cells also allows the development of B memory cells. B cell development is completed by somatic hypermutation, affinity maturation, and isotype switching, all of which result in the production of highly specific antibodies [209]. Upon viral infections, B cells secrete virus-specific antibodies, neutralizing the virus and preventing viral cell entrance by blocking the viral binding domain [210]. B cells and antibodies, therefore, are an important correlate of protection by eliciting virus-specific effector functions.
Following RSV infection in infants and adults, B cell numbers in the peripheral blood were increased compared to uninfected individuals. They displayed a differentiated phenotype by the expression of typical proliferation, differentiation, and survival markers, such as B cell-activating factor (BAFF) and A proliferation-inducing ligand (APRIL) (Figure 2) [211,212,213,214,215]. These observations are supported by studies in mice where RSV infection led to the upregulation of BAFF, APRIL, and CXCL13 on B cells [216,217]. These studies demonstrate the activation and circulation of B cells following RSV infection and indicate their migration to the site of infection. Although B cells are activated and recruited during RSV infection, studies suggest that these numbers rapidly decrease once the infection is cleared [218,219,220]. In blood samples of infected adults, low levels of RSV-specific B cells were detected in healthy ‘non-healthcare’ workers [219]. After experimental infection of the volunteers, the numbers of RSV-specific B cells rapidly decreased within months after exposure [219]. Ex vivo studies confirmed these observations after the experimental infection of adults in which RSV-specific B-cell responses generated in the volunteers were poorly maintained and returned to baseline levels one year after exposure, indicating a defective memory B-cell response in adults to a certain degree compared to samples from RSV-negative, hospitalized infants [220].
RSV-specific, neutralizing antibodies can be detected in humans as early as two days after infection [213]. They mainly target the RSV glyco- and fusion protein (preF and postF confirmation) and display a cross-reactivity for both RSV subtypes [212,221]. RSV-specific antibodies can mediate viral clearance through various mechanisms, like ADCC, antibody-dependent cellular phagocytosis, and the neutralization of viral particles [221]. Their protective role was confirmed in mice, where administering RSV-specific, neutralizing antibodies before RSV exposure reduced the morbidity and mortality of the animals compared to IgG-treated controls [222]. The presence of these neutralizing antibodies before the RSV challenge further restricted virus replication in the early stage of infection and thereby reduced the activation of CD8+ memory T cells that displayed immunopathogenic activity in control mice through the increased secretion of IFN-γ [222]. In earlier studies with older adults, low RSV-neutralizing serum and mucosal antibody levels prior to infection correlated with disease severity [21,223,224,225,226,227]. Upon RSV infection, older adults develop stronger nasal and serum antibody responses than younger subjects, which is dependent on the duration of viral shedding [213,228]. In a recent study, older adults mounted poor nasal IgA responses, but robust serum IgG responses that correlated with recovery from the RSV infection [226]. Levels of RSV-specific, neutralizing antibodies, however, were relatively short-lived and titers dropped more than 4-fold within a year after infection in the majority of study subjects [218]. Despite the fact that older adults have comparable antibody levels to younger subjects prior to infection, they are more susceptible to severe RSV infections, which indicates that other immunological parameters, like dysfunctional T cells, may account for this [229,230].
Although antibodies mediate viral clearance, they may also contribute to immunopathogenesis during RSV infections. Isolated RSV-specific antibodies and B cells of infected infants show significant differences from those of adults [231]. F-specific antibodies of infants under 3 months of age showed restricted affinity, neutralization capacity, and recognition of antigenic sites compared to adult samples [231,232]. The RSV F protein contains six major antigenic sites (ø, I–V), of which only antigenic sites I and III were recognized by antibodies isolated from infants. Antibodies directed against antigenic site I do not display neutralizing activity and their preferred production in infants could result in lower levels of neutralizing antibodies. A high abundance of non-neutralizing antibodies in infants could ultimately result in the formation of immune complexes that may contribute to lung pathology if not cleared in time [231]. The defective generation of neutralizing antibodies seems to be age-dependent in childhood as the recognition of additional epitopes increases with age [231]. A genetic limitation in infants under three months of age may be responsible for this defect as infant B cells have limited use of antibody variable genes and lack somatic hypermutation [233]. Maternally derived antibodies may provide an explanation for this limitation since several studies indicated a suppressive effect of maternal antibodies on the infant’s immune response in the first months of life during RSV infection in vivo [219,234,235]. Recent immunization experiments in infant mice indicate that maternal antibodies altered B cell differentiation and isotype switching, thereby limiting the B cell repertoire [236].
The virus itself further affects B-cell responses. Analysis of neonatal regulatory B cells (nBregs) isolated from cord blood demonstrated their infectability by RSV [237]. The interaction of the RSV F protein with the B cell receptor activated the regulatory B cells and increased the expression of CX3CR1, a potential viral receptor, via the CX3C motif of the RSV G protein. Therefore, the interaction of RSV F with BCRs on the surface of nBregs leads to increased expression of CX3CR1 on the surface of the cells, making them susceptible to direct infection. The infection of nBregs resulted in the secretion of IL-10, an immunoregulatory cytokine, which reduced the production of Th1-type cytokines [237]. This study, therefore, demonstrates the immune modulatory effects mediated by RSV F and G on nBregs, ultimately resulting in an immune response that favors viral survival (Figure 3) [237]. Besides the direct infection of nBregs, the secretion of soluble G may depict another immune modulatory effect on B cell activity. Soluble G acts as an antibody decoy during infections, thereby serving as an escape mechanism for viral progeny from G-specific antibodies and interfering with antibody-mediated viral clearance [238].
Antibodies are the major correlate of protection during RSV infection in infants, as shown by the prophylactic use of human polyclonal neutralizing antibodies [239,240,241] and the neutralizing monoclonal antibodies palivizumab and nirsevimab [242,243]. The presence of pre-existing, RSV-specific antibodies and, therefore, memory B cells during reinfections is crucial, as they are able to detect the virus before the infection of immune cells. Memory B cells thus have a relatively long-lasting effector function, while the effector functions of memory T cells are limited to the presence of antigen [244].
Direct modulation by RSV G and the genetic limitations of the immune system in infants under 3 months of age interfere with the generation of neutralizing antibodies, leaving the infant at risk of more severe infections. Direct infection of nBregs resulting in the secretion of immunoregulatory IL-10 demonstrates that interactions of viral proteins can shift the immune response, generating an environment that favors viral shedding. Although RSV-specific immune responses have been studied extensively in infants, the induction of these responses in older adults has been underinvestigated and warrants more research, especially in the context of age-related immunosenescence [245].

3.2. T Cells

Unlike B cells, T cells arise from a common lymphoid progenitor in the thymus [208]. T cells migrate to secondary lymphoid tissues upon maturation to encounter antigens [207,208]. Two major subsets of T cells have been described, characterized by the expression of either CD4 or CD8 on their surface. Both subsets are activated by the formation of a so-called ‘immunological synapse’ which describes the interaction of different T cell markers, e.g., CD4/CD8, CD3, or T cell receptor, with antigen-bearing MHCs on the surface of professional (MHC-II; DCs, macrophages, monocytes, and B cells) and non-professional (MHC-I; epithelial cells) antigen-presenting cells [207,246]. Activated T cells can exert various effector functions, such as the elimination of infected cells, activation of other immune cells, like B cells, through the secretion of cytokines or direct interaction, or regulation of immune responses to prevent excessive damage [247]. In the following part, the protective role of the individual T cell subsets as well as their contribution to immune pathogenesis during an RSV infection will be illustrated in more detail.

3.2.1. CD4+ T Cells

CD4+ T cells are pivotal in the host’s immune response as they orchestrate the activity of innate and adaptive immune cells through the secretion of several cytokines. They can be divided into different subsets (i.e., Th1, Th2, Th9, Th17, T-follicular helper, and Tregs), which mainly provide helper functions and are characterized by the secretion of specific cytokines [247]. For example, Th1-like cells are involved in activating mononuclear phagocytes or cytotoxic T cells by the secretion of IFN-γ, TNF-α, IL-2, IL-12, and IL-18. Th2-like cells, by contrast, are involved in eosinophil responses, and B cells maturation, and production of virus-specific antibodies by secreting IL-4, IL-5, IL-10, and IL-13 upon activation [247].
During RSV infection, CD4+ T cells are recruited to the airways and orchestrate the immune response via secretion of IL-17, IFN-γ, and TNF-α (Figure 2). The adoptive transfer of CD4+ T cells into nude mice demonstrated their beneficial role during infection and B cell differentiation as they were necessary to induce the production of RSV-specific antibodies [248]. Adoptive transfer of CD4+ T cells from the airways of RSV-infected mice into naïve mice reduced disease severity by suppressing the secretion of TNF-α compared to control mice, further supporting their protective role during infection [249]. In human experimental infection studies, numbers of RSV F- and G-specific, CD4+ T cells were increased in the airways of infected individuals [250]. By contrast, in infants with severe RSV infection with subsequent wheezing, numbers of CD4+ T cells were reduced and produced lower levels of TNF-α after in vitro restimulation compared to infants without wheezing, indicating a partial impairment of the CD4+ T-cell responses in these children [148,251,252]. The production of proinflammatory cytokines by CD4+ T cells during viral infections is crucial for the activation and regulation of innate and adaptive immune responses. However, the balance between pro- and anti-inflammatory cytokine responses is of great importance, and any alteration in this balance may have serious consequences, as illustrated by the use of TNF-α. The studies mentioned above show that an imbalanced TNF-α response during RSV infections can lead to more severe disease and prolonged viral shedding.
A dysfunctional CD4+ T-cell response during infection was linked to immunopathology after FI-RSV vaccination [253,254]. Studies in mice vaccinated with FI-RSV showed a Th2-skewed immune response characterized by the increased expression of Th2-type cytokines like IL-5 and IL-13, resulting in airway hyperresponsiveness (AHR) and increased mucus secretion compared to mock-immunized controls [255]. Depletion of CD4+ T cells in the mice significantly improved the condition of the mice during infection, indicating that a biased Th2-type immune response contributes to disease severity in FI-RSV-vaccinated mice [255]. Similarly, RSV infection of neonatal mice and their reinfection in the adult phase led to an exaggerated Th2-type immune response resulting in AHR, mucus hypersecretion, and eosinophilia compared to mice that were infected during adulthood [256]. The increased expression of IL4-Rα on the CD4+ Th cells of these mice indicates a preferential proliferation of CD4+ T cells to a Th2-type [256]. In experimental infection studies of adults, increased levels of CD4+ T-cell-derived IFN-γ, IL-2, IL-4, IL-10, and TNF-α and the number of CD8+IFN-γ+ T cells correlated with increased disease severity and hospitalization, emphasizing the potential immunopathogenic role of CD4+ T cells during RSV infection [257]. Depletion of CD4+ T cells, IL-10, or IL-4 before infection or vaccination ameliorated disease severity but consequently resulted in prolonged viral shedding [118,255,258,259,260]. It has been shown that RSV can productively infect CD4+ and CD8+ T cells, resulting in the decreased expression of IFN-γ and interfering with the T cell functions and development of Th1 cells, probably in an F-protein-dependent manner (Figure 3) [261,262]. This direct infection may hinder the generation of long-term protective immunity since RSV-specific lymphocyte responses only persist for about a year and do not boost virus-specific cell-mediated immunity upon reinfection [263].
A balanced CD4+ T-cell response is pivotal for developing protective immune responses by coordinating the recruitment and activity of innate and adaptive immune cells through the secretion of cytokines. Direct infection by RSV and partial genetic dysfunction of the infant’s CD4+ T-cell response, however, may interfere with this balance, leading to a Th2-skewed immune response and more severe infections.

3.2.2. CD8+ T Cells

CD8+ T cells can recognize and eliminate virus-infected cells through their cytotoxic activity. During RSV infection, naïve CD8+ T cells are activated through the interaction of the T cell receptor either with MHC-I on the surface of infected epithelial cells or with MHC-I on the surface of APCs in lung-draining lymph nodes (cross-presentation) [195]. Following activation, virus-specific effector CD8+ T cells expand and migrate to the site of infection in response to chemoattractants CCL5 and CXCL10 [264,265]. Activated CTLs directly interact with antigen-bearing MHC-I on the surface of RSV-infected cells and induce the apoptosis of the target cell by the secretion of perforin and granzymes [266,267,268].
Several studies revealed a protective role of CD8+ T cells during RSV infection. The depletion of CD8+ T cells in RSV-infected mice resulted in delayed viral clearance compared to control mice. The adoptive transfer of CTLs, isolated from RSV-infected mice, into naïve mice during RSV infection reduced both the viral load and weight loss [249]. These results are supported by human studies analyzing the blood and tissue samples of infected infants, in whom reduced CD8+ T cell counts were associated with delayed viral clearance and increased disease severity in comparison to uninfected infant samples, emphasizing their protective role during infection [148,252,269,270]. In an RSV-infected infant with severe immunodeficiency, bone marrow transplantation significantly reduced the nasal viral load [271]. This decrease in viral load began with the increasing activity of CTLs after transplantation [271]. CTLs mediate their effector functions during RSV infection by the secretion of several cytokines, e.g., IL-2, IL-10, INF-γ, TNF-α, and the serine protease granzyme B (Figure 2) [264,266,267,272,273,274,275]. The importance of CD8+ T-cell-derived cytokines is demonstrated by studies in mice that were primed for an RSV-specific CD8+ T-cell response prior to RSV challenge [276]. In the primed mice, CD8+ T cells suppressed the exaggerated Th2-type cytokine response characterized by the decrease in IL-4 and IL-5, and consequently led to the suppression of excessive recruitment of eosinophils compared to controls. Thus, it can be assumed that CD8+ T-cell-derived cytokines contribute to the regulation of CD4+ T-cell responses [276].
Despite their crucial role in viral clearance, CTLs are also involved in immunopathology following RSV infection. RSV-infected mice displayed increased levels of CTL-secreted IFN-γ that correlated with disease severity in comparison to control mice [277]. Depletion of CTLs in mice markedly reduced disease severity following RSV infection, although viral shedding was prolonged, indicating that CTLs are involved in both protective and harmful immunity [258]. In BAL samples of hospitalized, RSV-infected infants, increased levels of granzymes A and B were detected compared to children without a pulmonary disease (Figure 3) [278,279]. The increased levels of granzymes correlated with IL-8 levels in the airways and disease severity [278,279]. In experimental infections of adults, the peak of CTL numbers in the respiratory mucosa was linked to the reduction in viral load and an increase in symptoms, emphasizing the dual role of CTLs during RSV infection [266]. Several studies have implicated CTL-derived IFN-γ and TNF-α as a driving force in CTL-mediated immunopathogenesis during RSV infection. Increased levels of IFN-γ in the BAL of RSV-infected mice correlated with disease severity, while its depletion with neutralizing antibodies or absence in IFN-γ-KO mice led to ameliorated disease [277,280]. A similar role in immunopathogenesis was shown for TNF-α. Its neutralization in mice before RSV infection reduced weight loss [281].
These data emphasize the role of CTLs in both virus elimination and immunopathogenesis. While CTLs mediate viral clearance through their cytotoxic activity and are involved in the regulation of a balanced CD4+ T-cell response, their excessive activation and infiltration of the lung tissue may contribute to lung pathology and disease severity characterized by the increased secretion of granzymes.

3.2.3. Regulatory T Cells

Previously discussed immune cells, such as B and T cells, illustrate the importance of a balanced immune response during viral infection, like SARS-CoV-2 or hepatitis B virus [282,283]. Tregs are a subset of CD4+ T cells characterized by the expression of CD25 and the transcription factor forkhead box protein 3 (FoxP3), which is pivotal for their development and regulatory function [284]. Tregs can control immune cell recruitment and proliferation and prevent exaggerated immune responses during infections by secreting TGF-β and IL-10 [285,286,287,288].
During RSV infection, Tregs accumulate early in the lungs and lymph nodes of mice [275,289,290]. The depletion of Tregs in mice resulted in the delayed recruitment of CTLs and CD4+ T cells to the site of infection, followed by delayed viral clearance, increased weight loss, and slower recovery compared to naïve control mice, indicating that Tregs mediate the recruitment of T lymphocytes during RSV infection [117,290,291,292,293]. Despite the delayed recruitment of CTLs and CD4+ T cells after Treg depletion in mice, their numbers were significantly increased later. They were associated with an excessive inflammatory response later during infection, characterized by the increased secretion of IFN-γ and TNF-α (Figure 2) [117,290]. Exaggerated and persistent Th2-like responses, characterized by IL-13-expressing CD4+ T cells, were also noticed in the absence of Tregs, further illustrating the role of Tregs in the regulation and suppression of immunopathology during RSV infection [117]. In vivo studies showed an increased expression of granzyme B by lung-resident T cells compared to naïve mice, suggesting that the expression of granzyme B mediates the regulatory activity of Tregs and that Treg-derived granzyme B may eliminate activated lymphocytes, thereby regulating inflammatory responses [292]. Since Tregs are a major source of IL-10, their immunosuppressive function might be mediated through an IL-10-dependent mechanism. Studies in IL-10−/− mice show an increased accumulation of CTLs, increased levels of proinflammatory cytokines, like IFN-γ and TNF-α, and increased disease severity in the lungs of RSV-infected mice in comparison to control mice [294,295]. The absence of Tregs in mice further indirectly affected immunity to RSV as decreased concentrations of neutralizing and increased concentrations of non-neutralizing antibodies were measured compared to those in naïve mice [293]. These data underline the versatile role of Tregs during RSV infection.
In infants with severe infections, the numbers of activated Tregs were reduced in blood and nasal aspirates compared to uninfected children and remained at that level for several weeks after infection [296,297]. The decrease in IL-33 levels, a cytokine involved in the accumulation and homeostasis of Tregs in mucosal sites, in the nasal aspirates of RSV-infected infants, may explain the reduced numbers of Tregs in these samples (Figure 3) [296]. The reduced accumulation of Tregs and lower levels of IL-33 in these infants may be the consequence of a Th2-type cytokine milieu. The Th2-type environment, characterized by the increased secretion of IL-5 and IL-13, induced the Th2-like development of the Tregs effector phenotype and a loss of its immunosuppressive function [298]. Th2-like Tregs were characterized by the expression of GATA-3, a transcription factor that can inhibit the expression of FoxP3 in Tregs [298]. Since Tregs are involved in cell recruitment, especially during the early stage of infection, a decrease in their activity would mean a delayed infiltration of immune cells into the tissue, e.g., CTLs, and, consequently, a delay in viral clearance [290].
Tregs and their activity are crucial to limiting immunopathology during RSV infection, probably mediated by granzyme B and IL-10. Alteration in the host immune response to RSV infection towards a Th2-like phenotype indirectly interferes with Treg activity, resulting in delayed and increased recruitment of immune cells and exaggerated responses.

3.2.4. Memory T Cells

Memory T cells are essential for the rapid, antigen-specific immune response during virus reinfection. Following viral clearance, the virus-specific T cell population contracts, leaving only a small fraction of cells to survive, which differentiate into persisting memory T cells [299]. Memory T cells can be classified into four subsets based on surface marker expression [300]. Following infection, central memory T cells (CD45RA-CCR7+) home in on secondary lymphoid organs, while effector memory T cells (CD45RA-CCR7) migrate into the peripheral tissue to mediate effector functions [300]. Late effector memory T cells are considered terminally differentiated and have a reduced functional capacity [300]. Tissue-resident memory T cells (Trm), characterized by the increased expression of CD69 and CD103, represent the last subset and reside within the peripheral tissue [266,301]. Lung Trms are further characterized by the expression of CD11a and CD49a [302].
During secondary infections with RSV, memory cell populations rapidly expand and induce a virus-specific immune response in a MAVS- and IFN-α-dependent manner [195,303,304]. Memory T cells in mice are recruited and activated in three waves: the initial activation of tissue-resident memory T cells in the lungs is followed by the secondary recruitment of circulating, RSV-specific memory cells from the periphery. The third wave consists of memory T cells activated by antigen-presenting cells in the lung-draining lymph nodes [305]. The fast response mediated by memory T cells leads to early virus recognition and elimination, reducing the disease severity and viral load [266]. In blood samples of young and elderly volunteers, resting CD8+ memory T cells were identified that were characterized by an increased expression of CD27, CD28, and IL-7Rα (Figure 2) [303]. In vitro restimulation of these memory cells led to the production of IFN-γ and granzyme B, demonstrating that they can regain their effector functions and proliferate and expand during RSV infection [303]. Similar results were shown in RSV-infected mice [306]. The adoptive transfer of tissue-resident memory T cells into naïve mice before infection reduced weight loss and the viral load compared to control mice showing the protective effect of Trms during RSV infection [249].
Because repeated RSV infections are common, an impairment of RSV-specific memory T cell generation may be assumed (Figure 3). In mice, RSV infection induces RSV-specific Trms that correlate with viral clearance and reduced disease severity [306]. However, the number of RSV-specific Trms rapidly waned within five months, corresponding to the age of a young adult in humans [306]. Prime and challenge studies in mice showed similar results as RSV-specific Trms displayed an impaired IFN-γ response, and the numbers of Trms declined rapidly after the clearance of infection [307]. This defect in RSV-specific Trm response was restricted to the respiratory tract, indicating an immunosuppressive effect of RSV on the effector functions and the generation of memory of local T cells after infection [307]. Preexisting, RSV-specific CD8+ memory T cells mediate viral clearance in the absence of RSV-specific CD4+ T cells and antibodies in mice, emphasizing the protective role of CD8+ memory T cells in the absence of other adaptive immune cells [308]. Besides their protective role, CD8+ memory T cells were also involved in excessive weight loss and increased disease severity and immunopathology in infected mice mediated by the increased secretion of IFN-γ compared to control mice [308]. This study demonstrates that memory T cells mediate viral clearance upon reinfection but also lead to increased disease severity and immunopathogenesis in the absence of regulation factors like CD4+ T cells and antibodies. In a recent study, peripheral blood mononuclear cells were isolated from healthy children who encountered RSV infection during infancy. Restimulation of these PBMCs showed an impaired memory T-cell response irrespective of the disease severity during RSV infection when compared to children that were not infected during infancy [309]. The altered memory T-cell response was indicated by reduced IFN-γ, TNF-α, and IL-2 secretion. This study demonstrates that RSV infections during infancy can attenuate the memory Th1- and Th17-cell responses to RSV later in life regardless of the disease severity of the primary infection or reinfection [309].
The reduced survival of memory T cells may be due to intrinsic cell programming, as studies in infant mice infected with influenza virus showed an increased expression of T-bet and a reduced expression of survival factor CD127 on the surface of T cells compared to infected adult mice. This altered phenotype persisted even after the adoptive transfer of infant Trms into adult mice, indicating that the host environment is not responsible for the inefficient establishment of memory and that infant Trms are intrinsically programmed for short-term immunity [310].
Generating memory T cells is essential for long-term protection against recurrent infections. Fully functional memory T cells are produced during RSV infections that can mediate protection upon reinfection. The rapid decrease in RSV-specific memory T cells and impaired functionality in infants and older adults indicate that this protection is not long-lasting and predisposes the host to recurrent infections. Further studies will be necessary to better understand the role of memory T cells during RSV infection and immunopathogenesis, and the role of RSV in regulating these processes.

4. Vaccine Development and Treatment Options with Novel Insights into Immune-Mediated Protection

After the first isolation of RSV from chimpanzees in 1956 and infants in 1957, the first formalin-inactivated vaccine, FI-RSV, was developed in the 1960s [311,312] and used in a cohort of infants in the USA. Vaccination with FI-RSV led to an increased frequency and severity of ALRIs in seronegative infants following their first natural infection and even led to the death of two vaccine recipients [29]. Possible explanations for ERD in children were discussed above (see Section 2.2, Section 2.3, and Section 3.2.1). For over six decades, the search for an effective vaccine continued without success. RSV vaccine and human monoclonal antibody development changed drastically with the stabilization of the F protein in its prefusion conformation as antibodies directed against prefusion epitopes have a tenfold increase in neutralizing activity compared to antibodies directed to postfusion epitopes [313]. Recently, two stabilized, prefusion F-protein-based vaccines, Arexvy (GSK) and Abrysvo (Pfizer), were licensed for the prevention of ALRI in older adults. Abrysvo was recently approved for the vaccination of pregnant women in their third trimester for the prevention of RSV-induced ALRI in infants during their first RSV season. Since a broad range of RSV vaccines and treatment options are currently in clinical trials, we focus on the ones that are either licensed or are/were in phase III clinical trials (Figure 4).
Vector-based vaccines have been used for decades for several pathogens, showing their potential to induce protective immunity in recipients after vaccination. They are generated by integrating a viral sequence encoding a target antigen into a non-pathogenic, often replication-deficient viral vector, like modified vaccinia virus or adenoviruses [314]. Vector-based vaccines generally aim to induce a robust antibody and CTL response and are characterized by high immunogenicity. The advantages of vector-based vaccines are the simple and safe production of a high number of vaccine doses [314]. Among the candidates for RSV, a modified vaccinia virus Ankara (MVA)-based vaccine expressing the RSV N, F, G, and M2 proteins (MVA-BN-RSV) is currently in phase III trials for Bavaria Nordic. MVA-BN-RSV would be recommended for use in older adults since vaccination induces stronger T-cell responses than antibody responses. In clinical trials, vaccination induced moderate neutralizing antibody responses and enhanced Th1-type immune responses indicated by the low levels of IL-4 in vaccinated individuals compared to placebo controls. A booster shot after 12 months further increased the antibody titers and T-cell responses in vaccine recipients [315,316]. Phase III clinical trials for MVA-BN-RSV are currently ongoing (NCT05238025). Although vector-based vaccines elicit strong T-cell responses, preexisting immunity to adenoviruses or poxviruses (in smallpox-vaccinated adults) may interfere with vaccine efficacy. Ad26.RSV.preF is an adenovirus-vectored vaccine candidate developed by Janssen Pharmaceuticals which expresses the preF protein. Similar to MVA-BN-RSV, it is designed for use in older adults and is additionally suitable for seropositive children. First trials with Ad26.RSV.preF resulted in increased antibody titers, reduced risk of ALRI, and reduced viral load in comparison to the unvaccinated group [317,318,319]. In March 2023, Janssen Pharmaceuticals discontinued its phase III vaccine development strategy based on Ad26.RSV.preF.
In contrast to whole virus vaccines, subunit vaccines are based on one or more purified viral antigens [320]. Therefore, the choice of antigen is critical in generating protective immune responses. Subunit vaccines are relatively safe as reversion to virulence is not possible, and a spread to unvaccinated individuals can be ruled out [320]. Compared to whole-virus vaccines, however, subunit vaccines must generally be delivered together with an adjuvant, and multiple doses may be necessary because of reduced immunogenicity [320]. A protein-based, bivalent vaccine directed against the prefusion F protein of both RSV subtypes (RSVPreF) was designed by Pfizer for the vaccination of pregnant women and older adults. Intramuscular vaccination of healthy adult men and nonpregnant women (18–49 years) as well as older adults (50–85 years) with RSVPreF induced robust neutralizing antibody responses against both RSV subtypes, with elevated titers through 12 months post-vaccination, while being safe and well tolerated [321,322,323]. In phase III trials of RSVPreF in older adults, vaccination efficiently prevented RSV-associated ALRI in the vaccine recipients compared to the placebo group [324]. Consistent with previous studies, RSVPreF was safe and well-tolerated [324]. In May 2023, RSVPreF, marketed as Abrysvo, was approved by the FDA for the vaccination of older adults (≥60 years) for the prevention of RSV-associated ALRI. Phase III trials for RSVPreF in older adults are currently ongoing to evaluate the efficacy and safety of a second dose of RSVPreF throughout two RSV seasons (NCT05035212). Single-dose vaccination of pregnant women with RSVPreF, administered by the beginning of their third trimester, showed the induction of neutralizing antibodies and efficient transplacental migration of these antibodies to the fetus. Maternal vaccination effectively reduced the risk of ALRI and protected infants from RSV infection during the first six months of life compared to the placebo group [325,326]. The vaccine, marketed as Abrysvo, was recently approved by the FDA for the vaccination of pregnant women during their third trimester representing the first vaccine targeting the infant population. Phase III trials for maternal vaccination are currently ongoing to further evaluate the efficacy and safety of the vaccine in pregnant women and infants (NCT04424316).
A similar subunit vaccine, RSVPreF3, is based on a prefusion F protein antigen and was developed by GSK. Intramuscular vaccination of healthy pregnant women during their late second or third trimester with unadjuvanted RSVPreF3 was safe and induced neutralizing antibodies that were transferred to the fetus [327]. Transplacentally derived antibodies in newborns were high and had waned after approximately six months [327]. In February 2022, GSK stopped recruitment for their phase III trials due to safety concerns, as premature birth was more likely in vaccinated women compared to the placebo group (NCT04605159, NCT04980391, and NCT05229068). Single-dose vaccination of older adults with RSVPreF3 (RSVPreF3 OA) adjuvanted with AS01E, administered intramuscularly, significantly reduced the risk of ALRI in vaccine recipients compared to the placebo group [328]. The generated antibodies were detectable for at least six months. Vaccination was safe and displayed a similar efficacy regardless of RSV strain, age group, or comorbidities [328]. In May 2023, RSVPreF3 OA, marketed as Arexvy, was approved by the FDA for the vaccination of older adults (≥60 years) to reduce the risk of ALRI during an RSV season. Phase III clinical trials for RSVPreF OA are currently ongoing to monitor the effects of repeated vaccination and the long-term effects of vaccination in volunteers (NCT04886596).
During the SARS-CoV-2 pandemic, mRNA vaccines against COVID-19 attracted much interest. These mRNA-based vaccines, coding for a specific viral antigen, are administered intramuscularly using lipid nanoparticles [329]. After translation, mRNAs are degraded, reducing toxicity and the risk of side effects. The successful vaccination campaign during the pandemic also showed that this vaccine system could be safe and immunogenic in different age groups [329]. Although mRNA vaccines can be produced fast and on a large scale, they tend to be relatively unstable and degrade quickly if not stored properly [329]. mRNA-1345, which was developed by Moderna, is a nucleic-acid-based vaccine encoding a membrane-anchored RSV F protein stabilized in the prefusion conformation. Single-dose vaccination of older adults with mRNA-1345 resulted in increased antibody titers compared to the unvaccinated control group, which persisted for six months and effectively prevented RSV-associated ALRI in vaccine recipients [330]. Vaccinations were well-tolerated in older adults without safety concerns. Phase III studies for mRNA-1345 are currently ongoing to further evaluate the efficacy and safety of the vaccine as primary efficacy endpoints in older adults were recently met (NCT05127434).
Prophylactic treatment with monoclonal antibodies represents an alternative to vaccination. Palivizumab, a humanized IgG1-type monoclonal antibody, was approved in 1998 to treat high-risk infants during their first RSV season [243,331]. Palivizumab targets the antigenic site II of the F protein and prevents viral infection and subsequent replication. Although antigenic site II is present on both preF and postF confirmation, antibodies directed against this epitope display a reduced neutralizing activity compared to antigenic site Ø, which is only present in the preF confirmation [332,333]. Trials in high-risk infants, to whom palivizumab was administered every 30 days for five months, showed reduced RSV-associated hospitalization and disease severity following intramuscular injection while being well tolerated compared to the placebo group [243,331]. Due to palivizumab’s relatively short half-life and the necessity of monthly administrations during RSV season associated with high costs, the American Academy of Pediatrics recommended using palivizumab for high-risk infants during their first RSV season [334], advice that was followed in several other countries [335]. Recently, a new monoclonal antibody, nirsevimab, was approved. This recombinant human IgG1-type monoclonal antibody is directed against the antigenic site Ø in the RSV F prefusion confirmation. A triple amino acid substitution in the Fc domain increased nirsevimab’s half-life, giving it a significant advantage over palivizumab [336]. A single-dose administration of nirsevimab to preterm infants before the RSV season significantly reduced the risk of ALRI and hospitalization compared to placebo-treated infants [337,338]. This effect lasted the whole season and was protective against both RSV serotypes without any safety issues [337,338]. Nirsevimab thus has two critical advantages over palivizumab: the higher neutralization activity of the antibodies and the increased half-life, which means that a single dose before the start of the RSV season is sufficient for the protection of high-risk infants. This makes nirsevimab more cost-effective. The recent approval of nirsevimab offers at least a cost-effective and efficient alternative to vaccination for the infant population. However, it is unclear if RSV can accumulate resistance mutations towards the neutralizing activity of nirsevimab, which has been the case for palivizumab [339].
The stabilization of the prefusion F protein is a critical factor for the development of vaccines and human monoclonal antibodies for the prevention of severe RSV infections in infants, older adults, and the immunocompromised. The progress in vaccine development is best illustrated by the recent approval of Arexvy and Abrysvo, which offer protection for the older population from RSV-associated ALRI. For infants, the vaccination of pregnant women seems an attractive option as Pfizer’s PreF-protein vaccine was approved recently by the FDA based on promising data from phase III clinical trials.

5. Concluding Remarks

Despite decades of research, RSV remains a substantial global burden, mainly affecting infants, older adults, and immunocompromised individuals. After the early failure of FI-RSV-based vaccines in the 1960s, research has focused on understanding the host’s immune response and possible immunomodulatory effects of RSV that may counteract a protective response and may even enhance immunopathological effects. This review emphasizes that effective and well-balanced immune responses are fundamental for reducing RSV-associated disease. As summarized, RSV-altered immune cell activity and differentiation robustly impact the cellular and humoral immune response. This dysregulation is mainly characterized by a Th2-skewed immune response leading to more severe disease and immunopathology. In the end, altered RSV-mediated responses of the innate and adaptive immune system may affect and suppress the generation of a proper immunological memory leading to impaired and short-lived immunity. Comprehending these alterations is thereby of great interest, although further studies will be necessary to address this in more detail.
Solving the prefusion conformation of the RSV F protein and its stabilization demonstrated the immense progress made in vaccine development after decades of basic research. As a consequence of this breakthrough, two preF-based vaccines have recently received licensure for use in older adults as both safety and efficacy were demonstrated. Although the recently developed RSV vaccines seem highly efficacious in older adults, their transmission-reducing potential and durability of protection in this age group require further investigation. The risk of waning antibody levels, also seen after RSV infection, may call for annual vaccination to protect this vulnerable age group.
A preF-protein-based vaccine for the vaccination of pregnant women was licensed recently by the FDA. This way, infants can be protected by vaccine-induced maternally derived antibodies during their first RSV season. Furthermore, several vaccine candidates are currently in phase III clinical trials showing promising results for the vaccination of older adults and pregnant women. Maternal vaccination seems to be a promising approach for the infant population, at least during their first RSV season, by the transplacental transfer of neutralizing antibodies, as direct vaccination of newborns may result in a suboptimal immune response considering their immature immune system. Maternal vaccination, however, may bear certain risks, taking into account the current developments in relation to the RSVPreF3 vaccine and the higher abundance of premature births after vaccination with one candidate vaccine.
Although a significant amount of data has been collected recently, our understanding of the immunological mechanisms during a natural RSV infection is still incomplete. Considering the phenomena of immunosenescence and preexisting immunity of older adults on one side and the immaturity of the infant’s immune system on the other, analyzing the differences between the immune responses of the two main age groups at risk is critical for vaccine development. Further studies in infants and older adults will be necessary to fully comprehend the protective and harmful immune responses upon RSV infection and vaccination and to pave the way toward a better understanding of cell- and antibody-mediated immunity to RSV infection.

Author Contributions

A.A., M.L., R.M. and G.F.R. conceptualized and composed the manuscript; S.M.K., A.D.M.E.O. and M.L. reviewed the final manuscript; R.M. and G.F.R. oversaw all aspects of the manuscript preparation. All authors have read and agreed to the published version of the manuscript.

Funding

This work was financially supported by the Innovative Medicines Initiative 2 of the European Union’s Horizon 2020 research and innovation program (EU IMI101007799, Inno4Vac) and the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation)—398066876/GRK 2485/2, Research Training Group VIPER. Open Access publication was funded by the DFG—491094227 “Open Access Publication Funding” and the University of Veterinary Medicine Hannover Foundation.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Collins, P.L.; Fearns, R.; Graham, B.S. Respiratory syncytial virus: Virology, reverse genetics, and pathogenesis of disease. Curr. Top. Microbiol. Immunol. 2013, 372, 3–38. [Google Scholar] [CrossRef] [PubMed]
  2. Mufson, M.A.; Orvell, C.; Rafnar, B.; Norrby, E. Two distinct subtypes of human respiratory syncytial virus. J. Gen. Virol. 1985, 66, 2111–2124. [Google Scholar] [CrossRef] [PubMed]
  3. Yu, J.M.; Fu, Y.H.; Peng, X.L.; Zheng, Y.P.; He, J.S. Genetic diversity and molecular evolution of human respiratory syncytial virus A and B. Sci. Rep. 2021, 11, 12941. [Google Scholar] [CrossRef] [PubMed]
  4. Cantu-Flores, K.; Rivera-Alfaro, G.; Munoz-Escalante, J.C.; Noyola, D.E. Global distribution of respiratory syncytial virus A and B infections: A systematic review. Pathog. Glob. Health 2022, 116, 398–409. [Google Scholar] [CrossRef] [PubMed]
  5. Hall, C.B.; Weinberg, G.A.; Iwane, M.K.; Blumkin, A.K.; Edwards, K.M.; Staat, M.A.; Auinger, P.; Griffin, M.R.; Poehling, K.A.; Erdman, D.; et al. The burden of respiratory syncytial virus infection in young children. N. Engl. J. Med. 2009, 360, 588–598. [Google Scholar] [CrossRef]
  6. Falsey, A.R.; Hennessey, P.A.; Formica, M.A.; Cox, C.; Walsh, E.E. Respiratory Syncytial Virus Infection in Elderly and High-Risk Adults. N. Engl. J. Med. 2005, 352, 1749–1759. [Google Scholar] [CrossRef]
  7. Falsey, A.R.; Walsh, E.E. Respiratory syncytial virus infection in elderly adults. Drugs Aging 2005, 22, 577–587. [Google Scholar] [CrossRef]
  8. Nair, H.; Nokes, D.J.; Gessner, B.D.; Dherani, M.; Madhi, S.A.; Singleton, R.J.; O’Brien, K.L.; Roca, A.; Wright, P.F.; Bruce, N.; et al. Global burden of acute lower respiratory infections due to respiratory syncytial virus in young children: A systematic review and meta-analysis. Lancet 2010, 375, 1545–1555. [Google Scholar] [CrossRef]
  9. Li, Y.; Wang, X.; Blau, D.M.; Caballero, M.T.; Feikin, D.R.; Gill, C.J.; Madhi, S.A.; Omer, S.B.; Simoes, E.A.F.; Campbell, H.; et al. Global, regional, and national disease burden estimates of acute lower respiratory infections due to respiratory syncytial virus in children younger than 5 years in 2019: A systematic analysis. Lancet 2022, 399, 2047–2064. [Google Scholar] [CrossRef]
  10. Savic, M.; Penders, Y.; Shi, T.; Branche, A.; Pircon, J.Y. Respiratory syncytial virus disease burden in adults aged 60 years and older in high-income countries: A systematic literature review and meta-analysis. Influenza Other Respir. Viruses 2023, 17, e13031. [Google Scholar] [CrossRef]
  11. Chow, E.J.; Uyeki, T.M.; Chu, H.Y. The effects of the COVID-19 pandemic on community respiratory virus activity. Nat. Rev. Microbiol. 2023, 21, 195–210. [Google Scholar] [CrossRef] [PubMed]
  12. Chuang, Y.C.; Lin, K.P.; Wang, L.A.; Yeh, T.K.; Liu, P.Y. The Impact of the COVID-19 Pandemic on Respiratory Syncytial Virus Infection: A Narrative Review. Infect. Drug Resist. 2023, 16, 661–675. [Google Scholar] [CrossRef] [PubMed]
  13. Di Mattia, G.; Nenna, R.; Mancino, E.; Rizzo, V.; Pierangeli, A.; Villani, A.; Midulla, F. During the COVID-19 pandemic where has respiratory syncytial virus gone? Pediatr. Pulmonol. 2021, 56, 3106–3109. [Google Scholar] [CrossRef]
  14. Smyth, R.L.; Openshaw, P.J. Bronchiolitis. Lancet 2006, 368, 312–322. [Google Scholar] [CrossRef]
  15. Henderson, F.W.; Collier, A.M.; Clyde, W.A., Jr.; Denny, F.W. Respiratory-syncytial-virus infections, reinfections and immunity. A prospective, longitudinal study in young children. N. Engl. J. Med. 1979, 300, 530–534. [Google Scholar] [CrossRef] [PubMed]
  16. Glezen, W.P.; Taber, L.H.; Frank, A.L.; Kasel, J.A. Risk of primary infection and reinfection with respiratory syncytial virus. Am. J. Dis. Child. 1986, 140, 543–546. [Google Scholar] [CrossRef]
  17. Collins, P.L.; Graham, B.S. Viral and host factors in human respiratory syncytial virus pathogenesis. J. Virol. 2008, 82, 2040–2055. [Google Scholar] [CrossRef]
  18. Pickles, R.J.; DeVincenzo, J.P. Respiratory syncytial virus (RSV) and its propensity for causing bronchiolitis. J. Pathol. 2015, 235, 266–276. [Google Scholar] [CrossRef]
  19. McNamara, P.S.; Smyth, R.L. The pathogenesis of respiratory syncytial virus disease in childhood. Br. Med. Bull. 2002, 61, 13–28. [Google Scholar] [CrossRef]
  20. Hall, C.B. Respiratory syncytial virus and parainfluenza virus. N. Engl. J. Med. 2001, 344, 1917–1928. [Google Scholar] [CrossRef]
  21. Walsh, E.E.; Falsey, A.R. Respiratory syncytial virus infection in adult populations. Infect. Disord. Drug Targets 2012, 12, 98–102. [Google Scholar] [CrossRef] [PubMed]
  22. Shi, T.; Ooi, Y.; Zaw, E.M.; Utjesanovic, N.; Campbell, H.; Cunningham, S.; Bont, L.; Nair, H.; Investigators, R. Association Between Respiratory Syncytial Virus-Associated Acute Lower Respiratory Infection in Early Life and Recurrent Wheeze and Asthma in Later Childhood. J. Infect. Dis. 2020, 222, S628–S633. [Google Scholar] [CrossRef]
  23. Coutts, J.; Fullarton, J.; Morris, C.; Grubb, E.; Buchan, S.; Rodgers-Gray, B.; Thwaites, R. Association between respiratory syncytial virus hospitalization in infancy and childhood asthma. Pediatr. Pulmonol. 2020, 55, 1104–1110. [Google Scholar] [CrossRef] [PubMed]
  24. van Wijhe, M.; Johannesen, C.K.; Simonsen, L.; Jorgensen, I.M.; Investigators, R.; Fischer, T.K. A Retrospective Cohort Study on Infant Respiratory Tract Infection Hospitalizations and Recurrent Wheeze and Asthma Risk: Impact of Respiratory Syncytial Virus. J. Infect. Dis. 2022, 226, S55–S62. [Google Scholar] [CrossRef] [PubMed]
  25. McGinley, J.P.; Lin, G.L.; Oner, D.; Golubchik, T.; O’Connor, D.; Snape, M.D.; Gruselle, O.; Langedijk, A.C.; Wildenbeest, J.; Openshaw, P.; et al. Clinical and Viral Factors Associated With Disease Severity and Subsequent Wheezing in Infants With Respiratory Syncytial Virus Infection. J. Infect. Dis. 2022, 226, S45–S54. [Google Scholar] [CrossRef]
  26. Rosas-Salazar, C.; Chirkova, T.; Gebretsadik, T.; Chappell, J.D.; Peebles, R.S., Jr.; Dupont, W.D.; Jadhao, S.J.; Gergen, P.J.; Anderson, L.J.; Hartert, T.V. Respiratory syncytial virus infection during infancy and asthma during childhood in the USA (INSPIRE): A population-based, prospective birth cohort study. Lancet 2023, 401, 1669–1680. [Google Scholar] [CrossRef]
  27. Glezen, W.P.; Greenberg, S.B.; Atmar, R.L.; Piedra, P.A.; Couch, R.B. Impact of respiratory virus infections on persons with chronic underlying conditions. JAMA 2000, 283, 499–505. [Google Scholar] [CrossRef]
  28. Fixler, D.E. Respiratory syncytial virus infection in children with congenital heart disease: A review. Pediatr. Cardiol. 1996, 17, 163–168. [Google Scholar] [CrossRef] [PubMed]
  29. Kim, H.W.; Canchola, J.G.; Brandt, C.D.; Pyles, G.; Chanock, R.M.; Jensen, K.; Parrott, R.H. Respiratory syncytial virus disease in infants despite prior administration of antigenic inactivated vaccine. Am. J. Epidemiol. 1969, 89, 422–434. [Google Scholar] [CrossRef] [PubMed]
  30. Palacios-Pedrero, M.A.; Osterhaus, A.; Becker, T.; Elbahesh, H.; Rimmelzwaan, G.F.; Saletti, G. Aging and Options to Halt Declining Immunity to Virus Infections. Front. Immunol. 2021, 12, 681449. [Google Scholar] [CrossRef]
  31. Andrade, C.A.; Pacheco, G.A.; Galvez, N.M.S.; Soto, J.A.; Bueno, S.M.; Kalergis, A.M. Innate Immune Components that Regulate the Pathogenesis and Resolution of hRSV and hMPV Infections. Viruses 2020, 12, 637. [Google Scholar] [CrossRef] [PubMed]
  32. Ballegeer, M.; Saelens, X. Cell-Mediated Responses to Human Metapneumovirus Infection. Viruses 2020, 12, 542. [Google Scholar] [CrossRef] [PubMed]
  33. Farrag, M.A.; Almajhdi, F.N. Human Respiratory Syncytial Virus: Role of Innate Immunity in Clearance and Disease Progression. Viral Immunol. 2016, 29, 11–26. [Google Scholar] [CrossRef] [PubMed]
  34. Sun, Y.; Lopez, C.B. The innate immune response to RSV: Advances in our understanding of critical viral and host factors. Vaccine 2017, 35, 481–488. [Google Scholar] [CrossRef]
  35. Turvey, S.E.; Broide, D.H. Innate immunity. J. Allergy Clin. Immunol. 2010, 125, S24–S32. [Google Scholar] [CrossRef]
  36. Mogensen, T.H. Pathogen recognition and inflammatory signaling in innate immune defenses. Clin. Microbiol. Rev. 2009, 22, 240–273. [Google Scholar] [CrossRef]
  37. Openshaw, P.J.M.; Chiu, C.; Culley, F.J.; Johansson, C. Protective and Harmful Immunity to RSV Infection. Annu. Rev. Immunol. 2017, 35, 501–532. [Google Scholar] [CrossRef]
  38. Vareille, M.; Kieninger, E.; Edwards, M.R.; Regamey, N. The airway epithelium: Soldier in the fight against respiratory viruses. Clin. Microbiol. Rev. 2011, 24, 210–229. [Google Scholar] [CrossRef]
  39. Kuek, L.E.; Lee, R.J. First contact: The role of respiratory cilia in host-pathogen interactions in the airways. Am. J. Physiol. Lung Cell Mol. Physiol. 2020, 319, L603–L619. [Google Scholar] [CrossRef]
  40. Wallace, L.E.; Liu, M.; van Kuppeveld, F.J.M.; de Vries, E.; de Haan, C.A.M. Respiratory mucus as a virus-host range determinant. Trends Microbiol. 2021, 29, 983–992. [Google Scholar] [CrossRef]
  41. Ridley, C.; Thornton, D.J. Mucins: The frontline defence of the lung. Biochem. Soc. Trans. 2018, 46, 1099–1106. [Google Scholar] [CrossRef] [PubMed]
  42. Chatterjee, M.; van Putten, J.P.M.; Strijbis, K. Defensive Properties of Mucin Glycoproteins during Respiratory Infections-Relevance for SARS-CoV-2. mBio 2020, 11, e02374-20. [Google Scholar] [CrossRef] [PubMed]
  43. LeMessurier, K.S.; Tiwary, M.; Morin, N.P.; Samarasinghe, A.E. Respiratory Barrier as a Safeguard and Regulator of Defense Against Influenza A Virus and Streptococcus pneumoniae. Front. Immunol. 2020, 11, 3. [Google Scholar] [CrossRef] [PubMed]
  44. Sanders, C.J.; Doherty, P.C.; Thomas, P.G. Respiratory epithelial cells in innate immunity to influenza virus infection. Cell Tissue Res. 2011, 343, 13–21. [Google Scholar] [CrossRef]
  45. Zanin, M.; Baviskar, P.; Webster, R.; Webby, R. The Interaction between Respiratory Pathogens and Mucus. Cell Host Microbe 2016, 19, 159–168. [Google Scholar] [CrossRef]
  46. Bailey, K.L. Aging Diminishes Mucociliary Clearance of the Lung. Adv. Geriatr. Med. Res. 2022, 4, e220005. [Google Scholar] [CrossRef]
  47. Ioannidis, I.; McNally, B.; Willette, M.; Peeples, M.E.; Chaussabel, D.; Durbin, J.E.; Ramilo, O.; Mejias, A.; Flano, E. Plasticity and virus specificity of the airway epithelial cell immune response during respiratory virus infection. J. Virol. 2012, 86, 5422–5436. [Google Scholar] [CrossRef]
  48. Zhang, L.; Peeples, M.E.; Boucher, R.C.; Collins, P.L.; Pickles, R.J. Respiratory syncytial virus infection of human airway epithelial cells is polarized, specific to ciliated cells, and without obvious cytopathology. J. Virol. 2002, 76, 5654–5666. [Google Scholar] [CrossRef]
  49. Barnes, M.V.C.; Openshaw, P.J.M.; Thwaites, R.S. Mucosal Immune Responses to Respiratory Syncytial Virus. Cells 2022, 11, 1153. [Google Scholar] [CrossRef]
  50. Feng, Z.; Xu, L.; Xie, Z. Receptors for Respiratory Syncytial Virus Infection and Host Factors Regulating the Life Cycle of Respiratory Syncytial Virus. Front. Cell Infect. Microbiol. 2022, 12, 858629. [Google Scholar] [CrossRef]
  51. Tayyari, F.; Marchant, D.; Moraes, T.J.; Duan, W.; Mastrangelo, P.; Hegele, R.G. Identification of nucleolin as a cellular receptor for human respiratory syncytial virus. Nat. Med. 2011, 17, 1132–1135. [Google Scholar] [CrossRef] [PubMed]
  52. Mastrangelo, P.; Chin, A.A.; Tan, S.; Jeon, A.H.; Ackerley, C.A.; Siu, K.K.; Lee, J.E.; Hegele, R.G. Identification of RSV Fusion Protein Interaction Domains on the Virus Receptor, Nucleolin. Viruses 2021, 13, 261. [Google Scholar] [CrossRef] [PubMed]
  53. Green, G.; Johnson, S.M.; Costello, H.; Brakel, K.; Harder, O.; Oomens, A.G.; Peeples, M.E.; Moulton, H.M.; Niewiesk, S. CX3CR1 Is a Receptor for Human Respiratory Syncytial Virus in Cotton Rats. J. Virol. 2021, 95, e0001021. [Google Scholar] [CrossRef] [PubMed]
  54. Johnson, S.M.; McNally, B.A.; Ioannidis, I.; Flano, E.; Teng, M.N.; Oomens, A.G.; Walsh, E.E.; Peeples, M.E. Respiratory Syncytial Virus Uses CX3CR1 as a Receptor on Primary Human Airway Epithelial Cultures. PLoS Pathog. 2015, 11, e1005318. [Google Scholar] [CrossRef]
  55. Chirkova, T.; Lin, S.; Oomens, A.G.P.; Gaston, K.A.; Boyoglu-Barnum, S.; Meng, J.; Stobart, C.C.; Cotton, C.U.; Hartert, T.V.; Moore, M.L.; et al. CX3CR1 is an important surface molecule for respiratory syncytial virus infection in human airway epithelial cells. J. Gen. Virol. 2015, 96, 2543–2556. [Google Scholar] [CrossRef]
  56. Anderson, C.S.; Chu, C.Y.; Wang, Q.; Mereness, J.A.; Ren, Y.; Donlon, K.; Bhattacharya, S.; Misra, R.S.; Walsh, E.E.; Pryhuber, G.S.; et al. CX3CR1 as a respiratory syncytial virus receptor in pediatric human lung. Pediatr. Res. 2020, 87, 862–867. [Google Scholar] [CrossRef]
  57. Krusat, T.; Streckert, H.J. Heparin-dependent attachment of respiratory syncytial virus (RSV) to host cells. Arch. Virol. 1997, 142, 1247–1254. [Google Scholar] [CrossRef]
  58. Bourgeois, C.; Bour, J.B.; Lidholt, K.; Gauthray, C.; Pothier, P. Heparin-like structures on respiratory syncytial virus are involved in its infectivity in vitro. J. Virol. 1998, 72, 7221–7227. [Google Scholar] [CrossRef]
  59. Feldman, S.A.; Audet, S.; Beeler, J.A. The Fusion Glycoprotein of Human Respiratory Syncytial Virus Facilitates Virus Attachment and Infectivity via an Interaction with Cellular Heparan Sulfate. J. Virol. 2000, 74, 6442–6447. [Google Scholar] [CrossRef]
  60. Behera, A.K.; Matsuse, H.; Kumar, M.; Kong, X.; Lockey, R.F.; Mohapatra, S.S. Blocking intercellular adhesion molecule-1 on human epithelial cells decreases respiratory syncytial virus infection. Biochem. Biophys. Res. Commun. 2001, 280, 188–195. [Google Scholar] [CrossRef]
  61. Imai, T.; Hieshima, K.; Haskell, C.; Baba, M.; Nagira, M.; Nishimura, M.; Kakizaki, M.; Takagi, S.; Nomiyama, H.; Schall, T.J.; et al. Identification and molecular characterization of fractalkine receptor CX3CR1, which mediates both leukocyte migration and adhesion. Cell 1997, 91, 521–530. [Google Scholar] [CrossRef] [PubMed]
  62. Kim, K.W.; Vallon-Eberhard, A.; Zigmond, E.; Farache, J.; Shezen, E.; Shakhar, G.; Ludwig, A.; Lira, S.A.; Jung, S. In vivo structure/function and expression analysis of the CX3C chemokine fractalkine. Blood 2011, 118, e156–e167. [Google Scholar] [CrossRef] [PubMed]
  63. Jones, B.A.; Beamer, M.; Ahmed, S. Fractalkine/CX3CL1: A Potential New Target for Inflammatory Diseases. Mol. Interv. 2010, 10, 263–270. [Google Scholar] [CrossRef] [PubMed]
  64. Bazan, J.F.; Bacon, K.B.; Hardiman, G.; Wang, W.; Soo, K.; Rossi, D.; Greaves, D.R.; Zlotnik, A.; Schall, T.J. A new class of membrane-bound chemokine with a CX3C motif. Nature 1997, 385, 640–644. [Google Scholar] [CrossRef]
  65. Tripp, R.A.; Jones, L.P.; Haynes, L.M.; Zheng, H.; Murphy, P.M.; Anderson, L.J. CX3C chemokine mimicry by respiratory syncytial virus G glycoprotein. Nat. Immunol. 2001, 2, 732–738. [Google Scholar] [CrossRef]
  66. Ha, B.; Chirkova, T.; Boukhvalova, M.S.; Sun, H.Y.; Walsh, E.E.; Anderson, C.S.; Mariani, T.J.; Anderson, L.J. Mutation of Respiratory Syncytial Virus G Protein’s CX3C Motif Attenuates Infection in Cotton Rats and Primary Human Airway Epithelial Cells. Vaccines 2019, 7, 69. [Google Scholar] [CrossRef]
  67. Malik, G.; Zhou, Y. Innate Immune Sensing of Influenza A Virus. Viruses 2020, 12, 755. [Google Scholar] [CrossRef]
  68. Islamuddin, M.; Mustfa, S.A.; Ullah, S.; Omer, U.; Kato, K.; Parveen, S. Innate Immune Response and Inflammasome Activation During SARS-CoV-2 Infection. Inflammation 2022, 45, 1849–1863. [Google Scholar] [CrossRef]
  69. O’Connor, C.M.; Sen, G.C. Innate Immune Responses to Herpesvirus Infection. Cells 2021, 10, 2122. [Google Scholar] [CrossRef]
  70. Loo, Y.M.; Fornek, J.; Crochet, N.; Bajwa, G.; Perwitasari, O.; Martinez-Sobrido, L.; Akira, S.; Gill, M.A.; Garcia-Sastre, A.; Katze, M.G.; et al. Distinct RIG-I and MDA5 signaling by RNA viruses in innate immunity. J. Virol. 2008, 82, 335–345. [Google Scholar] [CrossRef]
  71. Meineke, R.; Rimmelzwaan, G.F.; Elbahesh, H. Influenza Virus Infections and Cellular Kinases. Viruses 2019, 11, 171. [Google Scholar] [CrossRef]
  72. Carty, M.; Guy, C.; Bowie, A.G. Detection of Viral Infections by Innate Immunity. Biochem. Pharmacol. 2021, 183, 114316. [Google Scholar] [CrossRef]
  73. Liu, P.; Jamaluddin, M.; Li, K.; Garofalo, R.P.; Casola, A.; Brasier, A.R. Retinoic acid-inducible gene I mediates early antiviral response and Toll-like receptor 3 expression in respiratory syncytial virus-infected airway epithelial cells. J. Virol. 2007, 81, 1401–1411. [Google Scholar] [CrossRef] [PubMed]
  74. Rudd, B.D.; Burstein, E.; Duckett, C.S.; Li, X.; Lukacs, N.W. Differential role for TLR3 in respiratory syncytial virus-induced chemokine expression. J. Virol. 2005, 79, 3350–3357. [Google Scholar] [CrossRef] [PubMed]
  75. Bueno, S.M.; Gonzalez, P.A.; Riedel, C.A.; Carreno, L.J.; Vasquez, A.E.; Kalergis, A.M. Local cytokine response upon respiratory syncytial virus infection. Immunol. Lett. 2011, 136, 122–129. [Google Scholar] [CrossRef] [PubMed]
  76. Noah, T.L.; Becker, S. Respiratory syncytial virus-induced cytokine production by a human bronchial epithelial cell line. Am. J. Physiol. 1993, 265, 472–478. [Google Scholar] [CrossRef]
  77. Noah, T.L.; Henderson, F.W.; Wortman, I.A.; Devlin, R.B.; Handy, J.; Koren, H.S.; Becker, S. Nasal cytokine production in viral acute upper respiratory infection of childhood. J. Infect. Dis. 1995, 171, 584–592. [Google Scholar] [CrossRef]
  78. Saito, T.; Deskin, R.W.; Casola, A.; Häeberle, H.; Olszewska, B.; Ernst, P.B.; Alam, R.; Ogra, P.L.; Garofalo, R. Respiratory Syncytial Virus Induces Selective Production of the Chemokine RANTES by Upper Airway Epithelial Cells. J. Infect. Dis. 1996, 175, 497–504. [Google Scholar] [CrossRef]
  79. Olszewska-Pazdrak, B.; Casola, A.; Saito, T.; Alam, R.; Crowe, S.E.; Mei, F.; Ogra, P.L.; Garofalo, R.P. Cell-Specific Expression of RANTES, MCP-1, and MIP-1a by Lower Airway Epithelial Cells and Eosinophils Infected with Respiratory Syncytial Virus. J. Virol. 1998, 72, 4756–4764. [Google Scholar] [CrossRef]
  80. Wang, S.-Z.; Hallsworth, P.G.; Dowling, K.D.; Alpers, J.H.; Bowden, J.J.; Forsyth, K.D. Adhesion molecule expression on epithelial cells infected with respiratory syncytial virus. Eur. Respir. J. 2000, 15, 358–366. [Google Scholar] [CrossRef]
  81. Guo, X.; Liu, T.; Shi, H.; Wang, J.; Ji, P.; Wang, H.; Hou, Y.; Tan, R.X.; Li, E. Respiratory Syncytial Virus Infection Upregulates NLRC5 and Major Histocompatibility Complex Class I Expression through RIG-I Induction in Airway Epithelial Cells. J. Virol. 2015, 89, 7636–7645. [Google Scholar] [CrossRef]
  82. Levitz, R.; Wattier, R.; Phillips, P.; Solomon, A.; Lawler, J.; Lazar, I.; Weibel, C.; Kahn, J.S. Induction of IL-6 and CCL5 (RANTES) in human respiratory epithelial (A549) cells by clinical isolates of respiratory syncytial virus is strain specific. Virol. J. 2012, 9, 190. [Google Scholar] [CrossRef]
  83. Touzelet, O.; Broadbent, L.; Armstrong, S.D.; Aljabr, W.; Cloutman-Green, E.; Power, U.F.; Hiscox, J.A. The Secretome Profiling of a Pediatric Airway Epithelium Infected with hRSV Identified Aberrant Apical/Basolateral Trafficking and Novel Immune Modulating (CXCL6, CXCL16, CSF3) and Antiviral (CEACAM1) Proteins. Mol. Cell Proteom. 2020, 19, 793–807. [Google Scholar] [CrossRef]
  84. Van Royen, T.; Rossey, I.; Sedeyn, K.; Schepens, B.; Saelens, X. How RSV Proteins Join Forces to Overcome the Host Innate Immune Response. Viruses 2022, 14, 419. [Google Scholar] [CrossRef]
  85. Moore, E.C.; Barber, J.; Tripp, R.A. 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). Virol. J. 2008, 5, 116. [Google Scholar] [CrossRef]
  86. Xu, X.; Zheng, J.; Zheng, K.; Hou, Y.; Zhao, F.; Zhao, D. Respiratory syncytial virus NS1 protein degrades STAT2 by inducing SOCS1 expression. Intervirology 2014, 57, 65–73. [Google Scholar] [CrossRef]
  87. Pei, J.; Beri, N.R.; Zou, A.J.; Hubel, P.; Dorando, H.K.; Bergant, V.; Andrews, R.D.; Pan, J.; Andrews, J.M.; Sheehan, K.C.F.; et al. Nuclear-localized human respiratory syncytial virus NS1 protein modulates host gene transcription. Cell Rep. 2021, 37, 109803. [Google Scholar] [CrossRef]
  88. Boyapalle, S.; Wong, T.; Garay, J.; Teng, M.; San Juan-Vergara, H.; Mohapatra, S.; Mohapatra, S. Respiratory syncytial virus NS1 protein colocalizes with mitochondrial antiviral signaling protein MAVS following infection. PLoS ONE 2012, 7, e29386. [Google Scholar] [CrossRef]
  89. Ling, Z.; Tran, K.C.; Teng, M.N. Human respiratory syncytial virus nonstructural protein NS2 antagonizes the activation of beta interferon transcription by interacting with RIG-I. J. Virol. 2009, 83, 3734–3742. [Google Scholar] [CrossRef]
  90. Spann, K.M.; Tran, K.C.; Collins, P.L. Effects of nonstructural proteins NS1 and NS2 of human respiratory syncytial virus on interferon regulatory factor 3, NF-kappaB, and proinflammatory cytokines. J. Virol. 2005, 79, 5353–5362. [Google Scholar] [CrossRef]
  91. Ren, J.; Liu, T.; Pang, L.; Li, K.; Garofalo, R.P.; Casola, A.; Bao, X. A novel mechanism for the inhibition of interferon regulatory factor-3-dependent gene expression by human respiratory syncytial virus NS1 protein. J. Gen. Virol. 2011, 92, 2153–2159. [Google Scholar] [CrossRef]
  92. Bitko, V.; Shulyayeva, O.; Mazumder, B.; Musiyenko, A.; Ramaswamy, M.; Look, D.C.; Barik, S. Nonstructural proteins of respiratory syncytial virus suppress premature apoptosis by an NF-kappaB-dependent, interferon-independent mechanism and facilitate virus growth. J. Virol. 2007, 81, 1786–1795. [Google Scholar] [CrossRef]
  93. Takeuchi, R.; Tsutsumi, H.; Osaki, M.; Haseyama, K.; Mizue, N.; Chiba, C. Respiratory syncytial virus infection of human alveolar epithelial cells enhances interferon regulatory factor 1 and interleukin-1beta-converting enzyme gene expression but does not cause apoptosis. J. Virol. 1998, 72, 4498–4502. [Google Scholar] [CrossRef]
  94. Simpson, J.; Loh, Z.; Ullah, M.A.; Lynch, J.P.; Werder, R.B.; Collinson, N.; Zhang, V.; Dondelinger, Y.; Bertrand, M.J.M.; Everard, M.L.; et al. Respiratory Syncytial Virus Infection Promotes Necroptosis and HMGB1 Release by Airway Epithelial Cells. Am. J. Respir. Crit. Care Med. 2020, 201, 1358–1371. [Google Scholar] [CrossRef]
  95. Lifland, A.W.; Jung, J.; Alonas, E.; Zurla, C.; Crowe, J.E., Jr.; Santangelo, P.J. Human respiratory syncytial virus nucleoprotein and inclusion bodies antagonize the innate immune response mediated by MDA5 and MAVS. J. Virol. 2012, 86, 8245–8258. [Google Scholar] [CrossRef]
  96. Rincheval, V.; Lelek, M.; Gault, E.; Bouillier, C.; Sitterlin, D.; Blouquit-Laye, S.; Galloux, M.; Zimmer, C.; Eleouet, J.F.; Rameix-Welti, M.A. Functional organization of cytoplasmic inclusion bodies in cells infected by respiratory syncytial virus. Nat. Commun. 2017, 8, 563. [Google Scholar] [CrossRef]
  97. Groskreutz, D.J.; Babor, E.C.; Monick, M.M.; Varga, S.M.; Hunninghake, G.W. Respiratory syncytial virus limits alpha subunit of eukaryotic translation initiation factor 2 (eIF2alpha) phosphorylation to maintain translation and viral replication. J. Biol. Chem. 2010, 285, 24023–24031. [Google Scholar] [CrossRef]
  98. Meineke, R.; Stelz, S.; Busch, M.; Werlein, C.; Kühnel, M.; Jonigk, D.; Rimmelzwaan, G.F.; Elbahesh, H. FDA-approved Abl/EGFR/PDGFR kinase inhibitors show potent efficacy against pandemic and seasonal influenza A virus infections of human lung explants. iScience 2023, 26, 106309. [Google Scholar] [CrossRef]
  99. Harcourt, J.L.; Haynes, L.M. Establishing a liquid-covered culture of polarized human airway epithelial Calu-3 cells to study host cell response to respiratory pathogens in vitro. J. Vis. Exp. 2013, 72, e50157. [Google Scholar] [CrossRef]
  100. Ito, K.; Daly, L.; Coates, M. An impact of age on respiratory syncytial virus infection in air-liquid-interface culture bronchial epithelium. Front Med. 2023, 10, 1144050. [Google Scholar] [CrossRef]
  101. Rajan, A.; Weaver, A.M.; Aloisio, G.M.; Jelinski, J.; Johnson, H.L.; Venable, S.F.; McBride, T.; Aideyan, L.; Piedra, F.A.; Ye, X.; et al. The human nose organoid respiratory virus model: An ex-vivo human challenge model to study RSV and SARS-CoV-2 pathogenesis and evaluate therapeutics. bioRxiv 2021. [Google Scholar] [CrossRef]
  102. Rothenberg, M.E.; Hogan, S.P. The eosinophil. Annu. Rev. Immunol. 2006, 24, 147–174. [Google Scholar] [CrossRef] [PubMed]
  103. Flores-Torres, A.S.; Salinas-Carmona, M.C.; Salinas, E.; Rosas-Taraco, A.G. Eosinophils and Respiratory Viruses. Viral Immunol. 2019, 32, 198–207. [Google Scholar] [CrossRef] [PubMed]
  104. Samarasinghe, A.E.; Melo, R.C.; Duan, S.; LeMessurier, K.S.; Liedmann, S.; Surman, S.L.; Lee, J.J.; Hurwitz, J.L.; Thomas, P.G.; McCullers, J.A. Eosinophils Promote Antiviral Immunity in Mice Infected with Influenza A Virus. J. Immunol. 2017, 198, 3214–3226. [Google Scholar] [CrossRef]
  105. Drake, M.G.; Bivins-Smith, E.R.; Proskocil, B.J.; Nie, Z.; Scott, G.D.; Lee, J.J.; Lee, N.A.; Fryer, A.D.; Jacoby, D.B. Human and Mouse Eosinophils Have Antiviral Activity against Parainfluenza Virus. Am. J. Respir. Cell Mol. Biol. 2016, 55, 387–394. [Google Scholar] [CrossRef]
  106. Handzel, Z.T.; Busse, W.W.; Sedgwick, J.B.; Vrtis, R.; Lee, W.M.; Kelly, E.A.B.; Gern, J.E. Eosinophils Bind Rhinovirus and Activate Virus-Specific T Cells. J. Immunol. 1998, 160, 1279–1284. [Google Scholar] [CrossRef]
  107. Ho, K.S.; Howell, D.; Rogers, L.; Narasimhan, B.; Verma, H.; Steiger, D. The relationship between asthma, eosinophilia, and outcomes in coronavirus disease 2019 infection. Ann. Allergy Asthma Immunol. 2021, 127, 42–48. [Google Scholar] [CrossRef]
  108. Becker, S.; Soukup, J.M. Airway epithelial cell-induced activation of monocytes and eosinophils in respiratory syncytial viral infection. Immunobiology 1999, 201, 88–106. [Google Scholar] [CrossRef]
  109. Soukup, J.M.; Becker, S. Role of monocytes and eosinophils in human respiratory syncytial virus infection in vitro. Clin. Immunol. 2003, 107, 178–185. [Google Scholar] [CrossRef]
  110. Harrison, A.M.; Bonville, C.A.; Rosenberg, H.F.; Domachowske, J.B. Respiratory syncytical virus-induced chemokine expression in the lower airways: Eosinophil recruitment and degranulation. Am. J. Respir. Crit. Care Med. 1999, 159, 1918–1924. [Google Scholar] [CrossRef]
  111. Phipps, S.; Lam, C.E.; Mahalingam, S.; Newhouse, M.; Ramirez, R.; Rosenberg, H.F.; Foster, P.S.; Matthaei, K.I. Eosinophils contribute to innate antiviral immunity and promote clearance of respiratory syncytial virus. Blood 2007, 110, 1578–1586. [Google Scholar] [CrossRef] [PubMed]
  112. Lindemans, C.A.; Kimpen, J.L.; Luijk, B.; Heidema, J.; Kanters, D.; van der Ent, C.K.; Koenderman, L. Systemic eosinophil response induced by respiratory syncytial virus. Clin. Exp. Immunol. 2006, 144, 409–417. [Google Scholar] [CrossRef] [PubMed]
  113. Stark, J.M.; Godding, V.; Sedgwick, J.B.; Busse, W.W. Respiratory syncytial virus infection enhances neutrophil and eosinophil adhesion to cultured respiratory epithelial cells. Roles of CD18 and intercellular adhesion molecule-1. J. Immunol. 1996, 156, 4774–4782. [Google Scholar] [CrossRef] [PubMed]
  114. Olszewska-Pazdrak, B.; Pazdrak, K.; Ogra, P.L.; Garofalo, R.P. Respiratory Syncytial Virus-Infected Pulmonary Epithelial Cells Induce Eosinophil Degranulation by a CD18-Mediated Mechanism. J. Immunol. 1998, 160, 4889–4895. [Google Scholar] [CrossRef]
  115. Castilow, E.M.; Legge, K.L.; Varga, S.M. Cutting edge: Eosinophils do not contribute to respiratory syncytial virus vaccine-enhanced disease. J. Immunol. 2008, 181, 6692–6696. [Google Scholar] [CrossRef]
  116. Prince, G.A.; Jenson, A.B.; Hemming, V.; Murphy, B.; Walsh, E.; Horswood, R.; Chanock, R. Enhancement of respiratory syncytial virus pulmonary pathology in cotton rats by prior intramuscular inoculation of formalin-inactiva ted virus. J. Virol. 1986, 57, 721–728. [Google Scholar] [CrossRef]
  117. Durant, L.R.; Makris, S.; Voorburg, C.M.; Loebbermann, J.; Johansson, C.; Openshaw, P.J. Regulatory T cells prevent Th2 immune responses and pulmonary eosinophilia during respiratory syncytial virus infection in mice. J. Virol. 2013, 87, 10946–10954. [Google Scholar] [CrossRef]
  118. Conners, M.; Giese, N.A.; Kulkarni, A.B.; Firestone, C.Y.; Morse, H.C.r.; Murphy, B.R. Enhanced pulmonary histopathology induced by respiratory syncytial virus (RSV) challenge of formalin-inactivated RSV-immunized BALB/c mice is abrogated by depletion of interleukin-4 (IL-4) and IL-10. J. Virol. 1994, 68, 5321–5325. [Google Scholar] [CrossRef]
  119. Garofalo, R.; Kimpen, J.L.L.; Welliver, R.C.; Ogra, P.L. Eosinophil degranulation in the respiratory tract during naturally acquired respiratory syncytial virus infection. J. Pediatr. 1992, 120, 28–32. [Google Scholar] [CrossRef]
  120. Dyer, K.D.; Percopo, C.M.; Fischer, E.R.; Gabryszewski, S.J.; Rosenberg, H.F. Pneumoviruses infect eosinophils and elicit MyD88-dependent release of chemoattractant cytokines and interleukin-6. Blood 2009, 114, 2649–2656. [Google Scholar] [CrossRef]
  121. Kimpen, J.L.; Garofalo, R.; Welliver, R.C.; Fujihara, K.; Ogra, P.L. An ultrastructural study of the interaction of human eosinophils with respiratory syncytial virus. Pediatr. Allergy Immunol. 1996, 7, 48–53. [Google Scholar] [CrossRef] [PubMed]
  122. Kimpen, J.L.L.; Garofalo, R.; Welliver, R.C.; Ogra, P.L. Activation of Human Eosinophils In vitro by Respiratory Syncytial Virus. Pediatr. Res. 1992, 32, 160–164. [Google Scholar] [CrossRef] [PubMed]
  123. Schwarze, J.; Cieslewicz, G.; Hamelmann, E.; Joetham, A.; Shultz, L.D.; Lamers, M.C.; Gelfand, E.W. IL-5 and Eosinophils Are Essential for the Development of Airway Hyperresponsiveness Following Acute Respiratory Syncytial Virus Infection. J. Immunol. 1999, 162, 2997–3004. [Google Scholar] [CrossRef]
  124. Zhang, D.; Yang, J.; Zhao, Y.; Shan, J.; Wang, L.; Yang, G.; He, S.; Li, E. RSV Infection in Neonatal Mice Induces Pulmonary Eosinophilia Responsible for Asthmatic Reaction. Front. Immunol. 2022, 13, 817113. [Google Scholar] [CrossRef]
  125. Ng, L.G.; Ostuni, R.; Hidalgo, A. Heterogeneity of neutrophils. Nat. Rev. Immunol. 2019, 19, 255–265. [Google Scholar] [CrossRef] [PubMed]
  126. Kruger, P.; Saffarzadeh, M.; Weber, A.N.; Rieber, N.; Radsak, M.; von Bernuth, H.; Benarafa, C.; Roos, D.; Skokowa, J.; Hartl, D. Neutrophils: Between host defence, immune modulation, and tissue injury. PLoS Pathog. 2015, 11, e1004651. [Google Scholar] [CrossRef]
  127. Ma, Y.; Zhang, Y.; Zhu, L. Role of neutrophils in acute viral infection. Immun. Inflamm. Dis. 2021, 9, 1186–1196. [Google Scholar] [CrossRef]
  128. McNamara, P.S.; Flanagan, B.F.; Hart, C.A.; Smyth, R.L. Production of chemokines in the lungs of infants with severe respiratory syncytial virus bronchiolitis. J. Infect. Dis. 2005, 191, 1225–1232. [Google Scholar] [CrossRef]
  129. Bataki, E.L.; Evans, G.S.; Everard, M.L. Respiratory syncytial virus and neutrophil activation. Clin. Exp. Immunol. 2005, 140, 470–477. [Google Scholar] [CrossRef]
  130. Deng, Y.; Herbert, J.A.; Smith, C.M.; Smyth, R.L. An in vitro transepithelial migration assay to evaluate the role of neutrophils in Respiratory Syncytial Virus (RSV) induced epithelial damage. Sci. Rep. 2018, 8, 6777. [Google Scholar] [CrossRef]
  131. Funchal, G.A.; Jaeger, N.; Czepielewski, R.S.; Machado, M.S.; Muraro, S.P.; Stein, R.T.; Bonorino, C.B.; Porto, B.N. Respiratory syncytial virus fusion protein promotes TLR-4-dependent neutrophil extracellular trap formation by human neutrophils. PLoS ONE 2015, 10, e0124082. [Google Scholar] [CrossRef] [PubMed]
  132. Souza, P.S.S.; Barbosa, L.V.; Diniz, L.F.A.; da Silva, G.S.; Lopes, B.R.P.; Souza, P.M.R.; de Araujo, G.C.; Pessoa, D.; de Oliveira, J.; Souza, F.P.; et al. Neutrophil extracellular traps possess anti-human respiratory syncytial virus activity: Possible interaction with the viral F protein. Virus Res. 2018, 251, 68–77. [Google Scholar] [CrossRef] [PubMed]
  133. Deng, Y.; Herbert, J.A.; Robinson, E.; Ren, L.; Smyth, R.L.; Smith, C.M. Neutrophil-Airway Epithelial Interactions Result in Increased Epithelial Damage and Viral Clearance during Respiratory Syncytial Virus Infection. J. Virol. 2020, 94, e02161-19. [Google Scholar] [CrossRef] [PubMed]
  134. McNamara, P.S.; Ritson, P.; Selby, A.; Hart, C.A.; Smyth, R.L. Bronchoalveolar lavage cellularity in infants with severe respiratory syncytial virus bronchiolitis. Arch. Dis. Child. 2003, 88, 922–926. [Google Scholar] [CrossRef] [PubMed]
  135. Everard, M.L.; Swarbrick, A.; Wrightham, M.; McIntyre, J.; Dunkley, C.; James, P.D.; Sewell, H.F.; Milner, A.D. Analysis of cells obtained by bronchial lavage of infants with respiratory syncytial virus infection. Arch. Dis. Child. 1994, 71, 428–432. [Google Scholar] [CrossRef]
  136. Wang, S.Z.; Xu, H.; Wraith, A.; Bowden, J.J.; Alpers, J.H.; Forsyth, K.D. Neutrophils induce damage to respiratory epithelial cells infected with respiratory syncytial virus. Eur. Respir. J. 1998, 12, 612–618. [Google Scholar] [CrossRef] [PubMed]
  137. Acosta, P.L.; Caballero, M.T.; Polack, F.P. Brief History and Characterization of Enhanced Respiratory Syncytial Virus Disease. Clin. Vaccine Immunol. 2015, 23, 189–195. [Google Scholar] [CrossRef] [PubMed]
  138. Halfhide, C.P.; Flanagan, B.F.; Brearey, S.P.; Hunt, J.A.; Fonceca, A.M.; McNamara, P.S.; Howarth, D.; Edwards, S.; Smyth, R.L. Respiratory syncytial virus binds and undergoes transcription in neutrophils from the blood and airways of infants with severe bronchiolitis. J. Infect. Dis. 2011, 204, 451–458. [Google Scholar] [CrossRef]
  139. Jaovisidha, P.; Peeples, M.E.; Brees, A.A.; Carpenter, L.R.; Moy, J.N. Respiratory syncytial virus stimulates neutrophil degranulation and chemokine release. J. Immunol. 1999, 163, 2816–2820. [Google Scholar] [CrossRef]
  140. Cortjens, B.; de Boer, O.J.; de Jong, R.; Antonis, A.F.; Sabogal Pineros, Y.S.; Lutter, R.; van Woensel, J.B.; Bem, R.A. Neutrophil extracellular traps cause airway obstruction during respiratory syncytial virus disease. J. Pathol. 2016, 238, 401–411. [Google Scholar] [CrossRef]
  141. Habibi, M.S.; Thwaites, R.S.; Chang, M.; Jozwik, A.; Paras, A.; Kirsebom, F.; Varese, A.; Owen, A.; Cuthbertson, L.; James, P.; et al. Neutrophilic inflammation in the respiratory mucosa predisposes to RSV infection. Science 2020, 370, eaba9301. [Google Scholar] [CrossRef] [PubMed]
  142. Vivier, E.; Tomasello, E.; Baratin, M.; Walzer, T.; Ugolini, S. Functions of natural killer cells. Nat. Immunol. 2008, 9, 503–510. [Google Scholar] [CrossRef] [PubMed]
  143. Lodoen, M.B.; Lanier, L.L. Natural killer cells as an initial defense against pathogens. Curr. Opin. Immunol. 2006, 18, 391–398. [Google Scholar] [CrossRef] [PubMed]
  144. Bhat, R.; Farrag, M.A.; Almajhdi, F.N. Double-edged role of natural killer cells during RSV infection. Int. Rev. Immunol. 2020, 39, 233–244. [Google Scholar] [CrossRef] [PubMed]
  145. Hussell, T.; Openshaw, P.J. Intracellular IFN-? expression in natural killer cells precedes lung CD8 T cell recruitment during respiratory syncytial virus infectio. J. Gen. Virol. 1998, 79, 2593–2601. [Google Scholar] [CrossRef]
  146. Hussell, T.; Openshaw, P.J. IL-12-activated NK cells reduce lung eosinophilia to the attachment protein of respiratory syncytial virus but do not enhance the severity of illness in CD8 T cell-immunodeficient conditions. J. Immunol. 2000, 165, 7109–7115. [Google Scholar] [CrossRef]
  147. Kaiko, G.E.; Phipps, S.; Angkasekwinai, P.; Dong, C.; Foster, P.S. NK cell deficiency predisposes to viral-induced Th2-type allergic inflammation via epithelial-derived IL-25. J. Immunol. 2010, 185, 4681–4690. [Google Scholar] [CrossRef]
  148. Welliver, T.P.; Garofalo, R.P.; Hosakote, Y.; Hintz, K.H.; Avendano, L.; Sanchez, K.; Velozo, L.; Jafri, H.; Chavez-Bueno, S.; Ogra, P.L.; et al. Severe human lower respiratory tract illness caused by respiratory syncytial virus and influenza virus is characterized by the absence of pulmonary cytotoxic lymphocyte responses. J. Infect. Dis. 2007, 195, 1126–1136. [Google Scholar] [CrossRef]
  149. Haynes, L.M.; Moore, D.D.; Kurt-Jones, E.A.; Finberg, R.W.; Anderson, L.J.; Tripp, R.A. Involvement of toll-like receptor 4 in innate immunity to respiratory syncytial virus. J. Virol. 2001, 75, 10730–10737. [Google Scholar] [CrossRef]
  150. Li, F.; Zhu, H.; Sun, R.; Wei, H.; Tian, Z. Natural killer cells are involved in acute lung immune injury caused by respiratory syncytial virus infection. J. Virol. 2012, 86, 2251–2258. [Google Scholar] [CrossRef]
  151. Long, X.; Xie, J.; Zhao, K.; Li, W.; Tang, W.; Chen, S.; Zang, N.; Ren, L.; Deng, Y.; Xie, X.; et al. NK cells contribute to persistent airway inflammation and AHR during the later stage of RSV infection in mice. Med. Microbiol. Immunol. 2016, 205, 459–470. [Google Scholar] [CrossRef] [PubMed]
  152. van Erp, E.A.; Feyaerts, D.; Duijst, M.; Mulder, H.L.; Wicht, O.; Luytjes, W.; Ferwerda, G.; van Kasteren, P.B. Respiratory Syncytial Virus Infects Primary Neonatal and Adult Natural Killer Cells and Affects Their Antiviral Effector Function. J. Infect. Dis. 2019, 219, 723–733. [Google Scholar] [CrossRef] [PubMed]
  153. Harker, J.A.; Godlee, A.; Wahlsten, J.L.; Lee, D.C.; Thorne, L.G.; Sawant, D.; Tregoning, J.S.; Caspi, R.R.; Bukreyev, A.; Collins, P.L.; et al. Interleukin 18 coexpression during respiratory syncytial virus infection results in enhanced disease mediated by natural killer cells. J. Virol. 2010, 84, 4073–4082. [Google Scholar] [CrossRef] [PubMed]
  154. Hettinger, J.; Richards, D.M.; Hansson, J.; Barra, M.M.; Joschko, A.C.; Krijgsveld, J.; Feuerer, M. Origin of monocytes and macrophages in a committed progenitor. Nat. Immunol. 2013, 14, 821–830. [Google Scholar] [CrossRef] [PubMed]
  155. Ziegler-Heitbrock, L. Monocyte subsets in man and other species. Cell Immunol. 2014, 289, 135–139. [Google Scholar] [CrossRef]
  156. Italiani, P.; Boraschi, D. From Monocytes to M1/M2 Macrophages: Phenotypical vs. Functional Differentiation. Front. Immunol. 2014, 5, 514. [Google Scholar] [CrossRef]
  157. Shi, C.; Pamer, E.G. Monocyte recruitment during infection and inflammation. Nat. Rev. Immunol. 2011, 11, 762–774. [Google Scholar] [CrossRef]
  158. Naidoo, K.K.; Ndumnego, O.C.; Ismail, N.; Dong, K.L.; Ndung’u, T. Antigen Presenting Cells Contribute to Persistent Immune Activation Despite Antiretroviral Therapy Initiation During Hyperacute HIV-1 Infection. Front. Immunol. 2021, 12, 738743. [Google Scholar] [CrossRef]
  159. Cao, W.; Taylor, A.K.; Biber, R.E.; Davis, W.G.; Kim, J.H.; Reber, A.J.; Chirkova, T.; De La Cruz, J.A.; Pandey, A.; Ranjan, P.; et al. Rapid differentiation of monocytes into type I IFN-producing myeloid dendritic cells as an antiviral strategy against influenza virus infection. J. Immunol. 2012, 189, 2257–2265. [Google Scholar] [CrossRef]
  160. Gill, M.A.; Long, K.; Kwon, T.; Muniz, L.; Mejias, A.; Connolly, J.; Roy, L.; Banchereau, J.; Ramilo, O. Differential recruitment of dendritic cells and monocytes to respiratory mucosal sites in children with influenza virus or respiratory syncytial virus infection. J. Infect. Dis. 2008, 198, 1667–1676. [Google Scholar] [CrossRef]
  161. Goritzka, M.; Makris, S.; Kausar, F.; Durant, L.R.; Pereira, C.; Kumagai, Y.; Culley, F.J.; Mack, M.; Akira, S.; Johansson, C. Alveolar macrophage-derived type I interferons orchestrate innate immunity to RSV through recruitment of antiviral monocytes. J. Exp. Med. 2015, 212, 699–714. [Google Scholar] [CrossRef]
  162. Kim, T.H.; Kim, C.W.; Oh, D.S.; Jung, H.E.; Lee, H.K. Monocytes Contribute to IFN-beta Production via the MyD88-Dependent Pathway and Cytotoxic T-Cell Responses against Mucosal Respiratory Syncytial Virus Infection. Immune Netw. 2021, 21, e27. [Google Scholar] [CrossRef] [PubMed]
  163. Barr, F.E.; Pedigo, H.; Johnson, T.R.; Shepherd, V.L. Surfactant protein-A enhances uptake of respiratory syncytial virus by monocytes and U937 macrophages. Am. J. Respir. Cell Mol. Biol. 2000, 23, 586–592. [Google Scholar] [CrossRef] [PubMed]
  164. Kurt-Jones, E.A.; Popova, L.; Kwinn, L.; Haynes, L.M.; Jones, L.P.; Tripp, R.A.; Walsh, E.E.; Freeman, M.W.; Golenbock, D.T.; Anderson, L.J.; et al. Pattern recognition receptors TLR4 and CD14 mediate response to respiratory syncytial virus. Nat. Immunol. 2000, 1, 398–401. [Google Scholar] [CrossRef] [PubMed]
  165. Ahout, I.M.; Jans, J.; Haroutiounian, L.; Simonetti, E.R.; van der Gaast-de Jongh, C.; Diavatopoulos, D.A.; de Jonge, M.I.; de Groot, R.; Ferwerda, G. Reduced Expression of HLA-DR on Monocytes During Severe Respiratory Syncytial Virus Infections. Pediatr. Infect. Dis. J. 2016, 35, e89–e96. [Google Scholar] [CrossRef]
  166. Besteman, S.B.; Phung, E.; Raeven, H.H.M.; Amatngalim, G.D.; Rumpret, M.; Crabtree, J.; Schepp, R.M.; Rodenburg, L.W.; Siemonsma, S.G.; Verleur, N.; et al. Recurrent Respiratory Syncytial Virus Infection in a CD14-Deficient Patient. J. Infect. Dis. 2022, 226, 258–269. [Google Scholar] [CrossRef]
  167. Bont, L.; Heijnen, C.J.; Kavelaars, A.; van Aalderen, W.M.; Brus, F.; Draaisma, J.T.; Geelen, S.M.; Kimpen, J.L. Monocyte IL-10 production during respiratory syncytial virus bronchiolitis is associated with recurrent wheezing in a one-year follow-up study. Am. J. Respir. Crit. Care Med. 2000, 161, 1518–1523. [Google Scholar] [CrossRef]
  168. Midulla, F.; Huang, Y.T.; Gilbert, I.A.; Cirino, N.M.; McFadden, E.R., Jr.; Panuska, J.R. Respiratory syncytial virus infection of human cord and adult blood monocytes and alveolar macrophages. Am. Rev. Respir. Dis. 1989, 140, 771–777. [Google Scholar] [CrossRef]
  169. Krilov, L.R.; Hendry, R.M.; Godfrey, E.; McIntosh, K. Respiratory virus infection of peripheral blood monocytes: Correlation with ageing of cells and interferon production in vitro. J. Gen. Virol. 1987, 68, 1749–1753. [Google Scholar] [CrossRef]
  170. Takeuchi, R.; Tsutsumi, H.; Osaki, M.; Sone, S.; Imai, S.; Chiba, S. Respiratory syncytial virus infection of neonatal monocytes stimulates synthesis of interferon regulatory factor 1 and interleukin-1beta (IL-1beta)-converting enzyme and secretion of IL-1beta. J. Virol. 1998, 72, 837–840. [Google Scholar] [CrossRef]
  171. Polack, F.P.; Irusta, P.M.; Hoffman, S.J.; Schiatti, M.P.; Melendi, G.A.; Delgado, M.F.; Laham, F.R.; Thumar, B.; Hendry, R.M.; Melero, J.A.; et al. The cysteine-rich region of respiratory syncytial virus attachment protein inhibits innate immunity elicited by the virus and endotoxin. Proc. Natl. Acad. Sci. USA 2005, 102, 8996–9001. [Google Scholar] [CrossRef] [PubMed]
  172. Mosser, D.M.; Edwards, J.P. Exploring the full spectrum of macrophage activation. Nat. Rev. Immunol. 2008, 8, 958–969. [Google Scholar] [CrossRef] [PubMed]
  173. Hou, F.; Xiao, K.; Tang, L.; Xie, L. Diversity of Macrophages in Lung Homeostasis and Diseases. Front. Immunol. 2021, 12, 753940. [Google Scholar] [CrossRef] [PubMed]
  174. Wang, Y.; Zheng, J.; Wang, X.; Yang, P.; Zhao, D. Alveolar macrophages and airway hyperresponsiveness associated with respiratory syncytial virus infection. Front. Immunol. 2022, 13, 1012048. [Google Scholar] [CrossRef]
  175. Makris, S.; Bajorek, M.; Culley, F.J.; Goritzka, M.; Johansson, C. Alveolar Macrophages Can Control Respiratory Syncytial Virus Infection in the Absence of Type I Interferons. J. Innate Immun. 2016, 8, 452–463. [Google Scholar] [CrossRef]
  176. Oh, D.S.; Kim, T.H.; Lee, H.K. Differential Role of Anti-Viral Sensing Pathway for the Production of Type I Interferon beta in Dendritic Cells and Macrophages Against Respiratory Syncytial Virus A2 Strain Infection. Viruses 2019, 11, 62. [Google Scholar] [CrossRef]
  177. Kolli, D.; Gupta, M.R.; Sbrana, E.; Velayutham, T.S.; Chao, H.; Casola, A.; Garofalo, R.P. Alveolar macrophages contribute to the pathogenesis of human metapneumovirus infection while protecting against respiratory syncytial virus infection. Am. J. Respir. Cell Mol. Biol. 2014, 51, 502–515. [Google Scholar] [CrossRef]
  178. Reed, J.L.; Brewah, Y.A.; Delaney, T.; Welliver, T.; Burwell, T.; Benjamin, E.; Kuta, E.; Kozhich, A.; McKinney, L.; Suzich, J.; et al. Macrophage impairment underlies airway occlusion in primary respiratory syncytial virus bronchiolitis. J. Infect. Dis. 2008, 198, 1783–1793. [Google Scholar] [CrossRef]
  179. Huang, S.; Zhu, B.; Cheon, I.S.; Goplen, N.P.; Jiang, L.; Zhang, R.; Peebles, R.S.; Mack, M.; Kaplan, M.H.; Limper, A.H.; et al. PPAR-gamma in Macrophages Limits Pulmonary Inflammation and Promotes Host Recovery following Respiratory Viral Infection. J. Virol. 2019, 93, e00030-19. [Google Scholar] [CrossRef]
  180. Shirey, K.A.; Pletneva, L.M.; Puche, A.C.; Keegan, A.D.; Prince, G.A.; Blanco, J.C.; Vogel, S.N. Control of RSV-induced lung injury by alternatively activated macrophages is IL-4R alpha-, TLR4-, and IFN-beta-dependent. Mucosal Immunol. 2010, 3, 291–300. [Google Scholar] [CrossRef]
  181. Franke-Ullmann, G.; Pförtner, C.; Walter, P.; Steinmüller, C.; Lohmann-Matthes, M.L.; Kobzik, L.; Freihorst, J. Alteration of pulmonary macrophage function by respiratory syncytial virus infection in vitro. J. Immunol. 1995, 154, 268–280. [Google Scholar] [CrossRef] [PubMed]
  182. Panuska, J.R.; Cirino, N.M.; Midulla, F.; Despot, J.E.; McFadden, E.R., Jr.; Huang, Y.T. Productive infection of isolated human alveolar macrophages by respiratory syncytial virus. J. Clin. Investig. 1990, 86, 113–119. [Google Scholar] [CrossRef] [PubMed]
  183. Santos, L.D.; Antunes, K.H.; Muraro, S.P.; de Souza, G.F.; da Silva, A.G.; Felipe, J.S.; Zanetti, L.C.; Czepielewski, R.S.; Magnus, K.; Scotta, M.; et al. TNF-mediated alveolar macrophage necroptosis drives disease pathogenesis during respiratory syncytial virus infection. Eur. Respir. J. 2021, 57, 2003764. [Google Scholar] [CrossRef] [PubMed]
  184. Haeberle, H.A.; Takizawa, R.; Casola, A.; Brasie, A.R.; Dieterich, H.-J.; van Rooijen, N.; Gatalica, Z.; Garofalo, R.P. Respiratory syncytial virus-induced activation of nuclear factor-kappaB in the lung involves alveolar macrophages and toll-like receptor 4-dependent pathways. J. Infect. Dis. 2002, 186, 1199–1206. [Google Scholar] [CrossRef] [PubMed]
  185. Harris, J.; Werling, D. Binding and entry of respiratory syncytial virus into host cells and initiation of the innate immune response. Cell Microbiol. 2003, 5, 671–680. [Google Scholar] [CrossRef]
  186. Becker, S.; Quay, J.; Soukup, J. Cytokine (tumor necrosis factor, IL-6, and IL-8) production by respiratory syncytial virus-infected human alveolar macrophages. J. Immunol. 1991, 147, 4307–4312. [Google Scholar] [CrossRef]
  187. Tsutsumi, H.; Matsuda, K.; Sone, S.; Takeuchi, R.; Chiba, S. Respiratory syncytial virus-induced cytokine production by neonatal macrophages. Clin. Exp. Immunol. 1996, 106, 442–446. [Google Scholar] [CrossRef]
  188. Eichinger, K.M.; Egana, L.; Orend, J.G.; Resetar, E.; Anderson, K.B.; Patel, R.; Empey, K.M. Alveolar macrophages support interferon gamma-mediated viral clearance in RSV-infected neonatal mice. Respir. Res. 2015, 16, 122. [Google Scholar] [CrossRef]
  189. Stockwin, L.H.; McGonagle, D.; Martin, I.G.; Blair, G.E. Dendritic cells: Immunological sentinels with a central role in health and disease. Immunol. Cell Biol. 2000, 78, 91–102. [Google Scholar] [CrossRef]
  190. Zaslona, Z.; Wilhelm, J.; Cakarova, L.; Marsh, L.M.; Seeger, W.; Lohmeyer, J.; von Wulffen, W. Transcriptome profiling of primary murine monocytes, lung macrophages and lung dendritic cells reveals a distinct expression of genes involved in cell trafficking. Respir. Res. 2009, 10, 2. [Google Scholar] [CrossRef]
  191. Boltjes, A.; van Wijk, F. Human dendritic cell functional specialization in steady-state and inflammation. Front. Immunol. 2014, 5, 131. [Google Scholar] [CrossRef] [PubMed]
  192. Geissmann, F.; Manz, M.G.; Jung, S.; Sieweke, M.H.; Merad, M.; Ley, K. Development of Monocytes, Macrophages, and Dendritic Cells. Science 2010, 327, 656–661. [Google Scholar] [CrossRef] [PubMed]
  193. Kim, T.H.; Oh, D.S.; Jung, H.E.; Chang, J.; Lee, H.K. Plasmacytoid Dendritic Cells Contribute to the Production of IFN-beta via TLR7-MyD88-Dependent Pathway and CTL Priming during Respiratory Syncytial Virus Infection. Viruses 2019, 11, 730. [Google Scholar] [CrossRef] [PubMed]
  194. Klouwenberg, P.K.; Tan, L.; Werkman, W.; van Bleek, G.M.; Coenjaerts, F. The role of Toll-like receptors in regulating the immune response against respiratory syncytial virus. Crit. Rev. Immunol. 2009, 29, 531–550. [Google Scholar] [CrossRef] [PubMed]
  195. Lukens, M.V.; Kruijsen, D.; Coenjaerts, F.E.; Kimpen, J.L.; van Bleek, G.M. Respiratory syncytial virus-induced activation and migration of respiratory dendritic cells and subsequent antigen presentation in the lung-draining lymph node. J. Virol. 2009, 83, 7235–7243. [Google Scholar] [CrossRef] [PubMed]
  196. Johnson, T.R.; Johnson, C.N.; Corbett, K.S.; Edwards, G.C.; Graham, B.S. Primary human mDC1, mDC2, and pDC dendritic cells are differentially infected and activated by respiratory syncytial virus. PLoS ONE 2011, 6, e16458. [Google Scholar] [CrossRef] [PubMed]
  197. Rudd, B.D.; Schaller, M.A.; Smit, J.J.; Kunkel, S.L.; Neupane, R.; Kelley, L.; Berlin, A.A.; Lukacs, N.W. MyD88-mediated instructive signals in dendritic cells regulate pulmonary immune responses during respiratory virus infection. J. Immunol. 2007, 178, 5820–5827. [Google Scholar] [CrossRef]
  198. Schijf, M.A.; Lukens, M.V.; Kruijsen, D.; van Uden, N.O.; Garssen, J.; Coenjaerts, F.E.; Van’t Land, B.; van Bleek, G.M. Respiratory syncytial virus induced type I IFN production by pDC is regulated by RSV-infected airway epithelial cells, RSV-exposed monocytes and virus specific antibodies. PLoS ONE 2013, 8, e81695. [Google Scholar] [CrossRef]
  199. Bartz, H.; Türkel, O.; Hoffjan, S.; Rothoeft, T.; Gonschorek, A.; Schauer, U. Respiratory syncytial virus decreases the capacity of myeloid dendritic cells to induce interferon-gamma in naïve T cells. Immunology 2003, 109, 49–57. [Google Scholar] [CrossRef]
  200. de Graaff, P.M.A.; de Jong, E.C.; van Capel, T.M.; van Dijk, M.E.A.; Roholl, P.J.M.; Boes, J.; Luytjes, W.; Kimpen, J.L.L.; van Bleek, G.M. Respiratory syncytial virus infection of monocyte-derived dendritic cells decreases their capacity to activate CD4 T cells. J. Immunol. 2005, 175, 5904–5911. [Google Scholar] [CrossRef]
  201. Cespedes, P.F.; Bueno, S.M.; Ramirez, B.A.; Gomez, R.S.; Riquelme, S.A.; Palavecino, C.E.; Mackern-Oberti, J.P.; Mora, J.E.; Depoil, D.; Sacristan, C.; et al. Surface expression of the hRSV nucleoprotein impairs immunological synapse formation with T cells. Proc. Natl. Acad. Sci. USA 2014, 111, E3214–E3223. [Google Scholar] [CrossRef] [PubMed]
  202. Lau-Kilby, A.W.; Turfkruyer, M.; Kehl, M.; Yang, L.; Buchholz, U.J.; Hickey, K.; Malloy, A.M.W. Type I IFN ineffectively activates neonatal dendritic cells limiting respiratory antiviral T-cell responses. Mucosal Immunol. 2020, 13, 371–380. [Google Scholar] [CrossRef] [PubMed]
  203. Malloy, A.M.; Ruckwardt, T.J.; Morabito, K.M.; Lau-Kilby, A.W.; Graham, B.S. Pulmonary Dendritic Cell Subsets Shape the Respiratory Syncytial Virus-Specific CD8+ T Cell Immunodominance Hierarchy in Neonates. J. Immunol. 2017, 198, 394–403. [Google Scholar] [CrossRef] [PubMed]
  204. Shrestha, B.; You, D.; Saravia, J.; Siefker, D.T.; Jaligama, S.; Lee, G.I.; Sallam, A.A.; Harding, J.N.; Cormier, S.A. IL-4Ralpha on dendritic cells in neonates and Th2 immunopathology in respiratory syncytial virus infection. J. Leukoc. Biol. 2017, 102, 153–161. [Google Scholar] [CrossRef] [PubMed]
  205. Johnson, T.R.; McLellan, J.S.; Graham, B.S. Respiratory syncytial virus glycoprotein G interacts with DC-SIGN and L-SIGN to activate ERK1 and ERK2. J. Virol. 2012, 86, 1339–1347. [Google Scholar] [CrossRef] [PubMed]
  206. Stambas, J.; Lu, C.; Tripp, R.A. Innate and adaptive immune responses in respiratory virus infection: Implications for the clinic. Expert. Rev. Respir. Med. 2020, 14, 1141–1147. [Google Scholar] [CrossRef]
  207. Bonilla, F.A.; Oettgen, H.C. Adaptive immunity. J. Allergy Clin. Immunol. 2010, 125, S33–S40. [Google Scholar] [CrossRef]
  208. Boehm, T.; Swann, J.B. Origin and evolution of adaptive immunity. Annu. Rev. Anim. Biosci. 2014, 2, 259–283. [Google Scholar] [CrossRef]
  209. Wang, Y.; Liu, J.; Burrows, P.D.; Wang, J.Y. B Cell Development and Maturation. Adv. Exp. Med. Biol. 2020, 1254, 1–22. [Google Scholar] [CrossRef]
  210. Lam, J.H.; Smith, F.L.; Baumgarth, N. B Cell Activation and Response Regulation During Viral Infections. Viral Immunol. 2020, 33, 294–306. [Google Scholar] [CrossRef]
  211. Roman, M.; Calhoun, W.J.; Hinton, K.L.; Avendano, L.F.; Simon, V.; Escobar, A.M.; Gaggero, A.; Diaz, P.V. Respiratory syncytial virus infection in infants is associated with predominant Th-2-like response. Am. J. Respir. Crit. Care Med. 1997, 156, 190–195. [Google Scholar] [CrossRef] [PubMed]
  212. Andreano, E.; Paciello, I.; Bardelli, M.; Tavarini, S.; Sammicheli, C.; Frigimelica, E.; Guidotti, S.; Torricelli, G.; Biancucci, M.; D’Oro, U.; et al. The respiratory syncytial virus (RSV) prefusion F-protein functional antibody repertoire in adult healthy donors. EMBO Mol. Med. 2021, 13, e14035. [Google Scholar] [CrossRef] [PubMed]
  213. Lee, F.E.; Falsey, A.R.; Halliley, J.L.; Sanz, I.; Walsh, E.E. Circulating antibody-secreting cells during acute respiratory syncytial virus infection in adults. J. Infect. Dis. 2010, 202, 1659–1666. [Google Scholar] [CrossRef]
  214. Raes, M.; Alliet, P.; Gillis, P.; Kortleven, J.; Magerman, K.; Rummens, J.L. Peripheral blood T and B lymphocyte subpopulations in infants with acute respiratory syncytial virus brochiolitis. Pediatr. Allergy Immunol. 1997, 8, 97–102. [Google Scholar] [CrossRef]
  215. Xiao, X.; Tang, A.; Cox, K.S.; Wen, Z.; Callahan, C.; Sullivan, N.L.; Nahas, D.D.; Cosmi, S.; Galli, J.D.; Minnier, M.; et al. Characterization of potent RSV neutralizing antibodies isolated from human memory B cells and identification of diverse RSV/hMPV cross-neutralizing epitopes. MAbs 2019, 11, 1415–1427. [Google Scholar] [CrossRef] [PubMed]
  216. Alturaiki, W.; McFarlane, A.J.; Rose, K.; Corkhill, R.; McNamara, P.S.; Schwarze, J.; Flanagan, B.F. Expression of the B cell differentiation factor BAFF and chemokine CXCL13 in a murine model of Respiratory Syncytial Virus infection. Cytokine 2018, 110, 267–271. [Google Scholar] [CrossRef]
  217. Reed, J.L.; Welliver, T.P.; Sims, G.P.; McKinney, L.; Velozo, L.; Avendano, L.; Hintz, K.; Luma, J.; Coyle, A.J.; Welliver, R.C., Sr. Innate Immune Signals Modulate Antiviral and Polyreactive Antibody Responses during Severe Respiratory Syncytial Virus Infection. J. Infect. Dis. 2009, 199, 1128–1138. [Google Scholar] [CrossRef]
  218. Falsey, A.R.; Singh, H.K.; Walsh, E.E. Serum antibody decay in adults following natural respiratory syncytial virus infection. J. Med. Virol. 2006, 78, 1493–1497. [Google Scholar] [CrossRef]
  219. Green, C.A.; Sande, C.J.; de Lara, C.; Thompson, A.J.; Silva-Reyes, L.; Napolitano, F.; Pierantoni, A.; Capone, S.; Vitelli, A.; Klenerman, P.; et al. Humoral and cellular immunity to RSV in infants, children and adults. Vaccine 2018, 36, 6183–6190. [Google Scholar] [CrossRef]
  220. Habibi, M.S.; Jozwik, A.; Makris, S.; Dunning, J.; Paras, A.; DeVincenzo, J.P.; de Haan, C.A.; Wrammert, J.; Openshaw, P.J.; Chiu, C.; et al. Impaired Antibody-mediated Protection and Defective IgA B-Cell Memory in Experimental Infection of Adults with Respiratory Syncytial Virus. Am. J. Respir. Crit. Care Med. 2015, 191, 1040–1049. [Google Scholar] [CrossRef]
  221. Cortjens, B.; Yasuda, E.; Yu, X.; Wagner, K.; Claassen, Y.B.; Bakker, A.Q.; van Woensel, J.B.M.; Beaumont, T. Broadly Reactive Anti-Respiratory Syncytial Virus G Antibodies from Exposed Individuals Effectively Inhibit Infection of Primary Airway Epithelial Cells. J. Virol. 2017, 91, e02357-16. [Google Scholar] [CrossRef] [PubMed]
  222. Schmidt, M.E.; Meyerholz, D.K.; Varga, S.M. Pre-existing neutralizing antibodies prevent CD8 T cell-mediated immunopathology following respiratory syncytial virus infection. Mucosal Immunol. 2020, 13, 507–517. [Google Scholar] [CrossRef] [PubMed]
  223. Falsey, A.R.; Walsh, E.E. Relationship of serum antibody to risk of respiratory syncytial virus infection in elderly adults. J. Infect. Dis. 1998, 177, 463–466. [Google Scholar] [CrossRef] [PubMed]
  224. Walsh, E.E.; Falsey, A.R. Humoral and mucosal immunity in protection from natural respiratory syncytial virus infection in adults. J. Infect. Dis. 2004, 190, 373–378. [Google Scholar] [CrossRef]
  225. Walsh, E.E.; Peterson, D.R.; Falsey, A.R. Risk factors for severe respiratory syncytial virus infection in elderly persons. J. Infect. Dis. 2004, 189, 233–238. [Google Scholar] [CrossRef]
  226. Ascough, S.; Dayananda, P.; Kalyan, M.; Kuong, S.U.; Gardener, Z.; Bergstrom, E.; Paterson, S.; Kar, S.; Avadhan, V.; Thwaites, R.; et al. Divergent age-related humoral correlates of protection against respiratory syncytial virus infection in older and young adults: A pilot, controlled, human infection challenge model. Lancet Healthy Longev. 2022, 3, e405–e416. [Google Scholar] [CrossRef]
  227. Falsey, A.R.; Walsh, E.E. Humoral immunity to respiratory syncytial virus infection in the elderly. J. Med. Virol. 1992, 36, 39–43. [Google Scholar] [CrossRef]
  228. Walsh, E.E.; Peterson, D.R.; Kalkanoglu, A.E.; Lee, F.E.; Falsey, A.R. Viral shedding and immune responses to respiratory syncytial virus infection in older adults. J. Infect. Dis. 2013, 207, 1424–1432. [Google Scholar] [CrossRef]
  229. Falsey, A.R.; Walsh, E.E.; Looney, R.J.; Kolassa, J.E.; Formica, M.A.; Criddle, M.C.; Hall, W.J. Comparison of respiratory syncytial virus humoral immunity and response to infection in young and elderly adults. J. Med. Virol. 1999, 59, 221–226. [Google Scholar] [CrossRef]
  230. Walsh, E.E.; Falsey, A.R. Age related differences in humoral immune response to respiratory syncytial virus infection in adults. J. Med. Virol. 2004, 73, 295–299. [Google Scholar] [CrossRef]
  231. Goodwin, E.; Gilman, M.S.A.; Wrapp, D.; Chen, M.; Ngwuta, J.O.; Moin, S.M.; Bai, P.; Sivasubramanian, A.; Connor, R.I.; Wright, P.F.; et al. Infants Infected with Respiratory Syncytial Virus Generate Potent Neutralizing Antibodies that Lack Somatic Hypermutation. Immunity 2018, 48, 339–349 e335. [Google Scholar] [CrossRef]
  232. van Erp, E.A.; Lakerveld, A.J.; de Graaf, E.; Larsen, M.D.; Schepp, R.M.; Hipgrave Ederveen, A.L.; Ahout, I.M.; de Haan, C.A.; Wuhrer, M.; Luytjes, W.; et al. Natural killer cell activation by respiratory syncytial virus-specific antibodies is decreased in infants with severe respiratory infections and correlates with Fc-glycosylation. Clin. Transl. Immunol. 2020, 9, e1112. [Google Scholar] [CrossRef] [PubMed]
  233. Williams, J.V.; Weitkamp, J.H.; Blum, D.L.; LaFleur, B.J.; Crowe, J.E., Jr. The human neonatal B cell response to respiratory syncytial virus uses a biased antibody variable gene repertoire that lacks somatic mutations. Mol. Immunol. 2009, 47, 407–414. [Google Scholar] [CrossRef]
  234. Murphy, B.R.; Graham, B.S.; Prince, G.A.; Walsh, E.E.; Chanock, R.M.; Karzon, D.T.; Wright, P.F. Serum and nasal-wash immunoglobulin G and A antibody response of infants and children to respiratory syncytial virus F and G glycoproteins following primary infection. J. Clin. Microbiol. 1986, 23, 1009–1014. [Google Scholar] [CrossRef] [PubMed]
  235. Kruijsen, D.; Bakkers, M.J.; van Uden, N.O.; Viveen, M.C.; van der Sluis, T.C.; Kimpen, J.L.; Leusen, J.H.; Coenjaerts, F.E.; van Bleek, G.M. Serum antibodies critically affect virus-specific CD4+/CD8+ T cell balance during respiratory syncytial virus infections. J. Immunol. 2010, 185, 6489–6498. [Google Scholar] [CrossRef] [PubMed]
  236. Vono, M.; Eberhardt, C.S.; Auderset, F.; Mastelic-Gavillet, B.; Lemeille, S.; Christensen, D.; Andersen, P.; Lambert, P.H.; Siegrist, C.A. Maternal Antibodies Inhibit Neonatal and Infant Responses to Vaccination by Shaping the Early-Life B Cell Repertoire within Germinal Centers. Cell Rep. 2019, 28, 1773–1784 e1775. [Google Scholar] [CrossRef]
  237. Zhivaki, D.; Lemoine, S.; Lim, A.; Morva, A.; Vidalain, P.O.; Schandene, L.; Casartelli, N.; Rameix-Welti, M.A.; Herve, P.L.; Deriaud, E.; et al. Respiratory Syncytial Virus Infects Regulatory B Cells in Human Neonates via Chemokine Receptor CX3CR1 and Promotes Lung Disease Severity. Immunity 2017, 46, 301–314. [Google Scholar] [CrossRef]
  238. Bukreyev, A.; Yang, L.; Fricke, J.; Cheng, L.; Ward, J.M.; Murphy, B.R.; Collins, P.L. The secreted form of respiratory syncytial virus G glycoprotein helps the virus evade antibody-mediated restriction of replication by acting as an antigen decoy and through effects on Fc receptor-bearing leukocytes. J. Virol. 2008, 82, 12191–12204. [Google Scholar] [CrossRef]
  239. Groothuis, J.R.; Levin, M.J.; Rodriguez, W.; Hall, C.B.; Long, C.E.; Kim, H.W.; Lauer, B.A.; Hemming, V.G. Use of intravenous gamma globulin to passively immunize high-risk children against respiratory syncytial virus: Safety and pharmacokinetics. The RSVIG Study Group. Antimicrob. Agents Chemother. 1991, 35, 1469–1473. [Google Scholar] [CrossRef]
  240. Groothuis, J.R.; Simoes, E.A.; Hemming, V.G. Respiratory syncytial virus (RSV) infection in preterm infants and the protective effects of RSV immune globulin (RSVIG). Respiratory Syncytial Virus Immune Globulin Study Group. Pediatrics 1995, 95, 463–467. [Google Scholar]
  241. Rodriguez, W.J.; Gruber, W.C.; Groothuis, J.R.; Simoes, E.A.; Rosas, A.J.; Lepow, M.; Kramer, A.; Hemming, V. Respiratory syncytial virus immune globulin treatment of RSV lower respiratory tract infection in previously healthy children. Pediatrics 1997, 100, 937–942. [Google Scholar] [CrossRef] [PubMed]
  242. Keam, S.J. Nirsevimab: First Approval. Drugs 2023, 83, 181–187. [Google Scholar] [CrossRef] [PubMed]
  243. Group, I.-R.S. Palivizumab, a Humanized Respiratory Syncytial Virus Monoclonal Antibody, Reduces Hospitalization From Respiratory Syncytial Virus Infection in High-risk Infants. Pediatrics 1998, 102, 531–537. [Google Scholar] [CrossRef]
  244. Kalia, V.; Sarkar, S.; Gourley, T.S.; Rouse, B.T.; Ahmed, R. Differentiation of memory B and T cells. Curr. Opin. Immunol. 2006, 18, 255–264. [Google Scholar] [CrossRef]
  245. Palacios-Pedrero, M.A.; Jansen, J.M.; Blume, C.; Stanislawski, N.; Jonczyk, R.; Molle, A.; Hernandez, M.G.; Kaiser, F.K.; Jung, K.; Osterhaus, A.; et al. Signs of immunosenescence correlate with poor outcome of mRNA COVID-19 vaccination in older adults. Nat. Aging 2022, 2, 896–905. [Google Scholar] [CrossRef]
  246. Nicholson, L.B. The immune system. Essays Biochem. 2016, 60, 275–301. [Google Scholar] [CrossRef]
  247. Benova, K.; Hanckova, M.; Koci, K.; Kudelova, M.; Betakova, T. T cells and their function in the immune response to viruses. Acta Virol. 2020, 64, 131–143. [Google Scholar] [CrossRef]
  248. Sealy, R.E.; Surman, S.L.; Hurwitz, J.L. CD4(+) T cells support establishment of RSV-specific IgG and IgA antibody secreting cells in the upper and lower murine respiratory tract following RSV infection. Vaccine 2017, 35, 2617–2621. [Google Scholar] [CrossRef]
  249. Kinnear, E.; Lambert, L.; McDonald, J.U.; Cheeseman, H.M.; Caproni, L.J.; Tregoning, J.S. Airway T cells protect against RSV infection in the absence of antibody. Mucosal Immunol. 2018, 11, 249–256. [Google Scholar] [CrossRef]
  250. Guvenel, A.; Jozwik, A.; Ascough, S.; Ung, S.K.; Paterson, S.; Kalyan, M.; Gardener, Z.; Bergstrom, E.; Kar, S.; Habibi, M.S.; et al. Epitope-specific airway-resident CD4+ T cell dynamics during experimental human RSV infection. J. Clin. Investig. 2020, 130, 523–538. [Google Scholar] [CrossRef]
  251. Kitcharoensakkul, M.; Bacharier, L.B.; Yin-Declue, H.; Boomer, J.S.; Sajol, G.; Leung, M.K.; Wilson, B.; Schechtman, K.B.; Atkinson, J.P.; Green, J.M.; et al. Impaired tumor necrosis factor-alpha secretion by CD4 T cells during respiratory syncytial virus bronchiolitis associated with recurrent wheeze. Immun. Inflamm. Dis. 2020, 8, 30–39. [Google Scholar] [CrossRef] [PubMed]
  252. Welliver, T.P.; Reed, J.L.; Welliver, R.C., Sr. Respiratory syncytial virus and influenza virus infections: Observations from tissues of fatal infant cases. Pediatr. Infect. Dis. J. 2008, 27, S92–S96. [Google Scholar] [CrossRef] [PubMed]
  253. De Swart, R.L.; Kuiken, T.; Timmerman, H.H.; van Amerongen, G.; Van Den Hoogen, B.G.; Vos, H.W.; Neijens, H.J.; Andeweg, A.C.; Osterhaus, A.D. Immunization of macaques with formalin-inactivated respiratory syncytial virus (RSV) induces interleukin-13-associated hypersensitivity to subsequent RSV infection. J. Virol. 2002, 76, 11561–11569. [Google Scholar] [CrossRef]
  254. Waris, M.E.; Tsou, C.; Erdman, D.D.; Zaki, S.R.; Anderson, L.J. Respiratory synctial virus infection in BALB/c mice previously immunized with formalin-inactivated virus induces enhanced pulmonary inflammatory response with a predominant Th2-like cytokine pattern. J. Virol. 1996, 70, 2852–2860. [Google Scholar] [CrossRef] [PubMed]
  255. Knudson, C.J.; Hartwig, S.M.; Meyerholz, D.K.; Varga, S.M. RSV vaccine-enhanced disease is orchestrated by the combined actions of distinct CD4 T cell subsets. PLoS Pathog. 2015, 11, e1004757. [Google Scholar] [CrossRef]
  256. You, D.; Marr, N.; Saravia, J.; Shrestha, B.; Lee, G.I.; Turvey, S.E.; Brombacher, F.; Herbert, D.R.; Cormier, S.A. IL-4Ralpha on CD4+ T cells plays a pathogenic role in respiratory syncytial virus reinfection in mice infected initially as neonates. J. Leukoc. Biol. 2013, 93, 933–942. [Google Scholar] [CrossRef] [PubMed]
  257. Roumanes, D.; Falsey, A.R.; Quataert, S.; Secor-Socha, S.; Lee, F.E.; Yang, H.; Bandyopadhyay, S.; Holden-Wiltse, J.; Topham, D.J.; Walsh, E.E. T-Cell Responses in Adults During Natural Respiratory Syncytial Virus Infection. J. Infect. Dis. 2018, 218, 418–428. [Google Scholar] [CrossRef]
  258. Graham, B.S.; Bunton, L.A.; Wright, P.F.; Karzon, D.T. Role of T lymphocyte subsets in the pathogenesis of primary infection and rechallenge with respiratory syncytial virus in mice. J. Clin. Investig. 1991, 88, 1026–1033. [Google Scholar] [CrossRef]
  259. Conners, M.; Kulkarni, A.B.; Firestone, C.Y.; Holmes, K.L.; Morse, H.C.r.; Sotnikov, A.V.; Murphy, B.R. Pulmonary histopathology induced by respiratory syncytial virus (RSV) challenge of formalin-inactivated RSV-immunized BALB/c mice is abrogated by depletion of CD4+ T cells. J. Virol. 1992, 66, 7444–7451. [Google Scholar] [CrossRef]
  260. Schneider-Ohrum, K.; Snell Bennett, A.; Rajani, G.M.; Hostetler, L.; Maynard, S.K.; Lazzaro, M.; Cheng, L.I.; O’Day, T.; Cayatte, C. CD4(+) T Cells Drive Lung Disease Enhancement Induced by Immunization with Suboptimal Doses of Respiratory Syncytial Virus Fusion Protein in the Mouse Model. J. Virol. 2019, 93, e00695-19. [Google Scholar] [CrossRef]
  261. Raiden, S.; Sananez, I.; Remes-Lenicov, F.; Pandolfi, J.; Romero, C.; De Lillo, L.; Ceballos, A.; Geffner, J.; Arruvito, L. Respiratory Syncytial Virus (RSV) Infects CD4+ T Cells: Frequency of Circulating CD4+ RSV+ T Cells as a Marker of Disease Severity in Young Children. J. Infect. Dis. 2017, 215, 1049–1058. [Google Scholar] [CrossRef] [PubMed]
  262. Schlender, J.; Walliser, G.; Fricke, J.; Conzelmann, K.K. Respiratory syncytial virus fusion protein mediates inhibition of mitogen-induced T-cell proliferation by contact. J. Virol. 2002, 76, 1163–1170. [Google Scholar] [CrossRef] [PubMed]
  263. Bont, L. Natural Reinfection with Respiratory Syncytial Virus Does Not Boost Virus-Specific T-Cell Immunity. Pediatr. Res. 2002, 52, 363–367. [Google Scholar] [CrossRef]
  264. Heidema, J.; Lukens, M.V.; van Maren, W.W.C.; van Dijk, M.E.A.; Otten, H.G.; van Vught, A.J.; van der Werff, D.B.M.; van Gestel, S.J.P.; Semple, M.G.; Smyth, R.L.; et al. CD8+ T cell responses in bronchoalveolar lavage fluid and peripheral blood mononuclear cells of infants with severe primary respiratory syncytial virus infections. J. Immunol. 2007, 179, 8410–8417. [Google Scholar] [CrossRef] [PubMed]
  265. Knudson, C.J.; Weiss, K.A.; Hartwig, S.M.; Varga, S.M. The pulmonary localization of virus-specific T lymphocytes is governed by the tissue tropism of infection. J. Virol. 2014, 88, 9010–9016. [Google Scholar] [CrossRef]
  266. Jozwik, A.; Habibi, M.S.; Paras, A.; Zhu, J.; Guvenel, A.; Dhariwal, J.; Almond, M.; Wong, E.H.C.; Sykes, A.; Maybeno, M.; et al. RSV-specific airway resident memory CD8+ T cells and differential disease severity after experimental human infection. Nat. Commun. 2015, 6, 10224. [Google Scholar] [CrossRef]
  267. Ruckwardt, T.J.; Luongo, C.; Malloy, A.M.; Liu, J.; Chen, M.; Collins, P.L.; Graham, B.S. Responses against a subdominant CD8+ T cell epitope protect against immunopathology caused by a dominant epitope. J. Immunol. 2010, 185, 4673–4680. [Google Scholar] [CrossRef]
  268. Taylor, G.; Stott, E.J.; Hayle, A.J. Cytotoxic lymphocytes in the lungs of mice infected with respiratory syncytial virus. J. Gen. Virol. 1985, 66, 2533–2538. [Google Scholar] [CrossRef]
  269. Aberle, J.H.; Aberle, S.W.; Dworzak, M.N.; Mandl, C.W.; Rebhandl, W.; Vollnhofer, G.; Kundi, M.; Popow-Kraupp, T. Reduced interferon-gamma expression in peripheral blood mononuclear cells of infants with severe respiratory syncytial virus disease. Am. J. Respir. Crit. Care Med. 1999, 160, 1263–1268. [Google Scholar] [CrossRef]
  270. De Weerd, W.; Twilhaar, W.N.; Kimpen, J.L. T cell subset analysis in peripheral blood of children with RSV bronchiolitis. Scand. J. Infect. Dis. 1998, 30, 77–80. [Google Scholar] [CrossRef]
  271. El Saleeby, C.M.; Suzich, J.; Conley, M.E.; DeVincenzo, J.P. Quantitative effects of palivizumab and donor-derived T cells on chronic respiratory syncytial virus infection, lung disease, and fusion glycoprotein amino acid sequences in a patient before and after bone marrow transplantation. Clin. Infect. Dis. 2004, 39, 17–20. [Google Scholar] [CrossRef]
  272. Heidema, J.; de Bree, G.J.; de Graaff, P.M.A.; van Maren, W.W.C.; Hoogerhout, P.; Out, T.A.; Kimpen, J.L.L.; van Bleek, G.M. Human CD8(+) T cell responses against five newly identified respiratory syncytial virus-derived epitopes. J. Gen. Virol. 2004, 85, 2365–2374. [Google Scholar] [CrossRef] [PubMed]
  273. Lukens, M.V.; Claassen, E.A.; de Graaff, P.M.; van Dijk, M.E.; Hoogerhout, P.; Toebes, M.; Schumacher, T.N.; van der Most, R.G.; Kimpen, J.L.; van Bleek, G.M. Characterization of the CD8+ T cell responses directed against respiratory syncytial virus during primary and secondary infection in C57BL/6 mice. Virology 2006, 352, 157–168. [Google Scholar] [CrossRef] [PubMed]
  274. Ostler, T.; Ehl, S. Pulmonary T cells induced by respiratory syncytial virus are functional and can make an important contribution to long-lived protective immunity. Eur. J. Immunol. 2002, 32, 2562–2569. [Google Scholar] [CrossRef] [PubMed]
  275. Ruckwardt, T.J.; Bonaparte, K.L.; Nason, M.C.; Graham, B.S. Regulatory T cells promote early influx of CD8+ T cells in the lungs of respiratory syncytial virus-infected mice and diminish immunodominance disparities. J. Virol. 2009, 83, 3019–3028. [Google Scholar] [CrossRef]
  276. Srikiatkhachorn, A.; Braciale, T.J. Virus-specific CD8+ T lymphocytes downregulate T helper cell type 2 cytokine secretion and pulmonary eosinophilia during experimental murine respiratory syncytial virus infection. J. Exp. Med. 1997, 186, 421–432. [Google Scholar] [CrossRef]
  277. Ostler, T.; Davidson, W.; Ehl, S. Virus clearance and immunopathology by CD8(+) T cells during infection with respiratory syncytial virus are mediated by IFN-gamma. Eur. J. Immunol. 2002, 32, 2117–2123. [Google Scholar] [CrossRef]
  278. Bem, R.A.; Bos, A.P.; Bots, M.; Wolbink, A.M.; van Ham, S.M.; Medema, J.P.; Lutter, R.; van Woensel, J.B.M. Activation of the granzyme pathway in children with severe respiratory syncytial virus infection. Pediatr. Res. 2008, 63, 650–655. [Google Scholar] [CrossRef]
  279. Siefker, D.T.; Vu, L.; You, D.; McBride, A.; Taylor, R.; Jones, T.L.; DeVincenzo, J.; Cormier, S.A. Respiratory Syncytial Virus Disease Severity Is Associated with Distinct CD8(+) T-Cell Profiles. Am. J. Respir. Crit. Care Med. 2020, 201, 325–334. [Google Scholar] [CrossRef]
  280. van Schaik, S.M.; Obot, N.; Enhorning, G.; Hintz, K.; Gross, K.; Hancock, G.E.; Stack, A.M.; Welliver, R.C. Role of interferon gamma in the pathogenesis of primary respiratory syncytial virus infection in BALB/c mice. J. Med. Virol. 2000, 62, 257–266. [Google Scholar] [CrossRef]
  281. Rutigliano, J.A.; Graham, B.S. Prolonged production of TNF-alpha exacerbates illness during respiratory syncytial virus infection. J. Immunol. 2004, 173, 3408–3417. [Google Scholar] [CrossRef] [PubMed]
  282. Meckiff, B.J.; Ramirez-Suastegui, C.; Fajardo, V.; Chee, S.J.; Kusnadi, A.; Simon, H.; Eschweiler, S.; Grifoni, A.; Pelosi, E.; Weiskopf, D.; et al. Imbalance of Regulatory and Cytotoxic SARS-CoV-2-Reactive CD4(+) T Cells in COVID-19. Cell 2020, 183, 1340–1353 e1316. [Google Scholar] [CrossRef] [PubMed]
  283. Lan, Y.T.; Wang, Z.L.; Tian, P.; Gong, X.N.; Fan, Y.C.; Wang, K. Treg/Th17 imbalance and its clinical significance in patients with hepatitis B-associated liver cirrhosis. Diagn. Pathol. 2019, 14, 114. [Google Scholar] [CrossRef] [PubMed]
  284. Chatila, T.A. Role of regulatory T cells in human diseases. J. Allergy Clin. Immunol. 2005, 116, 949–959, quiz 960. [Google Scholar] [CrossRef] [PubMed]
  285. Ghiringhelli, F.; Menard, C.; Terme, M.; Flament, C.; Taieb, J.; Chaput, N.; Puig, P.E.; Novault, S.; Escudier, B.; Vivier, E.; et al. CD4+CD25+ regulatory T cells inhibit natural killer cell functions in a transforming growth factor-beta-dependent manner. J. Exp. Med. 2005, 202, 1075–1085. [Google Scholar] [CrossRef] [PubMed]
  286. Lewkowicz, P.; Lewkowicz, N.; Sasiak, A.; Tchorzewski, H. Lipopolysaccharide-activated CD4+CD25+ T regulatory cells inhibit neutrophil function and promote their apoptosis and death. J. Immunol. 2006, 177, 7155–7163. [Google Scholar] [CrossRef]
  287. Rudensky, A.V. Regulatory T cells and Foxp3. Immunol. Rev. 2011, 241, 260–268. [Google Scholar] [CrossRef]
  288. Weiss, K.A.; Christiaansen, A.F.; Fulton, R.B.; Meyerholz, D.K.; Varga, S.M. Multiple CD4+ T cell subsets produce immunomodulatory IL-10 during respiratory syncytial virus infection. J. Immunol. 2011, 187, 3145–3154. [Google Scholar] [CrossRef]
  289. Demoulins, T.; Brugger, M.; Zumkehr, B.; Oliveira Esteves, B.I.; Mehinagic, K.; Fahmi, A.; Borcard, L.; Taddeo, A.; Jandrasits, D.; Posthaus, H.; et al. The specific features of the developing T cell compartment of the neonatal lung are a determinant of respiratory syncytial virus immunopathogenesis. PLoS Pathog. 2021, 17, e1009529. [Google Scholar] [CrossRef]
  290. Fulton, R.B.; Meyerholz, D.K.; Varga, S.M. Foxp3+ CD4 regulatory T cells limit pulmonary immunopathology by modulating the CD8 T cell response during respiratory syncytial virus infection. J. Immunol. 2010, 185, 2382–2392. [Google Scholar] [CrossRef]
  291. Lee, D.C.; Harker, J.A.; Tregoning, J.S.; Atabani, S.F.; Johansson, C.; Schwarze, J.; Openshaw, P.J. CD25+ natural regulatory T cells are critical in limiting innate and adaptive immunity and resolving disease following respiratory syncytial virus infection. J. Virol. 2010, 84, 8790–8798. [Google Scholar] [CrossRef] [PubMed]
  292. Loebbermann, J.; Thornton, H.; Durant, L.; Sparwasser, T.; Webster, K.E.; Sprent, J.; Culley, F.J.; Johansson, C.; Openshaw, P.J. Regulatory T cells expressing granzyme B play a critical role in controlling lung inflammation during acute viral infection. Mucosal Immunol. 2012, 5, 161–172. [Google Scholar] [CrossRef] [PubMed]
  293. Shao, H.Y.; Huang, J.Y.; Lin, Y.W.; Yu, S.L.; Chitra, E.; Chang, C.K.; Sung, W.C.; Chong, P.; Chow, Y.H. Depletion of regulatory T-cells leads to moderate B-cell antigenicity in respiratory syncytial virus infection. Int. J. Infect. Dis. 2015, 41, 56–64. [Google Scholar] [CrossRef]
  294. Loebbermann, J.; Schnoeller, C.; Thornton, H.; Durant, L.; Sweeney, N.P.; Schuijs, M.; O’Garra, A.; Johansson, C.; Openshaw, P.J. IL-10 regulates viral lung immunopathology during acute respiratory syncytial virus infection in mice. PLoS ONE 2012, 7, e32371. [Google Scholar] [CrossRef] [PubMed]
  295. Sun, J.; Cardani, A.; Sharma, A.K.; Laubach, V.E.; Jack, R.S.; Muller, W.; Braciale, T.J. Autocrine regulation of pulmonary inflammation by effector T-cell derived IL-10 during infection with respiratory syncytial virus. PLoS Pathog. 2011, 7, e1002173. [Google Scholar] [CrossRef]
  296. Christiaansen, A.F.; Syed, M.A.; Ten Eyck, P.P.; Hartwig, S.M.; Durairaj, L.; Kamath, S.S.; Varga, S.M. Altered Treg and cytokine responses in RSV-infected infants. Pediatr. Res. 2016, 80, 702–709. [Google Scholar] [CrossRef]
  297. Raiden, S.; Pandolfi, J.; Payaslian, F.; Anderson, M.; Rivarola, N.; Ferrero, F.; Urtasun, M.; Fainboim, L.; Geffner, J.; Arruvito, L. Depletion of circulating regulatory T cells during severe respiratory syncytial virus infection in young children. Am. J. Respir. Crit. Care Med. 2014, 189, 865–868. [Google Scholar] [CrossRef]
  298. Krishnamoorthy, N.; Khare, A.; Oriss, T.B.; Raundhal, M.; Morse, C.; Yarlagadda, M.; Wenzel, S.E.; Moore, M.L.; Peebles, R.S., Jr.; Ray, A.; et al. Early infection with respiratory syncytial virus impairs regulatory T cell function and increases susceptibility to allergic asthma. Nat. Med. 2012, 18, 1525–1530. [Google Scholar] [CrossRef]
  299. Badovinac, V.P.; Porter, B.B.; Harty, J.T. CD8+ T cell contraction is controlled by early inflammation. Nat. Immunol. 2004, 5, 809–817. [Google Scholar] [CrossRef]
  300. Mahnke, Y.D.; Brodie, T.M.; Sallusto, F.; Roederer, M.; Lugli, E. The who’s who of T-cell differentiation: Human memory T-cell subsets. Eur. J. Immunol. 2013, 43, 2797–2809. [Google Scholar] [CrossRef]
  301. Turner, D.L.; Bickham, K.L.; Thome, J.J.; Kim, C.Y.; D’Ovidio, F.; Wherry, E.J.; Farber, D.L. Lung niches for the generation and maintenance of tissue-resident memory T cells. Mucosal Immunol. 2014, 7, 501–510. [Google Scholar] [CrossRef]
  302. Zheng, M.Z.M.; Wakim, L.M. Tissue resident memory T cells in the respiratory tract. Mucosal Immunol. 2022, 15, 379–388. [Google Scholar] [CrossRef] [PubMed]
  303. De Bree, G.J.; Heidema, J.; van Leeuwen, E.M.M.; van Bleek, G.M.; Jonkers, R.E.; Jansen, H.M.; van Lier, R.A.W.; Out, T.A. Respiratory syncytial virus-specific CD8+ memory T cell responses in elderly persons. J. Infect. Dis. 2005, 191, 1710–1718. [Google Scholar] [CrossRef] [PubMed]
  304. Varese, A.; Nakawesi, J.; Farias, A.; Kirsebom, F.C.M.; Paulsen, M.; Nuriev, R.; Johansson, C. Type I interferons and MAVS signaling are necessary for tissue resident memory CD8+ T cell responses to RSV infection. PLoS Pathog. 2022, 18, e1010272. [Google Scholar] [CrossRef] [PubMed]
  305. Heidema, J.; Rossen, J.W.A.; Lukens, M.V.; Ketel, M.S.; Scheltens, E.; Kranendonk, M.E.G.; van Maren, W.W.C.; van Loon, A.M.; Otten, H.G.; Kimpen, J.L.L.; et al. Dynamics of human respiratory virus-specific CD8+ T cell responses in blood and airways during episodes of common cold. J. Immunol. 2008, 181, 5551–5559. [Google Scholar] [CrossRef] [PubMed]
  306. Luangrath, M.A.; Schmidt, M.E.; Hartwig, S.M.; Varga, S.M. Tissue-Resident Memory T Cells in the Lungs Protect against Acute Respiratory Syncytial Virus Infection. Immunohorizons 2021, 5, 59–69. [Google Scholar] [CrossRef] [PubMed]
  307. Chang, J.; Braciale, T.J. Respiratory syncytial virus infection suppresses lung CD8+ Tcell effector activity and peripheral CD8+ T-cell memory in the respiratory tract. Nat. Med. 2002, 8, 54–60. [Google Scholar] [CrossRef]
  308. Schmidt, M.E.; Knudson, C.J.; Hartwig, S.M.; Pewe, L.L.; Meyerholz, D.K.; Langlois, R.A.; Harty, J.T.; Varga, S.M. Memory CD8 T cells mediate severe immunopathology following respiratory syncytial virus infection. PLoS Pathog. 2018, 14, e1006810. [Google Scholar] [CrossRef]
  309. Chirkova, T.; Rosas-Salazar, C.; Gebretsadik, T.; Jadhao, S.J.; Chappell, J.D.; Peebles, R.S., Jr.; Dupont, W.D.; Newcomb, D.C.; Berdnikovs, S.; Gergen, P.J.; et al. Effect of Infant RSV Infection on Memory T Cell Responses at Age 2-3 Years. Front. Immunol. 2022, 13, 826666. [Google Scholar] [CrossRef]
  310. Zens, K.D.; Chen, J.K.; Guyer, R.S.; Wu, F.L.; Cvetkovski, F.; Miron, M.; Farber, D.L. Reduced generation of lung tissue-resident memory T cells during infancy. J. Exp. Med. 2017, 214, 2915–2932. [Google Scholar] [CrossRef]
  311. Chanock, R.; Roizman, B.; Myers, R. Recovery from infants with respiratory illness of a virus related to chimpanzee coryza agent (CCA). I. Isolation, properties and characterization. Am. J. Hyg. 1957, 66, 281–290. [Google Scholar] [CrossRef] [PubMed]
  312. Blount, R.E.J.; Morris, J.A.; Savage, R.E. Recovery of cytopathogenic agent from chimpanzees with coryza. Proc. Soc. Exp. Biol. Med. 1956, 92, 544–594. [Google Scholar] [CrossRef] [PubMed]
  313. Crank, M.C.; Ruckwardt, T.J.; Chen, M.; Morabito, K.M.; Phung, E.; Costner, P.J.; Holman, L.A.; Hickman, S.P.; Berkowitz, N.M.; Gordon, I.J.; et al. A proof of concept for structure-based vaccine design targeting RSV in humans. Science 2019, 365, 505–509. [Google Scholar] [CrossRef] [PubMed]
  314. Ura, T.; Okuda, K.; Shimada, M. Developments in Viral Vector-Based Vaccines. Vaccines 2014, 2, 624–641. [Google Scholar] [CrossRef]
  315. Jordan, E.; Lawrence, S.J.; Meyer, T.P.H.; Schmidt, D.; Schultz, S.; Mueller, J.; Stroukova, D.; Koenen, B.; Gruenert, R.; Silbernagl, G.; et al. Broad Antibody and Cellular Immune Response From a Phase 2 Clinical Trial With a Novel Multivalent Poxvirus-Based Respiratory Syncytial Virus Vaccine. J. Infect. Dis. 2021, 223, 1062–1072. [Google Scholar] [CrossRef]
  316. Samy, N.; Reichhardt, D.; Schmidt, D.; Chen, L.M.; Silbernagl, G.; Vidojkovic, S.; Meyer, T.P.; Jordan, E.; Adams, T.; Weidenthaler, H.; et al. Safety and immunogenicity of novel modified vaccinia Ankara-vectored RSV vaccine: A randomized phase I clinical trial. Vaccine 2020, 38, 2608–2619. [Google Scholar] [CrossRef]
  317. Falsey, A.R.; Williams, K.; Gymnopoulou, E.; Bart, S.; Ervin, J.; Bastian, A.R.; Menten, J.; De Paepe, E.; Vandenberghe, S.; Chan, E.K.H.; et al. Efficacy and Safety of an Ad26.RSV.preF-RSV preF Protein Vaccine in Older Adults. N. Engl. J. Med. 2023, 388, 609–620. [Google Scholar] [CrossRef]
  318. Stuart, A.S.V.; Virta, M.; Williams, K.; Seppa, I.; Hartvickson, R.; Greenland, M.; Omoruyi, E.; Bastian, A.R.; Haazen, W.; Salisch, N.; et al. Phase 1/2a Safety and Immunogenicity of an Adenovirus 26 Vector Respiratory Syncytial Virus (RSV) Vaccine Encoding Prefusion F in Adults 18-50 Years and RSV-Seropositive Children 12-24 Months. J. Infect. Dis. 2022, 227, 71–82. [Google Scholar] [CrossRef]
  319. Sadoff, J.; De Paepe, E.; DeVincenzo, J.; Gymnopoulou, E.; Menten, J.; Murray, B.; Rosemary Bastian, A.; Vandebosch, A.; Haazen, W.; Noulin, N.; et al. Prevention of Respiratory Syncytial Virus Infection in Healthy Adults by a Single Immunization of Ad26.RSV.preF in a Human Challenge Study. J. Infect. Dis. 2022, 226, 396–406. [Google Scholar] [CrossRef]
  320. Baxter, D. Active and passive immunity, vaccine types, excipients and licensing. Occup. Med. 2007, 57, 552–556. [Google Scholar] [CrossRef]
  321. Walsh, E.E.; Falsey, A.R.; Scott, D.A.; Gurtman, A.; Zareba, A.M.; Jansen, K.U.; Gruber, W.C.; Dormitzer, P.R.; Swanson, K.A.; Radley, D.; et al. A Randomized Phase 1/2 Study of a Respiratory Syncytial Virus Prefusion F Vaccine. J. Infect. Dis. 2022, 225, 1357–1366. [Google Scholar] [CrossRef] [PubMed]
  322. Falsey, A.R.; Walsh, E.E.; Scott, D.A.; Gurtman, A.; Zareba, A.; Jansen, K.U.; Gruber, W.C.; Dormitzer, P.R.; Swanson, K.A.; Jiang, Q.; et al. Phase 1/2 Randomized Study of the Immunogenicity, Safety, and Tolerability of a Respiratory Syncytial Virus Prefusion F Vaccine in Adults With Concomitant Inactivated Influenza Vaccine. J. Infect. Dis. 2022, 225, 2056–2066. [Google Scholar] [CrossRef] [PubMed]
  323. Schmoele-Thoma, B.; Zareba, A.M.; Jiang, Q.; Maddur, M.S.; Danaf, R.; Mann, A.; Eze, K.; Fok-Seang, J.; Kabir, G.; Catchpole, A.; et al. Vaccine Efficacy in Adults in a Respiratory Syncytial Virus Challenge Study. N. Engl. J. Med. 2022, 386, 2377–2386. [Google Scholar] [CrossRef] [PubMed]
  324. Walsh, E.E.; Pérez Marc, G.; Zareba, A.M.; Falsey, A.R.; Jiang, Q.; Patton, M.; Polack, F.P.; Llapur, C.; Doreski, P.A.; Ilangovan, K.; et al. Efficacy and Safety of a Bivalent RSV Prefusion F Vaccine in Older Adults. N. Engl. J. Med. 2023, 388, 1465–1477. [Google Scholar] [CrossRef]
  325. Kampmann, B.; Madhi, S.A.; Munjal, I.; Simões, E.A.F.; Pahud, B.A.; Llapur, C.; Baker, J.; Pérez Marc, G.; Radley, D.; Shittu, E.; et al. Bivalent Prefusion F Vaccine in Pregnancy to Prevent RSV Illness in Infants. N. Engl. J. Med. 2023, 388, 1451–1464. [Google Scholar] [CrossRef]
  326. Simoes, E.A.F. Respiratory Syncytial Virus Disease in Young Children and Older Adults in Europe: A Burden and Economic Perspective. J. Infect. Dis. 2022, 226, S1–S9. [Google Scholar] [CrossRef]
  327. Bebia, Z.; Reyes, O.; Jeanfreau, R.; Kantele, A.; De Leon, R.G.; Garcia Sanchez, M.; Banooni, P.; Gardener, G.J.; Bartha Rasero, J.L.; Encinas Pardilla, M.B.; et al. Safety and immunogenicity of an investigational respiratory syncytial virus vaccine (RSVPreF3) in mothers and their infants: A phase 2 randomized trial. J. Infect. Dis. 2023, 228, 299–310. [Google Scholar] [CrossRef]
  328. Papi, A.; Ison, M.G.; Langley, J.M.; Lee, D.G.; Leroux-Roels, I.; Martinon-Torres, F.; Schwarz, T.F.; van Zyl-Smit, R.N.; Campora, L.; Dezutter, N.; et al. Respiratory Syncytial Virus Prefusion F Protein Vaccine in Older Adults. N. Engl. J. Med. 2023, 388, 595–608. [Google Scholar] [CrossRef]
  329. Gebre, M.S.; Brito, L.A.; Tostanoski, L.H.; Edwards, D.K.; Carfi, A.; Barouch, D.H. Novel approaches for vaccine development. Cell 2021, 184, 1589–1603. [Google Scholar] [CrossRef]
  330. Chen, G.L.; Mithani, R.; Kapoor, A.; Lu, S.; El Asmar, L.; Panozzo, C.A.; Shaw, C.A.; Stoszek, S.K.; August, A. 234. Safety and Immunogenicity of mRNA-1345, an mRNA-Based RSV Vaccine in Younger and Older Adult Cohorts: Results from a Phase 1, Randomized Clinical Trial. Open Forum Infect. Dis. 2022, 9, ofac492.312. [Google Scholar] [CrossRef]
  331. Rodriguez-Fernandez, R.; Mejias, A.; Ramilo, O. Monoclonal Antibodies for Prevention of Respiratory Syncytial Virus Infection. Pediatr. Infect. Dis. J. 2021, 40, S35–S39. [Google Scholar] [CrossRef] [PubMed]
  332. Gilman, M.S.; Castellanos, C.A.; Chen, M.; Ngwuta, J.O.; Goodwin, E.; Moin, S.M.; Mas, V.; Melero, J.A.; Wright, P.F.; Graham, B.S.; et al. Rapid profiling of RSV antibody repertoires from the memory B cells of naturally infected adult donors. Sci. Immunol. 2016, 1, eaaj1879. [Google Scholar] [CrossRef] [PubMed]
  333. McLellan, J.S.; Chen, M.; Joyce, M.G.; Sastry, M.; Stewart-Jones, G.B.; Yang, Y.; Zhang, B.; Chen, L.; Srivatsan, S.; Zheng, A.; et al. Structure-based design of a fusion glycoprotein vaccine for respiratory syncytial virus. Science 2013, 342, 592–598. [Google Scholar] [CrossRef] [PubMed]
  334. American Academy of Pediatrics Committee on Infectious Diseases; American Academy of Pediatrics Bronchiolitis Guidelines Committee. Updated Guidance for Palivizumab Prophylaxis Among Infants and Young Children at Increased Risk of Hospitalization for Respiratory Syncytial Virus Infection. Pediatrics 2014, 134, 415–420. [Google Scholar] [CrossRef]
  335. Tulloh, R.M.; Feltes, T.F. The European Forum for Clinical Management: Prophylaxis against the respiratory syncytial virus in infants and young children with congenital cardiac disease. Cardiol. Young 2005, 15, 274–278. [Google Scholar] [CrossRef]
  336. Zhu, Q.; McLellan, J.S.; Kallewaard, N.L.; Ulbrandt, N.D.; Palaszynski, S.; Zhang, J.; Moldt, B.; Khan, A.; Svabek, C.; McAuliffe, J.M.; et al. A highly potent extended half-life antibody as a potential RSV vaccine surrogate for all infants. Sci. Transl. Med. 2017, 9, eaaj1928. [Google Scholar] [CrossRef]
  337. Hammitt, L.L.; Dagan, R.; Yuan, Y.; Baca Cots, M.; Bosheva, M.; Madhi, S.A.; Muller, W.J.; Zar, H.J.; Brooks, D.; Grenham, A.; et al. Nirsevimab for Prevention of RSV in Healthy Late-Preterm and Term Infants. N. Engl. J. Med. 2022, 386, 837–846. [Google Scholar] [CrossRef]
  338. Griffin, M.P.; Yuan, Y.; Takas, T.; Domachowske, J.B.; Madhi, S.A.; Manzoni, P.; Simoes, E.A.F.; Esser, M.T.; Khan, A.A.; Dubovsky, F.; et al. Single-Dose Nirsevimab for Prevention of RSV in Preterm Infants. N. Engl. J. Med. 2020, 383, 415–425. [Google Scholar] [CrossRef]
  339. Lin, G.L.; Drysdale, S.B.; Snape, M.D.; O’Connor, D.; Brown, A.; MacIntyre-Cockett, G.; Mellado-Gomez, E.; de Cesare, M.; Bonsall, D.; Ansari, M.A.; et al. Distinct patterns of within-host virus populations between two subgroups of human respiratory syncytial virus. Nat. Commun. 2021, 12, 5125. [Google Scholar] [CrossRef]
Figure 1. Overview of the intracellular mechanisms in airway epithelial cells (AECs) after respiratory syncytial virus (RSV) entry. RSV enters ciliated AECs by binding to receptors on the surface of the cells (e.g., CX3CR1), followed by internalization of the virus particle into the cytoplasm. Following infection, intracellular pattern recognition receptors, like RIG-I or TLR3, sense viral ssRNA or dsRNA intermediates resulting in the activation of transcription factors like NK-κB or IRF3. Nuclear translocation of transcription factors leads to the expression of several antiviral mediators, like cytokines, chemokines, and receptors for antigen presentation. RIG-I: retinoic-acid-inducible protein I; TLR3: toll-like receptor 3; IRF3: interferon regulatory factor 3; IFN: interferon; TNF-α: tumor necrosis factor α; IL: interleukin; MHC-I: major histocompatibility complex I. Created with BioRender.com.
Figure 1. Overview of the intracellular mechanisms in airway epithelial cells (AECs) after respiratory syncytial virus (RSV) entry. RSV enters ciliated AECs by binding to receptors on the surface of the cells (e.g., CX3CR1), followed by internalization of the virus particle into the cytoplasm. Following infection, intracellular pattern recognition receptors, like RIG-I or TLR3, sense viral ssRNA or dsRNA intermediates resulting in the activation of transcription factors like NK-κB or IRF3. Nuclear translocation of transcription factors leads to the expression of several antiviral mediators, like cytokines, chemokines, and receptors for antigen presentation. RIG-I: retinoic-acid-inducible protein I; TLR3: toll-like receptor 3; IRF3: interferon regulatory factor 3; IFN: interferon; TNF-α: tumor necrosis factor α; IL: interleukin; MHC-I: major histocompatibility complex I. Created with BioRender.com.
Viruses 15 01999 g001
Figure 2. Schematic representation of the antiviral responses of innate and adaptive immune cells during RSV infection. Innate immune cells mediate the control of infection by direct interaction with the infected AECs, followed by their phagocytosis, or by direct interaction with virus particles, leading to the neutralization of the virus, e.g., by NETosis. Non-professional and professional antigen-presenting cells are responsible for the activation and recruitment of adaptive immune cells to the site of infection to further drive viral clearance. Regulation of immune responses is thereby crucial to avoid exaggerated, potentially immunopathogenic responses. IL: interleukin; IFN: interferon; TNF: tumor necrosis factor; GrB: granzyme B; BAFF: B cell-activating factor; APRIL: A proliferation-inducing ligand; MHC: major histocompatibility complex; MMP9: matrix metalloprotease 9. Created with BioRender.com.
Figure 2. Schematic representation of the antiviral responses of innate and adaptive immune cells during RSV infection. Innate immune cells mediate the control of infection by direct interaction with the infected AECs, followed by their phagocytosis, or by direct interaction with virus particles, leading to the neutralization of the virus, e.g., by NETosis. Non-professional and professional antigen-presenting cells are responsible for the activation and recruitment of adaptive immune cells to the site of infection to further drive viral clearance. Regulation of immune responses is thereby crucial to avoid exaggerated, potentially immunopathogenic responses. IL: interleukin; IFN: interferon; TNF: tumor necrosis factor; GrB: granzyme B; BAFF: B cell-activating factor; APRIL: A proliferation-inducing ligand; MHC: major histocompatibility complex; MMP9: matrix metalloprotease 9. Created with BioRender.com.
Viruses 15 01999 g002
Figure 3. Overview of immunopathogenic responses mediated by innate and adaptive immune cells during RSV infection. Changes in the innate and adaptive immune response can be a consequence of microenvironmental changes, genetic predispositions, or interaction with the virus. RSV can directly infect a variety of immune cells or indirectly alter their immune response, e.g., by the secretion of soluble G, characterized by phenotype changes in immune cells, or the excessive secretion of immunosuppressive or proinflammatory cytokines. TNF-α: tumor necrosis factor α; IFN: interferon; IL: interleukin; ECP: eosinophil cationic protein; MIP-1α: macrophage inflammatory protein-1α; MPO: myeloperoxidase. Created with BioRender.com.
Figure 3. Overview of immunopathogenic responses mediated by innate and adaptive immune cells during RSV infection. Changes in the innate and adaptive immune response can be a consequence of microenvironmental changes, genetic predispositions, or interaction with the virus. RSV can directly infect a variety of immune cells or indirectly alter their immune response, e.g., by the secretion of soluble G, characterized by phenotype changes in immune cells, or the excessive secretion of immunosuppressive or proinflammatory cytokines. TNF-α: tumor necrosis factor α; IFN: interferon; IL: interleukin; ECP: eosinophil cationic protein; MIP-1α: macrophage inflammatory protein-1α; MPO: myeloperoxidase. Created with BioRender.com.
Viruses 15 01999 g003
Figure 4. Schematic overview of current vaccines and monoclonal antibody-based treatment options available for RSV. The mentioned vaccines and monoclonal antibodies are either licensed or currently in phase III clinical trials for the treatment of patients at risk. Created with BioRender.com.
Figure 4. Schematic overview of current vaccines and monoclonal antibody-based treatment options available for RSV. The mentioned vaccines and monoclonal antibodies are either licensed or currently in phase III clinical trials for the treatment of patients at risk. Created with BioRender.com.
Viruses 15 01999 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Agac, A.; Kolbe, S.M.; Ludlow, M.; Osterhaus, A.D.M.E.; Meineke, R.; Rimmelzwaan, G.F. Host Responses to Respiratory Syncytial Virus Infection. Viruses 2023, 15, 1999. https://doi.org/10.3390/v15101999

AMA Style

Agac A, Kolbe SM, Ludlow M, Osterhaus ADME, Meineke R, Rimmelzwaan GF. Host Responses to Respiratory Syncytial Virus Infection. Viruses. 2023; 15(10):1999. https://doi.org/10.3390/v15101999

Chicago/Turabian Style

Agac, Ayse, Sophie M. Kolbe, Martin Ludlow, Albert D. M. E. Osterhaus, Robert Meineke, and Guus F. Rimmelzwaan. 2023. "Host Responses to Respiratory Syncytial Virus Infection" Viruses 15, no. 10: 1999. https://doi.org/10.3390/v15101999

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop