key: cord-1016789-q24cqjer authors: Hwang, Hye Suk; Lee, Young-Tae; Kim, Ki-Hye; Seo, Ho Seong; Yang, Kap Seung; Cho, Hoonsung; Kang, Sang-Moo title: Roles of the Fc Receptor γ-Chain in Inducing Protective Immune Responses after Heterologous Vaccination against Respiratory Syncytial Virus Infection date: 2021-03-08 journal: Vaccines (Basel) DOI: 10.3390/vaccines9030232 sha: 385c46dd72f3f7a6d76abaa906afecf889a29810 doc_id: 1016789 cord_uid: q24cqjer The roles of the Fc receptor (FcR) in protection or inflammatory disease after respiratory syncytial virus (RSV) vaccination and infection remain unknown. Virus-like particles containing RSV fusion proteins (RSV F-VLPs) induce T-helper type 1 antibody responses and protection against RSV. Heterologous RSV F-VLP prime and formalin-inactivated RSV (FI-RSV) boost vaccination has been reported to be effective in providing protection without inflammatory disease. Here, we investigated whether the FcRγ-chain is important for immune protection by the heterologous F-VLP and FI-RSV vaccination using FcRγ-chain knockout (−/−) mice. RSV F-VLP-primed and FI-RSV-boosted FcRγ −/− mice displayed less protective efficacy, as shown by higher lung viral titers upon RSV challenge, compared to RSV F-VLP-primed and FI-RSV-boosted immunized wild-type mice. RSV F-VLP and FI-RSV immunization induced lower levels of neutralizing activity and interferon-γ-producing CD8 T-cells in the bronchoalveolar lavage cells of FcRγ −/− mice than in those of wild-type mice. In addition, FcRγ −/− mice displayed a trend of enhancing lung histopathology after RSV vaccination and infection. This study suggests that the FcRγ-chain plays an important role in inducing antiviral protection and CD8 T-cell responses in RSV F-VLP prime and FI-RSV boost vaccination after RSV infections. Respiratory syncytial virus (RSV) is a pathogen that is the leading cause of severe respiratory disease in infants, children, and the elderly. Despite decades of research, there is no licensed RSV vaccine. Formalin-inactivated RSV (FI-RSV) vaccines have failed because of vaccine-enhanced respiratory disease (ERD) in young vaccinee children after exposure to RSV infection during the following epidemic season [1, 2] . Virus-like particles (VLPs) are a unique delivery platform for virus structural proteins, mimicking viral morphology without genetic materials, and their advantages lie in their safety, immunogenicity, and antigen stability [3] . VLP vaccine platforms have been reported to avoid ERD in preclinical studies. VLP-based RSV vaccines assemble with the human metapneumovirus (hMPV) matrix protein (M) as the structural scaffold and the RSV fusion glycoprotein (F) in either post-fusion or pre-fusion conformation-induced T-helper type 1 (Th1)-mediated immune Influenza virus matrix-protein-derived RSV F-VLP was prepared as previously described [25] . A F-VLP containing RSV A2 fusion (F) protein was produced in insect cells using the recombinant baculovirus expression system as previously described. In brief, Sf9 insect cells were infected with recombinant baculoviruses expressing the influenza virus M1 matrix core protein and RSV A2 fusion (F) protein. Insect cell-culture supernatants containing released RSV F-VLP were harvested, cell debris was removed by low-speed Vaccines 2021, 9, 232 3 of 12 centrifugation, and finally, the RSV F-VLP vaccine was purified using sucrose-gradient ultracentrifugation. For FI-RSV, RSV was inactivated with formalin (1:4000 (vol/vol)) for 72 h at 37 • C. FI-RSV was pelleted by ultracentrifugation (100,800× g, 1 h) at 4 • C, resuspended in serum-free medium, and then adsorbed to an alum adjuvant (4 mg/mL, aluminum hydroxide, Sigma Aldrich) for use in the FI-RSV vaccination in this study [25] . BALB/c wild-type (WT) and FcRγ-deficient mice (FcRγ −/− encoded by Fcer1g on the BALB/c genetic background) were purchased (Taconic Farms, Hudson, NY) and maintained in the animal facility at Georgia State University (GSU). For immunization, 6-8-week-old female BALB/c WT mice (n = 5 mice per group) and FcRγ −/− mice (n = 5 per group) were intramuscularly immunized with the F-VLP (10 µg) prime and the FI-RSV (2 µg) + alum adjuvant (20 µg) boost for the heterologous (F-VLP/FI-RSV) group at weeks 0 and 4. Naïve control and immunized mice were infected with 0.5 × 10 6 plaque-forming units (PFU) of RSV A2 under isoflurane anesthesia 4 weeks after boost immunization. F-protein-specific antibody titers were determined in prime and boost immune sera using enzyme-linked immunosorbent assay (ELISA) with purified RSV F-protein (100 ng/mL, BEI resources) as a coating antigen [10] . Five hundred times-diluted prime or boost sera were added and incubated for 1.5 h at 37 • C; horseradish-peroxidase-conjugated goat anti-mouse IgG, IgG1, and IgG2a (Southern Biotechnology, Birmingham, AL) were used as secondary antibodies. To determine the RSV-neutralizing activity of immune sera, RSV expressing the red fluorescent monomeric Katushka 2 protein (A2-K-line19F) was used as previously described [25] . Briefly, 500 PFU of live A2-K-line19F RSV and complementinactivated immune sera at 56 • C were mixed for 30 min, added to the confluent HEp-2 cell monolayer plates, adsorbed for 2 h at 37 • C, and then the antibody-virus mixture was removed. The mean percentages of fluorescence reduction by sera from vaccinated mice and sera from naïve controls were determined and compared [26] . The bronchoalveolar lavage (BAL) fluid (BALF) of individual mice was collected by infusing 1 mL of PBS into the lungs via the trachea using a 25-gauge catheter at day 5 post-challenge. RSV titers were determined in the lung samples [10] . Briefly, serially diluted lung homogenates were added to the confluent HEp-2 cell monolayer plates, adsorbed for 2 h at 37 • C, and then incubated at 37 • C for 3 days. After fixation with 5% formaldehyde, the plate was developed using a mouse anti-RSV F-monoclonal antibody and HRP-conjugated anti-mouse IgG antibody using a 3,3 -diaminobenzidine tetrahydrochloride (DAB) substrate (Invitrogen). The detection limit of the viral titer from the lung samples was 50 PFU. For cell phenotype analysis in the mucosal compartment, BAL cells from individual mice (n = 5 per group) were prepared at day 5 post-challenge. BAL cells were stimulated with 4 µg/mL RSV peptides [25, 27, 28] , F 92-106 (ELQLLMQSTPATNNR), for CD8 T-cells for 5 h prior to assessing IFN-γ, TNF-α, or IL-4 cytokine-producing cells. Intracellular cytokines and surface markers for T-cells were stained with antibodies for IFN-γ, IL-4 (eBioscience), TNF-α (BioLegend), CD45, CD3, CD4, CD8, DX5, CD11c, and B220 (BD Biosciences). Stained cells were acquired on a FACSCanto flow cytometer (BD Biosciences) and analyzed using FlowJo software (Tree Star, Inc., Ashland, OR, USA). Lung tissues collected from mice at 5 days after RSV challenge were fixed with 10% neutral buffered formalin. Lung tissue histology was performed by staining with hematoxylin and eosin (H&E), periodic acid-Schiff (PAS), and hematoxylin and congo red (H&CR) and analyzed under light microscopy [10, 28, 29] . The sections around the airways were scored for the degree of inflammation according to the following scale: 0 Vaccines 2021, 9, 232 4 of 12 (none), 1 (mild), 2 (moderate), and 3 (severe) [30] . The areas of epithelium and PAS-positive areas within the airway epithelium were annotated using the magnetic lasso tool of Adobe Photoshop CS5.1 software. The degrees of pulmonary eosinophilia were identified by H&CR staining to enumerate eosinophils and were expressed as the number of eosinophils present per 400X field. All results are represented as the mean ± standard error of the mean (SEM). Significant differences among treatments were investigated by one-way analysis of variance (ANOVA), using Tukey's or Dunnett's multiple-comparison test, or two-way ANOVA with the Bonferroni post-test in GraphPad Prism version 5 (GraphPad Software, Inc., San Diego, CA, USA). p-Values ≤ 0.05 were considered statistically significant. To evaluate the possible roles of FcRγ in inducing protective immune responses to RSV, the groups of WT and FcRγ −/− mice were intramuscularly immunized via a heterologous F-VLP prime and FI-RSV boost regimen. As shown in Figure 1 , immunization of FcRγ −/− mice induced similar or slightly higher levels of RSV F-specific antibodies compared to those in WT mice at 3 weeks after the prime and boost vaccination ( Figure 1A ,B). Similarly, the F-VLP prime immunization of WT and FcRγ −/− mice elicited an IgG2a isotype-dominant response ( Figure 1A ,C) although IgG2a/IgG1 isotype ratios were slightly decreased in FcRγ −/− mice ( Figure 1C ). These results suggest that FcRγ −/− mice do not have a defect in inducing IgG antibody responses. Lung tissues collected from mice at 5 days after RSV challenge were fixed with 10% neutral buffered formalin. Lung tissue histology was performed by staining with hematoxylin and eosin (H&E), periodic acid-Schiff (PAS), and hematoxylin and congo red (H&CR) and analyzed under light microscopy [10, 28, 29] . The sections around the airways were scored for the degree of inflammation according to the following scale: 0 (none), 1 (mild), 2 (moderate), and 3 (severe) [30] . The areas of epithelium and PAS-positive areas within the airway epithelium were annotated using the magnetic lasso tool of Adobe Photoshop CS5.1 software. The degrees of pulmonary eosinophilia were identified by H&CR staining to enumerate eosinophils and were expressed as the number of eosinophils present per 400X field. All results are represented as the mean ± standard error of the mean (SEM). Significant differences among treatments were investigated by one-way analysis of variance (ANOVA), using Tukey's or Dunnett's multiple-comparison test, or two-way ANOVA with the Bonferroni post-test in GraphPad Prism version 5 (GraphPad Software, Inc., San Diego, CA, USA). p-Values ≤ 0.05 were considered statistically significant. To evaluate the possible roles of FcRγ in inducing protective immune responses to RSV, the groups of WT and FcRγ −/− mice were intramuscularly immunized via a heterologous F-VLP prime and FI-RSV boost regimen. As shown in Figure 1 , immunization of FcRγ −/− mice induced similar or slightly higher levels of RSV F-specific antibodies compared to those in WT mice at 3 weeks after the prime and boost vaccination ( Figure 1A ,B). Similarly, the F-VLP prime immunization of WT and FcRγ −/− mice elicited an IgG2a isotype-dominant response ( Figure 1A ,C) although IgG2a/IgG1 isotype ratios were slightly decreased in FcRγ −/− mice ( Figure 1C ). These results suggest that FcRγ −/− mice do not have a defect in inducing IgG antibody responses. To evaluate the possible roles of FcRγ in inducing virus-neutralizing antibodies by the RSV F-VLP prime and FI-RSV boost, serum samples collected at 2 weeks after the boost were tested for neutralizing activity. The representative fluorescence microscope images at 400× serum dilution shown in Figure 2A ,B show that both WT and FcRγ −/− mice with F-VLP/FI-RSV vaccination induced similar neutralizing activity at low 200x dilution of sera. At 1:400 dilution, antisera of FcRγ −/− mice showed a 26% reduction in fluorescence intensity versus a 75% reduction within antisera from WT mice after F-VLP/FI-RSV vaccination. At 1:800 dilution, antisera of the RSV F-VLP/FI-RSV immunization in WT mice showed a 23% reduction in fluorescence intensity. However, no fluorescence reduction was observed in antisera from FcRγ −/− mice. These results indicate that FcRγ plays a role in inducing neutralizing antibodies after heterologous RSV F-VLP/FI-RSV vaccination. the RSV F-VLP prime and FI-RSV boost, serum samples collected at 2 weeks after the boost were tested for neutralizing activity. The representative fluorescence microscope images at 400× serum dilution shown in Figure 2A ,B show that both WT and FcRγ −/− mice with F-VLP/FI-RSV vaccination induced similar neutralizing activity at low 200x dilution of sera. At 1:400 dilution, antisera of FcRγ −/− mice showed a 26% reduction in fluorescence intensity versus a 75% reduction within antisera from WT mice after F-VLP/FI-RSV vaccination. At 1:800 dilution, antisera of the RSV F-VLP/FI-RSV immunization in WT mice showed a 23% reduction in fluorescence intensity. However, no fluorescence reduction was observed in antisera from FcRγ −/− mice. These results indicate that FcRγ plays a role in inducing neutralizing antibodies after heterologous RSV F-VLP/FI-RSV vaccination. Naïve and immunized mice were challenged with live RSV A2 virus (0.5 × 10 6 PFU/mouse) 4 weeks after the boost, and lung viral loads at day 5 post-challenge were determined. FcRγ −/− naïve mice exhibited higher lung viral titers of RSV (3.5 log10) than WT mice (Figure 3 ). Consistent with the results of neutralizing activity, F-VLP/FI-RSVvaccinated WT mice controlled RSV lung viral loads more effectively to a level below the detection limit, compared to FcRγ −/− mice which displayed substantial levels of lung viral loads ( Figure 3 ). These results indicate that FcRγ plays a role in lung viral clearance after RSV infection in F-VLP/FI-RSV-immune mice. Naïve and immunized mice were challenged with live RSV A2 virus (0.5 × 10 6 PFU/mouse) 4 weeks after the boost, and lung viral loads at day 5 post-challenge were determined. FcRγ −/− naïve mice exhibited higher lung viral titers of RSV (3.5 log10) than WT mice ( Figure 3) . Consistent with the results of neutralizing activity, F-VLP/FI-RSV-vaccinated WT mice controlled RSV lung viral loads more effectively to a level below the detection limit, compared to FcRγ −/− mice which displayed substantial levels of lung viral loads ( Figure 3) . These results indicate that FcRγ plays a role in lung viral clearance after RSV infection in F-VLP/FI-RSV-immune mice. In addition to the potential necessity of neutralizing antibodies against live RSV infection, T-cell immune responses could contribute to the protection against RSV infection [23] . Therefore, we determined the induction of RSV F-specific CD8 T-cells by intracellular In addition to the potential necessity of neutralizing antibodies against live RSV infection, T-cell immune responses could contribute to the protection against RSV infection [23] . Therefore, we determined the induction of RSV F-specific CD8 T-cells by intracellular cytokine staining of BAL cells at day 5 post-challenge (Figure 4) . IFN-γ-TNF-α+ CD8 T-cells and IFN-γ-IL-4+ CD8 T-cells were detected at similarly low levels in BALF from both WT and FcRγ −/− mice with the F-VLP/FI-RSV vaccination after RSV challenge. Double positive IFN-γ+TNF-α+ CD8 T-cells were induced at higher levels in WT BALF than in FcRγ −/− BALF samples ( Figure 4A,B) . Double-positive IFN-γ+IL-4+ CD8 T-cells were not detected in the non-immunized groups. IFN-γ+TNF-α-and IFN-γ+IL4-CD8 T-cells were observed at higher levels in BAL cells from WT mice than those from FcRγ −/− mice with F-VLP/FI-RSV immunization ( Figure 4A-D) . Therefore, these results suggest that FcRγ plays a pivotal role in inducing effector CD8 T-cell responses, which might contribute to protection. . FcRγ −/− mice were less effective in clearing lung viral titers than WT mice after RSV infection. F-VLP/FI-RSV-immunized mice were challenged with RSV (0.5 × 10 6 PFU/mouse) at 4 weeks after boost vaccination. Lung viral loads in each mouse (PFU/mouse lung) were determined at 5 days after infection by an immuno-plaque assay. Naïve inf.: unimmunized naïve mice after RSV. n = 3 for naïve or naïve-infection of WT, n = 5 for the rest of the groups. Statistical significance: * p < 0.05. In addition to the potential necessity of neutralizing antibodies against live RSV infection, T-cell immune responses could contribute to the protection against RSV infection [23] . Therefore, we determined the induction of RSV F-specific CD8 T-cells by intracellular cytokine staining of BAL cells at day 5 post-challenge (Figure 4) . IFN-γ-TNF-α+ CD8 Tcells and IFN-γ-IL-4+ CD8 T-cells were detected at similarly low levels in BALF from both WT and FcRγ −/− mice with the F-VLP/FI-RSV vaccination after RSV challenge. Double positive IFN-γ+TNF-α+ CD8 T-cells were induced at higher levels in WT BALF than in FcRγ −/− BALF samples ( Figure 4A,B) . Double-positive IFN-γ+IL-4+ CD8 T-cells were not detected in the non-immunized groups. IFN-γ+TNF-α-and IFN-γ+IL4-CD8 T-cells were observed at higher levels in BAL cells from WT mice than those from FcRγ −/− mice with F-VLP/FI-RSV immunization ( Figure 4A-D) . Therefore, these results suggest that FcRγ plays a pivotal role in inducing effector CD8 T-cell responses, which might contribute to protection. [31, 32] . NK cells recognize particulate antibody-antigen immune complexes on infected cells that bind to receptors for the Fc portion of Ig (FcR) expressed on these cells and trigger antibody-dependent cell-mediated cytotoxicity (ADCC) [23, 33] . Furthermore, NK cells are important innate immune cells in the early response to and control of viral infections [34] , and their activity is tightly regulated, for example, via their interaction with antigen-specific antibodies [35] . The CD8 T-cells, NK cells, and dendritic cells (DCs) phenotype of BAL cells were analyzed to determine whether the deletion of FcRγ would affect the recruitment of CD8 T-cells, NK cells, and dendritic cells (DCs) into the airways after RSV infection in F-VLP/FI-RSV-immunized mice. The WT mice with immunization recruited higher levels of CD3+CD8+ T-cells, NK cells, and CD11c+B220+ DCs after RSV infection compared to FcRγ −/− mice ( Figure 5A-C) . Therefore, these results provide evidence that FcRγ plays a role in recruiting CD3+CD8+ T-cells, NK cells, and CD11c+B220+ DCs into the airway respiratory sites in F-VLP/FI-RSV-vaccinated mice after RSV infection. Furthermore, NK cells are important innate immune cells in the early response to and control of viral infections [34] , and their activity is tightly regulated, for example, via their interaction with antigen-specific antibodies [35] . The CD8 T-cells, NK cells, and dendritic cells (DCs) phenotype of BAL cells were analyzed to determine whether the deletion of FcRγ would affect the recruitment of CD8 T-cells, NK cells, and dendritic cells (DCs) into the airways after RSV infection in F-VLP/FI-RSV-immunized mice. The WT mice with immunization recruited higher levels of CD3+CD8+ T-cells, NK cells, and CD11c+B220+ DCs after RSV infection compared to FcRγ −/− mice ( Figure 5A-C) . Therefore, these results provide evidence that FcRγ plays a role in recruiting CD3+CD8+ T-cells, NK cells, and CD11c+B220+ DCs into the airway respiratory sites in F-VLP/FI-RSV-vaccinated mice after RSV infection. To assess differences in lung histopathology between WT and FcRγ −/− mice after RSV infection, histology tissues infiltrated with eosinophils and mucus-producing cells in the airways were visualized with H&E, H&CR, and periodic acid-Schiff (PAS) staining. In the groups vaccinated with the F-VLP prime and FI-RSV boost regimens, FcRγ −/− mice displayed higher levels of H&E-stained inflammation ( Figure 6A,B) , H&CR staining of eosinophils ( Figure 6C,D) , and PAS-positive histopathology ( Figure 6E ,F) than WT vaccinated mice ( Figure 6 ). WT mice that were immunized with F-VLP/FI-RSV showed slightly less pulmonary eosinophil infiltration and PAS-positive mucus production, compared to FcRγ −/− mice vaccinated with F-VLP/FI-RSV ( Figure 6) . These results suggest a role of FcRγ in attenuating histopathology in vaccinated mice after RSV infection. To assess differences in lung histopathology between WT and FcRγ −/− mice after RSV infection, histology tissues infiltrated with eosinophils and mucus-producing cells in the airways were visualized with H&E, H&CR, and periodic acid-Schiff (PAS) staining. In the groups vaccinated with the F-VLP prime and FI-RSV boost regimens, FcRγ −/− mice displayed higher levels of H&E-stained inflammation ( Figure 6A,B) , H&CR staining of eosinophils ( Figure 6C,D) , and PAS-positive histopathology ( Figure 6E ,F) than WT vaccinated mice ( Figure 6 ). WT mice that were immunized with F-VLP/FI-RSV showed slightly less pulmonary eosinophil infiltration and PAS-positive mucus production, compared to FcRγ −/− mice vaccinated with F-VLP/FI-RSV ( Figure 6) . These results suggest a role of FcRγ in attenuating histopathology in vaccinated mice after RSV infection. VLPs contain a high density of repetitive effective antigenic epitopes on their surfaces. DCs and B-cells function as antigen-presenting cells (APCs); therefore, APCs directly take up VLPs and present peptide-form antigens to activate T-cells [36] . Cross-linking of B-cell receptors by VLPs can be strong enough to prime B-cells and induce the production of antibodies even without the help of CD4+ T-cells [37] . Owing to their distinct multimeric-nature conformational advantage, VLP-based vaccines are capable of inducing both humoral and cellular immune responses, and adjuvant co-administration is not needed in most VLP vaccinations [38] . A previous study has shown that priming with RSV F-VLP shifts the immune response toward the Th1 pattern and effector CD8 T-cell responses under subsequent exposure to the ERD-prone FI-RSV vaccine against RSV infection [25] . However, Th1-type-biased RSV F-VLP immune mechanisms are not well understood. In this study, we attempted to better understand the roles and contributions of FcRγ by comparing immune responses and protection in WT and FcRγ −/− mice immunized with F-VLP/FI-RSV. Anti-RSV F IgG antibody and IgG isotype distribution in sera were found to be similar in WT and FcRγ −/− mice after F-VLP priming or F-VLP/FI-RSV vaccination. Therefore, this study suggests that FcRγ is not required for inducing vaccine-specific binding of IgG antibodies after F-VLP/FI-RSV. However, the F-VLP/FI-RSV WT mice induced higher levels of RSV-neutralizing activity in sera, compared to those in F-VLP/FI-RSV FcRγ −/− mice, providing evidence for a role of FcRγ in inducing protective neutralizing antibodies after vaccination against RSV. This role of FcRγ is further supported by the effective clearance of lung RSV loads in WT F-VLP/FI-RSV mice or the lower lung RSV titers in WT naïve mice after RSV infection, compared to those in FcRγ −/− mice. DCs are known to be professional APCs, highly effective in cross-presenting nonreplicating VLP antigens to CD8 T-cells to prime cytotoxic CD8 T-cell responses [31] . VLPspecific antibodies might contribute to more effective uptake via Fc and FcR interactions by DCs, promoting the cross-presentation of antigens to CD8 T-cells. Consistent, higher levels of effector CD8 T-cell responses were induced in WT F-VLP/FI-RSV mice, the levels of which were diminished in FcRγ −/− F-VLP/FI-RSV mice. Hemagglutinin-containing VLPs can prime influenza virus-specific CD8 T-cell responses that contribute to protection from influenza infection [39] . Interestingly, we found that FcRγ −/− naïve mice showed a defect in controlling lung RSV titers compared to WT naïve mice after RSV infection. FcR-mediated viral clearance appears to contribute to controlling viruses in naïve mice after infection. Particularly, FcRγ has been reported to play a critical role in providing broad cross-protection against conserved influenza epitopes in the presence of non-neutralizing antibodies [40] [41] [42] [43] . It is possible that FcRs play a role in inducing CD8 T-cells and mediating viral clearance, both of which contribute to protection. The Sf9 insect cell-system-based RSV F-or G-VLP has been shown to modulate the immune response to suppress Th2 immune-mediated pulmonary histopathology and eosinophilia [25] . In a subset of individuals, the bacterially produced recombinant VLPbased influenza vaccine shifted pre-vaccination-induced type-2 cytokines such as IL-5 and IL-13 to a post-vaccination type-1 cytokine signature characterized by IFN-γ [44] . In BALB/c mice, IgG2a responses reflect the establishment of Th1-type immune responses for viral infections [45] . We found that levels of IgG2a/IgG1 responses were significantly increased in BALB/c mice after the F-VLP prime vaccination but less so in FcRγ −/− mice. IgG2a antibodies are reported to have higher affinity for the activating receptors FcγRI, -III, and -IV, whereas mouse IgG1 only binds significantly to FcγRIII [46, 47] . In this study, we found that FcRγ plays a role in mediating CD8 T-cell cytotoxic immune responses in the local compartment. In the case of SARS-CoV-2 infection, FcRγ engagement on dendritic cells could help stimulate cytotoxic antiviral CD8 T-cell responses, which are often suppressed as a result of excessive viral replication and uncontrolled recruitment of monocytes, leading to Th2-cell-biased airway inflammation and acute lung damage [48, 49] . In addition to enhanced VLP antigen uptake and presentation, immune complex endocytosis through activation of type I FcRs is also accompanied by cellular activation and the induction of pro-inflammatory chemokine and cytokine expression, which in turn influence the differentiation, effector activities, and survival of APCs [50] [51] [52] . IgG antibodies are not required for FcγRIIB-mediated control of CD8 T-cell immunity, and a cell-intrinsic inhibitory function of FcγRIIB directly limited CD8 T-cell responses [53] . A previous study reported the contribution of accumulated immune complexes to enhanced RSV disease [15] . Antibody-dependent enhancement of microbial infection after ligation of macrophage FcRγ via infectious immune complexes has also been reported [16, 23] . In contrast, F-VLP/FI-RSV-vaccinated FcRγ −/− mice displayed a trend of increasing histopathology rather than attenuating inflammation after RSV infection. Therefore, this study provides evidence that FcRγ is an important host immune component in clearing lung viral loads and attenuating immune histopathology, at least in a BALB/c mouse model, after F-VLP/FI-RSV vaccination and RSV infection. Collectively, we have shown that FcRγ contributed to inducing RSV F-specific CD8 T-cells and neutralizing antibodies as well as providing effective viral clearance and protection against RSV. In future studies, it is important to determine the types of Fc receptors that mediate IFNγ-producing CD8 T-cell responses. Further studies are also required to understand and utilize Fc-mediated antibody effector functions for the development of safe and effective vaccines and antibody therapies against RSV. The present study demonstrated that the FcRγ-chain plays an important role in inducing antiviral protection and CD8 T-cell responses in RSV F-VLP prime and FI-RSV boost vaccination after RSV infections. Collectively, our data provide important information for FcRγ contributed to inducing RSV F-specific CD8 T-cells and neutralizing antibodies as well as providing effective viral clearance and protection against RSV. All animal experiments for the study were conducted according to accepted veterinary standards set by the Georgia State University (GSU) animal care center, and approved by the Ethics Committee on the use and care of animals at the GSU (The approval number is IACUC-A18001). Informed Consent Statement: Not applicable. The data that support the findings of this study are available on request from the corresponding author. Respiratory syncytial virus disease in infants despite prior administration of antigenic inactivated vaccine Immunological Lessons from Respiratory Syncytial Virus Vaccine Development Newcastle disease virus-like particles: Preparation, purification, quantification, and incorporation of foreign glycoproteins Novel Respiratory Syncytial Virus-Like Particle Vaccine Composed of the Postfusion and Prefusion Conformations of the F Glycoprotein Assembly and immunological properties of Newcastle disease virus-like particles containing the respiratory syncytial virus F and G proteins Newcastle disease virus-like particles containing respiratory syncytial virus G protein induced protection in BALB/c mice, with no evidence of immunopathology Priming immunization determines T helper cytokine mRNA expression patterns in lungs of mice challenged with respiratory syncytial virus Virus-Like Particle Vaccine Containing the F Protein of Respiratory Syncytial Virus Confers Protection without Pulmonary Disease by Modulating Specific Subsets of Dendritic Cells and Effector T Cells Virus-like nanoparticle and DNA vaccination confers protection against respiratory syncytial virus by modulating innate and adaptive immune cells Co-immunization with virus-like particle and DNA vaccines induces protection against respiratory syncytial virus infection and bronchiolitis Recognition of respiratory syncytial virus fusion protein by mouse cytotoxic T cell clones and a human cytotoxic T cell line Efficacy of a respiratory syncytial virus vaccine candidate in a maternal immunization model Priming Vaccination With Influenza Virus H5 Hemagglutinin Antigen Significantly Increases the Duration of T cell Responses Induced by a Heterologous H5 Booster Vaccination A role for immune complexes in enhanced respiratory syncytial virus disease Intrinsic antibody-dependent enhancement of microbial infection in macrophages: Disease regulation by immune complexes Contribution of Fcgamma Receptor-Mediated Immunity to the Pathogenesis Caused by the Human Respiratory Syncytial Virus Contribution of Fcgamma receptors to human respiratory syncytial virus pathogenesis and the impairment of T-cell activation by dendritic cells Review of palivizumab in the prophylaxis of respiratory syncytial virus (RSV) in high-risk infants 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 Prophylactic treatment with a G glycoprotein monoclonal antibody reduces pulmonary inflammation in respiratory syncytial virus (RSV)-challenged naive and formalininactivated RSV-immunized BALB/c mice A potent broadly neutralizing human RSV antibody targets conserved site IV of the fusion glycoprotein Fc-Mediated Antibody Effector Functions During Respiratory Syncytial Virus Infection and Disease Fc gamma receptors in respiratory syncytial virus infections: Implications for innate immunity Virus-like particle vaccine primes immune responses preventing inactivated-virus vaccine-enhanced disease against respiratory syncytial virus Viruslike particle vaccine induces protection against respiratory syncytial virus infection in mice Virus-specific CTL responses induced by an H-2K(d)-restricted, motif-negative 15-mer peptide from the fusion protein of respiratory syncytial virus Pulmonary immunity and immunopathology: Lessons from respiratory syncytial virus Comparison of histochemical methods for murine eosinophil detection in an RSV vaccine-enhanced inflammation model Differential pathogenesis of respiratory syncytial virus clinical isolates in BALB/c mice Critical role for activation of antigen-presenting cells in priming of cytotoxic T cell responses after vaccination with virus-like particles Cross-presentation of epitopes on virus-like particles via the MHC I receptor recycling pathway Antibody-Dependent Cellular Phagocytosis in Antiviral Immune Responses Viral modulation of NK cell immunity Natural killer cell activation by respiratory syncytial virus-specific antibodies is decreased in infants with severe respiratory infections and correlates with Fc-glycosylation Vaccine delivery: A matter of size, geometry, kinetics and molecular patterns Natural Killer and CD8 T Cells Contribute to Protection by Formalin Inactivated Respiratory Syncytial Virus Vaccination under a CD4-Deficient Condition Interaction Between Virus-Like Particles (VLPs) and Pattern Recognition Receptors (PRRs) From Dendritic Cells (DCs): Toward Better Engineering of VLPs. Front Protective CD8 T cell-mediated immunity against influenza A virus infection following influenza virus-like particle vaccination Broadly neutralizing anti-influenza antibodies require Fc receptor engagement for in vivo protection Multiple heterologous M2 extracellular domains presented on virus-like particles confer broader and stronger M2 immunity than live influenza A virus infection Supplementation of influenza split vaccines with conserved M2 ectodomains overcomes strain specificity and provides long-term cross protection Fc receptor is not required for inducing antibodies but plays a critical role in conferring protection after influenza M2 vaccination Induction of Human T-cell and Cytokine Responses Following Vaccination with a Novel Influenza Vaccine Expanding roles for CD4(+) T cells in immunity to viruses M2e-Based Universal Influenza A Vaccines Evaluation of a fully human monoclonal antibody against multiple influenza A viral strains in mice and a pandemic H1N1 strain in nonhuman primates The role of IgG Fc receptors in antibody-dependent enhancement Dysregulated Type I Interferon and Inflammatory Monocyte-Macrophage Responses Cause Lethal Pneumonia in SARS-CoV-Infected Mice Fcgamma Receptor Function and the Design of Vaccination Strategies Autonomous phagosomal degradation and antigen presentation in dendritic cells Fcgamma receptor-mediated induction of dendritic cell maturation and major histocompatibility complex class I-restricted antigen presentation after immune complex internalization Signaling through the Inhibitory Fc Receptor FcgammaRIIB Induces CD8(+) T Cell Apoptosis to Limit T Cell Immunity. Immunity The authors declare no conflict of interest.Vaccines 2021, 9, 232