Vagal-α7 nicotinic acetylcholine receptor signaling exacerbates influenza severity by promoting lung epithelial cell infection
Abstract
The vagus nerve circuit, operating through the alpha-7 nicotinic acetylcholine receptor (α7 nAChR), regulates the inflammatory response by influencing immune cells. However, the role of vagal-α7 nAChR signaling in influenza virus infection is unclear. In particular, does vagal-α7 nAChR signaling impact the infection of alveolar epithelial cells (AECs), the primary target cells of influenza virus? Here, we demonstrated a distinct role of α7 nAChR in type II AECs compared to its role in immune cells during influenza infection. We found that deletion of Chrna7 (encoding gene of α7 nAChR) in type II AECs or disruption of vagal circuits reduced lung influenza infection and protected mice from influenza-induced lung injury. We further unveiled that activation of α7 nAChR enhanced influenza infection through PTP1B-NEDD4L-ASK1-p38MAPK pathway. Mechanistically, activation of α7 nAChR signaling decreased p38MAPK phosphorylation during infection, facilitating the nuclear export of influenza viral ribonucleoproteins and thereby promoting infection. Taken together, our findings reveal a mechanism mediated by vagal-α7 nAChR signaling that promotes influenza viral infection and exacerbates disease severity. Targeting vagal-α7 nAChR signaling may offer novel strategies for combating influenza virus infections.
1 INTRODUCTION
The seasonal influenza pandemic has had a profound impact on human society for centuries. Influenza A virus (IAV) stands as the most prevalent subtype of influenza viruses. For instance, the 1918 “Spanish” flu, caused by an H1N1 influenza A strain, resulted in the loss of over 40 million lives worldwide. IAV predominantly targets and replicates within respiratory epithelial cells, particularly alveolar epithelial cells (AECs). IAV infection of AECs can trigger acute respiratory distress syndrome and acute lung injury (ALI), characterized by extensive lung inflammation and rapid onset pneumonia. The effective management of lung infection serves as a crucial aspect of clinical treatment for IAV-infected patients. However, there exists a scarcity of effective therapies to mitigate IAV infection in the lungs.
Although influenza infection is primarily known to cause respiratory symptoms, it has been associated with various neurological manifestations, from mild symptoms such as headache and dizziness to severe symptoms like acute necrotizing encephalopathy, encephalitis, Guillain–Barré syndrome, and acute disseminated encephalomyelitis.1 Remarkably, severe cases of influenza infection requiring intensive care unit treatment have often been linked with neurological manifestations. For instance, a recent study focusing on influenza A (H3N2) infection revealed that patients exhibiting neurological manifestations were at a significantly higher risk of hospitalization, with a ten-fold increase compared to those without such symptoms.2 However, much remains unknown regarding the underlying mechanisms of these associations between influenza and neurological conditions. Questions persist about how influenza infection leads to neurological disorders, the reciprocal relationship between them, and the specific role of the nervous system in the progression of influenza diseases.
The neuroimmune system evolves mechanisms to detect pathogens such as bacteria or viruses.3 When viral or bacterial infections occur, sensory neurons and their nerve endings sense changes in molecular signals within the infected area, leading to the release of neurotransmitters.4 These neurotransmitters then act on immune cells or pathogens, influencing the outcome of infection, inflammation, and immunity.4, 5 These neural regulatory mechanisms have been referred to as “the inflammatory reflex.”6 The vagus nerve is a vital component of the autonomic nervous system, responsible for regulating essential bodily functions such as respiratory rate, and is one crucial mediator of the inflammatory reflex, innervating the distal airways of the lung, including the alveoli.7 Vagus nerve controls innate immune responses and inflammation during infections through the alpha-7 nicotinic acetylcholine receptor (α7 nAChR), which is widely expressed in neuronal or nonneuronal cells, such as epithelial cells, endothelial cells, and macrophages.8 Recent studies have demonstrated that α7 nAChR plays a crucial role in various infections, including bacterial infections/lipopolysaccharide (LPS) stimulation,9, 10 SARS-CoV-2 infections,11 and human immunodeficiency virus (HIV) infections.12 In addition, activation of α7 nAChR promotes alveolar regeneration during ALI.13 Interestingly, in these cases, the effects mediated by α7 nAChR activation vary in different infections, either beneficial or detrimental, indicating that α7 nAChR functions with diverse mechanisms in the infections of different pathogens.
Intranasal inoculation of virulent strains of influenza viruses led to neural infection in mice, with viral antigens predominantly manifesting in the brain stem, olfactory bulbs, and thoracic cord.14, 15 Further evidence suggests that the influenza virus travels from the respiratory mucosa to the central nervous system (CNS) primarily via the vagus nerve.16 Influenza is known to induce cytokine storms, which directly contribute to the development of lung damage. In this regard, our recent study indicates that vagus-α7 nAChR signaling promotes vicarious activation of macrophages during influenza infection.17 However, whether activation of α7 nAChR affects the replication of influenza in lung epithelial cells remains unknown, posing an important scientific question that requires investigation. A previous study showed that nicotine, a nonspecific α7 nAChR agonist, promoted influenza infection.18 Thus, we extend our hypothesis to propose that hypervagal tone could lead to increased acetylcholine release, subsequently activating α7 nAChR and impacting influenza infection of AECs and disease severity. In fact, we have found that blood CHAT (acetylcholine synthase) expression was higher in patients with influenza compared to healthy controls (unpublished data). Hence, clarifying the association between vagus-α7 nAChR signaling and influenza infection holds paramount significance for both fundamental understanding and clinical implications.
To address this question and elucidate the underlying mechanism, we employed both in vivo and in vitro models to explore the function of vagal-α7 nAChR signaling in IAV infection. Our research revealed a distinct role of α7 nAChR in type II AECs compared to its role in immune cells. Specifically, we discovered that vagal-α7 nAChR signaling promotes IAV infection through the PTP1B-NEDD4L-ASK1-p38MAPK pathway in lung epithelial cells, ultimately exacerbating the severity of infection in the lungs. By uncovering the role of vagal-α7 nAChR signaling in enhancing influenza infection and worsening disease severity, our study identifies a potential target for therapeutic intervention. These findings hold promise for informing the development of innovative therapies aimed at bolstering host defenses against influenza viruses and improving patient outcomes in clinical settings.
2 MATERIALS AND METHODS
2.1 Animals
α7 nAChR deficient mice (C57BL/6J, B6.129S7-Chrna7tm1Bay) were from the Jackson Laboratory and housed in a pathogen-free condition. α7 nAChR floxed mice were generated in our lab with the method shown in Supporting Information S1: Figure S1. Spc-Cre+ mice were kindly provided as a gift from Prof. Kaifeng Xu (Chinese Academy of Medical Sciences). The surfactant protein C (SPC) is exclusively expressed in the type II AECs.19 Wildtype mice in C57BL/6J background were purchased from the Model Animal Research Center of Nanjing University. Anesthesia was induced with an intraperitoneal (i.p.) injection of a mixture of Pentobarbital sodium (50 mg/kg). The Committees on Animal Research of the Institut Pasteur of Shanghai, Chinese Academy of Sciences approved the protocols.
2.2 Unilateral vagotomy
As previously described,20 right or sham cervical vagotomy was performed with the animals under anesthesia. The procedure involved a longitudinal midline incision in the ventral region of the neck. Using blunt dissection, the overlying muscles and fascia were separated until the right vagus and carotid artery were visible. The vagus was carefully stripped away from the carotid artery and lightly cut off in the vagotomy group. The vagus was kept intact in the sham group. The wound was closed and sutured. The respiration rhythm was not affected by unilateral vagotomy.
2.3 Influenza viral propagation
The viruses (PR8, mouse adapted A/Puerto Rico/8/1934(H1N1), kindly provided by Dr. Ertl HC, Wistar Institute) were propagated in 10-day-old chicken embryo from specific-pathogen-free flocks (Beijing Merial). Each egg was injected with 0.1 mL of phosphate-buffered saline (PBS) containing ~103 infectious particles, incubated at 37°C with forced air circulation and egg rotation, and maintained at 4°C for 12 h before harvesting. The allantoic fluid was harvested separately from each egg. Those with high hemagglutinin activity were pooled, and aliquots were prepared and stored at −80°C. The hemagglutinin titer of the above subtype of influenza virus was measured with 1% chicken blood cells in a V-shaped plate. After quantification, the viruses were stored at −80°C for future use. H3N2 strain (kindly provided by Dr. Ertl HC) was also prepared in the same way.
2.4 Cell culture
Human lung carcinoma cell A549, human embryonic kidney 293T cell (HEK293T), and Madin–Darby Canine Kidney (MDCK) cell were purchased from ATCC and maintained in minimal essential medium (DMEM, Invitrogen/Ambion/Gibco/MP). All cell cultures were supplemented with 10% fetal calf serum (fetal bovine serum [FBS] qualified Australia origin, Invitrogen/Ambion/Gibco/MP), 100 U penicillin, and 100 μg streptomycin per milliliter (Invitrogen/Ambion/Gibco/MP). The cells were incubated at 37°C and 5% carbon dioxide.
2.5 Cell viral infection
For influenza virus infection, 5 × 105 low-passage A549 or HEK293T cells were seeded in designated plates. At 24 h post seeding, cells were washed with PBS and infected with PR8 at a multiplicity of infection (MOI) of 0.2 in DMEM containing 0.2% BSA and 1 μg/mL TPCK for the indicated time in a 5% CO2 incubator at 37°C. For mock infection, the same amount of allantoic fluid from noninoculated 10-day-old chicken embryos was used. The samples were collected at 24 h post infection (hpi).
2.6 Foci forming assay for determining viral load
For the cell culture study, the supernatant of PR8-infected cell culture media was collected and then plated in MDCK monolayer. After incubation, blocking, and immune staining, the plaques were quantified. The viral titer was expressed as log10-transformed Foci Forming Units (FFU) per mL. For determining lung viral load, mice were killed 6 days post infection (dpi), and the lungs were removed and homogenized in PBS containing 0.1% BSA. The supernatant was plated on confluent MDCK cells followed by the standard procedure of plaque assay. The viral titer was also expressed as FFU per mL with log10 transformation.
2.7 Animal infections and lung histology
Some 8–10 weeks male C57BL/6J and α7 nAChR deficient mice were used in the infection experiments. Mice were anesthetized and then given either 20 µL of PR8 (1.4 × 105 FFU) or PBS intranasally. Live virus experiments were conducted in Biosafety Level II facilities obeying governmental and institutional guidelines. After mice were anesthetized and killed with 80 mg/Kg pentobarbital sodium. The lungs were collected and fixed in 4% paraformaldehyde overnight at room temperature. The tissues were embedded in paraffin; 4 μm sections were cut by a Leica manual microtome (RM2125 RTS) and stained with hematoxylin and eosin. Lung injury score was graded independently in a double-blinded manner as we described previously.21
2.8 Flow cytometric analysis
As described,22 harvested lungs were inflated with 3 mL of a mixture of collagenase (150 units/mL of liberase Cl, Sigma/flu/Ald) and DNaseI (10 µg/mL, Sigma/flu/Ald) in RPMI-1640 containing 5% FBS and 20 mM HEPES. Lungs were chopped in the 3 mL enzyme mix and incubated for 35 min at 37°C. In total, 10 mM ethylenediaminetetraacetic acid (EDTA) was added and any remaining pieces were further dispersed by 12 passages through a 21 G needle. Suspensions were passed through a 100 µm nylon mesh, and cells were washed multiple times in RPMI-1640 with 5% FBS and 20 mM HEPES. red blood cell lysis was performed on cell preparations for cellular analysis by flow cytometry but not on preparations for flowcytometry. Unspecific staining was minimized through preincubation for 15 min with antimouse CD16/32. The antibodies for flow cytometry analysis were CF633 α-Bungarotoxin (Biotium, Cat#00009), mouse anti-CD326 (Ep-CAM), APC antibody (Biolegend, Cat#118213), anti-ProSPC antibody (Merck/Millipore, Cat# AB3786), and anti-Influenza A Virus Nucleoprotein (FITC) antibody (Abcam, Cat# ab20921). Fluorescent cells were analyzed after the exclusion of debris and aggregates with LSRFortessa (BD Biosciences). Data were analyzed by Flowjo 7.6 (Tree Star Inc.).
2.9 Chemicals and kits
The following chemicals and kits were used: MG-132 (SIGMA, Cat# 474790), GTS-21 (Abcam, Cat# ab120560), Collagenase (SIGMA, Cat# C9891-100MG), DNaseI (SIGMA, Cat# D4527), PTP1B activity Assay Kit (SIGMA, Cat# 539736), Pierce ECL Western blot analysis Substrate (Pierce, Cat# 32109), BCA Protein Assay Kit (Pierce, Cat# 23227), Trizol Reagent (Invitrogen/Ambion/Gibco/MP, Cat# 15596026), ACK Lysing Buffer (Invitrogen/Ambion/Gibco/MP, Cat# A1049201), Protease Inhibitor Cocktail (100X) (Cell Signaling Technology, Cat# 5871 S), Phosphatase inhibitor cocktail (Santa Cruz, Cat# SC-45044), Cytofix/cytoperm with Golgi plug kit (BD Pharmingen, Cat# 555028), Fixation and Permeabilization Solution (BD Pharmingen, Cat# 554722), and Perm Wash Buffer (BD Pharmingen, Cat# 554723).
2.10 Western blot analysis and immunoprecipitation
Cells were scraped in lysis buffer (20 mM Tris-HCl, pH 7.6, 2.5 mM EDTA, 1 mM EGTA, 1% Triton X-100, 0.5% sodium deoxycholate, 10% glycerol, 1 mM Na3VO4, 50 mM NaF, 1 mug/mL of aprotinin, 1 mg/mL of leupeptin, and 1 mM phenylmethyl sulfonyl fluoride), were sonicated for 10 s, and were centrifuged at 4°C for 20 min at 14 000g. Lysates preabsorbed to 20 μL protein A–protein G (Invitrogen) were incubated overnight with the appropriate antibodies and were immunoprecipitated with 40 μL protein A–protein G. Immunoprecipitates were recovered by centrifugation, were washed in ice–cold wash buffer (0.1% Triton X-100 and 1 mM phenylmethyl sulfonyl fluoride in Tris-buffered saline), and were “taken up” in sample buffer (125 mM Tris-HCl, pH 6.8, 2% SDS, 10% glycerol, and 0.5 mg/mL of bromophenol blue). Denatured proteins were loaded and run on a 10% Bis-Tris gel (Invitrogen). The proteins were then transferred to a polyvinylidene difluoride membrane and incubated with primary antibodies and secondary antibodies. Images were analyzed using Tanon Gel Image System 4200 (Shanghai Tanon Technology Co., Ltd.). The primary antibodies used for Western blot were anti-Influenza A Virus NP antibody (Abcam, Cat# ab128193), anti-Influenza A Virus PB2 antibody (Pierce, Cat# PA532220), anti-α7 nAChR antibody (H302) (Santa Cruz, sc-5544), anti-GAPDH antibody (EMAR, Cat# EM32010-02), anti-ASK1 antibody (Cell Signaling Technology, Cat# 8662), anti-p-p38MAPK (Thr 180/Tyr 182) antibody (Cell Signaling Technology, Cat# 9211 S), anti-p38MAPK antibody (Cell Signaling Technology, Cat# 54470), anti-β-actin antibody (EMAR, Cat# EM32011-02), anti-Influenza NS1 antibody (Santa Cruz, Cat# sc-17596), anti-NEDD4L antibody (Santa Cruz, Cat# sc-514954), anti-p-Tyr antibody (Santa Cruz, Cat# sc-7020), anti-PTP1B antibody (Cell Signaling Technology, Cat# 5311), anti-p-PTP1B Ser378 antibody (Abcam, Cat# ab76239), anti-Myc antibody (Santa Cruz, Cat# sc-40), anti-Lamin B1 (Proteintech, Cat# 66095-1-Ig), anti-HA antibody (Santa Cruz, Cat# sc-7392), anti-CRM1 antibody (Cell Signaling Technology, Cat# 46249), and anti-Flag antibody (Sigma, Cat# F1804). The secondary antibodies were mouse anti-rabbit-HRP (Cell Signaling Technology, Cat#5127), goat polyclonal anti-rabbit-HRP (Cell Signaling Technology, Cat#7074), and goat polyclonal anti-mouse-HRP (Cell Signaling Technology Cat#7076).
2.11 Quantitative real-time polymerase chain reaction (PCR)
Total ribonucleic acid (RNA) from lungs or cultured cells was homogenized and extracted using Trizol (Invitrogen), as described by the manufacturer. The total RNA obtained was suspended in RNase-free water and stocked at −80°C. Real-time polymerase chain reaction (PCR) was performed on an ABI PRISM Step-One sequence-detection system by using SYBR Green PCR Master Mix (Applied Biosystems) after a reverse transcription reaction of 2 µg of RNA by using M-MLV reverse transcriptase (TIANGEN). The relative expression levels of corresponding genes were determined by the 2-ΔΔCT method,23 normalized by ribosomal subunit 18 S or GAPDH. All human and mouse primers have been summarized in Supporting Information S1: Table 1.
2.12 Bulk RNA sequencing and analysis
Total RNA was extracted from A549 cells mock-infected or infected with/without GTS-21 (50 μM) and IAV at 24 hpi using TRIzol reagent (Invitrogen) based on the manufacturer's instructions. The RNA was treated with DNase I for 30 min at 37°C to remove residual deoxyribonucleic acid (DNA). mRNA was purified with oligo (dT) beads. Then, the purified mRNA was fragmented into small pieces with fragment buffer (provided by BGI) at the appropriate temperature. The first-strand and second-strand cDNA were generated. Afterward, A-Tailing Mix and RNA Index Adapters were added by incubating to end repair. The cDNA fragments were further amplified by PCR, and the products were purified by Ampure XP Beads, then dissolved in EB solution. The product was validated on the Agilent Technologies 2100 bioanalyzer for quality control. The amplified PCR products from the previous step were denatured and circularized by the splint oligo sequence to get the library. The single-strand circle DNA was formatted as the final library. The final library was amplified to make DNA nanoball (DNB), which had more than 300 copies of one molecular. DNBs were loaded into the patterned nanoarray, and single end 50 bases reads were generated on BGIseq500 platform (BGI). The gene expression levels were calculated in fragments per kilobase transcriptome per million mapped reads. The data were analyzed by R package DESeq2 R (1.30.0) and fgsea.
2.13 Plasmid constructs and transfection
FLAG-tagged NEDD4L expression plasmids were provided by Xin Ye (Institute of Microbiology, Chinese Academy of Sciences). Recombinant vectors encoding NEDD4L, PTPN1, MAPK14, and mutant MAPK14 were constructed by PCR-based amplification of cDNA from the human A549 cell line, then were subcloned into the PLKO eukaryotic expression vector (Invitrogen) through T4 DNA ligase. Transient transfection of HEK293T cells with indicated plasmids was performed routinely with polyethylenimine (PEI).
2.14 Lentivirus construction and infection
For the knockdown assay, short hairpin RNA (shRNA) sequences, synthesized by Nanjing Genewis Biotechnology, were inserted into pLKO.1 plasmid between the EcoRI and NheI restriction sites. PLKO.1 vector and packaging vectors were co-transfected into HEK293T cells with PEI reagent. For overexpression assay, the gene coding sequence was cloned into PLVX lentiviral vectors. PLVX vector and packaging vectors were co-transfected into HEK293T cells with PEI. The supernatants containing viruses were harvested after 48 h and were purified and enriched via filter and ultracentrifugation. The shRNA PLKO.1 construct was introduced into target cells via lentiviral transduction. The knockdown assay primer sequences were summarized in Supporting Information S1: Table 2.
2.15 His-pull-down assay
Myc-tagged ASK1, Flag-tagged NEDD4L, and His-tagged ubiquitin (Ubi) were co-transfected into HEK293T cells. MG-132 (10 μM) was added at 6 h before sample collection. About 48 h posttransfection, cells were harvested, washed with 1 × PBS, and lysed with lysis buffer. The lysates were incubated with nickel-nitrilotriacetic acid beads (Ni-NTA Beads, Qiagen 30210) at room temperature for 2 h. Beads were washed three times in Buffer 1 and twice in Buffer 2 (8 M urea, 100 mM Na2HPO4, 10 mM Tris-HCl, pH 6.3, 0.2% Triton X-100, and 10 mM imidazole) and once in Buffer 3 (100 mM NaCl, 20 mM Tris-HCl, pH 8.0, 20% glycerol, 1 mM dithiothreitol, and 10 mM imidazole). Then ubiquitination levels were detected by immunoblotting.
2.16 Immunofluorescent (IF) microscopy
A549 cells were infected with target lentiviruses. After 48 h of lentiviral infection, these cells were used for the following experiments. Following corresponding experimental treatment with PR8 infection, cells were fixed in 4% formaldehyde and permeabilized in 0.5% Triton-X 100. Cell nuclei were stained with DAPI dye and slides were imaged on a laser-scanning confocal microscope (Olympus FV-1200). Images were quantified by Image J Pro.
2.17 Statistical analysis
Statistics were done by SPSS software (SPSS Inc.) or GraphPad Prism software (GraphPad). An unpaired t test was used unless there were multiple comparisons in which case we used one-way ANOVA or two-way ANOVA. The log-rank test was used for comparing survival by GraphPad Prism software. The results are shown as mean ± SEM.
3 RESULTS
3.1 Deficiency of vagal-α7 nAChR signaling alleviates influenza viral disease severity and limits the infection of AECs
To figure out whether α7 nAChR signaling is involved in influenza viral infection, we first tested the effect of α7 nAChR knockout on IAV infection by infecting Chrna7+/+ (the coding gene of α7 nAChR) and Chrna7−/− mice with influenza virus A H1N1/PR8 (Figure 1A). The results showed that the survival of Chrna7−/− mice was significantly higher compared to Chrna7+/+ mice (Figure 1B), and Chrna7−/− mice had less body weight loss than Chrna7+/+ mice post PR8 infection (Figure 1C). Consistently, lung injury and inflammatory responses were also decreased in Chrna7−/− mice compared to Chrna7+/+ mice, manifested by reduced lung injury score (Figure 1D,E), lower protein levels and cell number in bronchoalveolar lavage fluid (BALF) (Figure 1F,G), and lower expression levels of inflammatory genes (Supporting Information S1: Figure S1A) at 6 dpi. Interestingly, we further identified a decreased PR8 viral titer in the lung of Chrna7−/− mice compared to Chrna7+/+ mice at 6 dpi (Figure 1H). Consistently, the levels of PR8 M gene were also dramatically decreased in Chrna7−/− mice compared to Chrna7+/+ mice at 6 dpi (Figure 1I). Additionally, we confirmed that Chrna7 gene was deleted in Chrna7−/− mice (Figure 1J). Notably, the levels of Chrna7 gene in the lung were significantly increased after PR8 infection (Figure 1J), supporting a possible role of α7 nAChR in regulating IAV infection. Given that AECs are major targets of IAV in the lung, we disassociated the lungs into single cells by enzyme digestion and detected the PR8-infected AECs by flow cytometry. The results showed that the infected AECs of Chrna7−/− mice were significantly decreased compared to Chrna7+/+ mice post PR8 infection (Figure 1K,L), consistent with the results of viral titer and viral gene expression. These results of α7 nAChR knockout mice indicate that deficiency of α7 nAChR dampens IAV infection in AECs of the lung.

3.2 Specific deletion of Chrna7 in type II AECs attenuates influenza viral infection
Given that α7 nAChR is ubiquitously expressed in many cell types and tissues, the conventional knockout mice are unspecific and insufficient to justify the role of α7 nAChR in AECs during influenza viral infection. Therefore, we further generated conditional Chrna7 floxed (Chrna7f/f) mice by CRISPR-Cas9 technology (Supporting Information S1: Figure S1B,C). By crossing with SftpcCre mice, Chrna7 gene was specifically deleted in type II AECs in the SftpcCreChrna7f/f mice as we previously showed.13 After PR8 infection, we found that SftpcCreChrna7f/f mice had improved survival and body weight loss compared to control littermates (Figure 1M–O). Similar to the results of whole-body knockout mice, SftpcCreChrna7f/f mice showed decreased viral loads in the lung compared to control mice (Figure 1P–S). In addition, PR8-induced inflammatory responses were also reduced in SftpcCreChrna7f/f mice compared to Chrna7f/f mice (Supporting Information S1: Figure S1D), consistent with the results of whole-body knockout mice (Supporting Information S1: Figure S1A). These results indicate that a deficiency of α7 nAChR in AECs is sufficient to render protection by reducing IAV infection in the lung.
Moreover, to test the role of vagal circuits in mediating α7 nAChR signaling during IAV infection, we performed unilateral vagotomy in the mice and then infected these mice with PR8 (Supporting Information S1: Figure S2A). However, the survival and lung viral load at 6 dpi had no significant differences between sham and vagotomized mice (data not shown), which may be due to the complexed effect on the body function after vagotomy. Interestingly, we found that the viral loads, BALF cell number, and infected AECs in the lung of vagotomized mice were significantly lower compared to those of sham mice at 2 dpi (Supporting Information S1: Figure S2B–E), indicating that loss of vagal circuits dampens influenza infection in AECs. Taken together, these findings support that deficiency of vagal-α7 nAChR signaling attenuates influenza infection in lung AECs and suggest that α7 nAChR activation in AECs may promote influenza infection.
3.3 Activation of α7 nAChR enhances influenza viral infection in vitro
Next, to test our hypothesis that α7 nAChR activation in AECs promotes influenza infection, we utilized A549 cells, a commonly used cell line of type II AECs, for the in vitro infection study. We first confirmed the expression of α7 nAChR in A549 cells by flow cytometry and staining with fluorescence-labeled α-Bungarotoxin (Figure 2A,B), a snake neurotoxin and competitively binds and blocks α7 nAChR. Furthermore, we treated A549 cell with GTS-21, a specific α7 nAChR agonist. The specificity of this agonist was previously validated through knockout mice in our study.24 Before IAV infection, A549 cells were treated with GTS-21 for 30 min to set up a context of α7 nAChR activation before the infection (Figure 2C). Furthermore, the results of Western blot showed that treatment of GTS-21 increased the amount of intracellular viral protein in A549 cells (Figure 2D,E), which was further confirmed by analysis of PR8 M gene expression in cells and viral titers in the supernatant (Figure 2F,G). Consistently, flow cytometry analysis of PR8 NP+ cells also supported the role of α7 nAChR activation in promoting IAV infection (Figure 2H,I). Additionally, treatment with a second α7 nAChR specific agonist, PHA568487, yielded similar effects in enhancing IAV infection (Figure 2J). On the contrary, when we treated A549 cells with a specific α7 nAChR antagonist, methyllycaconitine citrate, to antagonize its effect (Figure 2K), IAV infection was significantly inhibited (Figure 2L,M). Taken together, these results support our hypothesis regarding the function of α7 nAChR activation in promoting IAV infection.

3.4 Inactivation of ASK1-p38MAPK signaling mediated by α7 nAChR activation promotes influenza viral infection in vitro
To explore the underlying mechanism, we performed bulk RNA sequencing (RNA-seq), including three groups: 1. PBS-PBS: treated with vehicle and mock infection; 2. PBS-IAV: treated with vehicle and IAV infection; 3. GTS-IAV: treated with GTS-21 and IAV infection (Figure 2N). Influenza infection and activation of α7 nAChR caused a significant alteration of the epithelial transcriptome (Figure 2O; Supporting Information S1: Figure S3A,B). Probing deeper, we found that influenza infection induced markedly higher expression of inflammatory genes compared to noninfected cells (Supporting Information S1: Figure S3B), indicating an efficient establishment of IAV infection. By performing the pathway enrichment analysis of the differentially expressed genes (DEGs) induced by IAV, we identified a variety of virus- and infection-related pathways (Supporting Information S1: Figure S3C). Previous studies showed that in vitro influenza infection induced activation of p38 mitogen-activated protein kinase (p38MAPK), which functions as an important host factor for viral infection.25 Also, our results showed that multiple MAPK cascade-related pathways were enriched with the DEGs under influenza infection (Supporting Information S1: Figure S3C). Interestingly, during influenza infection, α7 nAChR activation further influenced MAPK cascade-related pathways (Figure 2P). Furthermore, gene set enrichment analysis showed that influenza infection induced significant activation of MAPK pathway compared to noninfected condition (Figure 2Q). Conversely, α7 nAChR activation suppressed MAPK pathway during influenza infection (Figure 2R). Taken together, these results indicate that MAPK cascade-related pathways are activated by influenza infection but suppressed by α7 nAChR activation, suggesting a potential role of MAPK pathways in α7 nAChR signaling-regulated influenza infection.
Next, to determine the role of p38MAPK, we first measured the changes of p-p38MAPK. In line with our RNA-seq data, the findings revealed that PR8 infection heightened p-p38MAPK levels in A549 cells, which were subsequently downregulated by GTS-21 treatment, without altering the total levels of p38MAPK (Figure 3A–C). To specifically identify the role of p38MAPK in influenza infection, we performed lentivirus-mediated knockdown of MAPK14 (the encoding gene of p38αMAPK, which is a major subtype of p38MAPK family) in A549 cells. The results showed that knockdown of MAPK14 significantly increased the levels of IAV infection (Figure 3E,F). On the contrary, overexpression of MAPK14 resulted in inhibition of the infection; however, this inhibitory effect was reversed when overexpressing MAPK14 with mutations at phosphorylation sites Thr180 and Tyr182, which are critical for p38MAPK activity (Figure 3G,H). These findings support the role of p38MAPK signaling mediated by α7 nAChR in regulating IAV infection. Next, we identified that α7 nAChR activation dose-dependently decreased the total amount of ASK1 (Figure 3A,D), which can mediate phosphorylation and activation of p38MAPK (Figure 3I). Furthermore, the knockdown of ASK1 gene reduced IAV-induced p38MAPK signaling (Figure 3J–L) and simultaneously enhanced IAV infection (Figure 3J,M), consistent with the results of MAPK14 knockdown. The effects of ASK1 and p38MAPK on IAV infection were further validated by IF antibody staining and flow cytometry measurement of IAV infected cells (Supporting Information S1: Figure S4A–D). Additionally, we detected the pattern of ASK1-p38MAPK signaling in vivo by measuring the protein levels in IAV-infected lung. Consistent with in vitro results, Chrna7−/− mice showed increased ASK1-p38MAPK signaling in the lung compared to Chrna7+/+ mice, accompanied by reduced IAV PB2 protein (Supporting Information S1: Figure S4E–I). Taken together, these results indicate that impaired ASK1-p38MAPK signaling mediated by α7 nAChR activation contributes to promoting influenza viral infection in AECs.

3.5 Activation of α7 nAChR facilitates NEDD4L-mediated ubiquitination and degradation of ASK1
Because the transcripts of MAPK14 and ASK1 were not affected by α7 nAChR activation during IAV infection (Supporting Information S1: Figure S5A,B), to further test how α7 nAChR regulates ASK1 levels, we tested the role of ubiquitin-proteasome pathway in posttranslational ASK1 degradation. We found that treatment of MG-132 (a proteasome inhibitor) reversed α7 nAChR-induced ASK1 degradation (Figure 4A), suggesting that activation of α7 nAChR may result in ASK1 ubiquitination. Of note, ASK1 possesses the “PPxY” motif (Supporting Information S1: Figure S5C), a catalytic target of E3 ubiquitination ligase NEDD4L (encoded by NEDD4L gene) (Supporting Information S1: Figure S5D).26 Furthermore, we observed that knockdown and overexpression of NEDD4L regulated ASK1 and p-p38MAPK in a dose-dependent manner (Figure 4B,C), making it possible that α7 nAChR may regulate ASK1-p38MAPK signaling through NEDD4L during IAV infection. Moreover, knockdown of NEDD4L enhanced infection-related ASK1-p38MAPK signaling and reduced IAV infection (Figure 4D–G); on the contrary, overexpression of NEDD4L attenuated ASK1-p38MAPK signaling but boosted IAV infection (Figure 4H–K). IF antibody staining and flow cytometry analysis confirmed the role of NEDD4L in IAV infection (Supporting Information S1: Figure S5E–H). In sum, these findings support the important role of NEDD4L in IAV infection by regulating ASK1-p38MAPK signaling. By performing co-immunoprecipitation (co-IP) in A549 cells, we confirmed the endogenous interaction between NEDD4L and ASK1 (Figure 4L). Furthermore, we overexpressed NEDD4L and ASK1 in 293 T cells and then performed co-IP. Similarly, the results supported the interaction between NEDD4L and ASK1 (Figure 4M). By his-pull-down analysis, we confirmed that NEDD4L can mediate significant ASK1 ubiquitination (Figure 4N) but not p38MAPK ubiquitination (Supporting Information S1: Figure S5I), supporting the role of NEDD4L in ASK1 ubiquitination and degradation. Of note, NEDD4L-mediated ASK1 ubiquitination was further enhanced by α7 nAChR activation (Figure 4N), indicating that α7 nAChR activation upregulates NEDD4L activity. Taken together, these results support the hypothesis that activation of α7 nAChR enhances NEDD4L-mediated ASK1 ubiquitination and degradation, thereby promoting IAV infection by limiting p-p38MAPK signaling.

3.6 Activation of α7 nAChR promotes the tyrosine phosphorylation of NEDD4L by inhibiting PTP1B
Levels of tyrosine (Tyr) phosphorylation of NEDD4L reflect its ubiquitin ligase activity.27 Detection of Tyr phosphorylation in immunoprecipitants of NEDD4L showed that activation of α7 nAChR increased Tyr phosphorylation of NEDD4L (Figure 5A), suggesting a role of α7 nAChR in modulating NEDD4L activity by promoting its Tyr phosphorylation. Activation of α7 nAChR in fibroblasts enhanced TGF-β signaling by regulating PTP1B (encoded by PTPN1 gene),24 an important negative regulator of tyrosine phosphorylation and interferon signaling pathway during influenza infection.28, 29 Moreover, we identified that activation of α7 nAChR dose-dependently increased phosphorylation levels of PTP1B at Ser378 site (Figure 5B,C), which is inversely correlated with PTP1B activity.30 PTP1B activity assays confirmed the inhibitory effect of α7 nAChR activation on its tyrosine phosphatase activity (Figure 5D). In line with the previous results, knockdown of PTPN1 decreased ASK1-p38MAPK signaling (Figure 5E–G) but increased IAV infection (Figure 5E,H), supporting the important role of PTP1B in mediating α7 nAChR and ASK1-p38MAPK signaling during IAV infection. Additionally, IF antibody staining and flow cytometry analysis confirmed the role of PTP1B in α7 nAChR signaling and IAV infection (Supporting Information S1: Figure S6A–D). Interestingly, we found that knockdown of PTPN1 significantly increased interferon signaling in response to IAV infection (Supporting Information S1: Figure S6E), consistent with previous findings.29 Of note, the boosted antiviral interferon signaling induced by PTPN1 knockdown failed to limit IAV infection (Figure 5E,H), further supporting the importance of ASK1-p38MAPK signaling in controlling IAV infection. By performing Co-IP assays, we confirmed the interaction between α7 nAChR and PTP1B (Figure 5I) and the interaction between PTP1B and NEDD4L (Figure 5J) in A549 cells. Notably, activation of α7 nAChR attenuated the interaction between PTP1B and NEDD4L (Figure 5J), further validating our hypothesis that α7 nAChR regulates NEDD4L activity through PTP1B. Additionally, knockdown and overexpression of PTPN1 significantly altered the Tyr phosphorylation levels of NEDD4L (Figure 5K), supporting the role of PTP1B in modulating NEDD4L activity. Taken together, these results suggest that activation of α7 nAChR promotes tyrosine phosphorylation of NEDD4L through inhibiting PTP1B activity.

3.7 Activation of α7 nAChR facilitates export of influenza viral ribonucleoproteins (vRNPs) by reducing p-p38MAPK levels in the nucleus
Our RNA-seq data supported the role of α7 nAChR in regulating the process of nuclear export (Figure 2P). Importantly, the export of IAV viral ribonucleoproteins (vRNP) complex from nucleus is a key step of influenza viral life cycle. To test whether p38MAPK signaling is involved in vRNP export, we first analyzed p-p38MAPK levels in the nucleus. The results confirmed the presence of p-p38MAPK in the nucleus by knockdown and overexpression of MAPK14 (Figure 6A). Furthermore, co-IP analysis revealed the interaction between p-38MAPK and IAV NP protein (Figure 6B), suggesting a possible role of p38-MAPK in regulating nuclear export of IAV vRNP complex. CRM1 (encoded by XPO1 gene) is critical for influenza viral vRNP nuclear transport and completing viral life cycle.31 Co-IP analysis confirmed the interaction between NP and CRM1 in nucleus (Figure 6C). To figure out the effect of p38MAPK on the interaction of NP and CRM1, we co-overexpressed p38MAPK, IAV NP, and CRM1 in 293 T cells followed by co-IP analysis of NP. We found that NP was able to interact with CRM1; however, overexpression of p38MAPK reduced the interaction of NP and CRM1 (Figure 6D), supporting the role of p38MAPK in reducing the NP-CRM1 interaction and nuclear export of IAV vRNP complex. Additionally, by comparing the effect of MAPK14 knockdown and overexpression, we confirmed the role of p38MAPK in negatively regulating the interaction of NP and CRM1 (Figure 6E). Finally, we showed that activation of α7 nAChR by GTS-21 and MAPK14 knockdown contributed to nuclear export of vRNPs staining in A549 cells; on the contrary, MAPK14 overexpression limited this process (Figure 6F,G). Taken together, these results support our hypothesis that activation of α7 nAChR contributes to the nuclear export of influenza vRNP complex by reducing p-p38MAPK signaling, which eventually results in increased influenza viral infection in AECs.

4 DISCUSSION
In this study, we found that activation of α7 nAChR led to a decrease in the phosphorylation levels of p38MAPK during influenza infection. The attenuated p38MAPK phosphorylation can facilitate influenza infection by promoting the nuclear export of influenza vRNP (Supporting Information S1: Figure S7). These findings unveil a novel mechanism through which influenza viruses exploit the host vagal system to enhance their infection. Our research sheds light on the intricate communication between the vagus nervous system and the immune system during influenza infection.
We observed that vagotomy resulted in a reduction of influenza replication in lung epithelial cells during the early stages, specifically 2–3 days post infection (Supporting Information S1: Figure S2). At this stage, acetylcholine-producing CD4 T cells13 have not been recruited into infected lungs (Supporting Information S1: Figure S2F,G), suggesting that the facilitating effect on influenza replication may mainly be attributed to the vagus nervous system itself. Furthermore, it is important to note that lung epithelial cells also express CHAT, enabling the synthesis of acetylcholine and the upregulation of Chrna7 expression in response to influenza infection (Figure 1J). Therefore, various factors may contribute to influenza replication under physiological or pathological conditions.
For certain patients, influenza viral infections can persist for extended durations and lead to severe disease, prompting exploration into whether vagus nerve and α7 nAChR signaling are implicated in this phenomenon. Blocking the vagus nerve and targeting α7 nAChR signaling could potentially help to limit infection during the initial 2 to 3 days following influenza infection. Conversely, during the later stages of infection (5–7 days), activation of α7 nAChR may exert anti-inflammatory effects. At this stage, inhibition of vagus nerve activity may not be conducive to reducing inflammation. As such, when bringing these findings from bench work to clinical bedside applications, the comprehensive effect of α7 nAChR signaling should be considered and balanced carefully.
The α7 nAChR receptor serves as a pivotal mediator in facilitating communication between the vagus nervous system and the lung. Our previous research has underscored the crucial role of vagal-α7 nAChR signaling in various lung diseases, including E. coli and LPS-induced ALI,9, 13 bleomycin-induced lung fibrosis,24 and allergic asthma.32 Remarkably, the impact of vagal-α7 nAChR signaling varies across different lung diseases, exhibiting unique effects. For instance, contrary to its role in promoting influenza infection, the activation of vagal-α7 nAChR signaling mitigates lung inflammation in LPS-induced ALI, showcasing an opposing effect. This disparity suggests that different pathogens elicit distinct responses due to substantial variations in the host factors involved in their life cycles. Moreover, our recent findings indicate that activation of α7 nAChR promotes HIV-1 transcription,33 but suppresses SARS-CoV-2 infection,11 further supporting our hypothesis. Overall, these findings complement the current theory of “the inflammatory reflex” in infections and inflammation.
As major respiratory pathogens, the activation of vagus-α7 nAChR signaling demonstrates opposing effects on SARS-CoV-2 replication compared to influenza in lung epithelial cells. This finding underscores the importance of focusing on patients concurrently infected with both viruses. A retrospective study revealed a remarkably high co-infection rate of SARS-CoV-2 and influenza viruses, reaching 57.3%, with 49.8% attributed to IAV co-infection.34 These patients should be carefully evaluated for vagal tone changes and nicotinic receptor interference. Additionally, it has been reported that GTS-21 is an antagonist for α4β235 and 5-HT3A.36 In our previous study, we confirmed that GTS-21 could specifically activate α7 nAChR to promote TGF-β1-induced lung profibrotic genes by using Chrna7 knockout mice.24 Thus, the specificity of GTS-21 on α7 nAChR has been validated.
When confronted with pathogen infections, the reaction of the nervous system represents the quickest and most direct influence on the infection process within the human body. Previous studies have shown that influenza viruses can invade the CNS via the vagus nerve in the lungs of mice.16 Considering our findings, it is plausible to speculate that the influenza virus may exploit the vagus nervous system to enhance the release of neurotransmitters, such as acetylcholine, thereby facilitating its replication and infection in the lungs. Understanding the mechanism of how the influenza virus exploits vagus nerve to facilitate its infection is important for revealing the evolvement trajectory of influenza–host interaction. Apart from the vagus nerve of parasympathetic reflex, what role of the other nervous components, such as sympathetic nerves, which are also important for the lung function, is a fascinating future research direction.
α7 nAChR has one of the highest permeabilities to calcium among all the nAChRs.37 Intracellular calcium signaling participates in the influenza viral life cycle by regulating viral internalization and infection.38 This prompts a significant question: can the activation of α7 nAChR elevate intracellular calcium levels in lung epithelial cells and thereby modulate the severity of influenza infection? Moreover, NEDD4L contains a C2 domain at N terminal (Supporting Information S1: Figure S5B), which can bind with calcium to enhance the activity of NEDD4L E3 ubiquitin ligases.39 Therefore, we hypothesize that α7 nAChR might regulate influenza infection by influencing intracellular calcium signaling, in addition to the PTP1B-NEDD4L-ASK1-p38MAPK pathway. The role of calcium signaling mediated by α7 nAChR activation needs future investigation.
A variety of viruses are known to activate p38MAPK signaling, with the consensus being that phosphorylation of p38MAPK is pivotal for both inflammatory responses and viral infections.25 However, previous studies utilizing different p38MAPK inhibitors have yielded conflicting results regarding the impact of p38MAPK on influenza infection in epithelial cells.40, 41 These discrepancies may stem from variations in the specificity and efficacy of different inhibitors. In our study, we employ genetic tools to precisely manipulate p38MAPK-related signaling, thereby elucidating the role of p38MAPK signaling in limiting influenza infection. Another explanation for this controversy is the existence of a threshold for influenza virus-induced p38MAPK signaling in epithelial cells, which can dictate its effect on infection levels. Moreover, the activation of α7 nAChR may alter the threshold of p38MAPK, consequently promoting infection by downregulating p38MAPK signaling.
In conclusion, our study reveals the mechanism by which influenza infection is facilitated via vagal-α7 nAChR signaling. These findings imply that the interaction between the influenza virus and the vagus nerve could influence the severity of the disease. The excitability of the vagus nerve and its receptor expression should be considered at different stages of influenza virus infection, which may be helpful in the treatment of influenza virus-induced ALI.
AUTHOR CONTRIBUTIONS
Caiqi Zhao, Mengyao Pan, Jie Chen, and Ling Li performed the experiments and analyzed data. Michael A. Matthay discussed, edited the manuscript, and provided funding. Wenjun Liu provided gene constructs for XPO1. Haichao Wang discussed and edited the manuscript. Yan Zhang built up α7 nAChR flox mice. Xia Jin edited the manuscript and provided funding. Xiao Su and Jin-fu Xu conceived and supervised the experiments, analyzed the results, provided funding, and wrote the manuscript.
ACKNOWLEDGMENTS
The authors thank Dr. K. Xu for providing SpcCre mouse line; D. Zhou for providing PR8 strain; and B. Lin, C. Wang, and X. Sun for their technical support and helpful discussions. This work is supported by the National Natural Science Foundation of China (82241042; 81730001; 81970075; 81900010), National Key Research and Development Program (YS2022YFC2304702), Science and Technology Commission of Shanghai Municipality (20DZ2261200), Innovative Research Team of High-level Local Universities in Shanghai (SHSMU-ZDCX20210602), and NHLBI (R01 HL134828).
CONFLICT OF INTEREST STATEMENT
The authors declare no conflict of interest.
Open Research
DATA AVAILABILITY STATEMENT
All data are available in the main text or the supporting information. All the cell lines generated in this study are available from the authors.