Abstract
The immune and sensory nervous systems, having evolved in parallel, communicate through shared receptors and transmitters to maintain homeostasis and respond to both external and internal disruptions. Although neural responses often confer protective benefits, they can also exacerbate inflammation during allergic reactions such as asthma. In our study, we modeled pollution-exacerbated asthma by exposing mice to ambient PM2.5 particles alongside ovalbumin. Compared to exposure to ovalbumin alone, this co-exposure significantly increased the numbers of neutrophils and γδ T cells in bronchoalveolar lavage fluid. We found that silencing nociceptor neurons at the peak of inflammation using intranasal QX-314 or ablating TRPV1-expressing neurons reduced lung neutrophil accumulation. Live in vivo intravital imaging confirmed that neuronal ablation reduced neutrophil numbers and increased their net displacement capacity. In neurons isolated from mice with pollution-exacerbated asthma, the chemical-sensing TRPA1 channel exhibited heightened sensitivity to its cognate ligand. Elevated levels of artemin were detected in the bronchoalveolar lavage fluid of pollution-exposed mice but returned to baseline in mice with ablated nociceptor neurons. Alveolar macrophages expressing the pollution-sensing aryl hydrocarbon receptor were identified as a putative source of artemin following exposure to PM2.5. This molecule enhanced TRPA1 responsiveness and, in turn, drove nociceptor-mediated neutrophil recruitment, revealing a novel mechanism by which lung-innervating neurons respond to air pollution in the context of allergy. Overall, our findings suggest that targeting artemin-driven pathways could provide a therapeutic strategy for controlling neutrophilic airway inflammation in asthma, a clinical condition typically refractory to treatment.
Introduction
Organisms have evolved sophisticated fail-safe systems to preserve homeostasis, integrating threat detection, reflex responses, and tailored immune mechanisms1. These systems are jointly orchestrated by the immune and sensory nervous systems, which are designed to sense external threats and internal disturbances. Both systems rely on shared metabolic pathways and signaling molecules—including receptors, cytokines, and neuropeptides 2. This common molecular framework supports constant interactions between these systems, which play pivotal roles in monitoring barrier tissues (e.g., skin 3, lung4,5, and gut6), activating anticipatory immune responses7, and managing pathologies such as allergies8,9, infections10, and malignancies11–14.
In the context of lung disease, vasoactive intestinal peptide (VIP), released by pulmonary sensory neurons following IL-5 stimulation, fosters allergic inflammation by acting on CD4⁺ T cells and innate lymphoid type 2 cells (ILC2s). This action elevates T helper type 2 (Th2) cytokines, which are central drivers of asthmatic responses15,16. Expanding on these insights, our work shows that vagal nociceptor neurons contribute to airway hyperresponsiveness1,17 mucus metaplasia18,19, and the detection of immunoglobulins E and G20,21. These neurons also mediate antibody class switching in B cells22 and promote IgG production23. More recently, we discovered that a subset of nociceptors is reprogrammed by the asthma-associated cytokine IL-13, rendering them susceptible to the inhibitory influence of neuropeptide Y (NPY) secreted by lung-innervating sympathetic neurons24.
Wildfire frequency and severity increased by 2.2-fold within the last 20 years 25. Between 2008 and 2018 in California, wildfire-attributable PM2.5 exposure caused an estimated ∼53,000 premature deaths26. Combined with urban pollution, these events have driven escalating levels of particulate matter (PM2.5), shifting asthma phenotypes from more responsive T helper 2 (Th2) / eosinophilic types to treatment-resistant neutrophilic and mixed Th17/Th1 variants27–32. Thus, recent estimates suggest that neutrophilic asthma accounts for 15–25% of all asthma cases, with about half of these resistant to standard therapies33.
To investigate whether neuronal silencing can mitigate neutrophilic airway inflammation, we used an innovative asthma model which combines ovalbumin with fine particulate matter (FPM). We further explored the molecular mechanisms underlying heightened neuronal sensitivity in this setting, identifying artemin—produced by alveolar macrophages via the pollution-sensing aryl hydrocarbon receptor (AhR)—as a critical mediator of this hypersensitivity.
Results
Heightened nociceptor sensitivity in pollution-exacerbated asthma
Prof. Ya-Jen Chang’s group developed a novel method to model pollution-exacerbated asthma and investigate the impact of fine particulate matter (FPM) on airway inflammation 34. Their work demonstrates that FPM exposure induces airway hyperreactivity and neutrophilic inflammation, promotes Th1 and Th17 immune responses, and increases epithelial cell apoptosis rates 34. Notably, γδ T cells significantly contribute to both the inflammation and airway hyperreactivity phenotypes by producing IL-17A 34. Building on this foundation, we explored whether lung-innervating nociceptor neurons become sensitized during such asthma exacerbations. To test this, male and female C57BL/6 mice (6–10 weeks old) were sensitized with an intraperitoneal injection of ovalbumin (OVA; 200 µg/dose) and aluminum hydroxide (1 mg/dose) emulsion on days 0 and 7, followed by intranasal challenges of OVA (50 µg/dose), with or without FPM (20 µg/dose), on days 14–16. On day 17, lung-innervating jugular-nodose-complex (JNC) neurons were harvested, cultured for 24 hours, and loaded with the calcium indicator Fura-2AM (Fig. 1A). Stimulation with the TRPA1 agonist AITC (10–100 µM) and the pan-neuronal activator KCl (40 mM) revealed increased responsiveness in neurons from mice exposed to both FPM and OVA, compared to OVA-only–exposed mice, whose sensitivity was similar to that of wild-type controls (Fig. 1B–C).

Air pollution exacerbates nociceptor neuronal activity
(A–C) Male and female C57BL/6 mice (6–10 weeks old) were sensitized via intraperitoneal injection with an emulsion of ovalbumin (OVA; 200 µg/dose) and aluminum hydroxide (1 mg/dose) on days 0 and 7. On days 14–16, mice were challenged intranasally with OVA (50 µg/dose), either alone or in combination with fine particulate matter (FPM; 20 µg/dose). Bronchoalveolar lavage fluid was collected, and jugular-nodose complex neurons were cultured on day 17 for 24 hours before being loaded with the calcium indicator Fura-2AM. Cells were sequentially stimulated with the TRPA1 agonist AITC (successively to 10 µM at 60–90 seconds, 30 µM at 90–120 seconds, 100 µM at 120–150 seconds) and then with KCl (40 mM at 420–435 seconds). Calcium flux was continuously monitored throughout the experiment. The amplitude of AITC responses was measured by calculating the ratio of peak F340/F380 fluorescence after stimulation to the baseline F340/F380 fluorescence measured 30 seconds prior to stimulation. Data are plots as the per dish average of AITC and KCl responsive neurons and show that AITC (10 µM) responses was higher in JNC neurons from OVA-FPM exposed mice when compared to vehicle or OVA alone (C). Data in are presented as means ± SEM (B-C). N are as follows: B: n = 35 neurons (control group), 19 neurons (OVA group), and 38 neurons (OVA + FPM group), C: n = 5 dishes totalling 35 neurons (control group), 8 dishes totalling 42 neurons (OVA group), and 10 dishes totalling 76 neurons (OVA + FPM group). P-values were determined by nested one-way ANOVA with post-hoc Bonferroni’s. P-values are shown in the figure.
Transcriptomic reprogramming of TRPV1⁺ neurons
Recent data indicate that the asthma-associated cytokines IL-4 and IL-13 can reprogram lung-innervating nociceptor neurons to adopt a pro-allergic phenotype 24. To investigate whether pollution-exacerbated asthma similarly reprograms these neurons, we used TRPV1cre::tdTomatofl/wt mice subjected to our standard OVA protocol, with or without FPM. We isolated and dissociated TRPV1⁺ JNC neurons, enriched them by excluding satellite glial and immune cells, and then purified them by FACS-sorting followed by RNA sequencing. Our analysis revealed that several differentially expressed genes (DEGs) were upregulated in the OVA-FPM group compared to naive mice (Lifr, Oprm1) (Fig. 2A–B), in OVA-FPM compared to OVA alone (Oprm1, Nefh, P2ry1, Prkcb, Gabra1, Kcnv1) (Fig. 2C–D), and in OVA alone compared to naive controls (Cntn1, Piezo1, Npy1r, Kcna1) (Fig. 2E–F). Collectively, these results suggest that pollution-exacerbated asthma both transcriptomically and functionally reprograms lung-innervating nociceptor neurons (Supplementary Table 1).

Air pollution reprograms the transcriptome of nociceptor neurons
(A–F) Naïve male and female TRPV1cre::tdTomatofl/wt mice (6–10 weeks old) underwent either a pollution-exacerbated asthma protocol, the classic OVA protocol, or remained naïve. On day 17 (peak inflammation), jugular-nodose-complex (JNC) neurons were harvested and dissociated, and TRPV1⁺ neurons (tdTomato⁺) were sorted via FACS to remove stromal cells and non-peptidergic neurons. RNA was then isolated for sequencing.
Volcano plots (A, C, E) and heatmaps (B, D, F) show differentially expressed genes (DEGs) for three comparisons: OVA + FPM vs. naïve (A-B), OVA + FPM vs. OVA alone (C-D), and OVA alone vs. naïve (E-F). Notable genes with increased expression include Lifr and Oprm3 in OVA + FPM vs. naïve, Oprm1, Nefh, P2ry1, Prkcb, Gabra1, and Kcnv1 in OVA + FPM vs. OVA, and Npy1r and Kcna1 in OVA alone vs. naïve.
Data are presented either as volcano plots (A, C, E), showing the log2 fold change of TPM between groups along with the corresponding –log10 p-values from DESeq2 analysis, or as heatmaps (B, D, F), showing the z-scores of rlog-transformed normalized counts. The experimental groups were naïve (n=2; A–B, E–F), OVA (n=3; C–F), and OVA-FPM (n=3; A–D). P-values were determined by DESeq2 (A, C, E) and are indicated in the figure.
Silencing nociceptor neurons reduces inflammation
To determine whether silencing nociceptor neurons affects pollution-exacerbated asthma, we adapted a previously established neuron-blocking strategy originally developed for pain and itch neurons. This method uses nonselective ion channels (TRPA1 and TRPV1) as a drug entry port for QX-314, a charged form of lidocaine. During inflammation, QX-314 enters sensory fibers, blocking sodium currents and producing a targeted, long-lasting (>9 hours) electrical blockade of nociceptors without impairing immune cell function35–38. Intranasal administration of QX-314 has already been shown to reverse allergic airway inflammation, coughing, mucus metaplasia, and hyperreactivity16,36,39–41.
In our model, an intranasal dose of QX-314 administered at the peak of inflammation (day 16, 5 nmol, 50 µL) significantly reduced neutrophil counts in bronchoalveolar lavage fluid (BALF) of OVA-FPM-exposed mice, restoring them to levels typically seen in asthmatic (OVA-only) mice (Fig. 3A–B). To validate these findings, we used mice genetically engineered to either retain (TRPV1wt::DTAfl/wt; denoted as TRPV1WT) or lack (TRPV1cre::DTAfl/wt; denoted as TRPV1DTA) TRPV1-expressing nociceptors11,42,43. Compared with controls, TRPV1DTA mice had markedly fewer BALF neutrophils and lung γδ T cells (Fig. 3C–E), underscoring the critical role nociceptor neurons play in shaping the inflammatory and immune responses in pollution-exacerbated asthma.

Nociceptor neurons control pollution-exacerbated asthma
(A–B) Male and female C57BL/6 mice (6–10 weeks old) were sensitized intraperitoneally with ovalbumin (OVA; 200 µg/dose in 200 µL) and aluminum hydroxide (1 mg/dose in 200 µL) on days 0 and 7. On days 14–16, mice were challenged intranasally with OVA (50 µg/dose in 50 µL) alone or with fine particulate matter (FPM; 20 µg/dose in 50 µL). On day 16, 30 minutes after the final challenge, mice received intranasal QX-314 (5 nmol/dose in 50 µL). Bronchoalveolar lavage fluid (BALF) was collected on day 17 and analyzed by flow cytometry. Compared with naïve or OVA-exposed mice, those co-challenged with OVA + FPM showed increased BALF neutrophils (A). QX-314 treatment normalized these levels, while BALF eosinophil levels remained comparable (B).
(C–E) Male and female littermate control (TRPV1WT) and nociceptor-ablated (TRPV1DTA) mice (6–10 weeks old) were sensitized and challenged under the same OVA ± FPM protocol (days 0, 7, and 14–16). BALF or lungs were collected on day 17 and assessed by flow cytometry. Compared with naïve or OVA-exposed mice, OVA + FPM co-challenged mice exhibited higher BALF neutrophils (C) and lung γδ T cells (E). Nociceptor ablation protected against these increases (C, E), while BALF eosinophil levels remained comparable (D).
Data are shown as mean ± SEM (A–E). Experiments were replicated twice, and animals pooled (A-E). N are as follows: A-B: control (n=6), OVA (n=7), OVA-FPM (n=12), OVA-FPM+QX-314 (n=10), C-D: TRPV1WT + control (n=9), TRPV1WT + OVA (n=13), TRPV1WT + OVA-FPM (n=18), TRPV1DTA + OVA-FPM (n=19), E: TRPV1WT + control (n=3), TRPV1WT + OVA (n=3), TRPV1WT + OVA-FPM (n=4), TRPV1DTA + OVA-FPM (n=5). P-values were determined by a one-way ANOVA with post-hoc Tukey’s (A-E). P-values are shown in the figure.
Impact on myeloid cell motility
Building on these observations, we used intravital microscopy44,45 to understand how pollution-exacerbated asthma and lung innervation influence the motility of alveolar macrophages (AMs) and neutrophils. Although the total number of AMs (labelled by i.n. PKH26) remained unchanged (Fig. 4A-B), their net displacement was lower in nociceptor-ablated (NaV1.8cre::DTAfl/wt; denoted as NaV1.8DTA) mice than in littermate controls (NaV1.8wt::DTAfl/wt; denoted as NaV1.8WT; Fig. 4C-F, Supplementary Video 1). In contrast, neutrophil accumulation (labelled by i.v. αLy6G) was observed in pollution-exposed, asthmatic littermate controls, but was absent in nociceptor-ablated mice (Fig. 4G-H). These data are consistent with our BALF flow cytometry data (Fig. 3A, 3C). Furthermore, total neutrophil displacement was higher in NaV1.8DTA mice (Fig. 4I–L, Supplementary Video 2), without any bias toward a specific behavior subtype (e.g. adherent, crawling, patrolling, or tethering; Fig. 4L). Taken together, these data suggest that nociceptor neurons modulate AM motility and neutrophil accumulation while limiting the motility of recruited neutrophils.

Vagal sensory neurons gatekeep alveolar macrophage motility and neutrophil numbers
(A–D) Male and female littermate control (NaV1.8wt::DTAfl/wt denoted as NaV1.8WT) and nociceptor-ablated (Nav1.8cre::DTAfl/wt denoted as NaV1.8DTA) mice (6–10 weeks old) were sensitized via intraperitoneal injection with an emulsion of ovalbumin (OVA; 200 µg/dose) and aluminum hydroxide (1 mg/dose) on days 0 and 7. Phagocytes were labeled by intranasal injection of PKH26 Red Fluorescent Cell Linker Kit (25 pmol/dose) on day 10. Mice were then challenged intranasally with OVA (50 µg/dose) alone or in combination with fine particulate matter (FPM; 20 µg/dose) on days 14–16, and images were acquired on day 17. (A) Representative maximum-intensity projection of alveolar macrophages (AMs, red). Scale bar: 20 µm. (B) Quantification of AM numbers per field of view (FOV). (C) Net displacement of AMs over 1 hour. (D-F) Representative tracks of individual AMs (each color represents a single AM) over 1 hour. While the AM numbers (A-B) were not impacted, their net displacement (C-F) was reduced in OVA-FPM-exposed NaV1.8DTA mice.
(G-L) Male and female littermate control (NaV1.8wt::DTAfl/wt denoted as NaV1.8WT) and nociceptor-ablated (NaV1.8cre::DTAfl/wt denoted as NaV1.8DTA) mice (6–10 weeks old) were sensitized via intraperitoneal injection with an emulsion of ovalbumin (OVA; 200 µg/dose) and aluminum hydroxide (1 mg/dose) on days 0 and 7. Mice were then challenged intranasally with OVA (50 µg/dose) alone or in combination with fine particulate matter (FPM; 20 µg/dose) on days 14–16, and images were acquired on day 17. Immediately prior to perform the intravital imaging, we administered an intravenous Ly6G antibody to label neutrophils. (G) Representative maximum-intensity projection of neutrophils (black). Scale bar: 20 µm. (H) Quantification of neutrophil numbers per FOV. (I) The total displacement of neutrophils over 20 minutes. (J-K) Representative tracks of individual neutrophils (each color represents a single neutrophil) over 20 minutes. (L) Frequency of neutrophil behaviors per FOV (adherent, crawling, patrolling, or tethering). Data show that OVA-FPM-exposed littermate control mice present an increase in neutrophil numbers per field of view (G-H), an effect absent in NaV1.8DTA. Interestingly, OVA-FPM-exposed NaV1.8DTA mice show an increase in neutrophil net displacement (I-K), an effect irrespective to one of the specific behaviors tested (L).
Data are presented as representative image (A, G; scale bar: 20µm), mean ± SEM (B, C, H), spider plot (D-F, J-K), violin plot showing median (I), and staked bar graph showing mean ± SEM (L). N are as follows: B: NaV1.8WT + control (n=6), NaV1.8WT + OVA-FPM (n=6), NaV1.8DTA + OVA-FPM (n=6), C: NaV1.8WT + control (n=42), NaV1.8WT + OVA-FPM (n=42), NaV1.8DTA + OVA-FPM (n=42), H: NaV1.8WT + control (n=6), NaV1.8WT + OVA-FPM (n=6), NaV1.8DTA + OVA-FPM (n=6), I: NaV1.8WT + OVA-FPM (n=385), NaV1.8DTA + OVA-FPM (n=469), J: NaV1.8WT + OVA-FPM (n=10), NaV1.8DTA + OVA-FPM (n=11). P-values were determined by a one-way ANOVA with post-hoc Tukey’s (B, C, H) or unpaired Student T-test (I, L). P-values are shown in the figure.
Elevated cytokines and artemin under pollution-exacerbated asthma
To further explore the role of nociceptor neurons in airway inflammation, we performed an unbiased multiplex cytokine array. This revealed elevated levels of the asthma-promoting cytokine IL-4 and TNFα, with TNFα levels returning to baseline following nociceptor ablation (Fig. 5A). In addition, targeted ELISA showed increased artemin levels, which returned to normal in the absence of nociceptor neurons (Fig. 5B). Artemin, a member of the glial cell line-derived neurotrophic factor (GDNF) family, is pivotal for the development and function of sympathetic46 and sensory47 neurons through its interaction with the GFRα3-RET receptor complex. Activation of this receptor supports neuronal survival and growth during embryogenesis and drives neuronal hyperactivity under inflammatory conditions48–50.

Artemin sensitizes TRPA1 activity in vagal sensory neurons
(A-B) Male and female littermate control (TRPV1WT) and nociceptor-ablated (TRPV1DTA) mice (6–10 weeks old) were sensitized and challenged under the same OVA ± FPM protocol (days 0, 7, and 14–16). BALF was collected on day 17 and assessed by multiplex array and ELISA. Compared with naïve or OVA-alone groups, OVA + FPM co-challenged mice exhibited levels of TNFα, and artemin. Notably, ablating nociceptors prevented these increases.
(C) In-silico analysis of the GSE124312 dataset51. The heatmap displays transcript expression levels for the pan neural-crest lineage transcription factor (Prdm12), voltage-gated sodium channels (Scn9a, Scn10a), jugular subset markers (Wfdc2, Mrgprd, Osmr, Sstr2, Nefh, Trpm8), peptidergic neuron markers (Trpa1, Trpv1, Calca, Tac1, Gfra3), and the pan placodal lineage marker (Phox2b). Gfra3 expression is enriched in the peptidergic neuron cluster labeled JG4. Experimental details and cell clustering are described by Kupari et al51.
(D) In-silico analysis of GSE19298752 showing co-expression of Gfra3 with Trpa1 and other inflammatory markers. Data are visualized as row z-scores in a heatmap or via UMAPs (TPTT > 1). Experimental details and cell clustering are described by Zhao et al52.
(E-G) Alveolar macrophages (3 × 10⁵ cells/well) from naïve male and female C57BL/6 mice were cultured overnight and then stimulated with vehicle (DMSO) or FPM (100 µg/mL). RNA was extracted 1- and 4-hours post-stimulation and Artn expression was assessed using qPCR. FPM exposure increased Artn transcript levels at both 1 and 4 hours (F, G).
(H-J) Naïve mice jugular-nodose-complex neurons were harvested, pooled, and cultured overnight with either vehicle or artemin (100 ng/mL). Cells were sequentially stimulated with AITC (TRPA1 agonist; 300 µM at 240–270 seconds), capsaicin (TRPV1 agonist; 300 nM at 320–335 seconds), and KCl (40 mM at 720–735 seconds). The percentage of AITC-responsive neurons (among all KCl-responsive cells) was normalized to vehicle-treated controls for each batch of experiments. Artemin-treated neurons showed increased responsiveness to AITC, while responses to capsaicin and KCl were unchanged (I-J).
Data are presented as means ± SEM (A-B, F-G, J), heatmap displaying the z-score of DESeq2 normalized counts (C), tSNE plots (D), schematics (E, H), means ± 95% CI of maximum Fura-2AM (F/F₀) fluorescence (I). N are as follows: A: TRPV1WT + control (n=2), TRPV1WT + OVA (n=3) TRPV1WT + OVA-FPM (n=3), TRPV1DTA + OVA-FPM (n=8), B: TRPV1WT + OVA (n=6) TRPV1WT + OVA-FPM (n=8), TRPV1DTA + OVA-FPM (n=14), F: n=2/time point, G: n=8/group, I: vehicle (n=107 neurons), Artemin (n=122 neurons); J: n=4/group. P-values were determined by a one-way ANOVA with post-hoc Tukey’s (A, B) or unpaired Student T-test (G, J). P-values are shown in the figure.
Mechanistic insights: artemin–nociceptor axis
To determine which vagal neuron subsets, express the artemin receptor GFRα3–RET, we performed an in-silico analysis of single-cell RNA sequencing data51,52 and found that Gfra3 is expressed by several nociceptor subtypes (Fig. 5C–D), including JG3 (OSM-R–expressing), JG4 (TRPA1/SSTR2-expressing), and JG6 (TRPM8-expressing). Concurrently, ImmGen data53 indicated robust Artn expression in macrophages, with Ahr levels slightly lower than those in ILC2s and eosinophils (SF. 1). We then conducted an in-silico analysis of data from Karlsson et al.54 (SF. 2A), Uhlén et al. 55 (SF. 2B) and Abdulla et al. 56 (SF. 2C), showing that Ahr is also expressed in human lung, that AHR protein is detected in human lung macrophages, and that Artn and Ahr are co-expressed in lung macrophages, respectively. Guided by these findings, we isolated alveolar macrophages from naive C57BL/6 mice and exposed them to fine particulate matter (FPM), observing a time-dependent increase in Artn transcripts (Fig. 5E–G). These results highlight key cellular interactions involving FPM, AhR and artemin in the context of lung inflammation.
A recent study49 linked artemin overexpression to increased mRNA levels of several nociceptor markers (Gfra3, TrkA, Trpv1, Trpa1), accompanied by heightened thermal sensitivity. Consistent with this, our experiments in JNC neurons from OVA-FPM co-exposed mice revealed amplified calcium responses to the TRPA1 agonist AITC (Fig. 1B–C). Follow-up tests in naive C57BL/6 mice showed elevated AITC responsiveness in artemin-exposed JNC neurons compared to vehicle-treated controls (Fig. 5H–J). Collectively, these data suggest that alveolar macrophages, through pollutant-driven production of artemin, sensitize nociceptor neurons and thereby potentiate allergic airway inflammation. This pathway underscores a critical link between environmental pollutants and the neurogenic exacerbation of allergic responses (SF. 3).
Discussion
Our research, in line with other studies, indicates that nociceptor neurons promote regulatory immunity in the context of bacterial57, viral58, or fungal59 infections, as well as malignancies11. To our knowledge, however, the impact of neuro-immunity on neutrophilic asthma is less understood. Using a model of pollution-exacerbated airway inflammation34, we found that ablation or silencing of nociceptor neurons prevented the induction of neutrophilic airway inflammation, revealing a potentially novel therapeutic avenue for treating refractory asthma. This result parallels our earlier findings that nociceptor neuron silencing via charged blockers of voltage-gated sodium channels 16,22,35, or calcium channels 36 can halt eosinophilic airway inflammation.
During bacterial infections, Pinho-Ribeiro et al. demonstrated that nociceptor neurons inhibit neutrophil influx and antimicrobial activity by releasing the neuropeptide CGRP60. In addition, Ugolini and colleagues showed that ablating NaV1.8+ nociceptor neurons during HSV-1 infection increases skin neutrophil infiltration, cytokine production, and lesion severity 58. Building on these findings, we used intravital imaging to study neuro–immune interactions in our model, we observed that the total number of alveolar macrophages remained unchanged in OVA-exposed mice lacking nociceptor neurons, but their displacement was significantly reduced. In contrast, pollution-exposed asthmatic mice lacking nociceptor neurons exhibited a marked decrease in neutrophil recruitment and a corresponding increase in their displacement. These shifts in cellularity occurred regardless of whether nociceptor neurons were genetically ablated (TRPV1+ or NaV1.8+ populations) or pharmacologically silenced with QX-314, as confirmed by flow cytometry. Although individual neutrophil behaviors—such as adherence, crawling, patrolling, and tethering—did not change, neuronal ablation effectively “removed the brakes” on neutrophil motility. This likely led to reduced neutrophil accumulation in the lung and overall dampening of lung inflammation in pollution-exacerbated asthma. These findings are consistent with the pioneering work by Baral et al., who showed that vagal TRPV1+ afferents suppress neutrophil recruitment and modulate lung γδ T cell numbers via CGRP signaling, thus supporting essential antibacterial clearance4. Related studies in influenza-infected mice revealed that vagal nociceptor deficiency significantly alters the lung immune landscape— expanding neutrophils and monocyte-derived macrophages 5—and transcriptional analyses further indicated disrupted interferon signaling as well as imbalances among neutrophil subpopulations in nociceptor-ablated mice. Collectively, these results highlight the bidirectional crosstalk between recruited and tissue-resident phagocytes and nociceptors in air-pollution–exacerbated allergic airway inflammation.
The aryl hydrocarbon receptor (AhR) is a crucial modulator of inflammation, as seen in psoriasis models, where its activation reduces inflammation, whereas its absence exacerbates disease61,62. Besides being expressed in alveolar macrophages, AhR is also present in ILC2s, eosinophils, and neurons. A recent preprint suggests AhR has a dual role in neural protection and axon regeneration: AhR activation in dorsal root ganglion neurons inhibits axon growth63. At the same time, its deletion lessens inflammation and stress signaling, driving pro-regenerative pathways63. Additional work indicates that AhR regulates the gut-brain axis64. Although we have not directly examined AhR in nociceptor neurons, our findings suggest that neurons, much like alveolar macrophages, may sense fine particulate matter via AhR. This possibility could explain the direct neuronal transcriptomic reprogramming we observed after pollutant exposure. Ongoing studies aim to probe this hypothesis using nociceptor-specific AhR knockout models.
A growing body of evidence indicates that airway-innervating sensory neurons undergo significant transcriptomic alterations in neutrophilic asthma. Although some lung innervation arises from the dorsal root ganglia (DRG)—especially for nociception in the upper thoracic segments (T1–T5)—the majority (70–80%, sometimes up to 90%) of afferent fibers originate from the JNC 65,66. Because the JNC mediates critical mechanosensory and chemosensory functions, our analysis focuses on this primary vagal pathway. While single-cell RNA sequencing has revealed that JNC neurons are highly heterogeneous and control breathing and tracheal reflexes 67–72, the molecular changes that occur during chronic inflammation remain poorly characterized.
Using an NaV1.8 reporter mice and intranasal retrograde tracing, we identified a subset of nociceptors innervating the lungs that are reprogrammed in allergic airway inflammation and by the asthma-driving cytokine IL-1324. Our transcriptomic data using sorted TRPV1+ neurons confirm the upregulation of Npy1r during allergic airway inflammation24 and highlight the upregulation of Cacna1c73 and Dmxl274 during pollution-exacerbated asthma, suggesting heightened vesicle trafficking and changes in synaptic release dynamics that may influence neuronal sensitivity and plasticity in asthmatic conditions. Other genes such as Oprm175 (mu-opioid receptor) and Hint176 (an opioid signaling modulator) suggest altered pain or irritant sensation in the lung, potentially shaping cough reflexes or bronchospasm. Changes in cytoskeletal regulators, including Map1b77 and Apc78, indicate that JNC axons may remodel their structure and synaptic architecture, potentially altering how airway signals are relayed to central circuits. Lifr79–82 (Leukemia inhibitory factor receptor alpha), which supports neuron survival and differentiation, may also be important in inflammatory or injury contexts in the lung.
Pathway analysis reveals gene clusters involved in synaptic structure, cytoskeletal organization, and epigenetic regulation. Enrichment in terms such as “synapse”, “presynaptic active zone,” and “postsynaptic density” indicates that both neurotransmitter release and postsynaptic excitability are being recalibrated, potentially amplifying neuro-immune feedback loops and leading to maladaptive reflexes like excessive bronchoconstriction or persistent cough in neutrophilic asthma. In parallel, pathways related to “cellular component organization,” “negative regulation of inclusion body assembly,” and “protein quality control” show that neurons may upregulate protective or adaptive mechanisms in response to chronic airway inflammation. These transcriptomic alterations point to a dynamic neuro-immune crosstalk in the inflamed lung, with neurons adapting their excitability and structural integrity in response to cytokines, environmental pollutants, and inflammatory cells, potentially exacerbating or perpetuating the pathophysiology of neutrophilic asthma.
Alveolar macrophages act as a critical early warning system in the lung45, detecting pollutants such as FPM and initiating protective reflexes by secreting artemin. This cytokine activates and sensitizes nociceptor neurons—particularly TRPA1-expressing fibers—to noxious stimuli. Single-cell RNA sequencing identified a subset of TRPA1⁺, SSTR2⁺ neurons that express the glial cell line-derived neurotrophic factor receptor GFRα3 and are thereby sensitized by artemin. This finding aligns with earlier data showing that keratinocyte-derived TSLP can sensitize skin-innervating nociceptor neurons, promoting itch and atopic dermatitis83. In the lungs, germline TRPA1 knockout reduces allergic airway inflammation84,85, and clinical data from Genentech suggest that TRPA1 agonists are elevated in asthmatic human airways, contributing to both inflammation and hyperreactivity. GDC-0334, a selective TRPA1 antagonist they develop, mitigates airway inflammation, cough, and allergic responses in preclinical models and reduces pain and itch in humans86. These findings strongly support the central role of TRPA1 sensitization and nociceptor activation in driving asthma pathology.
Our prior work showed that nodose ganglion neurons, which are highly TRPA1⁺, exhibit heightened thermal sensitivity and outgrowth responses to brain-derived neurotrophic factor (BDNF) but do not respond to nerve growth factor (NGF)43. Here, we demonstrate that vagal nociceptors express GFRα3 and respond to artemin, which sensitizes TRPA1 channels and promotes subsequent airway inflammation. Indeed, jugular-nodose-complex (JNC) neurons undergo substantial transcriptomic reprogramming when exposed to OVA plus FPM, mirroring the effects of pro-asthmatic cytokines. Parallel data from other disease models support this link between inflammation-induced sensitization and elevated artemin. For example, increased Artn and EGR1 levels in atopic dermatitis correlate with enhanced nerve density and scratching, both of which are attenuated in EGR1-deficient mice87. Similarly, artemin overexpression boosts sensory neuron thermal responses, underscoring its importance in driving neuronal outgrowth and sensitivity in atopic conditions49. These observations raise the possibility that blocking GFRα3 may offer a novel strategy to prevent maladaptive nociceptor involvement in pollution-exacerbated asthma.
Our previous work showed that vagal nociceptor neurons can detect allergen-IgE immune complexes and respond by releasing substance P (SP) and vasoactive intestinal peptide (VIP), but not CGRP20. These findings were corroborated by analyzing bronchoalveolar lavage fluid (BALF) from asthmatic mice²⁴. Moreover, we demonstrated that SP drives mucus metaplasia in asthmatic lungs18 and influences antibody class switching in B cells22, a result that other groups have replicated23,39,88,89. We also observed that TRPV1⁺ nociceptor ablation impairs antigen trafficking to lymph nodes and reduces IgG production 23,90,91, possibly decreasing the severity or onset of allergic responses. Emerging work highlights the varied immunomodulatory roles of different neuropeptides. For instance, CGRP impairs dendritic cell migration in psoriasis 9,92, whereas SP enhances dendritic cell migration to lymph nodes in atopic dermatitis8. VIP and neuromedin U (NMU) boost pro-asthmatic cytokines from lung ILC2s16,93–96, while CGRP can have similar97,98 or opposing effects99. In lung infections, CGRP reduces neutrophil and γδ T cell infiltration and protects against Staphylococcus aureus pneumonia4,100. Conversely, CGRP exacerbates psoriasis by inducing dendritic cells to release IL-23, which activates IL–17–producing γδ T cells, intensifying inflammation59. Likewise, during C. albicans infection, sensory neurons release CGRP that stimulates IL-23 production in CD301b⁺ dendritic cells, triggering Th17 and γδ T cell responses (marked by IL-17A and IL-22), enhancing host defense7. In our model, nociceptor neuron ablation diminishes γδ T cell activation, which we posit is chiefly driven by SP/VIP, given the absence of heightened CGRP release in our asthma models16,22. Future work will investigate whether neurons directly regulate γδ T cells and neutrophils through these or other neuropeptides.
Previous reports show that TRPV1⁺ neuron activation elicits a local type 17 immune response that enhances host defense against Candida albicans59,101 and Staphylococcus aureus7. While our mixed OVA-FPM was expected to induce a strong Th17/Th2 component, we did not observe an increase in IL-17A in our cytokine bead array. Similarly, increases in classic asthma-driving cytokines were modest (IL-4, IL-5) or absent (IL-13). One possible explanation is that these experiments were conducted in C57BL/6 mice, rather than BALB/c mice34. We also noted an increase in TNFα and artemin, both of which were reduced by nociceptor neuron silencing. While we found that TNFα can reprogram nociceptor neurons24, we propose that this TNFα may originate from activated neutrophils102,103, consistent with the reduced TNFα and neutrophil activation seen in neuron-ablated mice. It is worth noting that the PM₂₅ used in this study—Standard Reference Material (SRM) 2786—was obtained from an air intake system in the Czech Republic and that we cannot rule out that it may contain trace levels of LPS that could contribute to this neutrophil–TNFα hypothesis. Our rationale for choosing SRM2786 was that it is commercially available and represents a broad spectrum of ambient air pollutants, in contrast to more specialized sources such as diesel exhaust particles. Future studies will need to validate these findings using wildfire-collected particles104, LPS-free air intake particles, and replication in BALB/c mice.
In summary, our study highlights how alveolar macrophages, via AhR-dependent pollutant detection and artemin secretion, sensitize TRPA1⁺ nociceptor neurons, thereby amplifying neutrophilic asthma. We propose several potential therapeutic strategies to curb neutrophilic airway inflammation: (i) targeting AhR signaling in alveolar macrophages, (ii) blocking Artemin’s interaction with GFRα3, (iii) using TRPA1 antagonists such as GDC-0334, and (iv) silencing nociceptor neurons with charged lidocaine derivatives. Collectively, these approaches offer new directions to manage the increasingly prevalent yet treatment-resistant, neutrophilic variant of asthma.
Materials and methods
Animals
All procedures involving animals adhered to the guidelines of the Canadian Council on Animal Care (CCAC), McGill University (xxx) and the Queen’s University Animal Care Committee (UACC, protocol 2384). Mice were housed in individually ventilated cages with free access to water and food under 12-hour light cycles.
Parental strains C57BL/6 (# 000664), DTAfl/fl (# 010527, # 009669), tdTomatofl/fl (# 007914), TRPV1cre/cre (# 017769) and NaV1.8cre/cre (# 036564) were purchased from The Jackson Laboratory. Male and female mice were bred in-house and used at 6–12 weeks of age. Crosses were performed to generate the following genotypes: NaV1.8cre/wt::DTAfl/wt, TRPV1cre/wt::DTAfl/wt, TRPV1cre/wt::tdTomatofl/wt, and littermate control, namely: NaV1.8wt/wt::DTAfl/wt, TRPV1wt/wt::DTAfl/wt. Male and female progeny mice were used between 8 and 16 weeks of age.
Ovalbumin Model of Allergic Airway Inflammation
On days 0 and 7, mice were sensitized by intraperitoneal (i.p.) injections of 200 μL of a solution containing 1 mg/mL grade V ovalbumin (OVA; Sigma, #A5503) and 5 mg/mL aluminum hydroxide (Sigma, #239186) in phosphate-buffered saline (PBS, ThermoFisher, #10010023). On days 14–16, mice were anesthetized with isoflurane (2.5%; CDMV, #108737) and intranasally instilled daily with 50 μg OVA in 50 μL PBS with or without fine particulate matter (FPM; 20 µg/dose; NIST 2786). Control mice were sensitized but did not undergo challenges. Unless otherwise indicated, mice were sacrificed on day 17.
Neuron silencing
In some experiments, QX-314 (Tocris, #1014; 5 nmol in 50 µL) or PBS was administered intranasally on day 16 to control asthmatic mice (as detailed in the asthma protocol). The mice were euthanized on day 17, and bronchoalveolar lavage fluid, lung tissues, and the jugular-nodose complex were collected.
Bronchoalveolar lavage
Bronchoalveolar lavage fluid was harvested in anesthetized mice, following a tracheal incision, by lavaging twice with 1 ml of either PBS or FACS buffer (2% FBS and 1 mM EDTA in PBS) through a Surflo ETFE IV Catheter 20G × 1″ (Terumo Medical Products, # SR-OX2025CA). The lavage fluid was centrifuged at 350 × g for 6.5 minutes, and the supernatant was collected for ELISA analysis. The cell pellet was resuspended, subjected to RBC lysis (Cytek, # TNB-4300-L100 or Gibco, # A1049201), and stained for surface markers before flow cytometry.
Flow cytometry
Single-cell suspensions derived from bronchoalveolar lavage fluid or lung samples were stained with Ghost Dye Violet 510 (Cytek, # 13-0870-T100) and appropriate antibody cocktails in PBS. Cells were incubated at 4°C for 30 minutes, then fixed with 10% neutral buffered formalin (Sigma Aldrich, # HT501128) at room temperature for 15 minutes before data acquisition. To assess eosinophil and neutrophil infiltration in BALF, cells were stained with fluorochrome-conjugated antibodies against CD45 (clone: 30-F11), CD90.2 (clone: 53-2.1), CD11b (clone: M1/70), CD11c (clone: N418), Ly6C (clone: HK1.4), Ly6G (clone: 1A8), and Siglec-F (clone: 1RNM44N). For γδ T cell analysis in lung tissue, staining included CD45 (clone: 30-F11), TCRγδ (clone: GL3), CD90.2 (clone: 53-2.1), and lineage markers TCRβ (clone: H57-597), CD19 (clone: 1D3/CD19), NK1.1 (clone: PK136), CD11b (clone: M1/70), CD11c (clone: N418), F4/80 (clone: BM8), and FcεRIα (clone: MAR-1), obtained from Biolegend or Thermo Fisher Scientific. Data were acquired using a BD FACS Canto II system.
Lung tissue harvesting
After diaphragm incision and transcardial perfusion with 10 ml of PBS, lung tissues were dissected, minced with razor blades, and either placed in TRIzol™ Reagent (Invitrogen, # 15596026) for RNA extraction or transferred into a digestion buffer consisting of 1.6 mg/ml collagenase type 4 (Worthington LS004189) and 100 µg/ml DNase I (Roche, # 11284932001) in supplemented DMEM107. The tissues were digested for 45 minutes at 37°C with mechanical dissociation through 18-gauge needles after 30 minutes, followed by filtration through a 70 µm nylon mesh and RBC lysis. Cells were resuspended in FACS buffer for flow cytometry or fluorescence-activated cell sorting or in FBS-supplemented DMEM for in vitro stimulation in 96-well plates at 37°C with 5% CO₂, after which supernatants were collected.
Alveolar macrophage culture
Alveolar macrophages were obtained from the BALF of naïve mice, where they represented approximately 95% of the recovered cells. After centrifugation and RBC lysis, these cells were seeded at 3 × 105 per well in 96-well plates containing DMEM (Gibco 11965092) supplemented with 1 mM sodium pyruvate (Gibco, # 11360070), 2 mM GlutaMAX (Gibco, # 35050061), 100 U/mL penicillin and 100 µg/mL streptomycin (Corning, # 30-002-CI), 10 mM HEPES (Gibco, # 15630080), and 10% FB Essence (VWR 10805-184), and cultured overnight. They were then stimulated with 100 µg/ml FPM (NIST, # 2786) for 1–4 hours, followed by RNA extraction for quantitative PCR analysis.
Neuron culture
The jugular-nodose complex (JNC) was collected from anesthetized mice following exsanguination and placed in a digestion buffer containing 1 mg/ml (325 U/ml) collagenase type 4 (Worthington, # LS004189), 2 mg/ml (1.8 U/ml) Dispase II (Sigma, # 04942078001), and 250 µg/ml (735.25 U/ml) DNase I (Roche, # 11284932001) prepared in supplemented DMEM without FB Essence. This mixture was incubated at 37°C for 60 minutes to ensure enzymatic digestion, followed by mechanical dissociation through progressive pipetting with tips of decreasing diameter and final passage through a 25-gauge needle. The cell suspension underwent density gradient centrifugation at 200 × g for 20 minutes at low acceleration and deceleration, layering 150 mg/ml bovine serum albumin (BSA; Hyclone, # SH30574.02) in PBS to separate the cells. The bottom fraction was collected, RBC lysed and seeded onto glass-bottom dishes (Abidi, # 81218) coated with 50 µg/ml laminin (Sigma, # L2020) and 100 µg/ml poly-D-lysine (Sigma, # P6407). Cells were cultured overnight in Neurobasal-A medium (Gibco, # 10888022) supplemented with 1 mM sodium pyruvate (Gibco, # 11360070), 2 mM GlutaMAX™ (Gibco, # 35050061), 100 U/mL penicillin, 100 µg/mL streptomycin (Corning, # 30-002-CI), 10 mM HEPES (Gibco, # 15630080), B-27 supplement (Gibco 17504-044), 50 ng/ml mouse nerve growth factor (NGF; Gibco, # 13257-019), 2 ng/ml mouse glial-derived neurotrophic factor (GDNF; Novus, # NBP2-61336), and cytosine-β-D-arabinofuranose (Thermo Scientific, # J6567106). In some experiments, 100 ng/ml artemin or 50 µM HCl (as vehicle control) were added in vitro in place of NGF or GDNF. This culture system was subsequently used for calcium imaging.
Real-time quantitative PCR (qPCR)
qPCR was performed on stimulated alveolar macrophages that were lysed using TRIzol Reagent and stored at –80°C until RNA extraction. RNA from sorted cells was extracted using the PureLink RNA Micro Scale Kit (ThermoFisher, # 12183016). In contrast, RNA from lung tissues or lung cell suspensions was extracted with the E.Z.N.A.® Total RNA Kit I (Omega Bio-tek, # R6834). Extraction procedures followed the manufacturers’ instructions, including phenol-chloroform purification and mixing with an equal volume of isopropanol. Complementary DNA (cDNA) was synthesized using the SuperScript VILO Master Mix (Invitrogen, # 11755050), with 1–2 µg of RNA as the template in each reaction. Quantitative PCR was carried out with PowerUp SYBR Green Master Mix (Applied Biosystems, # A25742), using 50–100 ng of cDNA and 200 nM of each primer. The reactions were run on either a Mic qPCR Cycler (Bio Molecular Systems) or a CFX Opus Real-Time PCR System (Bio-Rad Laboratories). The primer pair for Artn was Forward: 5′-TGATCCACTTGAGCTTCGGG-3′ and Reverse: 5′-CTCCATACCAAAGGGGTCCTG-3′.
Calcium imaging recording
Cultured neurons were loaded with 5 µM fura-2 AM (Cayman Chemical Company, # 34993) and incubated at 37°C for 40 minutes. After incubation, the cells were washed four times with standard external solution (SES; Boston BioProducts, # C-3030F) and maintained in this solution during imaging. The Fura-2 signals were recorded and used for downstream analyses. Agonists, diluted in SES, were delivered using a ValveLink 8.2 system (AutomateScientific) equipped with 250 µm Perfusion Pencil® tips (Automate Scientific) and controlled by Macro Recorder (Barbells Media, Germany). Between drug injections, SES flow was maintained to ensure a complete washout of each agonist. Imaging for Fura-2 experiments was conducted with a NIKON ECLIPSE Ti2 Inverted Microscope using an S Plan Fluor ELWD 20X objective lens to optimize UV light transmission. Images were captured every 3 or 4 seconds using sCMOS cameras such as PCO.Edge 4.2 LT (Excelitas Technologies), Prime 95B (Teledyne Photometrics), or Orca Flash 4.0 v2 (Hamamatsu Photonics). Regions of interest were manually delineated in NIS-Elements (Nikon), and the F340/F380 ratios were exported to Excel (Microsoft 365) for further analysis108. The data were compressed by calculating a maximum value every 15 seconds for all subsequent evaluations.
Intravital microscopy
6–10 weeks old male and female littermate control (NaV1.8wt::DTAfl/wt denoted as NaV1.8WT) and nociceptor-ablated (NaV1.8cre::DTAfl/wt denoted as NaV1.8DTA) mice were sensitized via intraperitoneal injection of an emulsion containing ovalbumin (OVA; 200 µg/dose) and aluminum hydroxide (1 mg/dose) on days 0 and 7. On day 10, phagocytes were labeled by intranasal injection of PKH26 Red Fluorescent Cell Linker Kit (Sigma, # PKH26PCL) at 25 pmol/dose in Diluent B. Mice were then challenged intranasally with OVA (50 µg/dose) alone or in combination with fine particulate matter (FPM; 20 µg/dose) on days 14–16. Alveolar macrophage intravital imaging was performed on day 17 and is presented as a 1-hour time-lapse video.
Lung images were acquired using a Nikon CSU-X1 multichannel spinning-disk confocal upright microscope with a protocol adapted from previously published methods44,45. Mice were anesthetized with 10 mg/kg xylazine hydrochloride and 200 mg/kg ketamine hydrochloride, and body temperature was maintained at 37°C with a heating pad (World Precision Instruments). The right jugular vein was cannulated for additional anesthetic as needed and for injecting anti-mouse Ly6G Alexa Fluor 647 (BioLegend, clone: 1A8, # 127610) to label neutrophils. After exposing the trachea and inserting a catheter connected to a small rodent ventilator (Harvard Apparatus), the mouse was placed in a right lateral decubitus position. A small incision was made between ribs 4 and 5 to create an opening of about 1.5 cm, and an intercostal lung window was carefully fitted and stabilized by a vacuum of roughly 20 mmHg. Time-lapse images were acquired without delay using a 20X water-immersion objective (numerical aperture 1).
Images are presented as maximum-intensity projections of z-stacks. Alveolar macrophage movement was tracked for over 1 hour, and neutrophil behavior was recorded for 20 minutes. Cell displacement was quantified using the ICY software’s manual tracking plugin. Neutrophil behavior was classified into adherent, tethering, crawling, or patrolling using Imaris (Oxford Instruments) spot tracking. Track Duration and Track Speed Mean were used to define each behavior per field of view (FOV). Tethering was determined by a Track Duration under 150 seconds for cells that rapidly entered the FOV before arresting and exiting again. Adherent cells remained immobile for more than 150 seconds, with a Track Speed Mean of ≤ 0.03 µm/s. Crawling cells were motile with steady movement that persisted for at least half of the video’s duration; these had a Track Speed Mean > 0.03 µm/s and a Track Duration > 600 seconds. Patrolling cells shared the rapid entry observed with tethering cells but, instead of briefly arresting continued crawling and exited the FOV, showing a Track Speed Mean > 0.03 µm/s and a Track Duration > 150 seconds but < 600 seconds.
Bulk RNA sequencing
RNA-seq was performed on JNCs isolated from TRPV1cre::tdTomatofl/wt mice subjected to OVA sensitization and challenge, as previously described. tdTomato+ cells were sorted using a BD FACSAria cell sorter24, and total RNA was extracted following established protocols. Library preparation was carried out at the Institut de Recherche en Cancérologie et en Immunologie (IRIC) of the Université de Montréal. RNA quality was assessed using an Agilent Bioanalyzer, ensuring a minimum RNA Integrity Number (RIN) of 7.5. Libraries were prepared using a poly(A)-enrichment, single-stranded RNA-seq strategy (KapaBiosystems, KAPA RNA Hyperprep Kit, #KR1352) and sequenced on an Illumina NextSeq500 platform with 75-cycle single-end reads.
Basecalling was performed using Illumina RTA 2.4.11, and demultiplexing was conducted with bcl2fastq 2.20, allowing for one mismatch in the index. Trimmomatic was used to remove adapter sequences and low-quality bases from the 3′ end of each read. The remaining high-quality reads were aligned to the GRCm38 mouse genome using STAR v2.5.11, which also generated gene-level read counts. Differential expression analysis was performed using DESeq2 on these read counts, normalized by the DESeq2 pipeline. Log2 fold changes and –log10 p-values were calculated from the normalized data, and genes were considered differentially expressed if their adjusted p-value (false discovery rate, FDR) was below 0.05. Further data analysis and visualization were conducted in RStudio.
Enzyme-linked immunosorbent assay (ELISA)
ELISA was used to measure artemin levels in BALF (R&D Systems, # DY1085-05) following the manufacturer’s instructions. Inflammatory cytokines in BALF were detected with a Cytometric Bead Array Flex Set from BD Biosciences: Master Buffer Set (# 558266), IL-1β (# 560232), IL-4 (# 558298), IL-5 (# 558302), IL-6 (# 558301), IL-10 (# 558300), IL-13 (# 558349), IL-17A (# 560283), IFNγ (# 558296), MCP-1 (# 558342), and TNF (# 558299), also used according to the manufacturer’s guidelines.
In-silico analysis of mouse immune cells expression profile using the Immgen database
Using the publicly available Immgen database53 we proceed to an in-silico analysis of RNA-sequencing data (DESeq2 data) of various mouse immune cells. As per Immgen protocol, RNA-sequencing reads were aligned to the mouse genome GENCODE GRCm38/mm10 primary assembly (GenBank assembly accession GCA_000001635.2) and gene annotations vM16 with STAR 2.5.4a. The ribosomal RNA gene annotations were removed from general transfer format file. The gene-level quantification was calculated by featureCounts. Raw reads count tables were normalized by median of ratios method with DESeq2 package from Bioconductor and then converted to GCT and CLS format. Samples with less than 1 million uniquely mapped reads were automatically excluded from normalization. Experimental details are defined in www.immgen.org/Protocols/ImmGenULI_RNAseq_methods.pdf.
In silico analysis of RNA-Seq data
In silico analysis of RNA-Seq data involved extracting information from Kupari et al.’s supplementary materials (GSE124312)51, with clusters based on that publication’s designations. Additional data from Zhao et al., (GSE192987)52 were reanalyzed using R and plotted with UMAP to visualize the co-expression of relevant genes. Bulk JNC sequencing datasets were analyzed with DESeq2.
In-silico analysis of lung cancer patients’ tumor expression profile using single-cell RNA sequencing
We performed an in-silico analysis of single-cell RNA-sequencing data from mouse lung tumor–infiltrating CD45⁺ cells in non–small cell lung cancer (NSCLC) models (GSE127465)109, accessed via the publicly available Broad Institute Single-Cell Portal. The expression levels of Ahr, Artn, Calca, Vip, Tac1, and Trpa1 were plotted within CD45⁺ myeloid cell populations using RStudio. Additionally, we conducted a Spearman correlation analysis to explore relationships among these genes. Gene expression data are presented as normalized counts per ten thousand. We extracted the relevant expression values and used RStudio to generate dot plots for individual cells in each myeloid population, as well as to perform correlation analyses of the selected genes. Experimental and clustering details can be found at Zilionis et al., 109.
In-silico analysis of human lung-tissue gene expression using the Human Protein Atlas (HPA) Single Cell Type Atlas
We retrieved the lung dataset generated by Karlsson et al. 106 from the HPA Single Cell Type Atlas, which pools single-cell RNA-sequencing reads into annotated clusters and reports expression as per-gene z-scores after median-of-ratios normalization. Using R (v4.3.2), we downloaded the cluster-level expression matrix, isolated all clusters and extracted transcript values for Gfra3, Ahr, Artn, Calca, Tac1, Vip and Trpa1. The positive z-scores observed in the “lung” clusters confirm that Ahr mRNA is expressed in patient lung tissue. Because the Karlsson pipeline equalizes library depth before z-score transformation, these values permit direct comparison of Ahr abundance to the whole-body baseline captured by the same atlas. Additional, experimental and clustering details can be found at Karlsson et al., 106.
In-silico analysis of AHR protein abundance in human lung macrophages using Human Protein Atlas immunohistochemistry data
We retrieved the antibody-based tissue microarray summary from Uhlén et al. 55 via the Human Protein Atlas pathology portal, which lists per-tissue immunoreactivity for each antibody as categorical scores (“not detected,” “low,” “medium,” “high”). From the raw TSV file, we report protein expression (Ahr, Artn, Gfra3, Calca, Vip, Tac1) across cell types and tissue types. Because the HPA pipeline reports these scores after internal normalization of DAB signal across replicate tissue cores, the categorical value can be compared directly across tissues without further scaling. This processed result confirms constitutive AHR protein presence in lung macrophages in situ. Additional, experimental and clustering details can be found at Uhlén et al. 55.
In-silico co-expression analysis of AHR and ARTN in human lung macrophages using the CZ CELLxGENE Discover single-cell atlas
We accessed the lung single-cell RNA-seq data curated by Abdulla et al. 56 through the CZ CELLxGENE Discover platform and downloaded the cell-level expression matrix together with standardized cell-type annotations. Raw UMI counts were normalized to ln (CPTT + 1) values during ingestion into the atlas, a transformation that equalizes library depth and stabilizes variance while preserving the full gene–cell count matrix for downstream analyses. Additional, experimental and clustering details can be found at Abdulla et al. 56.
G-Profiler and Go-term
The top 50 differentially expressed genes (based on DESeq p-values) from each comparison were submitted to the web-based tool g:Profiler 110 for enrichment analysis (g:GOSt). The resulting pathway enrichments—derived from multiple databases, including Gene Ontologies (GO; covering Molecular Function, Biological Process, and Cellular Component sub-ontologies) 111, the Kyoto Encyclopedia of Genes and Genomes (KEGG) 112, Reactome (REAC) 113, WikiPathways (WP) 114, TRANSFAC (TF) 115, miRTarBase (MIRNA), the Comprehensive Resource of Mammalian Protein Complexes (CORUM) 116, and the Human Phenotype Ontology (HP) 117—are listed in Supplementary Table 1.
Data availability
Bulk RNA-sequencing raw and processed data have been deposited in the NCBI’s gene expression omnibus (GSE298583). Processed data can also be accessed in the supplementary Table 1. Additional information and raw data are available from the lead contact upon reasonable request.
Statistics
P values ≤ 0.05 were considered statistically significant. One-way ANOVA, and Student’s t-tests were conducted using GraphPad Prism, while DESeq2 and Seurat analyses, including their statistical tests, were performed in RStudio.
Replicates
The number of replicates (n) for each experiment is specified in the figure legends and represents the number of animals for in vivo data. For in vitro experiments, replicates may be culture wells or dishes, animals, fields of view during microscopy, or individual neurons in calcium imaging. All experiments included different preparations from distinct animals to ensure biological reproducibility.
Exclusion
One replicate in the naïve mice group of the RNA-sequencing analysis (Figure 2) was excluded because the PCA indicated it as an outlier. No other data were excluded from the study.
Supplementary figures

In silico analysis of Artn expression in mouse immune cells
(A) In-silico re-analysis of Artn expression in mouse immune cells using the ImmGen database53. Artn and Ahr are expressed in Itgam+ macrophages. Data are presented as per-gene z-scores of normalized gene expression, calculated by the median of ratios method.
(B) In-silico re-analysis of the single-cell RNA-seq dataset from Tavares-Ferreira et al., 105 (Sensoryomics; dbGaP accession phs001158) shows that Gfra3 is expressed in Trpa1-positive nociceptors, C-LTMRs, and silent nociceptors within the human dorsal root ganglion. Expression levels are reported as per-gene z-scores calculated with the median-of-ratios normalization method.

In-silico re-analysis of Ahr and Artn expression in human tissues
(A) In-silico re-analysis of data from Karlsson et al., 106 (Human Protein Atlas, proteinatlas.org) indicates that Ahr and Artn are expressed in human lung. Expression values are reported as per-gene z-scores using the median-of-ratios normalization method. Experimental details and cell clustering are described by Karlsson et al., 106
(B) In-silico re-analysis of data from Uhlén et al. 55 (Human Protein Atlas, proteinatlas.org) confirms Ahr protein expression in human lung macrophages, as shown by immunohistochemistry. These data are presented as protein-normalized expression levels. Experimental details and cell clustering are described by Uhlén et al. 55.
(C) In-silico re-analysis of data from Abdulla et al. 56 (CELLxGENE, CZI Single-Cell Biology) reveals co-expression of Artn and Ahr in lung and alveolar macrophages from patients. Expression values are provided as per-gene z-scores, calculated by the median-of-ratios normalization method. Experimental details and cell clustering are described by Abdulla et al. 56.

Schematic of nociceptor involvement in pollution-exacerbated allergic asthma
In our study, mice were exposed to PM₂₅ particles and ovalbumin (OVA) to model pollution-exacerbated asthma. Compared to mice exposed to OVA alone, co-exposure to PM₂₅ and OVA significantly increased bronchoalveolar lavage fluid (BALF) neutrophils and lung γδ T cells levels. To counteract this heightened airway inflammation, we administered intranasal QX-314—a charged lidocaine derivative—at the peak of inflammation, effectively normalizing BALF neutrophil levels. Ablation of TRPV1⁺ nociceptor neurons produced a similar effect.
Further analysis with calcium imaging revealed that neurons from the jugular-nodose complex in pollution-exposed asthmatic mice were more sensitive via their TRPA1 channels. Levels of TNFα and the growth factor artemin were also elevated in the BALF of these mice, returning to normal following nociceptor ablation.
We identified alveolar macrophages as the source of artemin, which they secrete upon sensing fine particulate matter (FPM) through aryl hydrocarbon receptors. Artemin, in turn, heightened TRPA1 responsiveness to its agonist (mustard oil), thereby exacerbating airway inflammation. Our findings suggest that silencing nociceptor neurons can disrupt this pathway, offering a novel therapeutic approach to mitigate neutrophilic airway inflammation driven by pollution.
Acknowledgements
ST’s work is supported by the Canadian Institutes of Health Research (193741, 407016, 461274, and 461275), the Canadian Foundation for Innovation (44135), the Canadian Cancer Society Emerging Scholar Research Grant (708096), the Knut and Alice Wallenberg Foundation (KAW 2021.0141, KAW 2022.0327), the Swedish Research Council (2022-01661), the Natural Sciences and Engineering Research Council of Canada (RGPIN-2019-06824), and the NIH/NIDCR (R01DE032712). AT’s work is supported by the Canadian Institute of Health Research (186176). Salary support for JCW was provided by the Fonds de recherche du Québec – Santé, the Canadian Allergy, Asthma, and Immunology Foundation, Asthma Canada, and CIHR. Salary support for AK was provided by the Canada Graduate Scholarship Masters.
Additional files
References
- 1Neuroimmunity: Physiology and PathologyAnnu Rev Immunol 34:421–447https://doi.org/10.1146/annurev-immunol-041015-055340Google Scholar
- 2CNS injury: IL-33 sounds the alarmImmunity 42:403–405https://doi.org/10.1016/j.immuni.2015.02.019Google Scholar
- 3Epineural optogenetic activation of nociceptors initiates and amplifies inflammationNature biotechnology 39:179–185Google Scholar
- 4Nociceptor sensory neurons suppress neutrophil and gammadelta T cell responses in bacterial lung infections and lethal pneumoniaNat Med 24:417–426https://doi.org/10.1038/nm.4501Google Scholar
- 5Vagal TRPV1 (+) sensory neurons regulate myeloid cell dynamics and protect against influenza virus infectionbioRxiv https://doi.org/10.1101/2024.08.21.609013Google Scholar
- 6Gut-innervating nociceptor neurons regulate Peyer’s patch microfold cells and SFB levels to mediate Salmonella host defenseCell 180:33–49Google Scholar
- 7Cutaneous TRPV1+ neurons trigger protective innate type 17 anticipatory immunityCell 178:919–932Google Scholar
- 8Substance P release by sensory neurons triggers dendritic cell migration and initiates the type-2 immune response to allergensImmunity 53:1063–1077Google Scholar
- 9Multimodal control of dendritic cell functions by nociceptorsScience 379:eabm5658Google Scholar
- 10Bacteria activate sensory neurons that modulate pain and inflammationNature 501:52–57Google Scholar
- 11Nociceptor neurons affect cancer immunosurveillanceNature 611:405–412Google Scholar
- 12Functional neuronal circuits promote disease progression in cancerScience Advances 9:eade4443Google Scholar
- 13.Tumor-infiltrating nerves functionally alter brain circuits and modulate behavior in a mouse model of head-and-neck cancerbioRxiv Google Scholar
- 14Next Directions in the Neuroscience of Cancers Arising outside the CNSCancer Discovery 14:669–673Google Scholar
- 15Sense and immunity: context-dependent neuro-immune interplayFrontiers in immunology 8:1463Google Scholar
- 16Silencing nociceptor neurons reduces allergic airway inflammationNeuron 87:341–354Google Scholar
- 17Population of sensory neurons essential for asthmatic hyperreactivity of inflamed airwaysProceedings of the National Academy of Sciences 111:11515–11520Google Scholar
- 18Vagal sensory neurons drive mucous cell metaplasiaJournal of Allergy and Clinical Immunology 145:1693–1696Google Scholar
- 19Nociceptor neurons direct goblet cells via a CGRP-RAMP1 axis to drive mucus production and gut barrier protectionCell 185:4190–4205Google Scholar
- 20FcεR1-expressing nociceptors trigger allergic airway inflammationJournal of Allergy and Clinical Immunology 147:2330–2342Google Scholar
- 21Cartilage-binding antibodies induce pain through immune complex–mediated activation of neuronsJournal of Experimental Medicine 216:1904–1924Google Scholar
- 22.Nociceptor neurons promote IgE class switch in B cellsJCI insight 6Google Scholar
- 23TRPV1 nociceptors are required to optimize antigen-specific primary antibody responses to novel antigensBioelectronic Medicine 10Google Scholar
- 24Cytokines reprogram airway sensory neurons in asthmaCell Rep 43https://doi.org/10.1016/j.celrep.2024.115045Google Scholar
- 25Increasing frequency and intensity of the most extreme wildfires on EarthNat Ecol Evol 8:1420–1425https://doi.org/10.1038/s41559-024-02452-2Google Scholar
- 26Mortality attributable to PM2. 5 from wildland fires in California from 2008 to 2018Science Advances 10:eadl1252Google Scholar
- 27Neutrophilic inflammation in asthma and association with disease severityTrends in immunology 38:942–954Google Scholar
- 28Characterization of Asthma by Age of Onset: A Multi-Database Cohort StudyJ Allergy Clin Immunol Pract 10:1825–1834https://doi.org/10.1016/j.jaip.2022.03.019Google Scholar
- 29Adult-onset asthma: is it really different?Eur Respir Rev 22:44–52https://doi.org/10.1183/09059180.00007112Google Scholar
- 30Acute effects of smoke exposure on airway and systemic inflammation in forest firefightersJ Asthma Allergy 11:81–88https://doi.org/10.2147/JAA.S136417Google Scholar
- 31The Effects of Wildfire Smoke on Asthma and AllergyCurr Allergy Asthma Rep 23:375–387https://doi.org/10.1007/s11882-023-01090-1Google Scholar
- 32Clearing the Air: Understanding the Impact of Wildfire Smoke on Asthma and COPDHealthcare (Basel) 12https://doi.org/10.3390/healthcare12030307Google Scholar
- 33Neutrophilic inflammation in severe persistent asthmaAmerican journal of respiratory and critical care medicine 160:1532–1539Google Scholar
- 34Identification of a PD-L1+Tim-1+ iNKT subset that protects against fine particulate matter-induced airway inflammationJCI Insight 7https://doi.org/10.1172/jci.insight.164157Google Scholar
- 35Inhibition of inflammatory pain and cough by a novel charged sodium channel blockerBr J Pharmacol 178:3905–3923https://doi.org/10.1111/bph.15531Google Scholar
- 36Novel charged sodium and calcium channel inhibitor active against neurogenic inflammationeLife 8https://doi.org/10.7554/eLife.48118Google Scholar
- 37Inhibition of nociceptors by TRPV1-mediated entry of impermeant sodium channel blockersNature 449:607–610https://doi.org/10.1038/nature06191Google Scholar
- 38Nanophotonics Enable Targeted Photothermal Silencing of Nociceptor NeuronsSmall 18:e2103364https://doi.org/10.1002/smll.202103364Google Scholar
- 39Sensory neurons regulate stimulus-dependent humoral immunity in mouse models of bacterial infection and asthmaNat Commun 15:8914https://doi.org/10.1038/s41467-024-53269-3Google Scholar
- 40Comorbid functional dyspepsia reflects IL-33-mediated airway neuronal dysfunction in asthmaJ Allergy Clin Immunol 154:1422–1433https://doi.org/10.1016/j.jaci.2024.06.008Google Scholar
- 41Evidence for vagal sensory neural involvement in influenza pathogenesis and diseasePLoS Pathog 20:e1011635https://doi.org/10.1371/journal.ppat.1011635Google Scholar
- 42Asthma: Methods and ProtocolsSpringer pp. 297–314Google Scholar
- 43Anatomical differences in nociceptor neurons sensitivityBioelectron Med 8https://doi.org/10.1186/s42234-022-00088-wGoogle Scholar
- 44Alveolar macrophage function is impaired following inhalation of berry e-cigarette vaporProc Natl Acad Sci U S A 121:e2406294121https://doi.org/10.1073/pnas.2406294121Google Scholar
- 45Patrolling Alveolar Macrophages Conceal Bacteria from the Immune System to Maintain HomeostasisCell 183:110–125https://doi.org/10.1016/j.cell.2020.08.020Google Scholar
- 46Artemin is a vascular-derived neurotropic factor for developing sympathetic neuronsNeuron 35:267–282https://doi.org/10.1016/s0896-6273(02)00774-2Google Scholar
- 47Artemin and an Artemin-Derived Peptide, Artefin, Induce Neuronal Survival, and Differentiation Through Ret and NCAMFront Mol Neurosci 12:47https://doi.org/10.3389/fnmol.2019.00047Google Scholar
- 48Artemin, a Glial Cell Line-Derived Neurotrophic Factor Family Member, Induces TRPM8-Dependent Cold PainJ Neurosci 33:12543–12552https://doi.org/10.1523/Jneurosci.5765-12.2013Google Scholar
- 49Artemin overexpression in skin enhances expression of TRPV1 and TRPA1 in cutaneous sensory neurons and leads to behavioral sensitivity to heat and coldJ Neurosci 26:8578–8587https://doi.org/10.1523/JNEUROSCI.2185-06.2006Google Scholar
- 50Overexpression of artemin in the tongue increases expression of TRPV1 and TRPA1 in trigeminal afferents and causes oral sensitivity to capsaicin and mustard oilBrain Res 1230:80–90https://doi.org/10.1016/j.brainres.2008.06.119Google Scholar
- 51An Atlas of Vagal Sensory Neurons and Their Molecular SpecializationCell Rep 27:2508–2523https://doi.org/10.1016/j.celrep.2019.04.096Google Scholar
- 52A multidimensional coding architecture of the vagal interoceptive systemNature 603:878–884https://doi.org/10.1038/s41586-022-04515-5Google Scholar
- 53ImmGen at 15Nat Immunol 21:700–703https://doi.org/10.1038/s41590-020-0687-4Google Scholar
- 54A singleБ─⌠cell type transcriptomics map of human tissuesScience advances 7:eabh2169Google Scholar
- 55A human protein atlas for normal and cancer tissues based on antibody proteomicsMol Cell Proteomics 4:1920–1932https://doi.org/10.1074/mcp.M500279-MCP200Google Scholar
- 56CZ CELLxGENE Discover: A single-cell data platform for scalable exploration, analysis and modeling of aggregated databioRxiv Google Scholar
- 57Bacteria hijack a meningeal neuroimmune axis to facilitate brain invasionNature 615:472–481https://doi.org/10.1038/s41586-023-05753-xGoogle Scholar
- 58Nociceptive sensory neurons promote CD8 T cell responses to HSV-1 infectionNat Commun 12:2936https://doi.org/10.1038/s41467-021-22841-6Google Scholar
- 59Nociceptive Sensory Fibers Drive Interleukin-23 Production from CD301b+ Dermal Dendritic Cells and Drive Protective Cutaneous ImmunityImmunity 43:515–526https://doi.org/10.1016/j.immuni.2015.08.016Google Scholar
- 60Blocking Neuronal Signaling to Immune Cells Treats Streptococcal Invasive InfectionCell 173:1083–1097https://doi.org/10.1016/j.cell.2018.04.006Google Scholar
- 61Tapinarof in the treatment of psoriasis: A review of the unique mechanism of action of a novel therapeutic aryl hydrocarbon receptor-modulating agentJ Am Acad Dermatol 84:1059–1067https://doi.org/10.1016/j.jaad.2020.10.085Google Scholar
- 62Activation of the aryl hydrocarbon receptor dampens the severity of inflammatory skin conditionsImmunity 40:989–1001https://doi.org/10.1016/j.immuni.2014.04.019Google Scholar
- 63Aryl hydrocarbon receptor restricts axon regeneration of DRG neurons in response to injurybioRxiv https://doi.org/10.1101/2023.11.04.565649Google Scholar
- 64The aryl hydrocarbon receptor and the gut-brain axisCell Mol Immunol 18:259–268https://doi.org/10.1038/s41423-020-00585-5Google Scholar
- 65Mapping of the Sensory Innervation of the Mouse Lung by Specific Vagal and Dorsal Root Ganglion Neuronal SubsetseNeuro 9https://doi.org/10.1523/ENEURO.0026-22.2022Google Scholar
- 66Evidence for multiple sensory circuits in the brain arising from the respiratory system: an anterograde viral tract tracing study in rodentsBrain Struct Funct 220:3683–3699https://doi.org/10.1007/s00429-014-0883-9Google Scholar
- 67An Airway Protection Program Revealed by Sweeping Genetic Control of Vagal AfferentsCell 181:574–589https://doi.org/10.1016/j.cell.2020.03.004Google Scholar
- 68Piezo2 senses airway stretch and mediates lung inflation-induced apnoeaNature 541:176–181https://doi.org/10.1038/nature20793Google Scholar
- 69Vagal Sensory Neuron Subtypes that Differentially Control BreathingCell 161:622–633https://doi.org/10.1016/j.cell.2015.03.022Google Scholar
- 70Unbiased classification of sensory neuron types by large-scale single-cell RNA sequencingNat Neurosci 18:145–153https://doi.org/10.1038/nn.3881Google Scholar
- 71A vagal reflex evoked by airway closureNature 627:830–838https://doi.org/10.1038/s41586-024-07144-2Google Scholar
- 72An airway-to-brain sensory pathway mediates influenza-induced sicknessNature 615:660–667https://doi.org/10.1038/s41586-023-05796-0Google Scholar
- 73Depolarization induces nociceptor sensitization by CaV1.2-mediated PKA-II activationJ Cell Biol 220https://doi.org/10.1083/jcb.202002083Google Scholar
- 74Rare copy number variations affecting the synaptic gene DMXL2 in neurodevelopmental disordersJ Neurodev Disord 11https://doi.org/10.1186/s11689-019-9263-3Google Scholar
- 75The A118G single nucleotide polymorphism of the mu-opioid receptor gene (OPRM1) is associated with pressure pain sensitivity in humansJ Pain 6:159–167https://doi.org/10.1016/j.jpain.2004.11.008Google Scholar
- 76HINT1 protein: a new therapeutic target to enhance opioid antinociception and block mechanical allodyniaNeuropharmacology 89:412–423https://doi.org/10.1016/j.neuropharm.2014.10.022Google Scholar
- 77Elevated levels of FMRP-target MAP1B impair human and mouse neuronal development and mouse social behaviors via autophagy pathwayNat Commun 14:3801https://doi.org/10.1038/s41467-023-39337-0Google Scholar
- 78Expression and Manipulation of the APC-beta-Catenin Pathway During Peripheral Neuron RegenerationSci Rep 8https://doi.org/10.1038/s41598-018-31167-1Google Scholar
- 79The role of the leukemia inhibitory factor receptor in neuroprotective signalingPharmacol Ther 183:50–57https://doi.org/10.1016/j.pharmthera.2017.08.008Google Scholar
- 80Nociceptor-immune interactomes reveal insult-specific immune signatures of painNat Immunol 25:1296–1305https://doi.org/10.1038/s41590-024-01857-2Google Scholar
- 81Defining the nociceptor transcriptomeFront Mol Neurosci 7https://doi.org/10.3389/fnmol.2014.00087Google Scholar
- 82Basophils Activate Pruriceptor-Like Vagal Sensory NeuronsbioRxiv https://doi.org/10.1101/2024.06.11.598517Google Scholar
- 83The epithelial cell-derived atopic dermatitis cytokine TSLP activates neurons to induce itchCell 155:285–295https://doi.org/10.1016/j.cell.2013.08.057Google Scholar
- 84A sensory neuronal ion channel essential for airway inflammation and hyperreactivity in asthmaProc Natl Acad Sci U S A 106:9099–9104https://doi.org/10.1073/pnas.0900591106Google Scholar
- 85Transient receptor potential ankyrin 1 antagonists block the noxious effects of toxic industrial isocyanates and tear gasesFASEB J 23:1102–1114https://doi.org/10.1096/fj.08-117812Google Scholar
- 86A TRPA1 inhibitor suppresses neurogenic inflammation and airway contraction for asthma treatmentJ Exp Med 218https://doi.org/10.1084/jem.20201637Google Scholar
- 87The EGR1-Artemin Axis in Keratinocytes Enhances the Innervation of Epidermal Sensory Neurons during Skin Inflammation Induced by House Dust Mite Extract from DermatophagoidesfarinaeJ Invest Dermatol 144:1817–1828https://doi.org/10.1016/j.jid.2024.01.017Google Scholar
- 88The effect of substance P on the regional lymph node antibody response to antigenic stimulation in capsaicin-pretreated ratsJ Immunol 139:3470–3473Google Scholar
- 89Overexpression of Substance P in pig airways increases MUC5AC through an NF-kbeta pathwayPhysiol Rep 9:e14749https://doi.org/10.14814/phy2.14749Google Scholar
- 90Neuronal Circuits Modulate Antigen Flow Through Lymph NodesBioelectron Med 3:18–28https://doi.org/10.15424/bioelectronmed.2016.00001Google Scholar
- 91Lymph nodes are innervated by a unique population of sensory neurons with immunomodulatory potentialCell 184:441–459https://doi.org/10.1016/j.cell.2020.11.028Google Scholar
- 92Nociceptive sensory neurons drive interleukin-23-mediated psoriasiform skin inflammationNature 510:157–161https://doi.org/10.1038/nature13199Google Scholar
- 93The neuropeptide neuromedin U stimulates innate lymphoid cells and type 2 inflammationNature 549:282–286https://doi.org/10.1038/nature23676Google Scholar
- 94Neuronal regulation of type 2 innate lymphoid cells via neuromedin UNature 549:277–281https://doi.org/10.1038/nature23469Google Scholar
- 95The neuropeptide NMU amplifies ILC2-driven allergic lung inflammationNature 549:351–356https://doi.org/10.1038/nature24029Google Scholar
- 96Type 2 innate lymphoid cells control eosinophil homeostasisNature 502:245–248https://doi.org/10.1038/nature12526Google Scholar
- 97Pulmonary neuroendocrine cells amplify allergic asthma responsesScience 360https://doi.org/10.1126/science.aan8546Google Scholar
- 98Calcitonin Gene-Related peptide receptor antagonist suppresses allergic asthma responses via downregulation of group 2 innate lymphoid cells in miceInt Immunopharmacol 122https://doi.org/10.1016/j.intimp.2023.110608Google Scholar
- 99Calcitonin Gene-Related Peptide Negatively Regulates Alarmin-Driven Type 2 Innate Lymphoid Cell ResponsesImmunity 51:709–723https://doi.org/10.1016/j.immuni.2019.09.005Google Scholar
- 100TRPV1(+) neurons alter Staphylococcus aureus skin infection outcomes by affecting macrophage polarization and neutrophil recruitmentBMC Immunol 24https://doi.org/10.1186/s12865-023-00584-xGoogle Scholar
- 101Cutaneous TRPV1(+) Neurons Trigger Protective Innate Type 17 Anticipatory ImmunityCell 178:919–932https://doi.org/10.1016/j.cell.2019.06.022Google Scholar
- 102TNF-alpha expression in neutrophils and its regulation by glycogen synthase kinase-3: a potentiating role for lithiumFASEB J 28:3679–3690https://doi.org/10.1096/fj.14-251900Google Scholar
- 103Neutrophils express tumor necrosis factor-alpha during mouse skin wound healingJ Invest Dermatol 105:120–123https://doi.org/10.1111/1523-1747.ep12313429Google Scholar
- 104Wildland fire smoke alters the composition, diversity, and potential atmospheric function of microbial life in the aerobiomeISME Commun 2https://doi.org/10.1038/s43705-022-00089-5Google Scholar
- 105Spatial transcriptomics of dorsal root ganglia identifies molecular signatures of human nociceptorsSci Transl Med 14:eabj8186https://doi.org/10.1126/scitranslmed.abj8186Google Scholar
- 106A single–cell type transcriptomics map of human tissuesScience advances 7:eabh2169Google Scholar
- 107Cigarette smoke-induced kinin B1 receptor promotes NADPH oxidase activity in cultured human alveolar epithelial cellsPeptides 32:1447–1456https://doi.org/10.1016/j.peptides.2011.05.005Google Scholar
- 108GRPR/PI3Kgamma: Partners in Central Transmission of ItchJ Neurosci 35:16272–16281https://doi.org/10.1523/JNEUROSCI.2310-15.2015Google Scholar
- 109Single-Cell Transcriptomics of Human and Mouse Lung Cancers Reveals Conserved Myeloid Populations across Individuals and SpeciesImmunity 50:1317–1334https://doi.org/10.1016/j.immuni.2019.03.009Google Scholar
- 110g: Profiler-interoperable web service for functional enrichment analysis and gene identifier mapping (2023 update)Nucleic acids research 51:W207–W212Google Scholar
- 111Gene ontology: tool for the unification of biologyNature genetics 25:25–29Google Scholar
- 112KEGG for taxonomy-based analysis of pathways and genomesNucleic acids research 51:D587–D592Google Scholar
- 113The reactome pathway knowledgebase 2022Nucleic acids research 50:D687–D692Google Scholar
- 114WikiPathways: connecting communitiesNucleic acids research 49:D613–D621Google Scholar
- 115The TRANSFAC project as an example of framework technology that supports the analysis of genomic regulationBriefings in bioinformatics 9:326–332Google Scholar
- 116.CORUM: the comprehensive resource of mammalian protein complexesБ─⌠2022Nucleic acids research 51:D539–D545Google Scholar
- 117The human phenotype ontology in 2021Nucleic acids research 49:D1207–D1217Google Scholar
Article and author information
Author information
Version history
- Sent for peer review:
- Preprint posted:
- Reviewed Preprint version 1:
- Reviewed Preprint version 2:
Cite all versions
You can cite all versions using the DOI https://doi.org/10.7554/eLife.101988. This DOI represents all versions, and will always resolve to the latest one.
Copyright
© 2024, Wang et al.
This article is distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use and redistribution provided that the original author and source are credited.
Metrics
- views
- 703
- downloads
- 5
- citations
- 0
Views, downloads and citations are aggregated across all versions of this paper published by eLife.