SUMMARY
Cytokines employ downstream Janus kinases (JAKs) to promote chronic inflammatory diseases. JAK1-dependent type 2 cytokines drive allergic inflammation, and patients with JAK1 gain-of-function (GOF) variants develop atopic dermatitis (AD) and asthma. To explore tissue-specific functions, we inserted a human JAK1 GOF variant (JAK1GOF) into mice and observed the development of spontaneous AD-like skin disease, but unexpected resistance to lung inflammation when JAK1GOF expression was restricted to the stroma. We identified a previously unrecognized role for JAK1 in vagal sensory neurons in suppressing airway inflammation. Additionally, expression of Calcb/CGRPβ, was dependent on JAK1 in vagal sensory neurons, and CGRPβ suppressed group 2 innate lymphoid cell function and allergic airway inflammation. Our findings reveal evolutionarily conserved but distinct functions of JAK1 in sensory neurons across tissues. This biology raises the possibility that therapeutic JAK inhibitors may be further optimized for tissue-specific efficacy to enhance precision medicine in the future.
Keywords: AAV, afferent nerves, allergic lung inflammation, atopic disorders, CGRP, ILC2, JAK1, neuropeptide, sensory neurons, vagus nerve
In Brief
Janus kinase 1 signaling exerts distinct effects across cell types and tissues, with JAK1 signaling in vagal sensory neurons promoting anti-inflammatory responses in the lung.
Graphical Abstract
INTRODUCTION
Cytokine receptors commonly employ the Janus kinase (JAK)-signal transducers and activators of transcription (STAT) pathway to influence effector cell programs. Indeed, JAK inhibition has revolutionized the treatment of a number of inflammatory, myeloproliferative, and even infectious disorders.1 Interest in the therapeutic potential of JAK inhibitors across the spectrum of allergic disorders is rapidly growing. In the past year, two JAK1-selective inhibitors were approved by the FDA for treatment of atopic dermatitis (AD), and several clinical trials for asthma treatment are underway.2–6 However, little is known about the pathophysiologic consequences of JAK inhibition. Indeed, the precise mechanisms underlying major adverse events of JAK inhibitor treatment, including cardiovascular events (MACE), cancer, and opportunistic infections, remain poorly defined.7 Thus, understanding the tissue-specific role of JAK signaling across organs can simultaneously inform understanding of biology but also foreshadow how JAK inhibitors may be used across the evolving medical landscape.
Type 2 cytokines critically rely on downstream JAK1 signaling to promote allergic inflammation.8 However, much of what we know about JAK inhibition has derived from our understanding of cytokine signaling within the hematopoietic compartment.9 Although, in immune cells, JAK1 phosphorylation drives a proinflammatory effector program,1 The role of JAK1 signaling within stromal cells (e.g., epithelial cells, fibroblasts, etc.) is just beginning to be uncovered. For example, sensory neuron-intrinsic JAK1 was found to play an unexpected role in promoting itch sensation, a difficult-to-treat component of AD pathology.10 Thus, probing the tissue- and cell-specific effects of JAK1 signaling is critical to understanding the molecular and cellular basis of allergic disease pathogenesis and treatment.
Patients with germline JAK1 gain-of-function (GOF) variants (JAK1GOF) develop atopic disorders such as AD and asthma, as well as eosinophilia.11,12 We sought to utilize a pathogenic human JAK1 GOF variant to probe the precise tissue-intrinsic mechanisms by which JAK1 signaling orchestrates various allergic pathologies. To do this, we generated a mouse line in which the endogenous murine Jak1 gene was replaced with a human JAK1 GOF mutation (JAK1GOF mice) that was reported in 2017.11 Although these mice exhibited spontaneous AD-like inflammation, as observed in patients, no lung pathology was evident at steady state. However, global JAK1GOF mice exhibited enhanced allergic lung inflammation in response to the classic aeroallergen Alternaria alternata. Strikingly, restriction of JAK1GOF expression to the stroma rendered mice resistant to allergic lung inflammation. Given recent reports that sensory neurons can suppress lung inflammation,13–18 we hypothesized that sensory neurons mediate a key regulatory circuit through JAK1 signaling.
Indeed, we found that chemical denervation of lung tissue as well as conditional deletion of endogenous murine Jak1 in sensory neurons aggravated allergic lung inflammation. We screened various neuropeptides associated with neuroinflammation and discovered that expression of Jak1 is required for proper expression of Calcb, which encodes the neuropeptide calcitonin gene-related peptide β (CGRPβ), within the vagal ganglia (VG). Additionally, we found that CGRPβ suppresses pathogenic group 2 innate lymphoid cell (ILC2) responses and allergic lung inflammation. Finally, retrograde viral delivery of JAK1GOF to the lung resulted in selective uptake into the VG and suppression of allergic lung inflammation. Similarly, conditional insertion of JAK1GOF into sensory neurons also promoted immune homeostasis in the lung.
Collectively, our study highlights that while JAK1 signaling promotes skin inflammation and is a major therapeutic target for AD and itch, sensory neuron-intrinsic signaling of JAK1 serves an immunoregulatory role in the lung. We reveal a previously unrecognized role for neuronal JAK1 in regulating neuropeptide expression and neuroinflammation. These findings underscore the importance of understanding tissue-specific JAK signaling across mammalian systems and the potential utility of JAK inhibitors across the disease spectrum.
RESULTS
Germline human JAK1 GOF promotes spontaneous allergic inflammation in the skin, but not in the lung
The first patients harboring a germline JAK1 GOF mutation were identified in 2017 as an alanine-to-aspartate substitution at position 634 (A634D) within the inhibitory pseudokinase domain of the JAK1 protein. These patients exhibited a number of allergic pathologies, including AD, asthma, and profound eosinophilia (Figure 1A).11 However, the severity of the patients’ AD and asthma were not reported in the original study. Thus, we examined the clinical characteristics of two patients when they originally presented with AD and asthma symptoms in more detail. Indeed, both patients with JAK1 GOF mutations presented with AD markedly earlier in life than with asthma (Figure 1B). Further, the overall relative severity of their AD (Figures 1C–E) was generally worse than the severity of their asthma as determined by quantitative and validated metrics such as Eczema Area and Severity Index (EASI) scores (Figure 1E), forced expiratory volume (FEV1) (Figure 1F), and forced vital capacity (FVC) (Figure 1G) measurements. These findings suggest that activation of JAK1 across multiple tissues results in differential tissue-specific outcomes.
Figure 1. Human germline JAK1 GOF mutation is associated with AD and asthma in patients and promotes spontaneous AD-like disease in mice.
(A) Human JAK1 c.1901C>A (p.A634D) GOF mutation within the pseudokinase domain.
(B) Timeline of AD and asthma diagnosis, EASI assessment, and PFTs.
(C) Baseline visual image of AD skin lesions from Patient A with a JAK1 GOF mutation.
(D) Baseline visual image of AD skin lesions from Patient B with a JAK1 GOF mutation.
(E) Baseline EASI scores from Patient A and Patient B.
(F) Baseline FEV1 of Patient A and Patient B.
(G) Baseline FVC of Patient A and Patient B.
(H) Schematic of the targeting strategy used to introduce the germline human JAK1 GOF mutation (JAK1GOF) into the murine Jak1 locus.
(I) Representative ear skin images of WT control and JAK1GOF mice.
(J) Ear thickness measurements of WT control and JAK1GOF mice.
(K) Representative skin histology images (H&E) of WT control and JAK1GOF mice. Scale bars indicate 100 μm.
(L-O) Flow cytometry of skin lymph nodes from WT control and JAK1GOF mice. Shown are the frequencies of (L) total CD45+ immune cells, (M) CD4+ T cells, (N) ILC2s, and (O) eosinophils.
(P-Q) Representative lung histology images of WT control and JAK1GOF mice, obtained by (P) H&E stain and (Q) PAS stain. Scale bars indicate 200 μm.
(R-U) Flow cytometry of lung tissue from WT control and JAK1GOF mice. Shown are the frequencies of (R) total CD45+ immune cells, (S) CD4+ T cells, (T) ILC2s, and (U) eosinophils.
(J, L-O, R-U) Data were pooled from three independent experiments, n = 8–15 pooled mice per group. †p < 0.0001, ***p < 0.001, **p < 0.01, *p < 0.05, and not significant (n.s.) (Unpaired t-test with Welch’s correction). Data are represented as mean ± standard deviation (SD).
AD – atopic dermatitis, EASI – eczema area and severity index, FEV1 – forced expiratory volume, FVC – forced vital capacity, GOF – gain-of-function, ILC2s – group 2 innate lymphoid cells, H&E – hematoxylin and eosin, JAK1 – Janus kinase 1, PAS – periodic acid-Schiff, PFT – pulmonary function test, WT – wild-type.
See also Figure S1
To examine how JAK1 confers unique effects across tissues, we generated mice in which the endogenous murine Jak1 gene was replaced with the mutant human JAK1 GOF variant (A634D) (Figure 1H). Strikingly, soon after birth, heterozygous JAK1GOF mice display spontaneous AD-like dermatitis with increased skin thickness compared to the skin of wild-type (WT) control mice (Figures 1I–J). Histologically, the JAK1GOF mice exhibited hyperkeratosis, epidermal hyperplasia, and dermal inflammatory immune cell infiltrate that were not observed in control mice (Figure 1K). Further, the JAK1GOF mice demonstrated a higher frequency of immune cell populations commonly associated with allergic inflammation, including total CD45+ immune cells (Figures 1L), CD4+ T cells (Figures 1M), ILC2s (Figures 1N), and eosinophils (Figures 1O) in the skin-draining lymph nodes compared to control mice at steady state. Taken together, these findings demonstrate that even one germline copy of the human JAK1 GOF allele is sufficient to drive AD-like pathology in mice, recapitulating the human skin phenotype.
Surprisingly, in contrast to the skin, the lungs of JAK1GOF mice did not display spontaneous inflammatory pathology when compared to those from age-matched WT control counterparts. In both JAK1GOF and control mice, assessment of airway remodeling and inflammatory infiltrate by hematoxylin and eosin (H&E) staining revealed no histopathologic evidence of lung inflammation (Figure 1P), and periodic acid-Schiff (PAS) staining revealed no evidence of goblet cell hyperplasia and mucus production (Figure 1Q). Further, the extent of CD45+ immune cell (Figures 1R and S1), CD4+ T cell (Figures 1S and S1), ILC2 (Figures 1T and S1), and eosinophil (Figures 1U and S1) enrichment was comparable if not reduced in the JAK1GOF mice compared to control mice. Collectively, these findings provoke the hypothesis that the lung harbors resistance to JAK1-mediated inflammation that is not observed in the skin.
Stromal JAK1GOF suppresses allergen-induced lung inflammation
We next examined whether challenging the airway with an allergen might provoke an enhanced inflammatory response not observed at steady state. To test this hypothesis, we treated both JAK1GOF and control mice intranasally with the asthma-associated fungal allergen Alternaria alternata (Figure 2A). In this well-established model of allergic lung inflammation, ILC2s promote eosinophil responses and allergic lung pathology.19–22 Indeed, histopathologic analysis revealed enhanced lung pathology in JAK1GOF mice compared to controls as demonstrated by increased airway remodeling, inflammatory infiltrate, goblet cell hyperplasia, and mucus production upon provocation by the allergen (Figures 2B–C). Further, we found that Alternaria alternata-treated JAK1GOF mice exhibit significantly elevated frequencies of ILC2s in their lungs compared to control mice (Figure 2D); however, we did not observe a difference in eosinophil frequency (Figure 2E). Taken together, these findings indicate that although global activation of JAK1 does not result in spontaneous allergic inflammation in the lung, provocation with an allergen renders JAK1GOF mice more susceptible to some features of allergic lung pathology.
Figure 2. Stroma-intrinsic expression of JAK1GOF is protective in allergic lung inflammation.
(A) Schematic of the allergic lung inflammation model using the protease allergen Alternaria alternata (ALT).
(B-C) Representative lung histology images of WT control and JAK1GOF mice challenged with intranasal ALT, obtained by (B) H&E stain and (C) PAS stain.
(D-E) Flow cytometry of lung tissue from WT control and JAK1GOF mice challenged with intranasal ALT. Shown are the frequencies of (D) ILC2s and (E) eosinophils. Data were obtained from three independent experiments, n = 5 mice per group.
(F) Schematic of experimental approach used to generate chimeric mice. BM derived from WT mice was transferred into WT control and experimental JAK1GOF mice.
(G) JAK1 genotype of the respective hematopoietic and stromal cell components in chimeric mice (WT→WT and WT→JAK1GOF).
(H) Schematic of experimental approach used to generate the allergic lung inflammation model in the BM chimeras.
(I-J) Representative lung histology images of chimeric WT→WT and WT→JAK1GOF mice challenged with intranasal ALT, obtained by (I) H&E stain and (J) PAS stain.
(K-L) Flow cytometry of lung tissue from chimeric WT→WT and WT→JAK1GOF mice challenged with intranasal ALT. Shown are the frequencies of (K) ILC2s and (L) eosinophils. Data were obtained from three independent experiments, n = 11–14 pooled mice per group.
(M) KEGG pathway overrepresentation analysis of RNA-seq data obtained from the lung tissues of chimeric WT→JAK1GOF vs. WT→WT mice (n = 2 mice/group). Overrepresentation analysis used Fisher’s exact test to determine the significance of enrichment. All terms shown were identified as the top four significantly suppressed pathways in terms of adjusted p-value (p-adjusted < 0.05, Benjamini-Hochberg procedure).
**p < 0.01, *p < 0.05, and not significant (n.s.) (Unpaired t-test with Welch’s correction). Data are represented as mean ± SD. Scale bars indicate 200 μm.
(A, F-G) Figure adapted from an image created with BioRender.com.
ALT- Alternaria alternata, BM – Bone marrow, BMT – Bone marrow transplantation.
See also Figure S1
Given the resistance to JAK1-mediated inflammation observed in the lung at steady state, we hypothesized that JAK1 activation within the lung stroma may confer tissue-specific resistance even upon allergen challenge. To test this hypothesis, we generated bone marrow (BM) chimeric mice in which we reconstituted both control and JAK1GOF mice with WT BM (WT→WT and WT→JAK1GOF) (Figures 2F–G). Upon airway challenge with Alternaria alternata (Figure 2H), WT→JAK1GOF mice exhibited markedly reduced lung pathology as evidenced by reduced peribronchial and perivascular inflammatory infiltrates (Figure 2I), goblet cell hyperplasia, and mucous production (Figure 2J) when compared to control WT→WT mice. Further, the frequencies of ILC2s (Figure 2K) and eosinophils (Figure 2L) were significantly reduced in WT→JAK1GOF mice compared to control WT→WT mice. Strikingly, KEGG pathway overrepresentation analysis of RNA-seq data obtained from lung tissue showed that expression of asthma- and inflammation-associated pathways were significantly suppressed in WT→JAK1GOF mice compared to control WT→WT mice (Figure 2M). While it is well-appreciated that JAK1GOF activates lymphocytes, these unanticipated findings indicate that stromal expression of JAK1GOF confers resistance to lung inflammation.
Sensory neurons suppress allergic lung inflammation
Recent studies demonstrated that neuropeptides can restrain ILC2 responses and lung inflammation.16,18 Thus, we hypothesized that sensory neurons in the lung may act to suppress inflammation. In contrast to the skin, which is almost entirely innervated by sensory neurons arising from the dorsal root ganglia (DRG), the lung is primarily innervated by sensory afferents that arise from the VG, with minor innervation from the DRG23 (Figure 3A). The majority of sensory neurons from the DRG express canonical nociceptive markers such as TRPV1 and Nav1.8.24–26 Sensory neurons in the vagus nerve exhibit expression of nociceptive ion channels comparable to that observed in DRG-derived neurons,27 as evidenced in our reanalysis of recently published single cell RNA-seq (scRNA-seq) datasets from vagal afferents28 (Figures 3B–C). To visualize the extent of TRPV1 and Nav1.8 expression within the VG and DRG, we generated reporter mice by crossing Trpv1Cre and Scn10aCre, respectively, to Rosa26STOPflox-tdTomato mice. Imaging of these reporter mice revealed widespread expression of both TRPV1 and Nav1.8 in both ganglia (Figures 3D–E). These findings suggested that sensory neurons within the VG can be chemically, genetically, and virally targeted.
Figure 3. Chemical denervation of TRPV1+ sensory neurons exacerbates allergic lung inflammation.
(A) Visual representation of the sensory innervation of the lung arising from the VG and DRG.
(B-C) Expression of Trpv1 and Scn10a (the gene encoding Nav1.8) within the VG, specifically the (B) JG, and (C) NG. Trpv1 expression is shown as the percentage of total neurons sequenced. Scn10a expression is shown as the percentage of Trpv1-expressing neurons. The full scRNA-seq dataset is available in Kupari et al.28
(D-E) Imaging of classical nociceptive markers (TRPV1 and Nav1.8) in the VG and DRG using Rosa26STOPflox-tdTomato mice crossed with the respective Trpv1Cre and Scn10aCre mice. (D) TRPV1 in the VG and DRG, (E) Nav1.8 in the VG and DRG.
(F) Schematic of the allergic lung inflammation model generated following systemic denervation of vagal and spinal visceral afferents.
(G) Withdrawal latency from noxious heat (hot plate test). Mice treated with s.c. vehicle control or RTX were placed on a hot plate set at 50 degrees Celsius, and withdrawal latency was measured as the time until the appearance of paw withdrawal behavior.
(H-I) Representative lung histology images from mice treated with s.c. vehicle control or RTX challenged with intranasal ALT, obtained by (H) H&E stain and (I) PAS stain.
(J-K) Flow cytometry of lung tissue from mice treated with s.c. vehicle control or RTX and challenged with intranasal ALT. Shown are the frequencies of (J) ILC2s and (K) eosinophils. Data were obtained from three independent experiments, n = 4–5 mice per group.
(L) Schematic of the allergic lung inflammation model generated following selective denervation of vagal afferents.
(M) Withdrawal latency from noxious heat (hot plate test). Mice treated with intraganglionic (i.g.) vehicle control or RTX were placed on a hot plate set at 50 degrees Celsius, and withdrawal latency was measured as the time until the appearance of paw withdrawal behavior.
(N-O) Representative lung histology images of mice treated with i.g. vehicle control or RTX challenged with intranasal ALT, obtained by (N) H&E stain and (O) PAS stain.
(P-Q) Flow cytometry of lung tissue from mice treated with i.g. vehicle control or RTX challenged with intranasal ALT. Shown are the frequencies of (P) ILC2s and (Q) eosinophils. Data were obtained from two independent experiments, n = 4–5 mice per group.
**p < 0.01, *p < 0.05, and not significant (n.s.) (Unpaired t-test with Welch’s correction). Data are represented as mean ± SD. Scale bars indicate 200 μm.
(A, F, L) Figure adapted from an image created with BioRender.com.
DRG – dorsal root ganglia, i.g. – intraganglionic, JG – jugular ganglia, NG – nodose ganglia, RTX – resiniferatoxin, s.c. – subcutaneous VG – vagal ganglia
See also Figures S1 and S2
TRPV1-expressing neurons can be chemically denervated by treatment with resiniferatoxin (RTX), an ultrapotent TRPV1 agonist.29,30 The majority of lung sensory innervation arises from vagal afferents while only a small proportion originates from DRG-derived spinal sensory neurons (i.e., spinal visceral afferents).23 Therefore, we first examined whether TRPV1+ neurons, including vagal and spinal sensory neurons, contribute to suppressing lung inflammation. WT C57BL/6 mice were subcutaneously (s.c.) injected with either vehicle or RTX; the latter leads to denervation of both vagal and spinal sensory neurons31 (Figure 3F). As expected, s.c. RTX treatment led to delayed paw withdrawal (enhanced withdrawal latency) in response to heat stimulation, indicating loss of noxious sensation (Figure 3G). Strikingly, mice chemically denervated of all sensory neurons, when compared to control mice, had increased peribronchial and perivascular inflammatory infiltrates (Figure 3H), goblet cell hyperplasia, and mucous production (Figure 3I). Further, the frequencies of ILC2s (Figure 3J) and eosinophils (Figure 3K) in the RTX-treated mice were significantly elevated as compared to vehicle-treated control mice. Additionally, transcriptomic analysis of lung tissue revealed upregulation of allergic inflammation-related genes (Figure S2A) and overrepresentation analysis of KEGG pathway revealed an enrichment of asthma- and inflammation-related pathways in the RTX-treated mice as compared to controls (Figure S2B).
We next investigated whether TRPV1+ sensory neurons, arising specifically from the VG, could regulate lung inflammation. To test this possibility, we performed intraganglionic (i.g.) injection of RTX or vehicle control to induce selective loss of sensory neurons from the VG (Figure 3L and S2C). Indeed, i.g. administration of RTX resulted in selective suppression of Trpv1 expression in the VG, while expression within DRG remained unchanged (Figure S2D–E). Further, chemical denervation of VG-specific sensory neurons did not affect paw withdrawal latency by the hot plate test (Figure 3M). However, denervation of VG-specific sensory neurons significantly affected Alternaria alternata-induced lung inflammation, which was evident from increased levels of peribronchial and perivascular inflammatory infiltrates (Figure 3N), goblet cell hyperplasia, and mucous production (Figure 3O) as well as increased frequencies of ILC2s (Figure 3P) and eosinophils (Figure 3Q) in the i.g. RTX-treated mice compared to controls.
We next tested the contribution of spinal sensory neurons from the DRG in regulating lung inflammation. RTX or vehicle was administered intrathecally (i.t.), (Figure S2F) to selectively impair spinal DRG sensory neurons.31 Indeed, i.t. RTX injection resulted in increased paw withdrawal latency to heat, indicating loss of sensation (Figure S2G). Impairment of DRG sensory neurons did not significantly alter the pathophysiology of Alternaria alternata-induced lung inflammation as histopathological analysis revealed no differences in the levels of peribronchial and perivascular inflammatory infiltrates (Figure S2H), goblet cell hyperplasia, and mucous production (Figure S2I) or in the frequencies of ILC2s (Figure S2J) and eosinophils (Figure S2K) between i.t. RTX- and vehicle-treated control mice. Transcriptomic analysis revealed very few differentially expressed genes (Figure S2L) and KEGG pathway overrepresentation analysis found no specific pathway enrichment induced by i.t. RTX treatment (Figure S2M). Collectively, these findings demonstrate that vagal sensory neurons critically suppress allergic lung inflammation.
Sensory neuron-intrinsic Jak1 regulates lung inflammation and levels of neuropeptides
Sensory neurons from the DRG express Jak1 to mediate critical functions such as itch.10 However, whether Jak1 is expressed in the VG has not been well addressed. Thus, we reanalyzed scRNA-seq datasets from the VG28 and found that the majority of Nav1.8+ sensory neurons co-express Jak1 (Figures 4A–B). We next hypothesized that Jak1 expression within vagal sensory neurons may be required for immune homeostasis in the lung. To test this, we employed mice that conditionally lack Jak1 in Nav1.8+ neurons (Jak1Δneuron mice; Scn10aCre:Jak1flox) (Figure 4C). Following allergen challenge in the airway, we found that all parameters of inflammation were significantly enhanced in Jak1Δneuron mice compared to controls, including histopathologic features of lung inflammation (Figures 4D–E) and frequencies of ILC2s (Figure 4F) and eosinophils (Figure 4G). These findings indicate that JAK1 signaling in Nav1.8+ sensory neurons regulates allergic inflammation in the lung.
Figure 4. Disruption of sensory neuron-intrinsic Jak1 exacerbates allergic lung inflammation and alters the expression levels of neuropeptides.
(A-B) Jak1 expression in Nav1.8+ vagal afferents, (A) JG and (B) NG. Gene expression is shown as the percentage of Nav1.8+ neurons that express Jak1. The full scRNA-seq dataset is available in Kupari et al.28
(C) Schematic of the allergic lung inflammation model using littermate control (Jak1flox) and experimental Jak1Δneuron mice.
(D-E) Representative lung histology images of control and Jak1Δneuron mice challenged with intranasal ALT, obtained by (D) H&E stain and (E) PAS stain.
(F-G) Flow cytometry of lung tissue from control and Jak1Δneuron mice challenged with intranasal ALT. Shown are the frequencies of (F) ILC2s and (G) eosinophils. Data were obtained from two independent experiments, n = 10–12 pooled mice per group.
(H-I) Profiling of neuropeptide expression in Nav1.8+ vagal afferents; Calca, Calcb, Tac1, Vip, and Nmu were evaluated in the (H) JG and (I) NG. Gene expression is shown as the percentage of Nav1.8+ neurons that express the indicated gene. The full scRNA-seq dataset is available in Kupari et al.28
(J-K) Imaging of the classical neuropeptides CGRPα and CGRPβ in the VG and DRG using Rosa26STOPflox-tdTomato mice crossed with CalcaCre and CalcbCre/ERT mice, respectively. Shown are (J) CGRPα in the VG and DRG and (K) CGRPβ in the VG and DRG.
(L) MotifMap (human)-predicted neuropeptide targets of STAT family members. Data were analyzed using the MotifMap Predicted Transcription Factor Targets dataset (see Methods).
(M-R) Quantitative PCR (qPCR) of transcripts from the VG and DRG from control and Jak1Δneuron mice challenged with intranasal ALT. Shown are (M, N) Calcb, (O, P) Calca, and (Q, R) Tac1 transcript levels. Data were obtained from two independent experiments, n = 3–4 mice per group.
**p < 0.01, *p < 0.05, and not significant (n.s.) (Unpaired t-test with Welch’s correction). Data are represented as mean ± SD. Scale bars indicate 200 μm.
(C) Figure adapted from an image created with BioRender.com.
See also Figures S1 and S3–5
We previously showed that both JAK1 and upstream IL-4Rα on spinal sensory neurons critically regulate itch in the setting of skin inflammation.10 To test whether these pathways exhibit similar overlapping neural functions in the airway, we also subjected Il4raΔneuron mice (conditional deletion of Il4ra on Nav1.8+ sensory neurons; Scn10aCre:Il4raflox) to allergic lung inflammation (Figure S3A). However, we found no effect of Il4ra deletion in this setting (Figures S3B–F). Thus, despite the presence of a neuronal IL-4Rα-JAK1 itch axis in the skin, IL-4Rα appears dispensable in lung sensory neurons in regulating allergic lung inflammation. Further, given the striking nature of the lung pathology following conditional deletion of Jak1 in sensory neurons, we sought to test whether neuronal JAK1 signaling may be relevant across multiple model systems. Thus, we employed an alternative mouse model of allergic lung inflammation induced by the protease allergen papain. Following papain challenge in the airway of Jak1Δneuron mice (Figure S3G), we observed enhanced histopathologic features of lung inflammation (Figures S3H–I) and increased frequencies of CD45+ immune cells (Figure S3J), ILC2s (Figure S3K), and eosinophils (Figure S3L) compared to control mice.
Although our findings demonstrate that neuron-intrinsic JAK1 signaling is required for homeostasis in the face of allergic lung inflammation, whether this function is impacted by JAK1-selective inhibitors in humans remains unclear. Strikingly, recent phase 1 and 2 clinical trials with multiple inhalational JAK1-selective inhibitors have failed in asthma (NCT03766399, NCT04150341, ACTRN12617001227381). Thus, we hypothesized that inhalational delivery of JAK1-selective inhibitors may display limited efficacy in vivo due to its restriction to the airway and thus preferential access to sensory neurons; in contrast, systemic delivery of JAK1-selective inhibitors may yet derive efficacy due to their capacity to potently target the hematopoietic compartment. To test this hypothesis, we compared the efficacy of upadacitinib, a JAK1-selective inhibitor, administered either intranasally (inhalational) or intraperitoneally (systemic) in the Alternaria alternata-induced allergic lung inflammation model (Figure S4A). Although both inhalational and systemic delivery of upadacitinib significantly reduced the number of eosinophils in the lung (Figure S4B), only systemic treatment resulted in suppression of ILC2s (Figure S4C). Further, only systemic delivery of upadacitinib resulted in an overall improvement of lung pathology (Figure S4D), goblet cell hyperplasia, and mucous content (Figures S4E). Collectively, these data suggest that inhalational delivery of JAK1-selective inhibitors may have limited efficacy.
Increasing evidence suggests that sensory neurons release neuropeptides to modulate inflammation at multiple barrier surfaces.13,16,18,32–34 However, the intracellular mechanisms guiding such neuronal responses remain poorly understood. Although JAK1 has been recently appreciated for its ability to relay sensory signals toward the spinal cord (afferent function), whether JAK1 controls expression of neuropeptides in sensory neurons to modulate neuroinflammation in tissues (efferent function) has not been explored. Thus, we sought to screen the expression of various neuropeptides within the VG using existing scRNA-seq datasets.28 Indeed, both the jugular ganglia (JG) and nodose ganglia (NG) express the classical neuropeptide Calca, which encodes the α form (CGRPα) of the neuropeptide used to identify peptidergic neurons (Figures 4H–I). Recent studies have demonstrated the capacity of CGRPα to suppress ILC2 responses in the lung.16,18 However, the expression of Calcb (the gene encoding CGRPβ) and the function of CGRPβ in the lung remain poorly understood. We also screened other neuropeptide genes, including Tac1 (the gene encoding Substance P [SP]), Vip (the gene encoding vasoactive intestinal peptide [VIP]), and Nmu (the gene encoding neuromedin U [NMU]) (Figures 4H–I). To visualize the extent of Calca and Calcb expression within the VG and DRG, we generated reporter mice by crossing CalcaCre and CalcbCre/ERT mice, respectively, to Rosa26STOPflox-tdTomato mice. Strikingly, imaging of these reporter mice revealed widespread expression of both Calca and Calcb in both the VG and DRG (Figures 4J–K).
The ability of JAK1 to alter cellular programs is largely dependent on downstream phosphorylation of STAT proteins, which enter the nucleus to alter transcription. Indeed, overactivity of STAT6 is strongly associated with allergic inflammatory processes, and STAT6 GOF was recently reported to be associated with similar atopic syndromes as observed for JAK1 GOF.35–40 We sought to examine whether STAT6 is associated with regulation of specific neuropeptide regulatory elements via in silico analysis of datasets from MotifMap Predicted Transcription Factor Targets (see Methods). We found that CALCB and TAC1, but not CALCA, NMU, and VIP, were predicted transcriptional targets of STAT6 (Figure 4L). Further, analysis of transcription factor DNA-binding motifs revealed a STAT6 DNA-binding motif ~500–2000 bp upstream of CALCB, TAC1, and CALCA, but not NMU, and VIP, suggesting that STAT6 may selectively regulate expression of genes that encode CGRP and SP. Thus, we hypothesized that JAK1 may regulate CGRPβ and/or SP.
We next subjected both Jak1Δneuron and control mice to airway allergen challenge and examined the VG for expression of neuropeptide transcripts (Figure S5A). Strikingly, we found that expression of Calcb was decreased within the VG (Figure 4M) of Jak1Δneuron mice compared to control mice. However, this Calcb deficiency was not observed within the DRG (Figure 4N). While no alterations were observed in Calca and Vip expression (Figures 4O–P and S5B–C), Tac1 was reduced in both the VG and DRG of Jak1Δneuron mice (Figures 4Q–R); Nmu was only reduced in the DRG of Jak1Δneuron mice (Figures S5D–E). In contrast to Jak1Δneuron mice, JAK1GOF mice exhibited increased expression of Calcb, but not Tac1, in their VG compared to control mice (Figures S5F–H). Taken together, these findings indicate that JAK1 regulates the expression of multiple neuropeptides, including Calcb, in the VG. Given these findings, along with our in silico analysis predicting CALCB as a transcriptional target of STAT6, we hypothesized that CGRPβ may be a key regulatory neuropeptide controlled by JAK1.
In light of our findings that both chemical denervation via i.g. RTX (Figures 3L–Q) and sensory neuron-intrinsic deletion of Jak1 (Figures 4C–G) results in enhanced allergic lung inflammation, we sought to test whether RTX-mediated denervation of TRPV1+ sensory neurons impact CGRPβ expression within the vagus nerve. To test this, we treated Calcb-reporter (CalcbCre/ERT:Rosa26STOPflox-tdTomato) mice with i.g. RTX and performed immunofluorescence imaging of total CGRPβ+ neurons (Figure S5I). Strikingly, i.g. RTX-mediated denervation demonstrated a marked reduction of CGRPβ+ neurons compared to the vehicle-treated mice (Figure S5J–K)
CGRPβ suppresses ILC2 function and allergic lung inflammation
Given that ILC2s are important drivers of allergic lung inflammation20,41 and the role of CGRPβ in this context is not well understood, we sought to test whether CGRPβ can directly suppress the function of ILC2s in the lungs. We sort-purified lung ILC2s from WT C57BL/6 mice and stimulated them in vitro with the alarmin cytokine IL-33, in combination with either CGRPβ or SP (Figure 5A). Strikingly, we found that CGRPβ potently suppressed IL-33-mediated stimulation of IL-5 and IL-13 production from ILC2s (Figures 5B–C). In contrast, the classical proinflammatory neuropeptide SP had no effect on IL-33-elicited activation of lung ILC2s (Figures 5B–C). Similar to CGRPβ, CGRPα also suppressed ILC2 function (Figures S6A–C), a well-known feature of this isoform.16,18 However, of these two isoforms, only Calcb was found to be differentially regulated in the VG of Jak1Δneuron mice (Figures 4M–P). Our findings demonstrate that CGRPβ can directly suppress ILC2 function and suggest its potential role in restraining allergic lung inflammation.
Figure 5. CGRPβ suppresses type 2 cytokine production from lung ILC2s and allergic lung inflammation.
(A) Schematic of the lung ILC2 suppression assay.
(B-C) Type 2 cytokine measurement from lung ILC2s stimulated with IL-33, SP, and/or CGRPβ.
Shown are data for (B) IL-5 and (C) IL-13.
(D) Schematic of the induction of allergic lung inflammation in Rag1−/− mice.
(E-F) Flow cytometry of lung tissue from Rag1−/− mice challenged with intranasal ALT as well as vehicle control or CGRPβ or CGRPβ 8–37. Shown are the frequencies of (E) ILC2s and (F) eosinophils. Data were obtained from two independent experiments, n = 4–5 mice per group.
(G-H) Representative lung histology images of Rag1−/− mice challenged with intranasal ALT as well as vehicle control or CGRPβ or CGRPβ 8–37, obtained by (G) H&E stain and (H) PAS stain.
***p < 0.001, **p < 0.01, *p < 0.05 (Unpaired t-test with Welch’s correction). Data are represented as mean ± SD. Scale bars indicate 200 μm.
(A, D) Figure adapted from an image created with BioRender.com.
ELISA – enzyme-linked immunosorbent assay, IL – interleukin, SP – substance P
See also Figures S1 and S6
Allergic lung inflammation is uniquely dependent on ILC2s in adaptive lymphocyte-deficient Rag1−/− mice.19,41 Thus, to specifically evaluate whether exogenous CGRPβ could suppress ILC2 responses in vivo and allergic lung inflammation, we challenged Rag1−/− mice with intranasal Alternaria alternata and simultaneously treated them with either intranasal CGRPβ or the CGRP receptor antagonist CGRPβ 8–37 (Figure 5D). Strikingly, while Alternaria alternata-treated and control vehicle-treated Rag1−/− mice developed robust ILC2 (Figure 5E) and eosinophil responses (Figure 5F) in association with lung pathology (Figures 5G–H), CGRPβ-treated mice exhibited attenuated immune cell responses (e.g., eosinophils) and tissue inflammation (Figures 5E–H). Conversely, treatment with the CGRPβ receptor antagonist resulted in aggravated allergic lung inflammation (Figures 5E–H). Consistent with our in vitro findings (Figures 5A–C), SP did not suppress allergic lung inflammation (Figures S6D–H). To test whether CGRPβ displayed a suppressive function in the presence of adaptive immunity, we also delivered it to the airways of WT mice with allergic lung inflammation and observed similar anti-inflammatory effects to those observed in Rag1−/− mice (Figures S6I–M). Collectively, these findings indicate that while neuron-intrinsic expression of Jak1 selectively regulates expression of Calcb in the VG (Figure 4M), CGRPβ, in turn, has the capacity to regulate lung inflammation.
Retrograde viral delivery and conditional insertion of human JAK1 GOF into sensory neurons are protective against allergic lung inflammation
Next, we sought to test whether the delivery of human JAK1 GOF specifically to lung-innervating neurons would be sufficient to suppress allergic lung inflammation. To test this hypothesis, we introduced retrograde Adeno-Associated Virus (AAV) co-expressing Cre recombinase and the human JAK1 GOF variant into Rosa26STOPflox-tdTomato mice and subjected them to Alternaria alternata allergen challenge in the airway (Figure 6A). We found that most reporter-labeled neurons were housed in the VG and were virtually undetectable in the corresponding thoracic DRG (Figures 6B–C and Videos S1–2). These findings indicate that vagal sensory neurons were infected with AAV and thus harbored the JAK1 GOF variant. Strikingly, histopathological analysis revealed that mice infected with AAV-Cre/JAK1GOF in the airway exhibited robust suppression of allergic lung inflammation (Figures 6D–E) and reduction in the frequencies of CD45+ immune cells (Figure 6F), ILC2s (Figure 6G), and eosinophils (Figure 6H).
Figure 6. Expression of human JAK1GOF in lung sensory neurons suppresses allergic inflammation.
(A) Schematic of the allergic lung inflammation model generated by delivery of AAV-Cre (control) or AAV-Cre/JAK1 GOF (experimental) into the airways of Rosa26STOPflox-tdTomato mice.
(B) Imaging of tdTomato in the DRG and VG following AAV-assisted gene delivery into lung-innervating sensory neurons. Shown are representative images of DRG and VG from mice treated with AAV-Cre/JAK1GOF.
(C) Numbers of tdTomato+ neurons in DRG (thoracic segments 4–8) and VG, n = 5 mice per group. Data were analyzed in mice treated with AAV-Cre/JAK1 GOF.
(D-E) Representative lung histology images of mice challenged with intranasal ALT following AAV-Cre control and AAV-Cre/JAK1GOF infection, obtained by (D) H&E stain and (E) PAS stain.
(F-H) Flow cytometry of lung tissue from mice challenged with intranasal ALT following AAV-Cre control and AAV-Cre/JAK1GOF infection. Shown are the frequencies of (F) total CD45+ immune cells, (G) ILC2s, and (H) eosinophils. Data were obtained from two independent experiments, n = 4–5 mice per group.
(I) Schematic of the allergic lung inflammation model generated by conditional insertion of JAK1GOF variant into sensory neurons.
(J-K) Representative lung histology images of mice challenged with intranasal ALT obtained by (J) H&E stain and (K) PAS stain.
(L-N) Flow cytometry of lung tissue from mice challenged with intranasal ALT. Shown are the frequencies of (L) total CD45+ immune cells, (M) ILC2s, and (N) eosinophils.
(O) Protein levels of CGRPβ in the bronchoalveolar lavage fluid (BALF). Data were obtained from two independent experiments, n = 4–5 pooled mice per group.
*p < 0.05, **p < 0.01, and not significant (n.s.) (Unpaired t-test with Welch’s correction). Data are represented as mean ± SD. Scale bars indicate 200 μm.
(A, I) Figure adapted from an image created with BioRender.com.
AAV – adeno-associated virus; specifically, AAV2-retro, BALF – Bronchoalveolar lavage fluid
Finally, we sought to test whether conditional insertion of the same human JAK1 GOF variant into sensory neurons would be sufficient to suppress allergic lung inflammation as well. Thus, we crossed sensory neuron-specific Scn10aCre mice with Jak1STOPflox-JAK1 GOF (JAK1GOF:neuron) mice. These mice are conditionally deficient in murine Jak1, but harbor human JAK1GOF only within sensory neurons. We then subjected these JAK1GOF:neuron mice to Alternaria alternata allergen challenge in the airway (Figure 6I). Indeed, similar to retrograde viral delivery of the JAK1 GOF variant, histopathological analysis exhibited robust suppression of allergic lung inflammation (Figures 6J–K) and reduction in the frequencies of CD45+ immune cells (Figure 6L), ILC2s (Figure 6M), and eosinophils (Figure 6N). Strikingly, the levels of CGRPβ protein in the airway bronchiolar lavage fluid (BALF) was significantly increased compared to control mice (Figure 6O). Collectively, these findings demonstrate that JAK1 regulates CGRPβ in sensory neurons and promotes immune homeostasis in the lung.
DISCUSSION
The canonical type 2 cytokines IL-4, IL-5, and IL-13 are universally elevated across both AD (eczema) and asthma.42 However, studies of targeted monoclonal antibody (mAb)-based therapies strongly suggest that although IL-13 is pathogenic in AD, it is a poor target in asthma.43–47 Conversely, while IL-5 is a driver of asthma, it has failed as a therapeutic target in AD.48–51 Taken together, these findings suggest that elevated signaling of the same cytokine pathways may imprint different phenotypes across tissues like the skin and lung. Further, while type 2 cytokines share and employ downstream JAK1 signaling to activate different cellular programs,52 the mechanisms by which JAK1 activity can result in different allergic phenotypes remain unclear. Strikingly, even JAK1 variants that are common in the population are associated with AD and eosinophil enrichment but not asthma (see GWAS-based analysis in Methods).53 Further, while multiple JAK1-selective inhibitors are FDA approved for AD, none are approved for asthma, and multiple inhalational JAK1-selective inhibitors have failed in clinical trials (NCT03766399, NCT04150341, ACTRN12617001227381). These findings suggest that JAK1 signaling has tissue-specific properties that uniquely impact particular diseases and their associated therapies.
By transposing the human JAK1 GOF allele into mice, we found that while AD-like disease develops spontaneously, activation of JAK1 in vagal sensory neurons confers resistance to allergic lung inflammation. Although patients with JAK1GOF spontaneously develop asthma,11,12 this phenotype was not observed in our JAK1GOF mice. We hypothesize that this difference may be due to limited exposure to environmental allergens in laboratory animals. Indeed, protease allergens such as house dust mite can directly stimulate sensory neurons to induce the release of neuropeptides.54,55 Indeed, only upon challenge with the allergen Alternaria alternata, did we observe unique susceptibility to allergic lung inflammation in JAK1GOF mice. These findings suggest that, independent of immune hypersensitivity, allergens may disrupt neuroimmune circuits to promote inflammation.
While JAK1 activation in lymphocytes is broadly proinflammatory, our findings suggest that JAK1 activation within the sensory nervous system serves a previously unrecognized immunoregulatory role. This observation may explain why JAK1-selective inhibitors, while highly successful in AD, have not advanced to treatment in asthma.1 Our findings highlight the need to better understand the cell-specific mechanisms by which cytokines imprint distinct JAK-STAT programs and pathology across different organ systems.
Employing a variety of different chemical, genetic, and viral neuronal manipulation approaches, we found that sensory neuron-intrinsic expression of murine Jak1 or exogenous human JAK1GOF is required to protect against allergen-induced lung inflammation. Further, we found that vagal sensory nerves are the key drivers of this homeostatic mechanism in the airway. By screening various neuropeptides associated with neuroinflammation, we find that JAK1 regulates Calcb expression in the VG, and CGRPβ directly suppresses lung inflammation. While recent studies have shown that a number of neuropeptides including NMU, SP, and VIP have proinflammatory functions in the airway,56–58 CGRPα has demonstrated anti-inflammatory properties.14,16–18 However, the role of CGRPβ is less understood. Thus, our findings highlight a previously unrecognized and selective role for the CGRPβ isoform in suppressing allergic lung inflammation. Future exploration into the differences between CGRPα and CGRPβ isoforms across tissues may yield deeper insights into barrier immunity and inflammation.
Understanding how various neuropeptides regulate inflammation in the skin, lung, and gut is a major emerging field of neuroimmunology.13,16,18,32–34 However, the cytokine signals that drive specific neuropeptide programs within these neuroimmune circuits remain poorly understood. The mechanisms by which JAK1 influences the transcription and release of CGRPβ remain unknown. Based on our in silico analyses, we suspect that downstream phosphorylation of STAT6 is likely a key mechanism that drives Calcb transcription. However, we also speculate that JAK1 may regulate the release of preformed CGRPβ from the synaptic vesicle in the axon terminal. Thus, JAK1 may have multiple functions within sensory neurons that are both STAT6-independent (CGRPβ protein release) and -dependent (Calcb transcription). Future studies aimed at understanding these pathways may reveal functions of JAK1 that are specific to sensory neurons and likely not observed in lymphocytes. Although our study reveals a mechanism by which sensory neurons employ JAK1 as a putative cytokine ‘sensor,’ discovery and characterization of additional JAK-STAT variants may reveal a highly dynamic landscape by which sensory neurons sense, process, and modulate inflammatory signals in tissues.
Sensory nerves consist of a highly sophisticated system that innervates virtually every organ in the body, including secondary lymphoid organs.59,60 Although traditionally thought to be only sensory in function, sensory neurons have been found in recent studies to promote barrier immunity to pathogens, regulate metabolism, and even contribute to cancer immunosurveillance.32,61–65 Thus, our observation that they suppress lung inflammation further underscores an emerging paradigm in which the sensory nervous system may regulate a variety of processes across the organism. Taken together, sensory neuromodulation may soon represent a therapeutic approach to treat a variety of inflammatory, infectious, metabolic, and even malignant disorders.
Limitations of the study
Although we identified a critical link between JAK1 within vagal sensory neurons and its ability to regulate Calcb and allergic lung inflammation, our study has limitations. First, the precise mechanisms by which neuron-intrinsic JAK1 modulates transcription, synthesis, and/or release of Calcb/CGRPβ remain unknown. Second, the neurons that express JAK1 within the human VG remain unknown. Third, whether inhibition of human vagal neuron-associated JAK1 alters neuropeptide expression has not been explored. Future studies in primary or iPSC-derived human vagal sensory neurons will be required to understand how our findings translate to human biology.
STAR METHODS
RESOURCE AVAILABILITY
Lead contact
Further information and requests for resources and reagents should be directed to and will be fulfilled by the lead contact, Brian S. Kim (itchdoctor@mountsinai.org).
Materials availability
JAK1 GOF mice will be made available upon request.
Data and code availability
No original codes were generated in this study.
Accession number is listed in the key resources table to account for all high throughput sequencing data.
KEY RESOURCES TABLE
REAGENT or RESOURCE | SOURCE | IDENTIFIER |
---|---|---|
Antibodies | ||
| ||
anti-mouse CD16/CD32 | Bio X Cell | Cat#: BE0307; RRID: AB_1107647 |
anti-mouse CD16/32 | Biolegend | Cat#: 101302; RRID: AB_312801 |
anti-mouse CD3 | BioLegend | Cat#: 100268; RRID: AB_2876392 |
anti-mouse CD3 | BioLegend | Cat#: 100247; RRID: AB_2572117 |
anti-mouse CD3e | BioLegend | Cat#: 100304; RRID: AB_312669 |
anti-mouse CD4 | BioLegend | Cat#: 100404; RRID: AB_312689 |
anti-mouse CD4 | BioLegend | Cat#: 100449; RRID: AB_2564587 |
anti-mouse CD8a | BioLegend | Cat#: 100704; RRID: AB_312743 |
anti-mouse CD8a | BioLegend | Cat#: 100782; RRID: AB_2819775 |
anti-mouse/human CD11b | BioLegend | Cat#: 101204; RRID: AB_312787 |
anti-mouse/human CD11b | BioLegend | Cat#: 101233; RRID: AB_10896949 |
anti-mouse CD11c | BioLegend | Cat#: 117304; RRID: AB_313773 |
anti-mouse CD11c | BioLegend | Cat#: 117326; RRID: AB_117326 |
anti-mouse CD11c | BioLegend | Cat#: 117339; RRID: AB_2562414 |
anti-mouse CD19 | Invitrogen | Cat#: 45-0193-82; RRID: AB_1106999 |
anti-mouse CD19 | Biolegend | Cat#: 115504; RRID: AB_313639 |
anti-mouse CD45 | Invitrogen | Cat#: 58-0451-82; RRID: AB_11218871 |
anti-mouse CD45 | Biolegend | Cat#: 103138; RRID: AB_2563061 |
anti-mouse CD45R/B220 | BioLegend | Cat#: 103275; RRID: AB_2860602 |
anti-mouse CD90.2 | BD Biosciences | Cat#: 740442; RRID: AB_2740169 |
anti-mouse CD117 (c-Kit) | BioLegend | Cat#: 105838; RRID: AB_2616739 |
anti-mouse CD117 (c-Kit) | Invitrogen | Cat#: 62-1171-82; RRID: AB_2637141 |
anti-mouse CD127 (IL-7Ra) | BioLegend | Cat#: 135013; RRID: AB_1937266 |
anti-mouse CD127 (IL-7Ra) | BioLegend | Cat#: 135035; RRID: AB_2564577 |
anti-mouse CD127 (IL-7Ra) | BioLegend | Cat#: 135037; RRID: AB_2565269 |
anti-mouse Siglec-F | BD Bioscience | Cat#: 562757; RRID: AB_2687994 |
anti-mouse Siglec-F | BioLegend | Cat#: 155506; RRID: AB_2750235 |
anti-mouse FceRIa | Biolegend | Cat#: 134304; RRID: AB_1626106 |
anti-mouse I-A/I-E | BioLegend | Cat#: 107622; RRID: AB_493727 |
anti-mouse/human KLRG1 (MAFA) | BioLegend | Cat#: 138418; RRID: AB_2563015 |
anti-mouse/human KLRG1 (MAFA) | BioLegend | Cat#: 138419; RRID: AB_2563357 |
anti-mouse Ly6A/E (Sca-1) | BD Biosciences | Cat#: 749199; RRID: AB_2873577 |
anti-mouse Ly6A/E (Sca-1) | Biolegend | Cat#: 108112; RRID: AB_313349 |
anti-mouse Ly-6G | Invitrogen | Cat#: 62-9668-82; RRID: AB_2762763 |
anti-mouse Ly-6G | BioLegend | Cat#: 127643; RRID: AB_2565971 |
anti-mouse Ly-6G/Ly-6C (Gr-1) | BioLegend | Cat#: 108404; RRID: AB_313369 |
| ||
Bacterial and virus strains | ||
| ||
AAV2-retro-Cre | VectorBuilder | Vector ID:VB220605-1075ztp |
AAV2-retro-Cre/JAK1 | VectorBuilder | Vector ID:VB220526-1322bxa |
| ||
Chemicals, peptides, and recombinant proteins | ||
| ||
Alternaria alternata | GREER | Cat#: NC1620293 |
β-CGRP (human) | Cayman Chemical | Cat#: 24725 |
β CGRP 8-37 human | MiliporeSigma | Cat#: SCP0060 |
Bovine Serum Albumin | MiliporeSigma | Cat#: A9418 |
Bovine Serum Albumin | MiliporeSigma | Cat#: A9576 |
CGRP (rat) | R&D | Cat#: 1161 |
Collagenase Type 5 | Worthington Biochemicals | Cat#: LS005282 |
DNase I | Roche | Cat#: 10104159001 |
DNase I | Worthington Biochemicals | Cat#: LS002139 |
Fetal Bovine Serum | MiliporeSigma | Cat#: F4135 |
Hialuronidase type 4 | MiliporeSigma | Cat#: H3384-500MG |
Histodenz | MiliporeSigma | Cat#: D2158 |
Liberase TM | Roche | Cat#: 5401127001 |
Papain from papaya latex | MiliporeSigma | Cat#: P4762-25MG |
Percoll™ | GE healthcare | Cat#: 17-0891-2 |
Quadrol | MiliporeSigma | Cat#: 122262 |
Resiniferatoxin | Alomone labs | Cat#: R-400 |
recombinant mouse IL-2 | R&D systems | Cat# 402-ML-020/CF |
recombinant mouse IL-33 | R&D systems | Cat#: 3626-ML-010/CF |
Substance P | Tocris | Cat#: 1156 |
Upadacitinib (ABT-494) | MedChemExpress | Cat#: HY-19569 |
| ||
Critical commercial assays | ||
| ||
Brilliant Violet 421 Streptavidin | Biolegend | Cat#: 405225 |
CountBrightTM Absolute Counting Beads, for flowcytometry | Invitrogen | Cat#: C36950 |
Fixation/Permeablization Kit | BD Biosciences | Cat#: 554714 |
Flexable Viability Dye eFluor™780 | Invitrogen | Cat#: 65-0865-18 |
IL-5 Mouse Uncoated ELISA Kit | Invitrogen | Cat#: 88-7054-88 |
IL-13 Mouse Uncoated ELISA Kit | Invitrogen | Cat#: 88-7137-22 |
Mouse CGRP2 ELISA Kit | MyBioSource | Cat#: MBS7241942 |
Power SYBR™ Green PCR Master Mix | Thermo Fisher Scientific | Cat#: 4367660 |
Streptavidin Microbeads | Miltenyi Biotec | Cat#: 130-048-101 |
UltraComp eBeads™ Compensation Beads | Invitrogen | Cat#: 01-2222-42 |
Viability Dye | Invitrogen | Cat#: 65-0865-14 |
Zombie NIR™ Fixable Viability Kit | BioLegend | Cat#: 423106 |
| ||
Deposited data | ||
| ||
Raw and analyzed bulk RNA-seq data | ISMMS BiNGS Core | GEO: GSE220497 |
| ||
Experimental models: Organisms/strains | ||
| ||
C57BL/6J | Jackson Laboratory | Stock No. 000664 |
C57BL/6J | Sankyo Labo Service Corpration, Inc. | N/A |
CalcaCre: Rosa26STOPflox-tdTomato | Hongzhen Hu | N/A |
CalcbCre/ERT: Rosa26STOPflox-tdTomato | Hongzhen Hu | N/A |
CalcbCre/ERT: Rosa26STOPflox-tdTomato | Brian Kim | N/A |
Il4ra flox | Frank Brombacher | N/A |
Jak1 flox | Nanjing Biomedical Research Institute | N/A |
Jak1 STOPflox-JAK1 GOF | Cyagen Biosciences Inc. | N/A |
JAK1 GOF | Cyagen Biosciences Inc. | N/A |
Scn10a Cre | Rohini Kuner | N/A |
Scn10aCre: Il4raflox | Brian Kim | N/A |
Scn10aCre: Jak1flox | Brian Kim | N/A |
Scn10aCre: Jak1STOPflox-JAK1 GOF | Brian Kim | N/A |
Scn10aCre: Rosa26STOPflox-tdTomato | Brian Kim | N/A |
Trpv1Cre: Rosa26STOPflox-tdTomato | Hongzhen Hu | N/A |
Rag1 −/− | Jackson Laboratory | Stock No. 002216 |
Rosa26 STOPflox-tdTomato | Jackson Laboratory | Stock No. 007909 |
| ||
Oligonucleotides | ||
| ||
Gapdh qRT-PCR primer 1 | integrated DNA Technologies | TCAAGAAGGTGGTGAAGCAGG |
Gapdh qRT-PCR primer 2 | integrated DNA Technologies | TATTATGGGGGTCTGGGATGG |
Calca qRT-PCR primer 1 | integrated DNA Technologies | GAGGGCTCTAGCTTGGACAG |
Calca qRT-PCR primer 2 | integrated DNA Technologies | AAGGTGTGAAACTTGTTGAGGT |
Calcb qRT-PCR primer 1 | integrated DNA Technologies | CTCTCAGCACGATATGGGTCC |
Calcb qRT-PCR primer 2 | integrated DNA Technologies | GCAAGAGATGTTTTTCCTGGTCG |
Nmu qRT-PCR primer 1 | integrated DNA Technologies | GAGGGAGCTTTGCCGTATAGT |
Nmu qRT-PCR primer 2 | integrated DNA Technologies | GATGCACAACAGAGGACACAA |
Tac1 qRT-PCR primer 1 | integrated DNA Technologies | TTTCTCGTTTCCACTCAACTGTT |
Tac1 qRT-PCR primer 2 | integrated DNA Technologies | GTCTTCGGGCGATTCTCTGC |
Trpv1 qRT-PCR primer 1 | Thermo Fisher Scientific | AGCTGCAGCGAGCCATCACCA |
Trpv1 qRT-PCR primer 2 | Thermo Fisher Scientific | ATCCTTGCCGTCCGGCGTGA |
Vip qRT-PCR primer 1 | integrated DNA Technologies | AGTGTGCTGTTCTCTCAGTCG |
Vip qRT-PCR primer 2 | integrated DNA Technologies | GCCATTTTCTGCTAAGGGATTCT |
| ||
Software and algorithms | ||
| ||
clusterProfiler R package (v4.2.2) | G Yu et. al | https://guangchuangyu.github.io/software/clusterProfiler/ |
DEGReport R package (v1.30.3) | L Pantano | https://github.com/lpantano/DEGreport |
DESeq2 (v1.34.0) R package | M Love et. al | https://github.com/mikelove/DESeq2 |
FastQC (v0.11.8) | Babraham Bioinformatics | https://www.bioinformatics.babraham.ac.uk/projects/fastqc/ |
FlowJo 10 | Tree Star | https://www.flowjo.com/ |
Galore! (v0.6.6) | Babraham Bioinformatics | https://www.bioinformatics.babraham.ac.uk/projects/trim_galore/ |
Glimma (v2.4.0) R packages | S Su et. al | https://github.com/Shians/Glimma |
NIS-Elements | Nikon Instruments | https://www.microscope.healthcare.nikon.com/products/software |
plotly R package (v4.9.4.1) | C Sievert et. al | https://plotly.com/r/ |
Prism 9 | GraphPad Software | https://www.graphpad.com/scientificsoftware/prism/ |
R (v.4.1.0) | R Core Team | https://www.r-project.org/ |
STAR aligner (v2.7.5b) | A Dobin et. al | https://github.com/alexdobin/STAR |
Salmon (v1.2.1) | R Patro et. al | https://combine-lab.github.io/salmon/ |
Ugent Center for Plant Systems Biology online tool | UCPSB: Bioinformatics & Evolutionary Genomics | https://bioinformatics.psb.ugent.be/webtools/Venn/ |
| ||
Other | ||
| ||
AutoMACS | Miltenyi Biotec | N/A |
Bead Homogenizer | BioSpec | Cat#: 112011 |
Cytek® Aurora | CYTEK | N/A |
FACSAria™ III Cell Sorter | BD | N/A |
FVMPE-RS | Olympus | N/A |
gentleMACS™ Octo Dissociator with Heaters | Miltenyi Biotec | Cat#: 130-096-427 |
Nikon 80i; CoolSnap ES camera | Nikon Instruments | N/A |
QuantStudio 6 Flex Real-Time PCR Systems | Thermo Fisher Scitntific | N/A |
Remote Infuse/Withdraw Pump 11 Elite Nanomite Programmable Syringe Pump | Harvard Apparatus | 70-4507 |
StepOnePlus qRT-PCR System | Thermo Fisher Scientific | N/A |
EXPERIMENTAL MODEL AND STUDY PARTICIPANT DETAILS
Human study participants
Human study participants study was approved by the institutional review board of the University of British Columbia under protocol number H15–00641. Patients with JAK1 GOF variants have been previously described in Del Bel et al.11 Diagnosis of AD and asthma followed guidelines of the European and North American consensus report66 and the Global Initiative for Asthma (https://ginasthma.org), respectively. Eczema Area and Severity Index (EASI) was used to score AD severity based on methodology described in Leshem et al.67 Written informed consent was obtained from parents as study participants were minors at the time of consent.
Animal studies
C57BL/6 wild-type (WT), Rag1−/−, and Rosa26STOPflox-tdTomato mice were purchased from Jackson Laboratory. Human JAK1 GOF (JAK1GOF) mice and Jak1STOPflox-JAK1 GOF mice were generated through Cyagen Biosciences, Inc. Schematic of the targeting strategy used to introduce the germline human JAK1 GOF mutation into the murine Jak1 locus is outlined in Figure 1H. Scn10aCre mice were provided by Dr. Rohini Kuner (Heidelberg University, Heidelberg, Germany). Jak1flox mice were purchased from Nanjing Biomedical Research Institute of Nanjing University. Il4raflox mice were generated by Dr. Frank Brombacher (International Center for Genetic Engineering and Biotechnology, Cape Town, South Africa). Trpv1Cre:Rosa26STOPflox-tdTomato, CalcaCre:Rosa26STOPflox-tdTomato, and CalcbCre/ERT:Rosa26STOPflox-tdTomato mice were provided by Dr. Hongzhen Hu (Icahn School of Medicine at Mount Sinai, New York, USA). Scn10aCre: Jak1flox (Jak1Δneuron) mice were generated by crossing Scn10aCre mice with Jak1flox mice. Scn10aCre:Il4raflox (Il4raΔneuron) mice were generated by crossing Scn10aCre mice with Il4raflox mice. Scn10aCre:Rosa26STOPflox-tdTomato mice were generated by crossing Scn10aCre mice with Rosa26STOPflox-tdTomato mice. Scn10aCre:Jak1STOPflox-JAK1 GOF (JAK1GOF:neuron) mice were generated by crossing Scn10aCre mice with Jak1STOPflox-JAK1 GOF mice. All mice were housed in specific-pathogenfree (SPF) conditions and environmentally controlled animal facilities with a 12-hour light-dark cycle and were given unrestricted access to food and water. All animal protocols and experiments were approved by the Institutional Animal Care and Use Committee (IACUC) of ISMMS, Washington University School of Medicine in St. Louis, and the National Research Institute for Child Health and Development. All mice (8–13 weeks old, male and/or female) were randomly assigned to experimental groups in an age- and gender-matched fashion, and littermates were used whenever possible. All experiments were performed following strict IACUC guidelines.
METHOD DETAILS
Intranasal administration of Alternaria alternata extract, papain, neuropeptides, and JAK inhibitor and lung inflammation assessment
Allergic lung inflammation was induced by intranasal administration of Alternaria alternata extract (Greer Laboratories, Inc.; 2 μg/dose in 40 μl of sterile phosphate-buffered saline [PBS]) for four consecutive days (Day 0, 1, 2, and 3) or papain (MilliporeSigma, 20 μg/dose in 40 μl of sterile PBS) for three consecutive days (Day 0, 1, and 2). The effect of exogenous neuropeptides was examined following twice daily (Day −1, 0, 1, 2, and 3) intranasal administration of neuropeptide CGRPβ (Cayman Chemical; 1 μg/dose in 40 μl of sterile PBS) or substance P (Tocris, 50 μg/dose in 40 μl of sterile PBS) prior to Alternaria alternata administration. Likewise, the effect of JAK1 inhibition was examined following twice daily (Day 0, 1, 2, and 3) intranasal administration of the JAK1 inhibitor, upadacitinib (MedChemExpress; 2.5 mg/kg in 40 μl of sterile PBS), prior to Alternaria alternata administration. Mice were sacrificed on Day 4 (or on Day 3 for papain) for clinical, histologic, and immunologic analysis. Lungs, collected following PBS perfusion, were processed for immune cell analysis by flow cytometry (right lungs) or fixed in 4% paraformaldehyde for histological analysis (left lungs). Histology slides and digital images were created by HistoWiz Inc. (histowiz.com).
Upadacitinib administration
In the Alternaria alternata-induced allergic lung model, upadacitinib was administered intranasally (2.5 mg/kg in 40 μl of sterile PBS) or intraperitoneally (5 mg/kg in 200 μl of sterile PBS) twice daily for four consecutive days (Day 0, 1, 2, and 3).
Tamoxifen administration
Tamoxifen (75 mg/kg in 100 μl corn oil) was administered intraperitoneally for 5 consecutive days, and experiments were performed 7 days post last administration.
Construction of Adeno-Associated Virus (AAV) vectors
AAV2-retro-Cre and AAV2-retro-Cre/hJAK1[NM_002227.4]*(A634D) were constructed by VectorBuilder Inc. (https://en.vectorbuilder.com/). AAV vector transduction protocols followed methods previously described by Su et al. 23 with minor modifications. Briefly, AAV (2 × 103 genome copies/ml) was diluted 10-fold in PBS and was administered intranasally (40 ul/mouse) for four consecutive days, and experiments were performed three weeks after the last administration,.
Flow cytometry
Preparation of lung tissue for flow cytometry followed a slightly modified protocol of Moro et al.,68. Briefly, three lobes of the right lung were placed into a solution containing 5 ml Hank’s Balanced Salt Solution (HBSS), Liberase TM (Roche; 50 μg/ml final concentration), and DNase I (Roche; 1 μg/ml final concentration) in a gentleMACS™ C-tube (Miltenyi Biotec). Lung tissues were homogenized using protocol “37_m_LDK1” on the gentleMACS™ Dissociator with Heaters (Miltenyi Biotec), then passed through a 70-μm cell strainer (Fisher Scientific) into a 50-ml tube. Any remaining tissue was manually homogenized on the strainer with the end of a 3-ml syringe plunger. The gentleMACS™ C tube and the strainer were washed with 2% (vol/vol) FBS/PBS. The tissue homogenate was centrifuged at 400g for 5 minutes at 4°C, and after discarding the cleared supernatant, the cell pellets were incubated with 1 ml red blood cell lysing buffer (MilliporeSigma) at room temperature for 1 minute followed by a 2% (vol/vol) FBS/PBS wash. Simultaneously, cervical skin lymph nodes from the same mice were processed by homogenizing manually with the end of a 3-ml syringe plunger on a 100-μm cell strainer (Fisher Scientific) directly into a 50-ml tube. The strainer was washed with 2% (vol/vol) FBS/PBS and centrifuged at 400g for 5 minutes at 4°C.
Both the lung and lymph node cell samples were stained with the Zombie NIR viability dye (Biolegend; 1:500) to exclude dead cells, followed by Fc-receptor blocking and cell-surface staining with specific antibodies (key resources table). ILC2s were defined as live CD45+ CD90+ CD127+ Sca-1+ KLRG-1+ cells that were negative for lineage markers CD3, CD4, CD8, CD11b, CD11c, B220 (or CD19), MHC-II, Ly6G, and Siglec F within a gate for lymphocytes. Eosinophils were defined as live CD45+ Siglec F+ cells within a gate for granulocytes. CD4+ T cells were defined as live CD45+ CD3+ CD4+ cells negative for B220 (or CD19), within a gate for lymphocytes. The cells were analyzed using a Cytek® Aurora (CYTEK) flow cytometer, and the absolute number of cells in each sample was determined using CountBright™ Absolute Counting Beads (Invitrogen).
Bone marrow (BM) transplantation
Recipient mice were supplied with 200 ml of drinking water containing 5 ml of Sulfatrim antibiotic (Sulfamethoxazole/trimethoprim) added into 200 ml of drinking water for one week starting from the day prior to irradiation (Day −1). At the end of Week 1, recipient mice were irradiated (950 cGy) using the X-RAD 320 (Precision X-Ray). Bone marrow was harvested from the femurs and tibias of donor mice, treated with red blood cell lysis buffer (MilliporeSigma) for two minutes, then passed through a 70-μm cell strainer (Fisher Scientific) into a 15-ml conical tube. The cell strainer and cells were washed with 2% (vol/vol) FBS/PBS. ViaStain™ AOPI staining (Nexcelom Bioscience) was used to determine the number of live cells on a Cellometer Auto 2000 (Nexcelom Bioscience). Each recipient mouse received 1 × 107 live BM cells by intravenous (i.v.) infusion within 24 hours following irradiation. Mice were used in experiments four weeks following BM transplantation.
Targeted ablation of TRPV1-expressing peripheral neurons
Chemical ablation of TRPV1-expressing peripheral neurons was performed using Resiniferatoxin (RTX) according to previously published protocols.14,31 Systemic ablation of TRPV1-expressing neurons was achieved by daily subcutaneously (s.c.) injection of increasing doses of RTX (Alomone Labs; 30, 70, 100 μg/kg for three consecutive days) or vehicle (100 μl saline) in the flanks of C57BL/6 WT mice. Targeted ablation of TRPV1-expressing neurons in the DRG was achieved with daily intrathecal (i.t.; in the L5-L6 region) injection of RTX (25 ng/mouse for two consecutive days) or vehicle (10 μl saline). RTX-treated mice were allowed to rest for 4–6 weeks before use. For targeted ablation of vagal ganglia (VG), TRPV1-expressing neurons in the VG were ablated based on a previously described protocol.69 Mice were assessed for heat sensitivity with a hot plate test to confirm the functional loss of TRPV1-expressing DRG neurons.14,31
C57BL/6 WT mice were injected with RTX intra-ganglionically (i.g.) into the right VG with RTX (25 ng/mouse) or vehicle (0.5 μl saline) using the Remote Infuse/Withdraw Pump 11 Elite Nanomite Programmable Syringe Pump (Harvard Apparatus). RTX-treated mice were allowed to rest for 4–6 weeks before use, and only the right lobes of the lung were used for analysis.
Fluorescent protein imaging in cleared tissue
Tissue samples containing fluorescent tdTomato protein were cleared as previously described.70 Thoracic segments of mouse DRG and VG were harvested from experimental mice and fixed in 4% paraformaldehyde. Fixed tissues were washed with PBS, placed directly on slides, and covered with the refractive index matching solution, RIMS (40 g Histodenz [MilliporeSigma], 30 ml Phosphate buffer (0.02 M), 0.1% Tween-20 [MilliporeSigma], 0.01% sodium azide). All images were captured using an upright microscope equipped for epifluorescence microscopy (Nikon 80i; CoolSnapES camera [Nikon Instruments] or FVMPE-RS [Olympus]).
GWAS (genome-wide association studies)-based analysis
GWAS were performed using data obtained from the publicly available GWAS Catalog (https://www.ebi.ac.uk/gwas/genes/JAK1) database, which includes the common JAK1 variants and their associated phenotypes.
Predicted STAT6 transcriptional targets
Data, obtained from the MotifMap Predicted Transcription Factor Targets dataset (https://maayanlab.cloud/Harmonizome/gene_set/STAT6/MotifMap+Predicted+Transcription+Factor+Targets), were used for analysis. The linked dataset of STAT6 transcriptional targets was used to search genes of interest (CALCA, CALCB, TAC1, NMU, and VIP are putative targets).
Isolation, culture, and analysis of lung ILC2s
Lung dissociation was performed as previously described.68 Briefly, pulmonary PBS perfusion through the heart was performed immediately after euthanasia in order to remove circulating cells from the lung vasculature prior to harvesting the lungs. Lung pairs were minced into small pieces using scissors and were placed directly into a gentleMACS™ C-tube (Miltenyi Biotec) containing 5 mL of 1% BSA in HBSS. The lungs were dissociated using the gentleMACS™ Dissociator programmed to protocol “m_lung-01–02.” Following dissociation, the lungs were incubated with collagenase type 5 (Worthington; 200 U/ml final concentration), DNase I (Worthington; 100 U/ml final concentration), and hyaluronidase type 4 (MilliporeSigma; 15000 U/ml final concentration) in a 37 ºC water bath with 40 shakes/minute for 30 minutes. The lungs were then dissociated for a second time using the gentleMACS™ Dissociator with the protocol “m_lung-01–02”. Next, the lung dissociation mixture was filtered through a 70-μm cell strainer (Fisher Scientific) into a 50-ml conical tube, and any pieces remaining on the strainer were manually homogenized with the end of a 3-ml syringe plunger. The conical tubes containing the mixture were centrifuged at 490g for 5 minutes at 4 °C. The supernatant was discarded, and the cell pellet was resuspended in 21 ml of 2% (vol/vol) FBS/PBS and mixed with 9 ml of 100% Percol™ (GE Healthcare). Then, 5 ml of 70% Percol™ was gently added to the bottom of the tube, and the tubes were centrifuged at 490g for 30 minutes at room temperature. After separation, the intermediate layer was carefully moved into a new tube and washed twice with 2% (vol/vol) FBS/PBS. During every wash cycle, the cells suspended in 2% (vol/vol) FBS/PBS were filtered through a 70-μm cell strainer after resuspension. The samples were then incubated in 800 μL Fc block containing 2% (vol/vol) FBS/PBS for 15 minutes. Following incubations, the samples were washed and then incubated with a biotin-conjugated antibody cocktail including lineage markers for 30 minutes. The samples were then washed and incubated with MicroBeads (Miltenyi Biotec) for 20 minutes. Next, the samples were depleted of lineage-positive cells using an AutoMACS (Miltenyi Biotec), and the negative fraction was collected for fluorescence-activated cell sorting (FACS) using a FACSAria™ III Cell Sorter (BD Biosciences). Sorted lung-derived ILC2s were defined as live, lineage (Lin)-, CD45+, CD127+, Sca-1+, and KLRG1+.
Sort-purified lung ILC2s were placed into a 96-well round-bottom plate (300 cells/well, 200 μl/well) and cultured with recombinant mouse IL-2 (R&D, 10 ng/ml final concentration) and/or recombinant mouse IL-33 (R&D; 50 ng/ml final concentration), Substance P (R&D; 10 ng/ml final concentration), CGRPα (R&D; 10 ng/ml final concentration), and CGRPβ (Cayman Chemicals, 10 ng/ml final concentration) for 72 hours. The final supernatant was collected, and levels of IL-5 and IL-13 were measured by ELISA kits (Invitrogen).
RNA isolation from mouse DRG and JG/NG (VG) and qRT-PCR
Mouse DRG or nodose ganglia (NG)/jugular ganglia (JG) (combined VG) were harvested and homogenized with a bead homogenizer (BioSpec) in lysis buffer RA1 (Macherey-Nagel). Total RNA was extracted with the NucleoSpin RNA isolation kit following manufacturer’s protocol. Equal amounts of cDNA were synthesized from total RNA extracts using the iScript cDNA Synthesis kit. Gene expression levels were determined using gene-specific primers (key resources table) and 2X qPCR Universal Green MasterMix (Lamda Biotech) or Power SYBR™ Green PCR Master Mix (Thermo Fisher Scientific). Reactions were cycled using a StepOnePlus™ Real-Time PCR System (Thermo Fisher Scientific) or QuantStudio 6 Flex Real-Time PCR System (Thermo Fisher Scientific) using the manufacturer’s protocol. Gene expression was normalized to that measured for Gapdh, and relative expression was calculated using the ΔΔCt method.
RNA-seq alignment and quality control
Library preparation and sequencing steps of RNA-seq were performed by GENEWIZ from Azenta Life Sciences (genewiz.com). All samples from all experiments were processed using the same pipeline for compatibility. Quality control was performed using FastQC (v0.11.8). Trim Galore! (v0.6.6) was used to trim the adapter sequences with a quality threshold of 20. The GRCm38 genome assembly from GENCODE release M25 was used as a genome and transcriptome reference. The alignment was performed using STAR aligner (v2.7.5b) 71. Gene-level read counts were obtained using Salmon (v1.2.1) for all libraries.72 All samples passed the alignment quality control requirements, with >90% of reads uniquely mapping (>10M uniquely mapped reads for each library) using STAR aligner.
Differential mRNA gene expression and pathway analysis
Differential expression analysis was performed using gene-level read counts and the DESeq2 (v1.34.0) R package.73 Genes with <5 reads in total across all samples were considered inactive genes. A gene was considered differentially expressed if the adjusted p-value (Benjamini-Hochberg procedure) was <0.05 and the absolute log2(fold change) was >1. Overrepresentation analysis of differentially expressed genes was performed using the clusterProfiler R package (v4.2.2).74,75 The gene sets used for functional analysis were obtained from the Molecular Signatures Database (MSigDB).76–78 The overlaps between the differentially expressed genes and the MSigDB gene sets were tested for statistical significance using Fisher’s exact test followed by the multiple test correction using the Benjamini-Hochberg procedure (adjusted p-value <0.05).
Single-cell RNA-seq analysis of the JG/NG (VG)
Normalized scRNA-seq expression data for JG and NG neurons was obtained from a previously published study on vagal sensory neurons.28 Cells from these datasets that were annotated as non-neuronal cells were filtered out from downstream analysis. The threshold for determining if a cell is positive for a gene was calculated using the method described by Usoskin et. al.79 and was performed by calculating the average value of the top three cells with the highest expression for that gene and multiplying by 0.05. The numbers of positive cells for the genes Trpv1, Scn10a, Jak1, Calca, Calcb, Vip, Nmu, and Tac1 were calculated using R (v.4.1.0). The numbers of cells positive for the genes Scn10a, Jak1, Calca, Calcb, Vip, Nmu, and Tac1 were calculated from Trpv1+ cells. The number of cells positive for Jak1, Calca, Calcb, Vip, Nmu, and Tac1 were also calculated from Scn10a+ cells. For both analyses, a global threshold (using all neuronal cells) was calculated.
QUANTIFICATION AND STATISTICAL ANALYSIS
The data are shown as the mean ± standard deviation (SD), unless otherwise indicated. Data from independent experiments are representative of at least two independent replicates or as pooled data. None of the data were excluded from statistical analyses, unless due to technical errors. Statistical significance was determined using unpaired Student’s t-tests with Welch’s correction, unless otherwise noted. Statistical evaluations were performed using GraphPad Prism 8.0 software (GraphPad Software). Significance is regarded as: †p < 0.0001, ***p < 0.001, **p < 0.01, *p < 0.05, and n.s. as not significant.
Supplementary Material
Representative gating strategy is shown using the lung from a steady-state WT mouse. ILC2 – group 2 innate lymphoid cell, WT – wild-type
(A) Volcano plot analysis of RNA-seq data obtained from lung tissues of mice treated with subcutaneous (s.c.) RTX vs. vehicle control challenged with intranasal ALT, n = 2–3 per group. The analysis identified 148 down-regulated and 177 up-regulated genes.
(B) KEGG pathway overrepresentation analysis of RNA-seq data obtained from lung tissues of mice treated with s.c. RTX vs. vehicle control challenged with intranasal ALT. Overrepresentation analysis used Fisher’s exact test to determine the significance of enrichment. All terms shown were identified as the top four significantly activated pathways in terms of adjusted p-value (p-adjusted < 0.05, Benjamini-Hochberg procedure), n = 2–3 mice per group.
(C) Schematic of the allergic lung inflammation model generated by selective denervation of vagal afferents.
(D-E) Expression levels of Trpv1 in the VG and DRG from vehicle control- and intraganglionic (i.g.) RTX-treated mice. Data were obtained from two independent experiments, (D) n = 7–10 ganglia per group, (E) n = 4–5 mice per group.
(F) Schematic of the allergic lung inflammation model generated by selective denervation of spinal visceral afferents.
(G) Withdrawal latency from noxious heat (hot plate test). Mice treated with intrathecal (i.t.) vehicle control or RTX were placed on a hot plate set at 50 degrees Celsius, and withdrawal latency was measured as the time until the appearance of paw withdrawal behavior.
(H-I) Representative lung histology images of mice treated with i.t. vehicle control or RTX challenged with intranasal ALT, obtained by (H) H&E stain and (I) PAS stain.
(J-K) Flow cytometry of lung tissue from mice treated with i.t. vehicle control or RTX challenged with intranasal ALT. Shown are the frequencies of (J) ILC2s and (K) eosinophils. Data were obtained from three independent experiments, n = 5–6 mice per group.
(L) Volcano plot analysis of RNA-seq data obtained from lung tissues of mice treated with i.t. RTX vs. vehicle control challenged with intranasal ALT, n = 2–3 per group. The analysis identified 20 up-regulated genes and 20 down-regulated genes.
(M) KEGG pathway overrepresentation analysis of RNA-seq data obtained from the lung tissues of mice treated with i.t. RTX vs. vehicle control challenged with intranasal ALT. Overrepresentation analysis used Fisher’s exact test to determine the significance of enrichment, n = 2–3 mice per group.
***p < 0.001 and not significant (n.s.) (Unpaired t-test with Welch’s correction). Data are represented as mean ± SD. Scale bars indicate 200 μm.
(C, F) Figure adapted from an image created with BioRender.com.
ALT – Alternaria alternata, DEG – differential gene expression, DRG – dorsal root ganglia, H&E – hematoxylin and eosin, i.g. – intraganglionic, ILC2s – group 2 innate lymphoid cells, i.t. – intrathecal, PAS – periodic acid-Schiff, RTX – resiniferatoxin, s.c. – subcutaneous, VG – vagal ganglia,
A) Schematic of the allergic lung inflammation model using littermate control (Il4raflox) and experimental Il4raΔneuron mice.
(B-C) Representative lung histology images of control and Il4raΔneuron mice challenged with intranasal ALT, obtained by (B) H&E stain and (C) PAS stain.
(D-E) Flow cytometry of lung tissue from control and Il4raΔneuron mice challenged with intranasal ALT. Shown are the frequencies of (D) CD45+ immune cells, (E) ILC2s, and (F) eosinophils. Data were obtained from two independent experiments, n = 4 mice per group.
(G) Schematic of the allergic lung inflammation model using littermate control (Jak1flox) and experimental Jak1Δneuron mice.
(H-I) Representative lung histology images of control and Jak1Δneuron mice challenged with intranasal papain obtained by (H) H&E stain and (I) PAS stain.
(J-L) Flow cytometry of lung tissue from control and Jak1Δneuron mice challenged with intranasal papain. Shown are the frequencies of (J) CD45+ immune cells, (K) ILC2s, and (L) eosinophils. Data were obtained from two independent experiments, n = 3–4 mice per group.
*p < 0.05 and not significant (n.s.) (Unpaired t-test with Welch’s correction). Data are represented as mean ± SD. Scale bars indicate 200 μm. (A, G) Figure adapted from an image created with BioRender.com.
(A) Schematic of vehicle (Veh) or upadacitinib (Upa) treatment of ALT-induced allergic lung inflammation in WT mice. (B-C) Flow cytometry of lung tissue from vehicle- or upadacitinib-treated mice challenged with intranasal ALT. Shown are the frequencies of (B) ILC2s and (C) eosinophils. Data were obtained from two independent experiments, n = 5 mice per group.
(D-E) Representative images of lung histology following (D) H&E and (E) PAS staining.
***p < 0.001, *p < 0.05, and not significant (n.s.) (Unpaired t-test with Welch’s correction). Data are represented as mean ± SD. Scale bars indicate 200 μm. (A) Figure adapted from an image created with BioRender.com.
(A) Schematic of approach used to measure expression changes by quantitative PCR (qPCR) in control and Jak1Δneuron mice. (B-E) Expression levels of neuropeptide transcripts in the VG and DRG from control and Jak1Δneuron mice challenged with intranasal ALT. Shown are (B, C) Vip and (D, E) Nmu transcript levels. Data were obtained from two independent experiments, n = 3–4 mice per group. (F) Schematic of approach used to measure expression changes by qPCR in control WT and JAK1GOF mice. (G-H) Expression levels of neuropeptide transcripts in the VG and DRG from control WT and JAK1GOF mice challenged with intranasal ALT. Shown are (G) Calcb and (H) Tac1 transcript levels. Data were obtained from two independent experiments, n = 8–9 mice per group. (I) Schematic of i.g. RTX injection following tamoxifen treatment in Calcb-reporter mice. (J) Imaging of CGRPβ+ neurons in Calcb-reporter mice in the VG. Shown are representative images of VG from mice treated with i.g. vehicle control or RTX. (K) Numbers of CGRPβ+ neurons in the VG. Data were obtained from two independent experiments, n = 3–5 ganglia per group. *p < 0.05 and not significant (n.s.) (Unpaired t-test with Welch’s correction). Data are represented as mean ± SD. Scale bars indicate 200 μm.
(A, F, I) Figure adapted from an image created with BioRender.com.
GOF – gain-of-function, JAK1 – Janus kinase 1
(A) Schematic of the lung ILC2 suppression assay.
(B-C) Type 2 cytokine measurement from lung ILC2s stimulated with IL-33 and/or CGRPα. Shown are levels of (B) IL-5 and (C) IL-13.
(D) Schematic of induction of allergic lung inflammation and SP treatment in Rag1−/− mice. (E-F) Flow cytometry of lung tissue from in Rag1−/− mice challenged with intranasal ALT as well as vehicle control or SP. Shown are the frequencies of (E) ILC2s and (F) eosinophils. Data were obtained from two independent experiments, n = 5 mice per group. (G-H) Representative lung histology images from Rag1−/− mice challenged with intranasal ALT as well as vehicle control or SP obtained by (G) H&E and (H) PAS staining. (I) Schematic of induction of allergic lung inflammation and CGRPβ treatment in WT mice. (J-K) Flow cytometry of lung tissue from WT mice challenged with intranasal ALT as well as vehicle control or CGRPβ. Shown are the frequencies of (J) ILC2s and (K) eosinophils. Data were obtained from two independent experiments, n = 4–5 mice per group.
(L-M) Representative lung histology images of WT mice challenged with intranasal ALT as well as vehicle control or CGRPβ obtained by (L) H&E stain and (M) PAS stain.
***p < 0.001, *p < 0.05 (Unpaired t-test with Welch’s correction). Data are represented as mean ± SD. Scale bars indicate 200 μm. (A, D, I) Figure adapted from an image created with BioRender.com. SP – substance P
Video S1. Lung-innervating spinal afferents in the DRG, related to Figure 6
Video S2. Lung-innervating vagal afferents in the VG, related to Figure 6
Highlights.
A human germline JAK1 GOF variant promotes spontaneous skin inflammation in mice
Vagal sensory neuron expression of JAK1 GOF promotes immune homeostasis in the lung
A neuronal JAK1-CGRPβ axis suppresses ILC2 responses and allergic lung inflammation
ACKNOWLEDGEMENTS
We thank all members of the Kim lab for their helpful comments and discussion. We thank Drs. Ivan E. de Araujo and Wenfei Han from the Nash Family Department of Neuroscience at Icahn School of Medicine at Mount Sinai (ISMMS) for sharing their mouse vagal nerve isolation protocol. We thank Dr. Rohini Kuner from Heidelberg University for providing Scn10aCre mice and Dr. Frank Brombacher from International Center for Genetic Engineering and Biotechnology in Cape Town for providing Il4raflox mice. This work is supported by the Allen Discovery Center program, a Paul G. Allen Frontiers Group advised program of the Paul G. Allen Family Foundation, the Doris Duke Charitable Foundation, LEO Pharma, and the National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS) (AR070116, AR077007, AR080392) and the National Institute of Allergy and Infectious Disease (NIAID) (AI167933, AI167047) of the National Institutes of Health (NIH). H.H. is supported by the National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) (DK103901), NIAMS (AR077183), and the National Institute on Alcohol Abuse and Alcoholism (NIAA) (AA027065) of the NIH. R.B.C. is supported by the NIH (AT012041). A.M.V.H. is supported by NIAMS (AR080219). H.Y. is supported by a Crohn’s and Colitis Foundation Research Fellowship Award (award number 937437). A.M.T. is supported by the NIAID (AI007163, AI154912). H.M. is supported by a Grant-in-Aid for Scientific Research (B) (20H01622) as well as the National Center for Child Health and Development (#2022B-11). M.T. is supported by a Japanese Society of Allergology International Scholarship.
This work was supported in part through the Bioinformatics for Next Generation Sequencing (BiNGS) shared resource facility within the Tisch Cancer Institute at the ISMMS. The development of this shared resource is partially supported by the NCI P30 (CA196521) Cancer Center support grant, and the ISMMS Skin Biology and Disease Resource-based Center NIAMS P30 award (AR079200). This work was also supported by the computational resources and staff expertise provided by Scientific Computing at ISMMS; the Office of Research Infrastructure of the NIH (S10OD026880); and the ISMMS Genomics Technology Facility.
Footnotes
DECLARATION OF INTERESTS
B.S.K. is founder of Klirna Biotech; he has served as a consultant for 23andMe, ABRAX Japan, AbbVie, Almirall, Amgen, Arcutis Biotherapeutics, Arena Pharmaceuticals, argenx, AstraZeneca, Boehringer Ingelheim, Bristol-Myers Squibb, Cara Therapeutics, Clexio Biosciences, Eli Lilly and Company, Escient Pharmaceuticals, Evommune, Galderma, Genentech, GlaxoSmithKline, Granular Therapeutics, Incyte Corporation, Innovaderm Research, Janssen, Kiniksa, LEO Pharma, Maruho, Novartis, Pfizer, Recens Medical, Regeneron Pharmaceuticals, Sanofi, Septerna, Triveni Bio, Vial, WebMD; he has stock in ABRAX Japan, KliRNA Biotech, Locus Biosciences, and Recens Medical; he holds a patent for the use of JAK1 inhibitors for chronic pruritus; he has a patent pending for the use of JAK inhibitors for interstitial cystitis. D.A. has contributed to scientific advisory boards at Pfizer, Takeda, FARE, and the KRF. D.B. is the founder of Lab11 Therapeutics. All other authors declare that they have no relevant conflicts of interests.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
2. REFERENCES
- 1.Philips RL, Wang Y, Cheon H, Kanno Y, Gadina M, Sartorelli V, Horvath CM, Darnell JE, Stark GR, and O’Shea JJ (2022). The JAK-STAT pathway at 30: Much learned, much more to do. Cell 185, 3857–3876. 10.1016/j.cell.2022.09.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Bieber T, Simpson EL, Silverberg JI, Thaçi D, Paul C, Pink AE, Kataoka Y, Chu C-Y, DiBonaventura M, Rojo R, et al. (2021). Abrocitinib versus Placebo or Dupilumab for Atopic Dermatitis. New England Journal of Medicine 384, 1101–1112. 10.1056/NEJMoa2019380. [DOI] [PubMed] [Google Scholar]
- 3.Guttman-Yassky E, Teixeira HD, Simpson EL, Papp KA, Pangan AL, Blauvelt A, Thaçi D, Chu C-Y, Hong H.C. h., Katoh N, et al. (2021). Once-daily upadacitinib versus placebo in adolescents and adults with moderate-to-severe atopic dermatitis (Measure Up 1 and Measure Up 2): results from two replicate double-blind, randomised controlled phase 3 trials. The Lancet 397, 2151–2168. 10.1016/S0140-6736(21)00588-2. [DOI] [PubMed] [Google Scholar]
- 4.Reich K, Teixeira HD, de Bruin-Weller M, Bieber T, Soong W, Kabashima K, Werfel T, Zeng J, Huang X, Hu X, et al. (2021). Safety and efficacy of upadacitinib in combination with topical corticosteroids in adolescents and adults with moderate-to-severe atopic dermatitis (AD Up): results from a randomised, double-blind, placebo-controlled, phase 3 trial. The Lancet 397, 2169–2181. 10.1016/S0140-6736(21)00589-4. [DOI] [PubMed] [Google Scholar]
- 5.Reich K, Thyssen JP, Blauvelt A, Eyerich K, Soong W, Rice ZP, Hong HC-H, Katoh N, Valenzuela F, DiBonaventura M, et al. (2022). Efficacy and safety of abrocitinib versus dupilumab in adults with moderate-to-severe atopic dermatitis: a randomised, double-blind, multicentre phase 3 trial. Lancet 400, 273–282. 10.1016/S0140-6736(22)01199-0. [DOI] [PubMed] [Google Scholar]
- 6.Simpson EL, Sinclair R, Forman S, Wollenberg A, Aschoff R, Cork M, Bieber T, Thyssen JP, Yosipovitch G, Flohr C, et al. (2020). Efficacy and safety of abrocitinib in adults and adolescents with moderate-to-severe atopic dermatitis (JADE MONO-1): a multicentre, double-blind, randomised, placebo-controlled, phase 3 trial. The Lancet 396, 255–266. 10.1016/S0140-6736(20)30732-7. [DOI] [PubMed] [Google Scholar]
- 7.Ytterberg SR, Bhatt DL, Mikuls TR, Koch GG, Fleischmann R, Rivas JL, Germino R, Menon S, Sun Y, Wang C, et al. (2022). Cardiovascular and Cancer Risk with Tofacitinib in Rheumatoid Arthritis. New England Journal of Medicine 386, 316–326. 10.1056/NEJMoa2109927. [DOI] [PubMed] [Google Scholar]
- 8.Schwartz DM, Bonelli M, Gadina M, and O’Shea JJ (2016). Type I/II cytokines, JAKs, and new strategies for treating autoimmune diseases. Nature reviews. Rheumatology 12, 25–36. 10.1038/nrrheum.2015.167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Villarino AV, Kanno Y, and O’Shea JJ (2017). Mechanisms and consequences of Jak-STAT signaling in the immune system. Nature immunology 18, 374–384. 10.1038/ni.3691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Oetjen LK, Mack MR, Feng J, Whelan TM, Niu H, Guo CJ, Chen S, Trier AM, Xu AZ, Tripathi SV, et al. (2017). Sensory Neurons Co-opt Classical Immune Signaling Pathways to Mediate Chronic Itch. Cell 171, 217–228.e213. 10.1016/j.cell.2017.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Del Bel KL, Ragotte RJ, Saferali A, Lee S, Vercauteren SM, Mostafavi SA, Schreiber RA, Prendiville JS, Phang MS, Halparin J, et al. (2017). JAK1 gain-of-function causes an autosomal dominant immune dysregulatory and hypereosinophilic syndrome. The Journal of allergy and clinical immunology 139, 2016–2020.e2015. 10.1016/j.jaci.2016.12.957. [DOI] [PubMed] [Google Scholar]
- 12.Gruber CN, Calis JJA, Buta S, Evrony G, Martin JC, Uhl SA, Caron R, Jarchin L, Dunkin D, Phelps R, et al. (2020). Complex Autoinflammatory Syndrome Unveils Fundamental Principles of JAK1 Kinase Transcriptional and Biochemical Function. Immunity 53, 672–684.e611. 10.1016/j.immuni.2020.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Talbot S, Abdulnour REE, Burkett PR, Lee S, Cronin SJF, Pascal MA, Laedermann C, Foster SL, Tran JV, Lai N, et al. (2015). Silencing Nociceptor Neurons Reduces Allergic Airway Inflammation. Neuron 87, 341–354. 10.1016/j.neuron.2015.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Baral P, Umans BD, Li L, Wallrapp A, Bist M, Kirschbaum T, Wei Y, Zhou Y, Kuchroo VK, Burkett PR, et al. (2018). Nociceptor sensory neurons suppress neutrophil and γδ T cell responses in bacterial lung infections and lethal pneumonia. Nature Medicine 24, 417–426. 10.1038/nm.4501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Rochlitzer S, Veres TZ, Kühne K, Prenzler F, Pilzner C, Knothe S, Winkler C, Lauenstein HD, Willart M, Hammad H, et al. (2011). The neuropeptide calcitonin gene-related peptide affects allergic airway inflammation by modulating dendritic cell function. Clinical and experimental allergy : journal of the British Society for Allergy and Clinical Immunology 41, 1609–1621. 10.1111/j.1365-2222.2011.03822.x. [DOI] [PubMed] [Google Scholar]
- 16.Wallrapp A, Burkett PR, Riesenfeld SJ, Kim SJ, Christian E, Abdulnour REE, Thakore PI, Schnell A, Lambden C, Herbst RH, et al. (2019). Calcitonin Gene-Related Peptide Negatively Regulates Alarmin-Driven Type 2 Innate Lymphoid Cell Responses. Immunity 51, 709–723.e706. 10.1016/j.immuni.2019.09.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Dakhama A, Kanehiro A, Mäkelä MJ, Loader JE, Larsen GL, and Gelfand EW (2002). Regulation of airway hyperresponsiveness by calcitonin gene-related peptide in allergen sensitized and challenged mice. American journal of respiratory and critical care medicine 165, 1137–1144. 10.1164/ajrccm.165.8.2109058. [DOI] [PubMed] [Google Scholar]
- 18.Nagashima H, Mahlakõiv T, Shih HY, Davis FP, Meylan F, Huang Y, Harrison OJ, Yao C, Mikami Y, Urban JF, et al. (2019). Neuropeptide CGRP Limits Group 2 Innate Lymphoid Cell Responses and Constrains Type 2 Inflammation. Immunity 51, 682–695.e686. 10.1016/j.immuni.2019.06.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Bartemes KR, Iijima K, Kobayashi T, Kephart GM, McKenzie AN, and Kita H. (2012). IL-33–Responsive Lineage − CD25 + CD44 hi Lymphoid Cells Mediate Innate Type 2 Immunity and Allergic Inflammation in the Lungs. The Journal of Immunology 188, 1503–1513. 10.4049/jimmunol.1102832. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Doherty TA, Khorram N, Chang JE, Kim H-K, Rosenthal P, Croft M, and Broide DH (2012). STAT6 regulates natural helper cell proliferation during lung inflammation initiated by Alternaria. Am J Physiol Lung Cell Mol Physiol 303, 577–588. 10.1152/ajplung.00174.2012.-Asthma. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Orimo K, Tamari M, Takeda T, Kubo T, Rückert B, Motomura K, Sugiyama H, Yamada A, Saito K, Arae K, et al. (2022). Direct platelet adhesion potentiates group 2 innate lymphoid cell functions. Allergy: European Journal of Allergy and Clinical Immunology 77, 843–855. 10.1111/all.15057. [DOI] [PubMed] [Google Scholar]
- 22.Salo PM, Arbes SJ, Sever M, Jaramillo R, Cohn RD, London SJ, and Zeldin DC (2006). Exposure to Alternaria alternata in US homes is associated with asthma symptoms. Journal of Allergy and Clinical Immunology 118, 892–898. 10.1016/j.jaci.2006.07.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Su Y, Barr J, Jaquish A, Xu J, Verheyden JM, and Sun X. (2022). Identification of lung innervating sensory neurons and their target specificity. American Journal of Physiology - Lung Cellular and Molecular Physiology 322, L50–L63. 10.1152/AJPLUNG.00376.2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Akopian AN, Sivilotti L, and Wood JN (1996). A tetrodotoxin-resistant voltage-gated sodium channel expressed by sensory neurons. Nature 379, 257–262. 10.1038/379257a0. [DOI] [PubMed] [Google Scholar]
- 25.Akopian AN, Souslova V, England S, Okuse K, Ogata N, Ure J, Smith A, Kerr BJ, McMahon SB, Boyce S, et al. (1999). The tetrodotoxin-resistant sodium channel SNS has a specialized function in pain pathways. Nature neuroscience 2, 541–548. 10.1038/9195. [DOI] [PubMed] [Google Scholar]
- 26.Caterina MJ, Schumacher MA, Tominaga M, Rosen TA, Levine JD, and Julius D. (1997). The capsaicin receptor: a heat-activated ion channel in the pain pathway. Nature 389, 816–824. 10.1038/39807. [DOI] [PubMed] [Google Scholar]
- 27.Mazzone SB, and Undem BJ (2016). Vagal Afferent Innervation of the Airways in Health and Disease. Physiological Reviews 96, 975–1024. 10.1152/physrev.00039.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Kupari J, Häring M, Agirre E, Castelo-Branco G, and Ernfors P. (2019). An Atlas of Vagal Sensory Neurons and Their Molecular Specialization. Cell Reports 27, 2508–2523.e2504. 10.1016/j.celrep.2019.04.096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.De Vries DJ, and Blumberg PM (1989). Thermoregulatory effects of resiniferatoxin in the mouse: Comparison with capsaicin. Life Sciences 44, 711–715. 10.1016/0024-3205(89)90382-2. [DOI] [PubMed] [Google Scholar]
- 30.Szallasi A, and Blumberg PM (1989). Resiniferatoxin, a phorbol-related diterpene, acts as an ultrapotent analog of capsaicin, the irritant constituent in red pepper. Neuroscience 30, 515–520. 10.1016/0306-4522(89)90269-8. [DOI] [PubMed] [Google Scholar]
- 31.Lai NY, Musser MA, Pinho-Ribeiro FA, Baral P, Jacobson A, Ma P, Potts DE, Chen Z, Paik D, Soualhi S, et al. (2020). Gut-Innervating Nociceptor Neurons Regulate Peyer’s Patch Microfold Cells and SFB Levels to Mediate Salmonella Host Defense. Cell 180, 33–49.e22. 10.1016/j.cell.2019.11.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Chu C, Artis D, and Chiu IM (2020). Neuro-immune Interactions in the Tissues. Immunity 52, 464–474. 10.1016/j.immuni.2020.02.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Chu C, Parkhurst CN, Zhang W, Zhou L, Yano H, Arifuzzaman M, and Artis D. (2021). The ChAT-acetylcholine pathway promotes group 2 innate lymphoid cell responses and anti-helminth immunity. Science Immunology 6, 10.1126/sciimmunol.abe3218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Galle-Treger L, Suzuki Y, Patel N, Sankaranarayanan I, Aron JL, Maazi H, Chen L, and Akbari O. (2016). Nicotinic acetylcholine receptor agonist attenuates ILC2-dependent airway hyperreactivity. Nature Communications 7. 10.1038/ncomms13202. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Daniel C, Salvekar A, and Schindler U. (2000). A gain-function mutation in STAT6. Journal of Biological Chemistry 275, 14255–14259. 10.1074/jbc.C000129200. [DOI] [PubMed] [Google Scholar]
- 36.Sharma M, Lu HY, Vaseghi-Shanjani M, Del Bel KL, van der Lee R, Richmond PA, Lin S, Dalmann J, Lee JJ, Matthews A, et al. (2022). Human Germline Heterozygous Gain-of-Function STAT6 Variants Cause Severe Allergic Disease 2 Running Title: Human germline STAT6 gain-of-function variants. medRxiv. 10.1101/2022.04.25.22274265. [DOI] [Google Scholar]
- 37.Sharma M, Leung D, Momenilandi M, Jones LCW, Pacillo L, James AE, Murrell JR, Delafontaine S, Maimaris J, Vaseghi-Shanjani M, et al. (2023). Human germline heterozygous gain-of-function STAT6 variants cause severe allergic disease. The Journal of experimental medicine 220. 10.1084/jem.20221755. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Takeuchi I, Yanagi K, Takada S, Uchiyama T, Igarashi A, Motomura K, Hayashi Y, Nagano N, Matsuoka R, Sugiyama H, et al. (2023). STAT6 gain-of-function variant exacerbates multiple allergic symptoms. The Journal of allergy and clinical immunology 151, 1402–1409.e1406. 10.1016/j.jaci.2022.12.802. [DOI] [PubMed] [Google Scholar]
- 39.Suratannon N, Ittiwut C, Dik WA, Ittiwut R, Meesilpavikkai K, Israsena N, Ingrungruanglert P, Dalm VASH, van Daele PLA, Sanpavat A, et al. (2023). A germline STAT6 gain-of-function variant is associated with early-onset allergies. The Journal of allergy and clinical immunology 151, 565–571.e569. 10.1016/j.jaci.2022.09.028. [DOI] [PubMed] [Google Scholar]
- 40.Minskaia E, Maimaris J, Jenkins P, Albuquerque AS, Hong Y, Eleftheriou D, Gilmour KC, Grace R, Moreira F, Grimbacher B, et al. (2023). Autosomal Dominant STAT6 Gain of Function Causes Severe Atopy Associated with Lymphoma. Journal of clinical immunology. 10.1007/s10875-023-01530-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Halim TYF, Krauß RH, Sun AC, and Takei F. (2012). Lung Natural Helper Cells Are a Critical Source of Th2 Cell-Type Cytokines in Protease Allergen-Induced Airway Inflammation. Immunity 36, 451–463. 10.1016/j.immuni.2011.12.020. [DOI] [PubMed] [Google Scholar]
- 42.Akdis CA, Arkwright PD, Brüggen MC, Busse W, Gadina M, Guttman-Yassky E, Kabashima K, Mitamura Y, Vian L, Wu J, and Palomares O. (2020). Type 2 immunity in the skin and lungs. Allergy: European Journal of Allergy and Clinical Immunology 75, 1582–1605. 10.1111/all.14318. [DOI] [PubMed] [Google Scholar]
- 43.Guttman-Yassky E, Blauvelt A, Eichenfield LF, Paller AS, Armstrong AW, Drew J, Gopalan R, and Simpson EL (2020). Efficacy and Safety of Lebrikizumab, a High-Affinity Interleukin 13 Inhibitor, in Adults with Moderate to Severe Atopic Dermatitis: A Phase 2b Randomized Clinical Trial. JAMA Dermatology 156, 411–420. 10.1001/jamadermatol.2020.0079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Hanania NA, Korenblat P, Chapman KR, Bateman ED, Kopecky P, Paggiaro P, Yokoyama A, Olsson J, Gray S, Holweg CTJ, et al. (2016). Efficacy and safety of lebrikizumab in patients with uncontrolled asthma (LAVOLTA I and LAVOLTA II): replicate, phase 3, randomised, double-blind, placebo-controlled trials. The Lancet Respiratory Medicine 4, 781–796. 10.1016/S2213-2600(16)30265-X. [DOI] [PubMed] [Google Scholar]
- 45.Panettieri RA, Sjöbring U, Péterffy AM, Wessman P, Bowen K, Piper E, Colice G, and Brightling CE (2018). Tralokinumab for severe, uncontrolled asthma (STRATOS 1 and STRATOS 2): two randomised, double-blind, placebo-controlled, phase 3 clinical trials. The Lancet Respiratory Medicine 6, 511–525. 10.1016/S2213-2600(18)30184-X. [DOI] [PubMed] [Google Scholar]
- 46.Simpson EL, Flohr C, Eichenfield LF, Bieber T, Sofen H, Taïeb A, Owen R, Putnam W, Castro M, DeBusk K, et al. (2018). Efficacy and safety of lebrikizumab (an anti-IL-13 monoclonal antibody) in adults with moderate-to-severe atopic dermatitis inadequately controlled by topical corticosteroids: A randomized, placebo-controlled phase II trial (TREBLE). Journal of the American Academy of Dermatology 78, 863–871.e811. 10.1016/j.jaad.2018.01.017. [DOI] [PubMed] [Google Scholar]
- 47.Wollenberg A, Blauvelt A, Guttman-Yassky E, Worm M, Lynde C, Lacour JP, Spelman L, Katoh N, Saeki H, Poulin Y, et al. (2021). Tralokinumab for moderate-to-severe atopic dermatitis: results from two 52-week, randomized, double-blind, multicentre, placebo-controlled phase III trials (ECZTRA 1 and ECZTRA 2)*. British Journal of Dermatology 184, 437–449. 10.1111/bjd.19574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Bel EH, Wenzel SE, Thompson PJ, Prazma CM, Keene ON, Yancey SW, Ortega HG, and Pavord ID (2014). Oral Glucocorticoid-Sparing Effect of Mepolizumab in Eosinophilic Asthma. New England Journal of Medicine 371, 1189–1197. 10.1056/nejmoa1403291. [DOI] [PubMed] [Google Scholar]
- 49.Kang EG, Narayana PK, Pouliquen IJ, Lopez MC, Ferreira-Cornwell MC, and Getsy JA (2020). Efficacy and safety of mepolizumab administered subcutaneously for moderate to severe atopic dermatitis. Allergy: European Journal of Allergy and Clinical Immunology 75, 950–953. 10.1111/all.14050. [DOI] [PubMed] [Google Scholar]
- 50.Oldhoff JM, Darsow U, Werfel T, Katzer K, Wulf A, Laifaoui J, Hijnen DJ, Plötz S, Knol EF, Kapp A, et al. (2005). Anti-IL-5 recombinant humanized monoclonal antibody (Mepolizumab) for the treatment of atopic dermatitis. Allergy: European Journal of Allergy and Clinical Immunology 60, 693–696. 10.1111/j.1398-9995.2005.00791.x. [DOI] [PubMed] [Google Scholar]
- 51.Pavord ID, Korn S, Howarth P, Bleecker ER, Buhl R, Keene ON, Ortega H, and Chanez P. (2012). Mepolizumab for severe eosinophilic asthma (DREAM): A multicentre, double-blind, placebo-controlled trial. The Lancet 380, 651–659. 10.1016/S0140-6736(12)60988-X. [DOI] [PubMed] [Google Scholar]
- 52.Morris R, Kershaw NJ, and Babon JJ (2018). The molecular details of cytokine signaling via the JAK/STAT pathway. Protein Science 27, 1984–2009. 10.1002/pro.3519. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Kichaev G, Bhatia G, Loh P-R, Gazal S, Burch K, Freund MK, Schoech A, Pasaniuc B, and Price AL (2019). Leveraging Polygenic Functional Enrichment to Improve GWAS Power. American journal of human genetics 104, 65–75. 10.1016/j.ajhg.2018.11.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Serhan N, Basso L, Sibilano R, Petitfils C, Meixiong J, Bonnart C, Reber LL, Marichal T, Starkl P, Cenac N, et al. (2019). House dust mites activate nociceptormast cell clusters to drive type 2 skin inflammation. Nature immunology 20, 1435–1443. 10.1038/s41590-019-0493-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Perner C, Flayer CH, Zhu X, Aderhold PA, Dewan ZNA, Voisin T, Camire RB, Chow OA, Chiu IM, and Sokol CL (2020). Substance P Release by Sensory Neurons Triggers Dendritic Cell Migration and Initiates the Type-2 Immune Response to Allergens. Immunity 53, 1063–1077.e1067. 10.1016/j.immuni.2020.10.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Wallrapp A, Riesenfeld SJ, Burkett PR, Abdulnour REE, Nyman J, Dionne D, Hofree M, Cuoco MS, Rodman C, Farouq D, et al. (2017). The neuropeptide NMU amplifies ILC2-driven allergic lung inflammation. Nature 549, 351–356. 10.1038/nature24029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Nussbaum JC, Van Dyken SJ, von Moltke J, Cheng LE, Mohapatra A, Molofsky AB, Thornton EE, Krummel MF, Chawla A, Liang H-E, and Locksley RM (2013). Type 2 innate lymphoid cells control eosinophil homeostasis. Nature 502, 245–248. 10.1038/nature12526. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Wang N, Wang J, Zhang Y, Hu S, Zhang T, Wu Y, Sun X, Zhang T, Yang S, and He L. (2022). Substance P-induced lung inflammation in mice is mast cell dependent. Clinical and experimental allergy : journal of the British Society for Allergy and Clinical Immunology 52, 46–58. 10.1111/cea.13902. [DOI] [PubMed] [Google Scholar]
- 59.Huang S, Ziegler CGK, Austin J, Mannoun N, Vukovic M, Ordovas-Montanes J, Shalek AK, and von Andrian UH (2021). Lymph nodes are innervated by a unique population of sensory neurons with immunomodulatory potential. Cell 184, 441–459.e425. 10.1016/j.cell.2020.11.028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Rosas-Ballina M, Olofsson PS, Ochani M, Valdés-Ferrer SI, Levine YA, Reardon C, Tusche MW, Pavlov VA, Andersson U, Chavan S, et al. (2011). Acetylcholine-synthesizing T cells relay neural signals in a vagus nerve circuit. Science (New York, N.Y.) 334, 98–101. 10.1126/science.1209985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Balood M, Ahmadi M, Eichwald T, Ahmadi A, Majdoubi A, Roversi K, Roversi K, Lucido CT, Restaino AC, Huang S, et al. (2022). Nociceptor neurons affect cancer immunosurveillance. Nature 611, 405–412. 10.1038/s41586-022-05374-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Cohen JA, Edwards TN, Liu AW, Hirai T, Jones MR, Wu J, Li Y, Zhang S, Ho J, Davis BM, et al. (2019). Cutaneous TRPV1+ Neurons Trigger Protective Innate Type 17 Anticipatory Immunity. Cell 178, 919–932.e914. 10.1016/j.cell.2019.06.022. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Huh JR, and Veiga-Fernandes H. (2020). Neuroimmune circuits in inter-organ communication. Nature Reviews Immunology. Nature Research 20, 217–228. 10.1038/s41577-019-0247-z. [DOI] [PubMed] [Google Scholar]
- 64.Kashem SW, Riedl MS, Yao C, Honda CN, Vulchanova L, and Kaplan DH (2015). Nociceptive Sensory Fibers Drive Interleukin-23 Production from CD301b+ Dermal Dendritic Cells and Drive Protective Cutaneous Immunity. Immunity 43, 515–526. 10.1016/j.immuni.2015.08.016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Wang Y, Leung VH, Zhang Y, Nudell VS, Loud M, Servin-Vences MR, Yang D, Wang K, Moya-Garzon MD, Li VL, et al. (2022). The role of somatosensory innervation of adipose tissues. Nature 609, 569–574. 10.1038/s41586-022-05137-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Akdis CA, Akdis M, Bieber T, Bindslev-Jensen C, Boguniewicz M, Eigenmann P, Hamid Q, Kapp A, Leung DYM, Lipozencic J, et al. (2006). Diagnosis and treatment of atopic dermatitis in children and adults: European Academy of Allergology and Clinical Immunology/American Academy of Allergy, Asthma and Immunology/PRACTALL Consensus Report. The Journal of allergy and clinical immunology 118, 152–169. 10.1016/j.jaci.2006.03.045. [DOI] [PubMed] [Google Scholar]
- 67.Leshem YA, Hajar T, Hanifin JM, and Simpson EL (2015). What the Eczema Area and Severity Index score tells us about the severity of atopic dermatitis: an interpretability study. The British journal of dermatology 172, 1353–1357. 10.1111/bjd.13662. [DOI] [PubMed] [Google Scholar]
- 68.Moro K, Ealey KN, Kabata H, and Koyasu S. (2015). Isolation and analysis of group 2 innate lymphoid cells in mice. Nature Protocols 10, 792–806. 10.1038/nprot.2015.047. [DOI] [PubMed] [Google Scholar]
- 69.Han W, and de Araujo IE (2021). Dissection and surgical approaches to the mouse jugular-nodose ganglia. STAR protocols 2, 100474–100474. 10.1016/j.xpro.2021.100474. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Yang B, Treweek JB, Kulkarni RP, Deverman BE, Chen CK, Lubeck E, Shah S, Cai L, and Gradinaru V. (2014). Single-cell phenotyping within transparent intact tissue through whole-body clearing. Cell 158, 945–958. 10.1016/j.cell.2014.07.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Dobin A, Davis CA, Schlesinger F, Drenkow J, Zaleski C, Jha S, Batut P, Chaisson M, and Gingeras TR (2013). STAR: ultrafast universal RNA-seq aligner. Bioinformatics (Oxford, England) 29, 15–21. 10.1093/bioinformatics/bts635. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Patro R, Duggal G, Love MI, Irizarry RA, and Kingsford C. (2017). Salmon provides fast and bias-aware quantification of transcript expression. Nature methods 14, 417–419. 10.1038/nmeth.4197. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Love MI, Huber W, and Anders S. (2014). Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome biology 15, 550–550. 10.1186/s13059-014-0550-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Wu T, Hu E, Xu S, Chen M, Guo P, Dai Z, Feng T, Zhou L, Tang W, Zhan L, et al. (2021). clusterProfiler 4.0: A universal enrichment tool for interpreting omics data. Innovation (Cambridge (Mass.)) 2, 100141–100141. 10.1016/j.xinn.2021.100141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Yu G, Wang L-G, Han Y, and He Q-Y (2012). clusterProfiler: an R Package for Comparing Biological Themes Among Gene Clusters. OMICS: A Journal of Integrative Biology 16, 284–287. 10.1089/omi.2011.0118. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Liberzon A, Birger C, Thorvaldsdóttir H, Ghandi M, Mesirov JP, and Tamayo P. (2015). The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell systems 1, 417–425. 10.1016/j.cels.2015.12.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Liberzon A, Subramanian A, Pinchback R, Thorvaldsdóttir H, Tamayo P, and Mesirov JP (2011). Molecular signatures database (MSigDB) 3.0. Bioinformatics 27, 1739–1740. 10.1093/bioinformatics/btr260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, and Mesirov JP (2005). Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proceedings of the National Academy of Sciences of the United States of America 102, 15545–15550. 10.1073/pnas.0506580102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Usoskin D, Furlan A, Islam S, Abdo H, Lönnerberg P, Lou D, Hjerling-Leffler J, Haeggström J, Kharchenko O, Kharchenko PV, et al. (2015). Unbiased classification of sensory neuron types by large-scale single-cell RNA sequencing. Nature neuroscience 18, 145–153. 10.1038/nn.3881. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
Supplementary Materials
Representative gating strategy is shown using the lung from a steady-state WT mouse. ILC2 – group 2 innate lymphoid cell, WT – wild-type
(A) Volcano plot analysis of RNA-seq data obtained from lung tissues of mice treated with subcutaneous (s.c.) RTX vs. vehicle control challenged with intranasal ALT, n = 2–3 per group. The analysis identified 148 down-regulated and 177 up-regulated genes.
(B) KEGG pathway overrepresentation analysis of RNA-seq data obtained from lung tissues of mice treated with s.c. RTX vs. vehicle control challenged with intranasal ALT. Overrepresentation analysis used Fisher’s exact test to determine the significance of enrichment. All terms shown were identified as the top four significantly activated pathways in terms of adjusted p-value (p-adjusted < 0.05, Benjamini-Hochberg procedure), n = 2–3 mice per group.
(C) Schematic of the allergic lung inflammation model generated by selective denervation of vagal afferents.
(D-E) Expression levels of Trpv1 in the VG and DRG from vehicle control- and intraganglionic (i.g.) RTX-treated mice. Data were obtained from two independent experiments, (D) n = 7–10 ganglia per group, (E) n = 4–5 mice per group.
(F) Schematic of the allergic lung inflammation model generated by selective denervation of spinal visceral afferents.
(G) Withdrawal latency from noxious heat (hot plate test). Mice treated with intrathecal (i.t.) vehicle control or RTX were placed on a hot plate set at 50 degrees Celsius, and withdrawal latency was measured as the time until the appearance of paw withdrawal behavior.
(H-I) Representative lung histology images of mice treated with i.t. vehicle control or RTX challenged with intranasal ALT, obtained by (H) H&E stain and (I) PAS stain.
(J-K) Flow cytometry of lung tissue from mice treated with i.t. vehicle control or RTX challenged with intranasal ALT. Shown are the frequencies of (J) ILC2s and (K) eosinophils. Data were obtained from three independent experiments, n = 5–6 mice per group.
(L) Volcano plot analysis of RNA-seq data obtained from lung tissues of mice treated with i.t. RTX vs. vehicle control challenged with intranasal ALT, n = 2–3 per group. The analysis identified 20 up-regulated genes and 20 down-regulated genes.
(M) KEGG pathway overrepresentation analysis of RNA-seq data obtained from the lung tissues of mice treated with i.t. RTX vs. vehicle control challenged with intranasal ALT. Overrepresentation analysis used Fisher’s exact test to determine the significance of enrichment, n = 2–3 mice per group.
***p < 0.001 and not significant (n.s.) (Unpaired t-test with Welch’s correction). Data are represented as mean ± SD. Scale bars indicate 200 μm.
(C, F) Figure adapted from an image created with BioRender.com.
ALT – Alternaria alternata, DEG – differential gene expression, DRG – dorsal root ganglia, H&E – hematoxylin and eosin, i.g. – intraganglionic, ILC2s – group 2 innate lymphoid cells, i.t. – intrathecal, PAS – periodic acid-Schiff, RTX – resiniferatoxin, s.c. – subcutaneous, VG – vagal ganglia,
A) Schematic of the allergic lung inflammation model using littermate control (Il4raflox) and experimental Il4raΔneuron mice.
(B-C) Representative lung histology images of control and Il4raΔneuron mice challenged with intranasal ALT, obtained by (B) H&E stain and (C) PAS stain.
(D-E) Flow cytometry of lung tissue from control and Il4raΔneuron mice challenged with intranasal ALT. Shown are the frequencies of (D) CD45+ immune cells, (E) ILC2s, and (F) eosinophils. Data were obtained from two independent experiments, n = 4 mice per group.
(G) Schematic of the allergic lung inflammation model using littermate control (Jak1flox) and experimental Jak1Δneuron mice.
(H-I) Representative lung histology images of control and Jak1Δneuron mice challenged with intranasal papain obtained by (H) H&E stain and (I) PAS stain.
(J-L) Flow cytometry of lung tissue from control and Jak1Δneuron mice challenged with intranasal papain. Shown are the frequencies of (J) CD45+ immune cells, (K) ILC2s, and (L) eosinophils. Data were obtained from two independent experiments, n = 3–4 mice per group.
*p < 0.05 and not significant (n.s.) (Unpaired t-test with Welch’s correction). Data are represented as mean ± SD. Scale bars indicate 200 μm. (A, G) Figure adapted from an image created with BioRender.com.
(A) Schematic of vehicle (Veh) or upadacitinib (Upa) treatment of ALT-induced allergic lung inflammation in WT mice. (B-C) Flow cytometry of lung tissue from vehicle- or upadacitinib-treated mice challenged with intranasal ALT. Shown are the frequencies of (B) ILC2s and (C) eosinophils. Data were obtained from two independent experiments, n = 5 mice per group.
(D-E) Representative images of lung histology following (D) H&E and (E) PAS staining.
***p < 0.001, *p < 0.05, and not significant (n.s.) (Unpaired t-test with Welch’s correction). Data are represented as mean ± SD. Scale bars indicate 200 μm. (A) Figure adapted from an image created with BioRender.com.
(A) Schematic of approach used to measure expression changes by quantitative PCR (qPCR) in control and Jak1Δneuron mice. (B-E) Expression levels of neuropeptide transcripts in the VG and DRG from control and Jak1Δneuron mice challenged with intranasal ALT. Shown are (B, C) Vip and (D, E) Nmu transcript levels. Data were obtained from two independent experiments, n = 3–4 mice per group. (F) Schematic of approach used to measure expression changes by qPCR in control WT and JAK1GOF mice. (G-H) Expression levels of neuropeptide transcripts in the VG and DRG from control WT and JAK1GOF mice challenged with intranasal ALT. Shown are (G) Calcb and (H) Tac1 transcript levels. Data were obtained from two independent experiments, n = 8–9 mice per group. (I) Schematic of i.g. RTX injection following tamoxifen treatment in Calcb-reporter mice. (J) Imaging of CGRPβ+ neurons in Calcb-reporter mice in the VG. Shown are representative images of VG from mice treated with i.g. vehicle control or RTX. (K) Numbers of CGRPβ+ neurons in the VG. Data were obtained from two independent experiments, n = 3–5 ganglia per group. *p < 0.05 and not significant (n.s.) (Unpaired t-test with Welch’s correction). Data are represented as mean ± SD. Scale bars indicate 200 μm.
(A, F, I) Figure adapted from an image created with BioRender.com.
GOF – gain-of-function, JAK1 – Janus kinase 1
(A) Schematic of the lung ILC2 suppression assay.
(B-C) Type 2 cytokine measurement from lung ILC2s stimulated with IL-33 and/or CGRPα. Shown are levels of (B) IL-5 and (C) IL-13.
(D) Schematic of induction of allergic lung inflammation and SP treatment in Rag1−/− mice. (E-F) Flow cytometry of lung tissue from in Rag1−/− mice challenged with intranasal ALT as well as vehicle control or SP. Shown are the frequencies of (E) ILC2s and (F) eosinophils. Data were obtained from two independent experiments, n = 5 mice per group. (G-H) Representative lung histology images from Rag1−/− mice challenged with intranasal ALT as well as vehicle control or SP obtained by (G) H&E and (H) PAS staining. (I) Schematic of induction of allergic lung inflammation and CGRPβ treatment in WT mice. (J-K) Flow cytometry of lung tissue from WT mice challenged with intranasal ALT as well as vehicle control or CGRPβ. Shown are the frequencies of (J) ILC2s and (K) eosinophils. Data were obtained from two independent experiments, n = 4–5 mice per group.
(L-M) Representative lung histology images of WT mice challenged with intranasal ALT as well as vehicle control or CGRPβ obtained by (L) H&E stain and (M) PAS stain.
***p < 0.001, *p < 0.05 (Unpaired t-test with Welch’s correction). Data are represented as mean ± SD. Scale bars indicate 200 μm. (A, D, I) Figure adapted from an image created with BioRender.com. SP – substance P
Video S1. Lung-innervating spinal afferents in the DRG, related to Figure 6
Video S2. Lung-innervating vagal afferents in the VG, related to Figure 6
Data Availability Statement
No original codes were generated in this study.
Accession number is listed in the key resources table to account for all high throughput sequencing data.
KEY RESOURCES TABLE
REAGENT or RESOURCE | SOURCE | IDENTIFIER |
---|---|---|
Antibodies | ||
| ||
anti-mouse CD16/CD32 | Bio X Cell | Cat#: BE0307; RRID: AB_1107647 |
anti-mouse CD16/32 | Biolegend | Cat#: 101302; RRID: AB_312801 |
anti-mouse CD3 | BioLegend | Cat#: 100268; RRID: AB_2876392 |
anti-mouse CD3 | BioLegend | Cat#: 100247; RRID: AB_2572117 |
anti-mouse CD3e | BioLegend | Cat#: 100304; RRID: AB_312669 |
anti-mouse CD4 | BioLegend | Cat#: 100404; RRID: AB_312689 |
anti-mouse CD4 | BioLegend | Cat#: 100449; RRID: AB_2564587 |
anti-mouse CD8a | BioLegend | Cat#: 100704; RRID: AB_312743 |
anti-mouse CD8a | BioLegend | Cat#: 100782; RRID: AB_2819775 |
anti-mouse/human CD11b | BioLegend | Cat#: 101204; RRID: AB_312787 |
anti-mouse/human CD11b | BioLegend | Cat#: 101233; RRID: AB_10896949 |
anti-mouse CD11c | BioLegend | Cat#: 117304; RRID: AB_313773 |
anti-mouse CD11c | BioLegend | Cat#: 117326; RRID: AB_117326 |
anti-mouse CD11c | BioLegend | Cat#: 117339; RRID: AB_2562414 |
anti-mouse CD19 | Invitrogen | Cat#: 45-0193-82; RRID: AB_1106999 |
anti-mouse CD19 | Biolegend | Cat#: 115504; RRID: AB_313639 |
anti-mouse CD45 | Invitrogen | Cat#: 58-0451-82; RRID: AB_11218871 |
anti-mouse CD45 | Biolegend | Cat#: 103138; RRID: AB_2563061 |
anti-mouse CD45R/B220 | BioLegend | Cat#: 103275; RRID: AB_2860602 |
anti-mouse CD90.2 | BD Biosciences | Cat#: 740442; RRID: AB_2740169 |
anti-mouse CD117 (c-Kit) | BioLegend | Cat#: 105838; RRID: AB_2616739 |
anti-mouse CD117 (c-Kit) | Invitrogen | Cat#: 62-1171-82; RRID: AB_2637141 |
anti-mouse CD127 (IL-7Ra) | BioLegend | Cat#: 135013; RRID: AB_1937266 |
anti-mouse CD127 (IL-7Ra) | BioLegend | Cat#: 135035; RRID: AB_2564577 |
anti-mouse CD127 (IL-7Ra) | BioLegend | Cat#: 135037; RRID: AB_2565269 |
anti-mouse Siglec-F | BD Bioscience | Cat#: 562757; RRID: AB_2687994 |
anti-mouse Siglec-F | BioLegend | Cat#: 155506; RRID: AB_2750235 |
anti-mouse FceRIa | Biolegend | Cat#: 134304; RRID: AB_1626106 |
anti-mouse I-A/I-E | BioLegend | Cat#: 107622; RRID: AB_493727 |
anti-mouse/human KLRG1 (MAFA) | BioLegend | Cat#: 138418; RRID: AB_2563015 |
anti-mouse/human KLRG1 (MAFA) | BioLegend | Cat#: 138419; RRID: AB_2563357 |
anti-mouse Ly6A/E (Sca-1) | BD Biosciences | Cat#: 749199; RRID: AB_2873577 |
anti-mouse Ly6A/E (Sca-1) | Biolegend | Cat#: 108112; RRID: AB_313349 |
anti-mouse Ly-6G | Invitrogen | Cat#: 62-9668-82; RRID: AB_2762763 |
anti-mouse Ly-6G | BioLegend | Cat#: 127643; RRID: AB_2565971 |
anti-mouse Ly-6G/Ly-6C (Gr-1) | BioLegend | Cat#: 108404; RRID: AB_313369 |
| ||
Bacterial and virus strains | ||
| ||
AAV2-retro-Cre | VectorBuilder | Vector ID:VB220605-1075ztp |
AAV2-retro-Cre/JAK1 | VectorBuilder | Vector ID:VB220526-1322bxa |
| ||
Chemicals, peptides, and recombinant proteins | ||
| ||
Alternaria alternata | GREER | Cat#: NC1620293 |
β-CGRP (human) | Cayman Chemical | Cat#: 24725 |
β CGRP 8-37 human | MiliporeSigma | Cat#: SCP0060 |
Bovine Serum Albumin | MiliporeSigma | Cat#: A9418 |
Bovine Serum Albumin | MiliporeSigma | Cat#: A9576 |
CGRP (rat) | R&D | Cat#: 1161 |
Collagenase Type 5 | Worthington Biochemicals | Cat#: LS005282 |
DNase I | Roche | Cat#: 10104159001 |
DNase I | Worthington Biochemicals | Cat#: LS002139 |
Fetal Bovine Serum | MiliporeSigma | Cat#: F4135 |
Hialuronidase type 4 | MiliporeSigma | Cat#: H3384-500MG |
Histodenz | MiliporeSigma | Cat#: D2158 |
Liberase TM | Roche | Cat#: 5401127001 |
Papain from papaya latex | MiliporeSigma | Cat#: P4762-25MG |
Percoll™ | GE healthcare | Cat#: 17-0891-2 |
Quadrol | MiliporeSigma | Cat#: 122262 |
Resiniferatoxin | Alomone labs | Cat#: R-400 |
recombinant mouse IL-2 | R&D systems | Cat# 402-ML-020/CF |
recombinant mouse IL-33 | R&D systems | Cat#: 3626-ML-010/CF |
Substance P | Tocris | Cat#: 1156 |
Upadacitinib (ABT-494) | MedChemExpress | Cat#: HY-19569 |
| ||
Critical commercial assays | ||
| ||
Brilliant Violet 421 Streptavidin | Biolegend | Cat#: 405225 |
CountBrightTM Absolute Counting Beads, for flowcytometry | Invitrogen | Cat#: C36950 |
Fixation/Permeablization Kit | BD Biosciences | Cat#: 554714 |
Flexable Viability Dye eFluor™780 | Invitrogen | Cat#: 65-0865-18 |
IL-5 Mouse Uncoated ELISA Kit | Invitrogen | Cat#: 88-7054-88 |
IL-13 Mouse Uncoated ELISA Kit | Invitrogen | Cat#: 88-7137-22 |
Mouse CGRP2 ELISA Kit | MyBioSource | Cat#: MBS7241942 |
Power SYBR™ Green PCR Master Mix | Thermo Fisher Scientific | Cat#: 4367660 |
Streptavidin Microbeads | Miltenyi Biotec | Cat#: 130-048-101 |
UltraComp eBeads™ Compensation Beads | Invitrogen | Cat#: 01-2222-42 |
Viability Dye | Invitrogen | Cat#: 65-0865-14 |
Zombie NIR™ Fixable Viability Kit | BioLegend | Cat#: 423106 |
| ||
Deposited data | ||
| ||
Raw and analyzed bulk RNA-seq data | ISMMS BiNGS Core | GEO: GSE220497 |
| ||
Experimental models: Organisms/strains | ||
| ||
C57BL/6J | Jackson Laboratory | Stock No. 000664 |
C57BL/6J | Sankyo Labo Service Corpration, Inc. | N/A |
CalcaCre: Rosa26STOPflox-tdTomato | Hongzhen Hu | N/A |
CalcbCre/ERT: Rosa26STOPflox-tdTomato | Hongzhen Hu | N/A |
CalcbCre/ERT: Rosa26STOPflox-tdTomato | Brian Kim | N/A |
Il4ra flox | Frank Brombacher | N/A |
Jak1 flox | Nanjing Biomedical Research Institute | N/A |
Jak1 STOPflox-JAK1 GOF | Cyagen Biosciences Inc. | N/A |
JAK1 GOF | Cyagen Biosciences Inc. | N/A |
Scn10a Cre | Rohini Kuner | N/A |
Scn10aCre: Il4raflox | Brian Kim | N/A |
Scn10aCre: Jak1flox | Brian Kim | N/A |
Scn10aCre: Jak1STOPflox-JAK1 GOF | Brian Kim | N/A |
Scn10aCre: Rosa26STOPflox-tdTomato | Brian Kim | N/A |
Trpv1Cre: Rosa26STOPflox-tdTomato | Hongzhen Hu | N/A |
Rag1 −/− | Jackson Laboratory | Stock No. 002216 |
Rosa26 STOPflox-tdTomato | Jackson Laboratory | Stock No. 007909 |
| ||
Oligonucleotides | ||
| ||
Gapdh qRT-PCR primer 1 | integrated DNA Technologies | TCAAGAAGGTGGTGAAGCAGG |
Gapdh qRT-PCR primer 2 | integrated DNA Technologies | TATTATGGGGGTCTGGGATGG |
Calca qRT-PCR primer 1 | integrated DNA Technologies | GAGGGCTCTAGCTTGGACAG |
Calca qRT-PCR primer 2 | integrated DNA Technologies | AAGGTGTGAAACTTGTTGAGGT |
Calcb qRT-PCR primer 1 | integrated DNA Technologies | CTCTCAGCACGATATGGGTCC |
Calcb qRT-PCR primer 2 | integrated DNA Technologies | GCAAGAGATGTTTTTCCTGGTCG |
Nmu qRT-PCR primer 1 | integrated DNA Technologies | GAGGGAGCTTTGCCGTATAGT |
Nmu qRT-PCR primer 2 | integrated DNA Technologies | GATGCACAACAGAGGACACAA |
Tac1 qRT-PCR primer 1 | integrated DNA Technologies | TTTCTCGTTTCCACTCAACTGTT |
Tac1 qRT-PCR primer 2 | integrated DNA Technologies | GTCTTCGGGCGATTCTCTGC |
Trpv1 qRT-PCR primer 1 | Thermo Fisher Scientific | AGCTGCAGCGAGCCATCACCA |
Trpv1 qRT-PCR primer 2 | Thermo Fisher Scientific | ATCCTTGCCGTCCGGCGTGA |
Vip qRT-PCR primer 1 | integrated DNA Technologies | AGTGTGCTGTTCTCTCAGTCG |
Vip qRT-PCR primer 2 | integrated DNA Technologies | GCCATTTTCTGCTAAGGGATTCT |
| ||
Software and algorithms | ||
| ||
clusterProfiler R package (v4.2.2) | G Yu et. al | https://guangchuangyu.github.io/software/clusterProfiler/ |
DEGReport R package (v1.30.3) | L Pantano | https://github.com/lpantano/DEGreport |
DESeq2 (v1.34.0) R package | M Love et. al | https://github.com/mikelove/DESeq2 |
FastQC (v0.11.8) | Babraham Bioinformatics | https://www.bioinformatics.babraham.ac.uk/projects/fastqc/ |
FlowJo 10 | Tree Star | https://www.flowjo.com/ |
Galore! (v0.6.6) | Babraham Bioinformatics | https://www.bioinformatics.babraham.ac.uk/projects/trim_galore/ |
Glimma (v2.4.0) R packages | S Su et. al | https://github.com/Shians/Glimma |
NIS-Elements | Nikon Instruments | https://www.microscope.healthcare.nikon.com/products/software |
plotly R package (v4.9.4.1) | C Sievert et. al | https://plotly.com/r/ |
Prism 9 | GraphPad Software | https://www.graphpad.com/scientificsoftware/prism/ |
R (v.4.1.0) | R Core Team | https://www.r-project.org/ |
STAR aligner (v2.7.5b) | A Dobin et. al | https://github.com/alexdobin/STAR |
Salmon (v1.2.1) | R Patro et. al | https://combine-lab.github.io/salmon/ |
Ugent Center for Plant Systems Biology online tool | UCPSB: Bioinformatics & Evolutionary Genomics | https://bioinformatics.psb.ugent.be/webtools/Venn/ |
| ||
Other | ||
| ||
AutoMACS | Miltenyi Biotec | N/A |
Bead Homogenizer | BioSpec | Cat#: 112011 |
Cytek® Aurora | CYTEK | N/A |
FACSAria™ III Cell Sorter | BD | N/A |
FVMPE-RS | Olympus | N/A |
gentleMACS™ Octo Dissociator with Heaters | Miltenyi Biotec | Cat#: 130-096-427 |
Nikon 80i; CoolSnap ES camera | Nikon Instruments | N/A |
QuantStudio 6 Flex Real-Time PCR Systems | Thermo Fisher Scitntific | N/A |
Remote Infuse/Withdraw Pump 11 Elite Nanomite Programmable Syringe Pump | Harvard Apparatus | 70-4507 |
StepOnePlus qRT-PCR System | Thermo Fisher Scientific | N/A |