Abstract A reduced sense of smell is a common condition in people with cystic fibrosis (CF) that negatively affects their quality of life. While often attributed to nasal mucosa inflammation, the underlying causes of the olfactory loss remain unknown. Here, we characterized gene expression in olfactory epithelium cells from patients with CF using single-nuclei RNA sequencing and found altered expression of olfactory receptors (ORs) and genes related to progenitor cell proliferation. We confirmed these findings in newborn, inflammation-free samples of a CF animal model and further identified ultrastructural alterations in the olfactory epithelium and bulbs of these animals. We established that CFTR, the anion channel whose dysfunction causes CF, is dispensable for odor-evoked signaling in sensory neurons, yet CF animals displayed defective odor-guided behaviors consistent with the morphological and molecular alterations. Our study highlights CF’s major role in modulating epithelial structure and OR expression, shedding light on the mechanisms contributing to olfactory loss in CF. __________________________________________________________________ Cystic fibrosis alters olfactory structures and olfactory receptor expression, causing smell loss beyond chronic inflammation. INTRODUCTION Smell deficits, such as hyposmia or anosmia, are common symptoms in patients with cystic fibrosis (CF) ([52]1–[53]7), although the underlying causes of these deficits are not yet understood. CF-associated olfactory deficits mainly affect odor thresholds but not odor identification ([54]1, [55]7), suggesting a dysfunction of the olfactory periphery. Smell loss in CF is usually interpreted as a consequence of chronic rhinosinusitis (CRS), a frequent condition in CF. However, several pieces of evidence suggest a more direct role of the pathology in olfactory loss, independent of chronic inflammation: (i) Patients with CF exhibit more persistent olfactory deficits compared to non-CF patients with CRS ([56]1, [57]8), even after undergoing sinonasal surgery ([58]3, [59]7) or in the absence of nasal obstruction ([60]1). (ii) Olfactory impairment has also been reported in children with CF when the long-term effects of sinus disease and chronic inflammation are not yet pronounced ([61]2, [62]5). (iii) Treatment with elexacaftor-tezacaftor-ivacaftor (ETI; also known as Trikafta or Kaftrio), which improves a number of respiratory CF symptoms including CRS, does not ameliorate olfactory performance in patients with CF ([63]9–[64]11). (iv) Cystic fibrosis transmembrane conductance regulator (CFTR), the anion channel whose dysfunction causes CF, is present in the olfactory epithelium as both mRNA and protein ([65]12–[66]15). Considering the strong link between olfactory loss and reduced quality of life ([67]16), gaining a deeper understanding of the causes of olfactory impairment in CF and developing therapeutic strategies can significantly enhance the quality of life for individuals with CF. Volatile odorant molecules are detected by olfactory receptors (ORs) expressed by olfactory sensory neurons (OSNs) located in the olfactory mucosa, which then send odor information to the olfactory bulbs (OBs) ([68]17). Each OSN expresses one of hundreds of distinct ORs, and OSNs expressing the same OR converge on different sets of OB glomeruli that encode odor information ([69]18). Canonical OSNs use a cyclic adenosine 3′,5′-monophosphate (cAMP)–mediated cascade to transduce odor stimuli and a Ca^2+-activated chloride conductance for response amplification ([70]19, [71]20). Whether CFTR chloride conductance may play a role in olfactory signal transduction has not been fully determined. The olfactory epithelium also retains lifetime regenerative capacity mediated by stem and progenitor cells—horizontal and globose basal cells (HBCs and GBCs)—which reside in the basal layer of the epithelium and are able to replenish lost OSNs ([72]12, [73]21, [74]22). Conditions encountered in CF such as persistent inflammation, mechanical stress, or dehydration of airway surface mucus may induce deleterious effects on epithelial cell proliferation and regeneration. However, whether CFTR plays a more direct role in epithelial cell proliferation to alter tissue regeneration and olfactory function has not been yet investigated. Here, we characterized gene expression in olfactory epithelial cells collected from healthy and CF human subjects and found altered expression of genes encoding ORs, genes involved in cell cycle and tissue development in progenitor cells, as well as extensive CFTR expression in actively proliferating cells. We confirmed these results in newborn, inflammation-free samples of a CF animal model (CFTR^−/− pig) ([75]23) and further identified structural alterations in the olfactory epithelium and OBs in these animals. We identified a previously unrecognized role of CFTR in modulating epithelial proliferation and OR expression, thereby shedding light on the mechanisms through which CF causes olfactory loss independent of infection and inflammation. RESULTS Patients with CF show smell loss and altered gene expression in the olfactory neuroepithelium We performed an initial olfactory assessment on 10 CF individuals (age range 18 to 52 years; five female), eight of which had at least one copy of F508del mutation, and seven were under ETI treatment, which improves a number of CF symptoms. We assessed symptoms of CRS using the SNOT-22 sino-nasal test and their sense of smell by the Sniffin’ Sticks test battery ([76]Fig. 1, A and B), a standardized smell test to categorize odor threshold and identification ([77]24, [78]25). When compared to healthy control individuals, patients with CF showed poorer outcomes in SNOT-22, subjective smelling ability, threshold, and threshold/identification index, but not in odor identification ([79]Fig. 1B), confirming hyposmia in these individuals. Statistical comparison between ETI-treated versus nontreated groups revealed no significant differences (P values: 0.09 to 0.98), which is consistent with previous reports showing that ETI treatment did not seem to improve olfactory dysfunction ([80]9, [81]11). Fig. 1. CF individuals show smell loss and altered gene expression in olfactory cells. [82]Fig. 1. [83]Open in a new tab (A) Schematic representation illustrating olfactory testing with the Sniffin’ Sticks test and collection of olfactory epithelium cells by nasal brushing from CF and healthy donors. (B) Scores of the SNOT-22 test (left) and different parameters of the Sniffin’ Sticks test battery showing subjective smelling ability, threshold, identification scores, and the threshold/identification (TI) index (right). Blue-filled circles indicate ETI-treated individuals. *P < 0.05; ns, not significant (Student’s t test); n = 10 CF and 3 controls; n = 10 CF and 17 controls for SNOT-22 test. (C) UMAP dimensionality reduction plot of gene expression in 26,493 integrated CF and control cell nuclei (n = 7 CF and 9 control individuals). (D) UMAP depicting expression of CFTR (top) and violin plots of CFTR normalized expression in the different cell types (bottom). (E) Percentage of each cell type that express CFTR. (F) Volcano plot and heatmap of the 25 most significantly DE genes (up- and down-regulated) in the GBC cell cluster. CF samples correspond to patients not treated with ETI. Two of the control samples contained no GBCs and are not displayed in the analysis. (G) Average expression of the 16 most expressed ORs in the OSN cluster of the CF sample. pDCs, plasmacytoid dendritic cells. We next collected olfactory epithelium samples from the same patients via nasal brushing on the olfactory cleft under constant endoscopic control. Samples were snap-frozen, and later nuclei were extracted and analyzed by single-nuclei RNA sequencing (snRNA-seq) ([84]Fig. 1A). We generated an integrated single-cell sequencing dataset from a total of 16 individuals (9 healthy controls and 7 CF individuals), permitting robust cluster annotation from >26,000 cells ([85]Fig. 1C). Cell types annotation was performed using known markers from ([86]12) and the Human Lung Cell Atlas (fig. S1) ([87]26). Uniform manifold approximation projection (UMAP) plots captured the distribution of the different olfactory, respiratory and immune cells ([88]Fig. 1C). We then identified all cells expressing CFTR ([89]Fig. 1D), a previously reported marker of olfactory microvillar cells ([90]12), which share many features with ionocytes from the airways ([91]27–[92]29). As expected, CFTR was expressed in 95.5% of olfactory microvillar/ionocyte cells, whereas OSNs showed little or no expression of CFTR ([93]Fig. 1E). Unexpectedly, a substantial fraction of other olfactory and respiratory cell types also expressed CFTR, including sustentacular cells (SUS) (27%), olfactory HBCs (11.1%), GBCs (21.3%), and Bowman’s cells (23.5%) ([94]Fig. 1E). To identify CF-specific differences in gene expression, we performed a differential expression (DE) analysis between the CF and control datasets. We analyzed cells from CF subjects not treated with ETI to exclude any potential pharmacological interference with CFTR function. DE analysis identified marked transcriptional alterations between CF and control GBCs: 813 up-regulated and 197 down-regulated genes ([95]Fig. 1F). Many of the up-regulated genes were related to the immune response, such as cytokines and interferon-induced proteins, which is consistent with chronic inflammation and recurring viral infections common in patients with CF ([96]Fig. 1F). Among the most down-regulated genes, we identified genes implicated in differentiation, replication, and proliferation of undifferentiated stem cells—such as ZNF365, ADRA1B, RADX, NMBR, NSG1, and GPER1— suggesting a potential role of CF in modulating olfactory cell renewal. We further performed an analysis of the expression of ORs in CF samples. OR expression was not restricted to OSN, and it was observed in 18 different olfactory, respiratory, and immune cell types (fig. S1). Within the OSN cluster, we analyzed the expression of the 16 most expressed OR genes in the CF sample ([97]Fig. 1G). Only 5 of these 16 ORs (OR5A1, OR5A2, OR6C4, OR5AN1, and OR9K2) have been reported previously as among the most frequently expressed in healthy controls ([98]12), suggesting that CF individuals may express a distinctive set of ORs. Together, these findings show that CF individuals display olfactory loss and marked transcriptional alterations related to the immune response, stem cell homeostasis and OR expression, suggesting a potential more direct CF role in olfactory loss in addition to CRS-related hyposmia. CFTR is expressed in proliferating cells of the human olfactory neuroepithelium To study a potential role of CFTR in olfactory cell proliferation, we assessed the cell cycle pathway based on the expression of G[2]M and S cell cycle marker expression ([99]Fig. 2A). We found that the large majority of proliferative activity concentrates on GBCs ([100]Fig. 2, A and B). The analysis of CFTR expression on the GBC cluster revealed that CFTR is expressed in cells from G[1]-S and G[2]M phases ([101]Fig. 2C), being significantly more expressed in G[1]-S phase (P < 0.005, Mann-Whitney test; [102]Fig. 2D). Differential analysis of CF versus control groups revealed a significantly higher G[2]M score in the CF GBC cell cluster (P < 0.001, Mann-Whitney test; [103]Fig. 2D), suggesting an active role of CFTR in GBC proliferation. Fig. 2. CFTR is expressed in proliferating human olfactory cells. [104]Fig. 2. [105]Open in a new tab (A) UMAP projection of the computationally assigned cell cycle scores S (left) and G[2]M (right). (B) Phase scoring shows an elevated G[2]M score in the GBC cluster. (C) UMAP projection of the G[2]M, G[1], and S scores (left) and CFTR expression (right) in the GBC cell cluster. (D) CFTR is significantly more expressed in the G[1]-S phase in the GBC cluster, and G[2]M score is significantly higher in the CF group; **P < 0.005 and ***P < 0.001 (Mann-Whitney test); n = 41 G[1]-S and 26 G[2]M that express CFTR; n = 214 CF and 101 control. (E) Olfactory cell culture after nasal brushing from healthy donors showing CFTR (green) RNA coexpression with DCX, PCNA, and Ki67 antibodies (magenta) visualized by dual in situ hybridization and immunostaining. Scale bars, 20 μM. (F) Effect of the CFTR inhibitor CFTR[inh]172 on PCNA and Ki67 immunolabeling showing an increase on the percentage of positive cells. Gaussian probability density functions show a shift toward an increased PCNA and Ki67 fluorescence intensity in CFTR[inh]172-treated cells. Vertical dashed line indicates the background threshold. n = 1298 (mock), 890 (10 μM), 748 (20 μM), and 885 cells (30 μM); P < 0.001 (Kolmogorov-Smirnov test) in all comparisons. a.u., arbitrary unit; NK, natural killer; DAPI, 4′,6-diamidino-2-phenylindole. To further explore CFTR role and expression, we cultured human olfactory cells collected by nasal brushing from 10 healthy control donors ([106]Fig. 2E). We characterized cell composition by immunoreactivity for 10 markers specific for the different olfactory cell types. Labeling of olfactory marker protein (OMP), G-protein subunit Gαolf, microtubule-associated protein doublecortin (DCX), proliferating cell nuclear antigen (PCNA), Nestin, and SRY-box 2 (SOX2) ([107]Fig. 2E and fig. S2) is consistent with the presence of OSNs, GBCs, and HBCs at different stages of differentiation ([108]12, [109]30). Quantitative reverse transcription polymerase chain reaction (RT-PCR) revealed consistent CFTR and neuroblast marker expression (PAX6, DCX, and NEUROD1) among donors (fig. S2). In situ hybridization revealed CFTR expression in 47% of all cells (fig. S2), which combined with immunolabeling, showed CFTR colocalization with the proliferation markers DCX and PCNA and the proliferation marker protein Ki-67 (Ki67) ([110]Fig. 2E and fig. S2). To better understand a potential functional CFTR role on proliferation, we treated the human olfactory cell cultures with CFTR[inh]172, a selective CFTR blocker. We observed an increase on both the percentage of PCNA^+ and Ki67^+ cells and the labeling intensity ([111]Fig. 2F), suggesting that cells were arrested at a premitotic stage after treatment. Together, these results indicate that CFTR is expressed in undifferentiated, actively proliferating cells and that CFTR activity may have an impact on olfactory cell proliferation. A CFTR-null animal model shows deficits on olfactory-driven behavior We hypothesized that CF-associated olfactory loss may be consequence of a combination of indirect factors, such as CRS, and direct factors intrinsic to CFTR function. However, assessing olfactory function in CF out of the context of chronic infection and inflammation is challenging: patients with CF start to develop CRS during childhood becoming persistent during their whole lifetime, likely affecting the integrity of the olfactory tissue. In this context, CF animal models appear as a suitable alternative to study olfactory function. However, common model species such as rodents do not develop many of the human alterations in the case of CF, particularly in the respiratory tract ([112]31–[113]33). For these reasons, we decided to use a CFTR knockout pig model that better recapitulates the different traits of the human anatomy and CF pathology, including intestinal and lung pathologies similar to the alterations described in CF patients ([114]23). We performed all experiments with newborn piglets ([115]Fig. 3A), which are devoid of any inflammatory condition ([116]34–[117]36). To assess olfactory function in the newborns, we developed a robust behavioral assay by quantifying the ability of suckling in nonexperienced (newborn) piglets (movie S1). In most mammal species, including humans and pigs, the first suckling episode involves a strong maternally guided olfactory component ([118]37–[119]41). We thus quantified the latency to the initial suckling episode in 17 newborn CFTR homozygous mutants (CFTR^−/−) and 15 control (CFTR^+/+) littermates, which were macroscopically indistinguishable ([120]Fig. 3, A and E). No noticeable motor activity issues were observed, as initial mobility after birth was normal, with both genotypes showing similar times to stand (fig. S3). However, we observed that mutant piglets exhibited significant delays in initiating suckling, taking more time to locate the nipple and begin feeding ([121]Fig. 3, B to D), which suggests a deficiency consistent with reduced olfactory function. Fig. 3. Newborn CFTR^−/− piglets display deficits in an olfactory-driven behavior. [122]Fig. 3. [123]Open in a new tab (A) Litter containing newborn piglets of all three genotypes: CFTR^+/+, CFTR^+/−, and CFTR^−/−. (B) Latency-to-suckle in CFTR^−/− piglets (179.7 min ± 33.9 SEM; n = 17) and CFTR^+/+ (73.9 min ± 18.6 SEM; n = 15) (**P < 0.01, Mann-Whitney U test). (C) Percentage of pigs suckling and their latency to locate the nipple over the 400-min assay period (***P < 0.001, Kolmogorov-Smirnov test). (D) Data from (C) binned in 25-min windows [did not suckle (DNS)]. A total of 23.6% (4 of 17) of CFTR mutants fail to locate the nipple after 400 min. (E) Birth weights (P = 0.26, Mann-Whitney U test). (F) Immunolabeling for OMP in freshly dissociated pig olfactory epithelium cells (28.9% CFTR^+/+ and 26.9% CFTR^−/−; n = 1932 CFTR^+/+ cells and 1917 CFTR^−/− cells from three animals per genotype; P = 1, Mann-Whitney U test). Scale bar, 20 μM. (G) Example of Ca^2+ imaging showing an activated cell (left, arrow) and the corresponding time course (right) evoked by stimulation with 1-octanol (10 μM). Scale bar, 20 μM. (H) Mean Ca^2+ peak amplitudes (ΔR/R[0]) in CFTR^+/+ (gray) versus CFTR^−/− (blue) cells after stimulation. (I) Proportion of activated OSNs (F[1,14] = 1.64, P = 0.27, two-way ANOVA). (J) Peak amplitudes (F[1,558] = 0.13, P = 0.72, two-way ANOVA). n = 10 to 149 activated cells of a total of 18,781 cells analyzed (10479 CFTR^+/+ and 8302 CFTR^−/− cells from five animals/genotype). CFTR is not required for sensory function in OSNs Next, we investigated the basic physiological properties of OSNs, which detect small volatile odor molecules, in CF animals. We collected cells from the olfactory mucosa of CFTR^−/− animals by freshly dissociating the pig olfactory epithelium using an adapted protocol originally developed for mouse olfactory cells (see Materials and Methods). We confirmed the presence of mature OSNs (mOSNs) via OMP immunolabeling ([124]Fig. 3F). We then performed live-cell Ca^2+ imaging in 18,781 olfactory cells ([125]Fig. 3, G to J) testing three odorant molecules: 1-octanol, 2-heptanone, and octanal. Subsets of OSNs from CFTR^−/− and control animals responded reliably to each stimulus with similar response rates (0.59 to 0.85% versus 0.41 to 0.9%) and amplitudes ([126]Fig. 3, H to J), indicating intact sensory activity. Similarly, Ca^2+ responses to olfactory signaling pathways activators forskolin, 3-isobutyl-1-methylxanthine (IBMX), and 8-bromoguanosine 3′,5′-cyclic monophosphate (8-Br-cGMP) also remained unaffected ([127]Fig. 3, H to J, and fig. S3). Together, these data establish that CFTR^−/− OSNs can still detect small organic molecules and that CFTR is not required for olfactory transduction in OSNs. CFTR deletion induces transcriptional changes related to cell cycle and development To explore whether the core transcriptional signature of the olfactory mucosa is affected by CFTR deletion, we performed bulk RNA sequencing (RNA-seq) and compared gene expression in the whole olfactory mucosa between CFTR^−/− and CFTR^+/+ pig samples. Genes (211) displayed a significant DE at a false discovery rate of 5%; 81 of these were up-regulated in CFTR^−/− samples, and 130 were down-regulated ([128]Fig. 4A and fig. S4). On the basis of published RNA-seq data in mice ([129]30), we determined that the largest fraction (49%) of down-regulated genes were not neuron specific ([130]Fig. 4, B and C). Gene Ontology analysis on the DE genes revealed a significant enrichment of terms related to organic compound processes, nucleic acid processes, DNA replication, cellular aromatic processes, and nucleobase-containing processes ([131]Fig. 4D). Many of these DE genes have been reported to interact and form a regulatory network likely to underlie alterations in cell cycle and development (fig. S4). Gene annotation enrichment analysis highlighted cell cycle/DNA signaling pathways, nucleotide metabolism, and development among the top hits of broad transcriptional changes ([132]Fig. 4E and fig. S4). We did not detect DE molecules involved in inflammation or immune response signaling pathways. Most cell cycle/development–related genes (41 of 56) were down-regulated ([133]Fig. 4, F and G), suggesting altered cell proliferation. By contrast, the largest fraction (56%) of up-regulated genes was neuron specific ([134]Fig. 4C). The analysis of cell-specific marker genes revealed significant down-regulation of genes expressed by undifferentiated GBCs (PCNA, NES, KRT7, and PAX6), but not in genes specific for other cell types ([135]Fig. 4H). Marker genes for more differentiated GBCs such as ASCL1, NEUROG1, and NEUROD1 were not significantly DE ([136]Fig. 4H), suggesting that CFTR effect is specific to progenitor cells in an early stage of differentiation. We also observed altered expression of 16 OR genes, 13 being up-regulated and 3 down-regulated ([137]Fig. 4I). These results showing altered transcription of proliferation marker genes and ORs align with the expression changes observed in human CF samples. Fig. 4. Transcriptome analysis of the CFTR^−/− piglet olfactory epithelium shows alterations related to cell cycle and development. [138]Fig. 4. [139]Open in a new tab Differential gene expression of the whole olfactory mucosa transcriptome of CFTR^−/−compared to control CFTR^+/+ piglets. (A) Statistically significant DE genes are highlighted in blue. (B) DE genes classified as OSN- and non-OSN–specific according to previously published mouse RNA-seq datasets in Saraiva et al. ([140]30). A majority (48.5%, 63 genes) of the 130 downregulated (DR) genes are non-OSN. (C) Of the 81 up-regulated genes, 55.6% (45 genes) were specific to OSNs. (D) Pathway enrichment analysis of all genes that were significantly DE (P < 0.05) using Panther Classification System. (E) Gene Ontology analysis of all DE genes (P < 0.05) using UniProt database for classification. (F) DE genes related to cell cycle/DNA or development. A total of 41 of 58 (70.7%) were down-regulated. n = 4 per genotype. (G) Heatmap of cell cycle and development-related DE genes. (H) Heatmap of cell type–specific markers: GBCs, HBCs, mOSNs, iOSNs, microvillar cells (microvilli), sustentacular cells (sus) and Bowman’s gland cells (Bow). Significantly DE genes (in green) correspond to GBCs markers. (I) Heatmap and RNA-seq normalized counts from the 16 DE OR genes. DR, down-regulated; UR, up-regulated. Olfactory structures exhibit morphological defects in CF pigs To better understand the impact of CFTR deletion and subsequent transcriptional changes, we performed a detailed histological characterization of the newborn nasal neuroepithelium in the CFTR-knockout pig. The macroscopic nasal anatomy appeared normal (fig. S5), yet examination by microscopy revealed a marked reduction in the olfactory epithelium thickness (79.7 versus 131.9 μm, P < 0.01) ([141]Fig. 5, A and B). CF preparations presented significant global cell loss (135.3 versus 236.4 cells/100 μm, P < 0.01) and reduced cell density (15.9 versus 19 cells/1000 μm^2, P < 0.05) ([142]Fig. 5B). Remaining OSNs displayed otherwise intact cell morphology and presence of Gαolf in the cilia (fig. S5). However, layer-specific quantifications determined major cell loss in mature OMP^+ neurons (41.3 versus 85.3 cells/100 μm, P < 0.01) ([143]Fig. 5C). We also observed significant cell reduction in nerve growth factor receptor positive (NGFR^+) cells (8.4 versus 16.8 cells/100 μm, P < 0.05), immature OMP^−/NGFR^− neurons (46.3 versus 76.6 cells/100 μm, P < 0.05), and SUS cell layer (39.2 versus 57.7 cells/100 μm, P < 0.05) ([144]Fig. 5C). Fig. 5. Morphological alterations in the CFTR^−/− piglet olfactory epithelium. [145]Fig. 5. [146]Open in a new tab (A) Immunostaining for NGFR and OMP in olfactory epithelium sections. Epithelium thickness is delimited by the arrows. (B) Quantification of the olfactory epithelium (OE) thickness, cell number, and density. (C) Quantification of the cell number of each layer: OMP^+, NGFR^+, sustentacular cells (OMP^− cells above OMP^+ layer) and iOSNs (OMP^− NGFR^− cells between layers). (D) Representative images of OB glomeruli immunolabeled for OMP in CFTR^+/+ and CFTR^−/− piglets. (E) Quantification of the number and size of the OB glomeruli. *P < 0.05 and **P < 0.01 (Mann-Whitney U test); n = 6 animals per genotype. Scale bars, 50 μm. We then assessed the morphology of 819 OB glomeruli (469 CFTR^+/+ and 350 CFTR^−/−) in the brain, which receive axons from specific OSN subsets. CFTR mutants displayed significantly fewer (59 glomeruli/mm^2 versus 108.9 glomeruli/mm^2, P < 0.05) and larger (7892 μm^2 versus 4686.8 μm^2, P < 0.01) glomeruli ([147]Fig. 5, D and E), characteristic of a less developed olfactory system ([148]42). Thus, we also assessed the olfactory epithelium and bulb morphology in 75-day pig fetuses, over a total gestation period of 114 days. CFTR^−/− fetuses still displayed lower number of OMP^+ cells in the epithelium and larger, less abundant glomeruli (fig. S5), indicating that alterations observed at birth emerge earlier in development. We next investigated the subpopulations of actively proliferating basal cells in the newborn olfactory epithelium using KRT5, NGFR, or SOX2 antibodies to discriminate between HBCs and GBCs ([149]Fig. 6, A to C). We observed fewer KRT5^−/NGFR^+ and KRT5^−/SOX2^+ cells (1.8 versus 5.5 cells/100 μm, P < 0.01) ([150]Fig. 6C), consistent with a reduced number of GBCs. By contrast, KRT5^+/SOX2^+ cells remained unaltered (14.5 versus 14.2 cells/100 μm, P = 1) ([151]Fig. 6C), indicating that quiescent HBCs are less affected by CFTR deletion. Fig. 6. CFTR deficiency alters the GBC proliferative phenotype. [152]Fig. 6. [153]Open in a new tab Double immunostainings for KRT5/NGFR (A) and KRT5/SOX2 (B). Arrowheads indicate NGFR^+/KRT5^− and basal SOX2^+/KRT5^− labeling specific for GBCs. (C) Quantification of SOX2^+/KRT5^− (GBCs) and SOX2^+/KRT5^+ (HBCs) cells. **P < 0.01 (Mann-Whitney U test); ns, not significant. n = 7 CFTR^+/+ and 5 CFTR^−/− animals. (D) Dual fluorescent in situ hybridization with probes for CFTR and NGFR in control CFTR^+/+. CFTR labeling is not only robust in microvillar-like cells in the apical epithelium (white arrowheads, top) but is also detected in other cell types in the OSN cell layer (gray arrowhead, middle), basal cells (black arrowheads, bottom, colocalized with NGFR), and Bowman glands (right panels). (E) Fluorescent in situ hybridization for CFTR and immunostaining for SOX2 in the basal portion of the epithelium in CFTR^+/+ showing colabeling. (F) Dual fluorescent in situ hybridization with probes for CFTR and NGFR in CFTR^−/− showing no evident CFTR labeling. Bottom panels show magnified views of boxed area. Scale bars, 20 μm. CFTR is expressed by different cell types in the olfactory epithelium Our RNA-seq results in human samples revealed CFTR expression in different olfactory cell types ([154]Fig. 1E). We analyzed CFTR localization in epithelial tissue from CFTR^+/+ newborn piglets by in situ hybridization. CFTR probes showed robust labeling in scattered cells in the apical part of the epithelium, consistent with microvillar cell location and morphology ([155]Fig. 6D). Notably, we also observed abundant basal CFTR labeling in NGFR^+ ([156]Fig. 6D) and SOX2^+ cells ([157]Fig. 6E), consistent with expression in GBCs and HBCs. CFTR labeling was also present in the SUS and OSN cell layers, as well as in Bowman’s glands ([158]Fig. 6D). This broad expression pattern of CFTR in the olfactory mucosa of pigs is in line with our findings on human cells ([159]Fig. 1). CFTR labeling was absent in the epithelium of CFTR^−/− piglets, confirming the loss of CFTR mRNA expression ([160]Fig. 6F). Previous studies in mice have suggested a role of CFTR-expressing microvillar cells, which share many features with ionocytes from the airways ([161]27–[162]29), in modulating stem cell activity by the release of neuropeptide Y (NPY) ([163]43). However, in our samples, NPY was absent in microvillar cells and did not overlap with CFTR labeling (fig. S6), indicating distinct properties between species. Olfactory mucosa snRNA-seq reveals GBC and OR alterations To further investigate the presence of CFTR transcripts in neurogenic progenitors, we analyzed the pig olfactory mucosa by snRNA-seq on a total of 8923 nuclei, comprising 4085 CFTR^+/+ and 4838 CFTR^−/−. Data were displayed on UMAP plots to analyze cellular heterogeneity after cluster identification based on known murine and human marker genes ([164]12) ([165]Fig. 7A and fig. S7). CFTR^−/− and control piglets contained a similar cell type repertoire, yet CFTR^−/− samples displayed higher proportions of non-olfactory cells ([166]Fig. 7B). When focusing exclusively on olfactory cell populations (a total of 2863 CFTR^+/+ and 2697 CFTR^−/− nuclei), we observed a reduced proportion of iOSNs (−32.4%), GBCs (−27.5%), and HBCs (−11.8%) ([167]Fig. 7C). UMAP projection in control CFTR^+/+ cells confirmed CFTR expression across many cell types ([168]Fig. 7, D and E), consistent with our in situ hybridizations ([169]Fig. 6C) and human snRNA-seq data ([170]Fig. 1E). Similar to humans, the percentage of CFTR-expressing cells (95.6%) was highest in microvillar/ionocyte cells (fig. S7), but we also detected expression in Bowman’s cells (56.5%), SUS cells (17.6%), HBCs (15.3%), GBCs (7%), and some mature (4.8%) and immature (3.8%) OSNs (fig. S7). Fig. 7. snRNA-seq analysis of CFTR^−/− piglet olfactory mucosa. [171]Fig. 7. [172]Open in a new tab (A) UMAP dimensionality reduction plot of gene expression in 8923 integrated CFTR^+/+ and CFTR^−/− olfactory and respiratory mucosal cell nuclei (n = 73 to 1835 cells). (B) Cell proportions for every cell cluster. (C) Olfactory cell types, as a percentage of the total olfactory cells. (D) UMAP depicting expression of CFTR in the CFTR^+/+ cell sample. (E) UMAP projection of the computationally assigned cell cycle (G[2]M, G[1], S; left) and CFTR expression (right) in the GBC cell cluster from the CFTR^+/+. (F) Ratio of G[2]M versus G[1] + S cells in the CFTR^+/+ GBC cell sample. G[2]M cells are 3.8 times more abundant among the CFTR-expressing GBCs . (G) DotPlot visualization of the expression of 11 GBC marker genes. (H) Percentage of GBCs that express the genes in (G) that are in G[2]M phase. We further assessed the G[2]M cell cycle pathway score as a proxy for proliferation based on the expression of G[2]-M and G[1]-S phase cell cycle markers, confirming that the bulk of proliferative activity occurs in GBCs (fig. S7). In control CFTR^+/+ samples, GBCs in G[2]M phase were 3.4 times more abundant among the CFTR-expressing versus CFTR-nonexpressing population ([173]Fig. 7F and fig. S7), indicating that most (77%) CFTR-expressing GBCs are actively proliferating. We observed that CFTR expression was most prominent in the less differentiated GBC cluster comprising cells in G[2]-M phase, whereas more differentiated GBCs in G[1]-S phase in transition to immature neurons, expressed little or no CFTR ([174]Fig. 7E). We also observed a consistent reduction in gene expression among specific GBC marker genes ([175]Fig. 7G), but not in other cell type–specific markers (fig. S7). Furthermore, GBC cell subsets expressing specific markers were consistently less frequent in G[2]-M phase (29.6% average reduction) ([176]Fig. 7H), indicating less proliferation in the CFTR^−/− epithelium. We assessed the expression of 200 ORs across mature and immature OSNs ([177]Fig. 8). Two genes annotated in the pig database as ORs, ENSSSCG00000033601 and ENSSSCG00000013696, were the most frequently expressed ORs (detected in 1190 distinct OSNs). However, the expression pattern of these two ORs did not match typical OR expression as they were coexpressed with other ORs and were not specific to neurons. We therefore consider unlikely that these two genes encode for real ORs. After exclusion of these two genes from the analysis, our data indicated a lower global OR expression in the CFTR^−/− sample ([178]Fig. 8B). Of the 198 identified ORs, 151 were detected in CFTR^+/+ cells, while 93 were detected in CFTR^−/− cells. We compared the composition of the two OR populations ([179]Fig. 8C and fig. S8). Some OR genes such as OR4E1 were largely underrepresented in the CFTR^−/− samples (nine CFTR^+/+ versus zero CFTR^−/− cells), and others such as OR51E2 were overrepresented (zero CFTR^+/+ versus four CFTR^−/− cells) ([180]Fig. 8A). Subsequent analysis identified three OR populations in mOSNs: 53% of the detected ORs were specific to CFTR^+/+, 23.7% to CFTR^−/−, and 23.2% common to both ([181]Fig. 8C). We then analyzed the expression of OR51E2 alongside another related OR, OR51E1, which showed no expression differences, using in situ hybridization in the pig olfactory epithelium. Consistent with our RNA-seq data, the OR51E2 probe labeled more cells in CFTR^−/−, while OR51E1 labeling remained similar across both genotypes ([182]Fig. 8D). We also detected differences in OR expression in immature OSNs ([183]Fig. 8, A and B). At this stage, it may be difficult to conclude that CFTR deficiency directly affected OR gene choice on early differentiation stages. Overall, our findings provide strong support for a model in which CFTR modulates the proliferative activity, and its deficiency induces structural changes in the olfactory epithelium and OB, as well as OR repertoire expression influencing odor responses. Fig. 8. snRNA-seq analysis of CFTR^−/− piglet olfactory mucosa. [184]Fig. 8. [185]Open in a new tab (A) Specific expression of 33 ORs in individual iOSNs and mOSNs. (B) Relative OR expression in mature and immature OSNs. (C) Expression map identifying 198 ORs, specific to either CFTR^+/+ (red) or CFTR^−/− (blue) or coexpressed in both (gray). Each row represents an individual cell and each column is a single OR. Gene identities and cell numbers can be found in data S1. (D) Dual fluorescent in situ hybridization with probes for the OR genes OR51E2 [indicated in red in (A)] and OR51E1 in the olfactory epithelium of CFTR^+/+ and CFTR^−/− piglets. Right: Ratio of OR51E2/OR51E1 cells per section. *P < 0.05 (Mann-Whitney U test); n = 6 per genotype. Scale bars, 50 μm. DISCUSSION The molecular pathology of olfactory loss in CF is poorly understood. While airflow obstruction and tissue inflammation contribute to olfactory deficits in many cases, a reduced number of OSNs and OB glomeruli as well as differential OR expression may also play major roles. In the current study, our findings elucidate the capacity of CFTR to influence progenitor cell proliferation and OR gene expression. CFTR deficiency reduces neurogenesis in the olfactory epithelium and alters the expression of subsets of OR genes, leading to olfactory deficits. The contribution of CFTR to local proliferation and tissue development has been established in other peripheral tissues ([186]44–[187]48). Emerging evidence suggests that defects in CFTR function alter airways development and epithelia differentiation ([188]49). Congenital defects in the trachea and mainstem bronchi have been recognized in both neonatal CFTR null pigs and young CF children ([189]47). The olfactory epithelium maintains a stable population of mOSNs via continual neurogenesis ([190]21), normally sustained through differentiation of actively proliferating neurogenic progenitor cells ([191]22, [192]50). We found CFTR expression in actively proliferating cells, in both human and pig samples ([193]Figs. 1 and [194]2). We also observe in human and pig CF olfactory tissue a significant down-regulation of progenitor cell genes together with other markers involved in cell cycle and tissue development ([195]Figs. 1 and [196]4) as well as a reduction of epithelium thickness and abnormal OB glomeruli organization in CFTR^−/− pigs. Together, these observations are consistent with a model in which CFTR^−/− olfactory epithelium exhibits fewer proliferation, resulting in a thinner epithelium and fewer OB glomeruli. Our results highlight the importance of CFTR in epithelial development and may indicate potential strategies for therapeutic intervention to improve olfactory function in CF individuals. Our results may provide an explanation for some abnormalities affecting the olfactory system function in patients with CF ([197]1–[198]7). Altered olfactory epithelium structure, and hence olfactory function, can occur as a primary defect, independently of secondary consequences of the disease. By studying newborn animals, we excluded the possibility of secondary CF-related conditions such as chronic inflammation, bacterial infection, or persistent desiccation of the sensory surface layer. Our analysis of RNA-seq data showed no evidence of inflammation in newborn animals, indicating that the epithelium largely remains under uninjured, steady-state conditions. The reasons for poor olfactory function in both CF humans and pigs are likely related to the lower number of sensory neurons in the olfactory epithelium, which may result in fewer electrical signals in response to odorants. In addition, we also observed that human CF individuals express a different OR repertoire compared to controls [[199]Fig. 1 and ([200]12)], which may further alter olfactory performance. Similar results were observed in pig CFTR^−/− samples, indicating that altered OR expression is an intrinsic consequence of the pathology. Changes in OR expression are bidirectional, as a different set of ORs is up-regulated in the CFTR^−/−, providing support for improved olfactory perception of some odors. This would imply that patients with CF may show not only a reduced smell sensitivity but also differences in perceiving certain odors. Excessive representation of certain ORs in CF individuals may lead to a larger number of fibers from a specific OSN type reaching the OB, resulting in enlarged glomeruli, as seen in CF pigs. Whether these enlarged glomeruli arise from differences in specific OSN types or a more general delayed OB development remains unclear, but in both cases, they likely contribute to altered olfactory function. CF pigs better recapitulate human CF than the mouse, particularly in the upper airways ([201]31–[202]33, [203]36, [204]51), allowing the collection of newborn samples void of infection and inflammation. Using this model, we found that CFTR channel function is not necessary for odor transduction signaling in OSNs as they exhibit normal odor-induced calcium transients ([205]Fig. 3). Yet, CFTR^−/− piglets display a defective suckling behavior, which is compatible with reduced olfactory function. In most mammals, the initiation of the milk suckling behavior is critically dependent on olfactory signals ([206]37–[207]39). Pups from mice mutant for key olfactory transduction genes (Gnal, Adcy3, Cnga2, Scn9a) die at birth because of an inability to locate the mother’s nipple ([208]52–[209]55). In pigs, anesthesia delivery into the nose of newly born piglets or washing the mother’s nipples strongly impairs the ability to initiate sucking ([210]40, [211]41). Although other sensory modalities may also play a role, this evidence supports the involvement of olfaction in the control of initial sucking performance in pigs. We also validated previous observations showing high CFTR expression in olfactory microvillar cells ([212]12, [213]14, [214]43), a cell population that shares many traits with ionocytes ([215]27–[216]29). The function of CFTR in olfactory microvillar cells is not well understood, but, in the mouse, it has been suggested to play a role in maintaining tissue homeostasis by stimulating stem cell activity via release of NPY ([217]43). However, it is unlikely that they share the same function in pigs and humans because NPY is not expressed in microvillar cells nor coexpressed with CFTR in these species [fig. S6 and ([218]12)]. Moreover, NPY expression remains unchanged in the CFTR^−/−. Limitations of this study include the restriction of our biopsy samples to a limited number of human subjects resulting in low cell numbers, particularly OSNs, and the lack of access to intact (post-mortem) tissue for immunohistochemistry or RNA in situ hybridization. Despite the substantial similarities in RNA-seq data between human and pig CF samples, we cannot exclude the relevance of species-specific differences, especially given the much larger OR gene repertoire in pigs. In addition, our data from newborn animals do not address the role of CFTR in the adult olfactory system, and therefore, we are not able to exclude the possibility that CFTR contribution to the adult olfactory neurogenesis and OR expression differs to our observations in newborn individuals. In summary, our findings elucidate the capacity of CFTR to regulate progenitor cell proliferation and OR expression in the olfactory epithelium. The structural abnormalities in the sensory epithelium and OBs altered OR expression, and olfactory deficits reveal CF-related smell deficits as an intrinsic consequence of CFTR loss, independent from chronic inflammation of the upper airways. MATERIALS AND METHODS Experimental design and participants The objective of this study was to establish the causes of smell alterations observed in patients with CF. In a prospective study design, patients who presented themselves with CF were recruited from CF outpatient clinic consultations in the sense of regular disease-specific follow-up. Healthy participants were recruited for comparison. For the Sniffin’ Sticks test, a total of three healthy participants (aged 23 to 52 years, one woman) and 10 patients with CF (aged 18 to 52 years, five women) participated in the study from 4 to 25 July 2023. For the SNOT-22 test, 17 healthy controls (23 to 52 years old, 10 women) and the previous 10 patients with CF were used. For snRNA-seq, samples from seven CF and nine controls were sequenced. The information on the genetic form of the disease comes from the documentation of earlier DNA tests. For healthy participants, exclusion criteria were neurological diseases, systemic diseases associated with smell disorders such as chronic renal failure, subjective smell impairment, CRS, allergic rhinitis, alcohol or drug abuse, and pregnancy. The study was conducted according to the Declaration of Helsinki and had been approved by the Ethics Committee at the TU Dresden (EK 552122022). All participants gave written informed consent. Olfactory testing A detailed medical history was taken including age, gender, CF symptoms, medication, surgery, and questions regarding olfactory function. Further, the questionnaire sino-nasal outcome test (SNOT-22) was filled out to focus on lists of symptoms and social/emotional consequences of rhinosinusitis. Patients underwent olfactory tests using the Sniffin’ Sticks test battery (Burghart Messtechnik, Holm, Germany) to categorize the olfactory function regarding odor threshold and identification ([219]24, [220]25). A nasal endoscopy by an ear, nose, and throat specialist was performed to categorize endonasal diseases such as polyps or any form of CRS according to the Lildholdt score ([221]56). Nasal brushing was performed on the olfactory cleft on both sides under constant endoscopic control using a sterile cotton swab (CLASSIQSwabs, Brescia, Italy). The tips of the brushes were stored in CryoStor cell cryopreservation media (~1 to 2 ml) for 10 min at 4°C, then transferred to a cooling device (Thermo Fisher Scientific, Dreieich, Germany), and stored at −80°C until further processing. Patient records were assigned to codes and anonymized. Human snRNA-seq Sample preparation Cellular heterogeneity of human CF olfactory epithelium was analyzed by snRNA-seq. Frozen individual olfactory epithelium swabs were thawed and pooled for nuclei extraction in four pools containing three distinct samples from three individuals each (16 total individuals, 9 controls and 7 CF). This pooling ensured a sufficient amount of material for nuclei isolation. Genetic polymorphisms between samples were then used to discriminate individual samples (see below). Nuclei extraction was performed according to ([222]57), with some modifications. Briefly, each cryotube was thawed at 37°C supplemented with 400 μl of preheated saline medium. Brushes were cleared on ice by pipetting, transferred into 15-ml tubes, and centrifuged for 5 min at 290g at 4°C. After the removal of the supernatant, the cell pellet was resuspended into 1 ml of a buffer containing 25 mM citric acid and 0.25 M sucrose. The suspension was homogenized by 15 strokes with loose and then tight pestles, interrupted by 2× 3-min incubation on ice. After a filtration on a 20-μm filter (Miltenyi, Paris, France), nuclei were centrifuged for 5 min at 500g, resuspended in 500 μl of previous citric buffer, and centrifuged for 5 min at 500g. Nuclei were resuspended in a buffer containing 25 mM KCl, 3 mM MgCl[2], 5 mM tris-buffer (pH 8), RNAsin Plus ribonuclease inhibitor (0.4 U/μl), SUPERaseIn RNase inhibitor (0.4 U/μl), and 1 mM dithiothreitol (DTT). After a control of morphology on a Floid cell imaging station (PicoQuant, Berlin, Germany), nuclei were counted with a Countess II FL Automated Cell Counter (Thermo Fisher Scientific, Waltham, USA). Nuclei were then diluted at a concentration of 1200 nuclei/μl to target a capture of ~10,000 nuclei, and the suspension was rapidly loaded on a 10X Genomics Next GEM Single Cell 3′ v3.1 system (10X Genomics, Pleasanton, USA). Libraries were prepared according to the constructor specifications, with sequencing on a NextSeq 2000 (Illumina, San Diego, USA). snRNA-seq processing and analysis Human raw data were analyzed using Cell Ranger Single Cell software v6.0.0, with inclusion of the intron sequences. The raw gene-barcode matrices were aligned to the hg38 genome. Demultiplexing was performed using Demuxafy within a Singularity container to accurately assign reads to individual samples ([223]58). After demultiplexing, all sample data were processed using Seurat package v.4.0 ([224]59) in R v.4.0.2. Cells with less than 200 genes or more than 5% mitochondrial content, and genes expressed in fewer than three cells were excluded from the analysis. Each individual Seurat object was normalized, and highly variable features were identified using the variance-stabilizing transformation (vst) method, selecting the top 2000 variable features. Integration features were selected from the variable features identified in each object. Each Seurat object was then scaled and subjected to principal components analysis (PCA) using the selected integration features. Integration anchors were identified using reciprocal PCA (rPCA), and the data were integrated on the basis of these anchors. The integrated data were further processed by setting the default assay to “integrated,” scaling the data, running PCA, and performing UMAP dimensionality reduction on the first 30 principal components (PCs). A neighbor graph was then constructed, and clustering was performed at a resolution of 1.0. Differentially expressed genes were identified for each cluster using the FindMarkers function. Pseudobulk analysis was conducted using the edgeR-LRT from Libra R package ([225]60) to aggregate single-cell data into bulk-like samples for DE analysis between CF and control. Codes used for all analyses can be found at [226]https://github.com/ymbouamboua/Human_Pig_Olfactory_CF. Numbers for all cell types are: B cells (n = 106), Bowman’s gland (n = 84), club (n = 2600); deuterosomal cells (n = 405), GBCs (n = 315), Goblet cells (n = 1785), ionocytes/microvillar cells (n = 430), macrophages (n = 166), monocytes (n = 345), neutrophils (n = 32), natural killer cells (n = 308), olfactory HBCs (n = 4104), OSNs (n = 31), plasmacytoid dendritic cells (n = 192), respiratory HBCs (n = 183), respiratory multiciliated cells (n = 10298), sustentacular cells (n = 383), T cells (n = 1147), and Tuft cells (n = 18). Pig snRNA-seq Sample preparation Pig main olfactory epithelia were snap frozen in liquid nitrogen and included in OCT. Sections (300 μm) were made using a cryostat and kept at −80 °C. In a glass dounce tissue grinder previously cooled on ice, seven tissue slices and 1 ml of citric acid–based buffer (0.25 M sucrose and 25 mM citric acid) were added. Tissue samples were homogenized with three to five strokes of loose pestle and incubated on ice for 5 min. After five more strokes using a loose pestle, samples were incubated on ice for 5 min, homogenized again with three strokes using a loose pestle and five strokes using a tight pestle ([227]57). The suspension was filtered through a 40-μm cell strainer, and 1 ml of citric acid–based buffer was used to wash the containers. After a centrifugation for 5 min at 500g at 4°C, the supernatant was carefully removed and the sample was resuspended in 1 ml of wash buffer [10 mM tris-HCl (pH 7.4), 10 mM NaCl, 3 mM MgCl[2], 1% bovine serum albumin, 0.1% Tween 20, 1 mM DTT, RNaseIn (0.6 U/μl), and SuperaseIn (0.2 U/μl)]. Sample was filtered through a 5-μm cell strainer and centrifuged for 5 min at 500g at 4°C. Nuclei were resuspended in 50 to 100 μl of diluted nuclei buffer [Nuclei Buffer 1X; Multiome kit, 10X Genomics), 1 mM DTT, RNaseIn (0.6 U/μl), and SuperaseIn (0.2 U/μl)]. The nuclei morphology was verified using a Floid cell imaging station. Nuclei were counted on a Countess II FL Automated Cell Counter, diluted to the desired concentration (for a target capture of 10,000 nuclei). An RNA quality test was performed for each preparation before the 10x sequencing library was made. To do so, an aliquot of purified nuclei was placed in QIAzol Lysis Reagent, and RNA was isolated using miRNeasy Micro kit. The RNA integrity number was checked using the Agilent Bioanalyzer System. snRNA-seq was performed according to the protocol provided by 10X Genomics. snRNA-seq processing and analysis Pig raw data were analyzed using Cell Ranger Single Cell software v6.0.0. The raw gene-barcode matrices were aligned to the Sscrofa11-1 genome. Cell Ranger output was analyzed with the Seurat package v.4.0 ([228]59) using R v.4.0.2. All individual data were filtered to keep cells with 200 to 7000 genes, less than 99% of dropouts, and less than 5% of mitochondrial sequences. Doublet cells were removed with DoubletFinder ([229]61). After normalization and variance stabilization with SCTransfor and dimensionality reduction by PCA, data were visualized using UMAP embedding based on the first 20 PCs. Global integration was performed after normalization and identification of the 4000 most variable features of each dataset. Features that varied across datasets were selected, and experiments were integrated with the matching mutual nearest neighbors method as described ([230]62). Cell clusters were annotated according to canonical gene markers: Bowman’s gland (SOX9, SLC6A11, and TMEM163), endothelial (MMRN1, CLVS1, and CCL21), fibroblast/stromal (DCN, LUM, and PTPRD), GBCs (EZH2, CXCR4, and HES6), immature neurons (GNG8, GAP43, and TPD52), immune (CD163, ARHGAP15, and MRC1), mature neurons (GNG13, STXBP5L, and PEX5L), olfactory ensheathing glia (SORCS1 and ZNF536), olfactory HBCs (CCDC129, CAPN13, and MGAM2), olfactory microvillar (STAP1, CLNK, and SLC35F3), pericytes (NOSTRIN, LRFN5, and CXCL2), respiratory ciliated (CFAP126 and FOXJ1), respiratory epithelial (NDAL, GPS, and EIF1), respiratory HBCs (KRT5, MET, and TP63), respiratory secretory (TTC6 and EIF2AK2), sustentacular (MOCOS, GLDN, and ERMN), and vascular smooth muscle (TAGLN, TPM2, and MYL9). Bowman’s gland cells (n = 460), endothelial cells (n = 167), fibroblasts/stromal cells (n = 1475), GBCs (n = 313), immature neurons (n = 1241), immune cells (n = 144), mature neurons (n = 1835), olfactory ensheathing glia (n = 360), olfactory HBCs (n = 108), olfactory microvillar/ionocyte cells (n = 128), pericytes (n = 129), respiratory multiciliated cells (n = 314), respiratory epithelial cells (n = 73), respiratory HBCs (n = 222), respiratory secretory cells (n = 546), sustentacular cells (n = 1115), and vascular smooth muscle cells (n = 293). Bulk RNA-seq processing and analysis Olfactory epithelia from CFTR-null and control piglets were dissected and immediately frozen on liquid N[2]. RNA was extracted using the RNeasy Mini Kit (QIAGEN) with on-column deoxyribonuclease (DNase) digestion. mRNA was prepared for sequencing using the TruSeq RNA sample preparation kit (Illumina). All samples were multiplexed together and sequenced on four lanes on the Illumina HiSeq 2500 platform to generate 100–base pair paired-end reads. Sequencing reads were mapped using STAR 2.3 to the 11.1 Sus scrofa reference genome, annotation version 11.1 in the Ensembl pig genome database. The number of fragments aligned to each gene was counted using the HTSeq package, with the script htseq-count (mode intersection-nonempty). Any read that maps to multiple locations in the genome (also called multireads) was not counted toward the expression estimates as it cannot be assigned to any gene unambiguously. To compare the expression values across samples, raw count data were normalized to account for the depth of sequencing. Size factors were calculated using DESeq2’s function estimateSizeFactorsForMatrix, and raw counts were divided by the corresponding size factor for each sample. To test for DE, DESeq2 was used with standard parameters. Genes were considered to be differentially expressed if they had an adjusted P value of ≤0.05 (equivalent to a false discovery rate of 5%). A total of 11 DE genes located on the Y chromosome were excluded from the analysis to prevent sex-specific bias. To find terms that are enriched in our list of DE genes the over/under-representation algorithm from GeneTrail ([231]http://genetrail.bioinf.uni-sb.de/) was used. The background provided were all those genes tested for DE. To assess whether the DE genes form putative regulatory networks, STRING ([232]http://string-db.org/) was used with default settings for the 212 DE genes only. All normalized data and detailed results of the DE and enrichment analyses can be found in the data S1. Heatmaps of genes with significantly changed expression were generated using OriginPro 2021b program. Functional classification was analyzed by Gene Ontology Consortium (Panther Classification System) and UniProt database ([233]www.uniprot.org) using the biological process terms. Classification into OSN- or non-OSN–specific genes in [234]Fig. 4 (B and C) was performed by direct comparison with the dataset published by Saraiva et al. ([235]30) obtained by RNA-seq of mouse OMP^+ mOSNs. Cell-type assignments for specific marker genes were based on previously published mouse and human expression studies ([236]12, [237]30). Human olfactory neuroepithelium cell culture Human olfactory cell samples were obtained independently from snRNA-seq samples using a slightly different technique. Cells from the olfactory mucosa were collected by nasal brushings from 10 healthy control, nonsmoker, severe acute respiratory syndrome coronavirus 2–negative subjects between 18 and 45 years old (both males and females) as previously described ([238]63). Every subject gave written informed consent for the study and the procedures involved. The study was approved by the local institutional ethics committee CEIm Ethical Committee Parc de Salut MAR, IMIM-Hospital del Mar Research Institute, Barcelona (study no. 2018/7942/I). Samples from the middle and upper turbinates were maintained in 250 μl of cold Dulbecco’s modified Eagle’s medium/Ham F-12 (DMEM/F12) enriched with 10% fetal bovine serum (FBS), 2% glutamine, and 1% streptomycin-penicillin (GibcoBRL), as previously described ([239]63). At 80% confluence, cells were expanded using 0.25% trypsin (GibcoBRL) and replated in 75-cm^2 flasks. Cells were then expanded until a maximum of five passages. Cells from two healthy individuals were treated with 0, 10, 20, or 30 μM CFTR[inh]172 selective CFTR blocker (Tocris) on the culture media and incubated for 24 hours. Cells were washed with phosphate-buffered saline (PBS) fixed with 4% paraformaldehyde (PFA) for 15 min, and in situ hybridization and/or immunolabeling was performed as described below. Quantitative RT-PCR RNA from cultured human olfactory cells from 10 controls was purified with the PureLink RNA Mini Kit (Invitrogen) and stored at −20°C. Extracted total RNA was converted to cDNA by reverse transcription of 20 ng of RNA using the Nucleospin RNA XS kit (Macherey-Nagel, Düren, Germany) according to the manufacturer’s protocol. The cDNA was applied in TakyonTM No ROX SYBR 2X MasterMix blue dTTP (Eurogentec), and the primers used are listed in data S1. All RT-qPCR reactions were performed in 96-microwell plates using The LightCycler 480 Real-Time PCR System (Roche Applied Science). Three technical replicates were averaged, and the quantitative RT-PCR data were analyzed using the 2ΔΔCt method. GAPDH and RPL19 genes were used as a control for normalization. The results are expressed as relative fold change. CFTR-deficient pigs Male and female CFTR^+/− heterozygous transgenic pigs ([240]23) were provided by the LMU Munich, Germany, transferred to France. Pigs (large white breed) were housed together and had access to a standard grain-based diet and water ad libitum. Heterozygous pigs were mated to generate CFTR^+/+, CFTR^+/−, and CFTR^−/− piglets. Newborn piglets (0.8 to 1.5 kg) were genotyped within 6 hours after birth and euthanized for olfactory tissue collection. E75 pig fetuses were collected from amniotic sacs by cesarean section in pregnant dams. Pig littermates homozygous for CFTR (CFTR^+/+) served as controls. Littermates were randomly assigned to experimental groups keeping the same sex ratio between groups. All experiments were conducted in accordance with the guidelines of the Institutional Animal Care and Use Committee at the French National Research Institute for Agriculture, Food and Environment (INRAE). All experimental procedures were evaluated by the Ethics Committee of the Val de Loire (CEEA VdL, committee no.19) and approved by the Ministry of Higher Education and Research (APAFIS#1166-2015071615392426 Notification and APAFIS#10125-20170602162555 Notification). Neonate suckling assay Newborn piglets were tested for suckling latency right after birth. Delivery was induced after 112 days of gestation by injection of 175 μg of cloprostenol and 20 units (IU) of oxytocin, and each piglet was marked right after birth and monitored, and latencies to stand up after birth and to suckle were scored (see movie S1). Testing lasted until the piglet reached the mother’s nipple and started to suckle or for a maximum of 400 min after the birth of each piglet. Each test group contained piglets from one single litter. Each litter was tested only once to ensure no learning occurred. Time scoring was performed by an experimenter blind to the genotype of the piglets, as biopsies for genotyping were collected after behavior testing. Calcium imaging Ca^2+ imaging was performed in freshly dissociated pig olfactory epithelium cells adapting protocols previously developed for mouse olfactory cells ([241]64, [242]65). The olfactory neuroepithelium was detached from the cartilage and minced in PBS at 4°C. The tissue was incubated (20 min at 37°C) in PBS supplemented with papain (0.22 U/ml; Worthington), DNase I (10 U/ml; Fermentas), and urea (40 mM; Sigma-Aldrich) gently extruded in DMEM (Invitrogen) supplemented with 10% FBS and centrifuged at 100g (5 min). Dissociated cells were plated on coverslips previously coated with concanavalin-A type IV (0.5 mg/ml; overnight at 4°C; Sigma-Aldrich). Cells were used immediately for imaging after loading with Fura-2/AM (5 μM; Invitrogen) for 60 min. Coverslips containing cells were placed in a laminar-flow chamber (Warner Instruments) and constantly perfused at 22°C with extracellular solution Hank’s balanced salt solution (Invitrogen) supplemented with 10 mM Hepes. Cells were alternately illuminated at 340 and 380 nm, and the light emitted above 510 nm was recorded using a C10600-10B Hamamatsu camera installed on an Olympus IX71 microscope. Images were acquired at 0.25 Hz and analyzed using ImageJ [National Institutes of Health (NIH)], including background subtraction, region of interest (ROI) detection, and signal analyses. ROIs were selected manually and always included the whole-cell body. Peak signals were calculated from the temporal profiles of image ratio/fluorescent values. Results are based on recordings from five piglets for each condition and genotype (n = 10 to 149 activated cells of a total of 18,781 cells analyzed; 10,479 CFTR^+/+ and 8302 CFTR^−/− cells). Cells were stimulated successively and in random order using bath application. The following criteria for stimulus-induced Ca^2+ responses were applied: (i) A response was defined as a stimulus-dependent deviation of fluorescence ratio that exceeded twice the SD of the mean of the baseline fluorescence noise. (ii) Cells showing a response to control buffer were excluded from analysis. (iii) A response had to occur within 1 min after stimulus application. In time-series experiments, ligand application was repeated to confirm the repeatability of a given Ca^2+ response. Chemostimuli were freshly prepared each day and diluted in extracellular solution to give the following final concentrations: 10 μM 1-octanol (Sigma-Aldrich); 10 μM 2-heptanone (Sigma-Aldrich), 10 μM octanal (Sigma-Aldrich), 50 μM forskolin (Sigma-Aldrich), 100 μM IBMX (Sigma-Aldrich), 500 μM 8-Br-cGMP (Sigma-Aldrich), and 90 mM KCl. Volatile odorants were initially prepared in dimethyl sulfoxide (Sigma-Aldrich) and further diluted in extracellular solution. Immunostaining Human olfactory cells were fixed in PBS containing 4% PFA for 15 min at room temperature, washed 3X in PBS, and incubated in blocking solution (PBS solution containing 0.1% Triton X-100 and 3% horse serum) for 30 min at room temperature. For newborn pigs, olfactory tissues were removed, postfixed overnight in 0.1 M phosphate buffer (PB) containing 4% PFA, and later cryoprotected in 0.1 M PB buffer containing 30% sucrose. Samples were embedded in Tissue-Tek OCT compound, snap-frozen in cold isopentane, and processed on a Leica CM 3050S cryostat. Olfactory epithelia were cut in 16-μm-thick coronal sections and were directly mounted on SuperFrost Plus slides glasses (Thermo Fisher Scientific). Olfactory bulbs were cut in 30-μm serial free-floating sagittal sections in a PBS solution. Sections were treated with 10 mM sodium citrate for 5 min at 95° to 100°C for antigen retrieval, washed (3× 5 min) with PBS, incubated in blocking solution (PBS solution containing 0.1% Triton X-100 and 5% normal horse serum) for 2 hours at room temperature (RT), incubated overnight at 4°C in blocking solution supplemented with the primary antibody, washed in PBS solution, and incubated in blocking solution supplemented with secondary antibody for 1 hour at RT. Nuclei were counterstained with 4′,6-diamidino-2-phenylindole (DAPI; 0.5 μg/ml; Sigma-Aldrich) for 5 min. We used the following primary antibodies: goat anti-SOX2 (1:300; R&D Systems, AF2018), rabbit anti-KRT5 (1:800; BioLegend, 905501), goat anti-OMP (1:2000; Wako, 019-22291), mouse anti-NGFR (1:1000; Sigma-Aldrich, N5408), mouse anti-PCNA (1:1000; Sigma-Aldrich, P8825), rabbit anti-DCX (1:2000; Abcam, AB18723), rabbit anti-Ki67 (1:100; Abcam, ab9021), mouse anti-Gαolf (1:500; Santa Cruz Biotechnology, sc-55545), rabbit anti-ITPR3 (1:500; Millipore, AB9076), rabbit anti-TRPM5 (1:400; Alomone Labs, ACC-045), and rat anti-NES (1:600; Santa Cruz Biotechnology, sc-33677). Secondary antibodies used for the corresponding target species were conjugated with Alexa Fluor 488, Alexa Fluor 546, and Alexa Fluor 647 (1:500; Thermo Fisher Scientific). Epithelium thickness, cell density, areas, and OB glomeruli quantifications were preformed from images acquired on a Zeiss LSM-780 confocal laser-scanning microscope. Image regions were analyzed in the entire z axis with 3-μm step intervals, and images were reconstituted using the maximum intensity projection tool of Zen software. Images were analyzed with Fiji/ImageJ (NIH). Demarcations of epithelium limits and cell-specific layers were based on OMP and NGFR immunoreactivities, and DAPI^+ cells were counted using the particle analyzer plug-in of Fiji. Counts of the number of cells were evaluated blindly for each animal. Five slices per animal and genotype were used. Cells were counted from five to seven independent animals. For each sample, images were acquired at least from five different anatomical levels. Olfactory bulb glomeruli demarcation was based on OMP immunoreactivity in 5 to 17 slices per animal from six independent animals per genotype. A total of 819 glomeruli (469 CFTR^+/+ and 350 CFTR^−/−) covering all main OB topography (dorsal, ventral, lateral, and medial) were analyzed. In situ hybridization Staining for CFTR, NGFR, OR51E2, OR51E1, and NPY mRNAs was performed using multiplex fluorescent in situ hybridization (ISH). Human olfactory cells and olfactory tissue sections were prepared as described above. RNAscope Fluorescent MultiplexV2 labeling kit (ACDBio 323110) was used to perform the ISH assays according to the manufacturer’s recommendations. Probes used for staining are hs-CFTR (ACDBio 603291), ss-CFTR (ACDBio 541401-C2), ss-NGFR (ACDBio 828841), ss-LOC100625684 (ACDBio 1216061-C2), ss-LOC100737531 (ACDBio 1216070-C3), and ss-NPY (ACDBio 318751). Negative control slides were performed in parallel. After incubation with fluorescent-labeled probes, slides were counterstained with DAPI and mounted with antifade fluorescent mounting medium (Dako). Fluorescent images were captured using sequential laser scanning confocal microscopy (Zeiss LSM-780). CFTR^+ cells were counted from four independent human subjects. Combined ISH and immunohistochemistry After a standard ISH protocol, cells and tissue sections were directly incubated in blocking solution followed by primary and secondary antibodies for DCX, PCNA, Ki67, and SOX2 as described above. Quantification and statistical analysis Statistical analyses were performed using the statistical package R version 3.6.0 and the packages ggplot2, drc and ggpubr (R-studio Software, 4.1.1), and OriginPro 2021b (OriginLab Corporation, Northampton, MA, USA). Statistical details of experiments can be found in the figure legends, including statistical test used, the exact value of n, as well as dispersion and precision measures. Assumptions of normality and homogeneity of variance were tested before conducting the following statistical approaches. Mann-Whitney U test was used to measure the significance of the differences between two distributions. Multiple groups were compared using a two-way analysis of variance (ANOVA). Kolmogorov-Smirnov test was used to compare cumulative distributions. The probability of error level (alpha) was chosen to be 0.05. P values and the specific statistical test performed for each experiment are included in the appropriate figure legend or main text. Unless otherwise stated, results are presented as individual data points combined with boxplot indicating median lines and 25 to 75% ranges. Acknowledgments