Attenuation of antigen-induced airway hyperresponsiveness in CGRP-deficient mice

Tomoko Aoki-Nagase1, Takahide Nagase1, Yoshio Oh-Hashi2, Takayuki Shindo2, Yukiko Kurihara2, Yasuhiro Yamaguchi1, Hiroshi Yamamoto1, Tetsuji Tomita1, Eijiro Ohga1, Ryozo Nagai2, Hiroki Kurihara3, and Yasuyoshi Ouchi1

1 Departments of Geriatric Medicine and 2 Cardiovascular Medicine, Graduate School of Medicine, University of Tokyo, Tokyo 113-8655; and 3 Division of Integrative Cell Biology, Department of Embryogenesis, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 862-0976, Japan


    ABSTRACT
TOP
ABSTRACT
INTRODUCTION
METHODS
RESULTS
DISCUSSION
REFERENCES

Bronchial hyperresponsiveness and eosinophilia are major characteristics of asthma. Calcitonin gene-related peptide (CGRP) is a neuropeptide that has various biological actions. In the present study, we questioned whether CGRP might have pathophysiological roles in airway hyperresponsiveness and eosinophilia in asthma. To determine the exact roles of endogenous CGRP in vivo, we chose to study antigen-induced airway responses using CGRP gene-disrupted mice. After ovalbumin sensitization and antigen challenge, we assessed airway responsiveness and measured proinflammatory mediators. In the sensitized CGRP gene-disrupted mice, antigen-induced bronchial hyperresponsiveness was significantly attenuated compared with the sensitized wild-type mice. Antigen challenge induced eosinophil infiltration in bronchoalveolar lavage fluid, whereas no differences were observed between the wild-type and CGRP-mutant mice. Antigen-induced increases in cysteinyl leukotriene production in the lung were significantly reduced in the CGRP-disrupted mice. These findings suggest that CGRP could be involved in the antigen-induced airway hyperresponsiveness, but not eosinophil infiltration, in mice. The CGRP-mutant mice may provide appropriate models to study molecular mechanisms underlying CGRP-related diseases.

asthma; bronchial hyperreactivity; eosinophilia; leukotriene; knockout mouse


    INTRODUCTION
TOP
ABSTRACT
INTRODUCTION
METHODS
RESULTS
DISCUSSION
REFERENCES

BRONCHIAL HYPERRESPONSIVENESS and inflammation, including eosinophilia, are major characteristics of asthma (11, 12, 23). Recent studies have shown that various mediators, including cytokines, eicosanoids, and adhesion molecules, are involved in the development of asthma. Genetic features are also potentially associated with the etiology of asthma. On the basis of the inheritance pattern, a number of genes could have substantial roles in the pathogenesis of bronchial asthma (43). However, the exact molecular mechanisms of bronchial asthma remain to be elucidated.

Calcitonin gene-related peptide (CGRP), a 37-amino acid neuropeptide, has various biological actions, including responses to sensory stimuli, cardiovascular regulation, and vasodilation (2, 3, 19, 20). CGRP belongs to the calcitonin family of peptides, which includes calcitonin, amylin, and adrenomedullin. The calcitonin receptor-like receptor functions as a CGRP receptor in the presence of receptor activity-modifying protein 1 (RAMP1) (24). There are two CGRP isoforms: alpha -CGRP, which is present in the central and peripheral nervous system (42), and beta -CGRP, which is expressed in specific neuronal sites (1). It has been shown that CGRP, a potent vasodilator (22), modulates hypoxic pulmonary vasoconstriction (17). Recent studies using genetically engineered mice have shown that CGRP-knockout mice exhibit increased blood pressure and overactivation of the sympathetic nervous system (38).

In the respiratory system, CGRP is synthesized by sensory C-fibers throughout the respiratory tree (47). CGRP is also found in neuroepithelial cells of the lung and coexists with tachykinins in many airway sensory nerves (20), and CGRP receptors have been found to densely populate lung vessels (17). In terms of its physiological role, it has been reported that CGRP potently constricts airway smooth muscle in humans (39) and guinea pigs (41). In addition, it has been shown that CGRP has a significant role in eosinophilia in allergic inflammation (7, 37). On the basis of these observations, it is assumed that CGRP might be involved in the pathogenesis of bronchial asthma.

In the present study, we questioned whether alpha -CGRP might have pathophysiological roles in airway hyperresponsiveness and eosinophil infiltration, which are hallmarks of bronchial asthma. To determine the exact roles of alpha -CGRP in vivo, we chose to study the airway responsiveness and eosinophilia in alpha -CGRP gene-disrupted mice, which have been recently established (38). After sensitization and antigen challenge, we assessed airway responsiveness and measured proinflammatory mediators.


    METHODS
TOP
ABSTRACT
INTRODUCTION
METHODS
RESULTS
DISCUSSION
REFERENCES

Mice. alpha -CGRP-null mice were established as previously reported (38). Briefly, the mouse CT/alpha -CGRP genomic DNA was cloned from a BALB/c mouse genomic library in EMBL3 using synthetic oligonucleotide probes derived from the mouse CT/alpha -CGRP cDNA sequence. A 7.0-kb fragment containing exons 3-5 of the mouse CT/alpha -CGRP gene was subcloned into pBluescript (Stratagene). A targeting vector was constructed by replacing the 1.6-kb XbaI-XbaI fragment encompassing exon 5, which is specific for alpha -CGRP, with the neomycin resistance gene, and flanking the thymidine kinase gene. This plasmid was linearized with NotI and introduced into 129/Sv-derived SM-1 ES cells by electroporation; then the cells were selected in medium containing G418 and ganciclovir. Homologous recombinants were identified by PCR and Southern blot analysis. Targeted ES cell clones were injected into C57BL/6 mouse blastocysts to generate chimeric mice. Male chimeras were then cross bred with C57BL/6 females, and germline transmission was achieved. Mice heterozygous for alpha -CGRP-mutant allele with the genetic background of the 129/Sv × C57BL/6 hybrid were mated. Offspring were genotyped at 4 wk of age. For genotyping, genomic DNAs were isolated from biopsied tail and subjected to PCR amplification. The animals were maintained on a 7:20-h light-dark cycle at 23°C. Mice were fed a standard laboratory diet and water ad libitum. Mutant mice (alpha -CGRP-/-) and their littermate controls (alpha -CGRP+/+) were used in the present study.

Sensitization and antigen challenge. Mice were sensitized with an intraperitoneal injection of 0.5 ml of a solution containing 0.1 mg of ovalbumin (OA) complexed with aluminum hydroxide (2 mg/ml). On day 8, the mice were boosted with the same mixture. On day 12, these sensitized mice were challenged for 30 min with 1% OA in saline aerosol generated with an ultrasonic nebulizer (Ultra-Neb100, DeVilbiss, Somerset, PA). Control mice received an intraperitoneal injection of saline and saline aerosols in the same manner. Three days after the aerosol challenge, we measured bronchial responsiveness or performed bronchoalveolar lavage (BAL).

Animal preparation. Animals were anesthetized with pentobarbital sodium (25 mg/kg ip) and ketamine hydrochloride (25 mg/kg ip) in combination and then paralyzed with pancuronium bromide (0.3 mg/kg ip). Anesthesia and paralysis were maintained by supplemental administration of 10% of the initial dose every hour. After tracheostomy, a metal endotracheal tube (1 mm ID, 8 mm long) was inserted in the trachea. Animals were mechanically ventilated (model 683, Harvard Apparatus, South Natick, MA) with tidal volumes of 10 ml/kg and frequencies of 2.5 Hz. The thorax was widely opened by means of a midline sternotomy, and a positive end-expiratory pressure of 2 cmH2O was applied by placing the expired line under water. During the experiments, O2 gas was continuously supplied to the ventilatory system. Under these ventilatory conditions, arterial pH, PO2, and PCO2 were 7.35-7.45, 100-180 mmHg, and 30-45 mmHg, respectively, at the end of experiments (Compact 3 blood gas analyzer, AVL Medical Systems). A heating pad was used to maintain the body temperature of animals.

Tracheal pressure was measured with a piezoresistive microtransducer (model 8510B-2, Endevco, San Juan Capistrano, CA) placed in the lateral port of the tracheal cannula. Tracheal flow was measured by means of a Fleisch pneumotachograph (Metabo, Lausanne, Switzerland). All signals were amplified, filtered at a cutoff frequency of 100 Hz, and converted from analog to digital with a converter (model DT2801-A, Data Translation, Marlborough, MA). The signals were sampled at a rate of 200 Hz and stored on an IBM-AT-compatible computer. Lung resistance (RL) and elastance (EL) were measured as previously described (27, 31, 32).

Airway responsiveness to methacholine administration. At the start of the protocol, two deep inhalations (3 times tidal volume) were delivered to standardize volume history. All animals were then challenged with saline aerosol for 2 min. Aerosols were generated by an ultrasonic nebulizer and delivered through the inspiratory line into the trachea. Measurements of 10-s duration were sampled during tidal ventilation 1 min after administration of saline aerosol. This represented the baseline measurement. Then each dose of methacholine (MCh) aerosol was administered for 2 min in a dose-response manner (0.625-20 mg/ml). Airway responsiveness was assessed using the concentration of MCh required to increase RL to 200% of baseline values (30).

BAL fluid. BAL was performed [5 times with 1 ml of phosphate-buffered saline (PBS)] in each group. In each animal, 90% (4.5 ml) of the total injected volume was consistently recovered. After BAL fluid (BALF) was centrifuged at 450 g for 10 min, the total and differential cell counts of the BALF were determined from the cell fraction (29, 34, 35). The supernatant was stored at -70°C until assays were performed. The concentration of protein was measured by Lowry's method, with bovine serum albumin as a standard.

Assay of IgE in BALF. IgE levels in the BALF were determined using ELISA kits (Amersham Pharmacia Biotech, Piscataway, NJ). The detection limit of the ELISA assays for IgE was 10 ng/ml.

Assay of thromboxane and leukotriene in BALF. Thromboxane (Tx) A2 (TxA2, measured as TxB2) and leukotriene (LT) C4/D4/E4 were determined using enzyme immunoassay (EIA) kits (Amersham Pharmacia Biotech). The detection limits of the EIA assays for TxB2 and LTC4/D4/E4 were 3.6 and 10 pg/ml, respectively.

Assay of endothelin-1 in BALF. Endothelin-1 (ET-1) levels in the BALF were determined using EIA kits (IBL, Fujioka, Japan). The detection limit of the EIA assays for ET-1 was 0.78 pg/ml.

Assessment of CGRP immunoreactivity. In each group, the lungs of the mouse were removed intact and fixed with 10% formalin. After fixation, the tissue blocks obtained from midsagittal slices of the lungs were embedded in paraffin. Blocks were cut 4 µm thick by using a microtome. The preparations were processed for immunostaining by means of the avidin-biotin-peroxidase complex method. The slides were deparaffinized in a xylene bath and dehydrated in ethanol. Endogenous peroxidase activities were blocked by treatment with 0.6% H2O2 in 100% methanol for 30 min at room temperature. The primary antibody, rabbit anti-CGRP (rat) IgG (Peninsula Laboratories, San Carlos, CA), was diluted 1:200 in 10% FCS-PBS and added to the preparations overnight in a cold room. After six washes with PBS, the preparations were exposed to biotin-bound goat antiserum against rabbit IgG as the second antibody for 60 min at room temperature (Histofine, Nichirei, Tokyo, Japan). After the slides were washed with PBS, the tissues were incubated in horseradish peroxidase-bound streptavidin for 45 min (Histofine). The peroxidase reaction was performed with 3,3'-diaminobenzidine tetrahydrochloride as a chromogen (Vector Laboratories, Burlingame, CA).

Sections were screened and graded using the immunoreactivity score by two observers who were blind to the status of the specimen (5, 21). Visual assessments of the density of CGRP immunoreactivity were graded from none (score = 0) to abundant (score = 4). To examine the location of CGRP immunoreactivity, we analyzed large airways (airway diameter >0.2 mm), small airways (airway diameter <0.1 mm), and lung parenchyma. The scores represent the density of CGRP-immunoreactive cells and tissues that might include nerves, ganglia, and neuroepithelial bodies (NEBs) (5). In terms of the reproducibility of this assessment, inter- or intraobserver variances were not significant.

Materials and chemicals. Materials and chemicals were obtained from Sigma Chemical (St. Louis, MO) unless otherwise specified.

Data analysis. Comparisons of data among the experimental groups were carried out with ANOVA (Scheffé's test). Values are means ± SE. P < 0.05 was taken as significant.


    RESULTS
TOP
ABSTRACT
INTRODUCTION
METHODS
RESULTS
DISCUSSION
REFERENCES

Airway responsiveness to MCh administration. There were no significant differences in baseline RL and EL among each group. MCh dose-response curves for RL and EL are demonstrated in Figs. 1 and 2, respectively.


View larger version (18K):
[in this window]
[in a new window]
 
Fig. 1.   Methacholine (MCh) dose-response curves for lung resistance (RL) in wild-type (+/+) mice and mice deficient in the alpha -isoform (-/-) of calcitonin gene-related peptide (alpha -CGRP) (n = 7-9). # P < 0.05 compared with other groups. OA, ovalbumin.



View larger version (18K):
[in this window]
[in a new window]
 
Fig. 2.   MCh dose-response curves for lung elastance (EL) in wild-type and alpha -CGRP-deficient mice (n = 7-9). # P < 0.05 compared with other groups.

Airway responsiveness was assessed using the MCh concentration required to increase RL to 200% of baseline: 17.4 ± 1.8 and 17.6 ± 1.4 mg/ml for saline-treated alpha -CGRP+/+ and alpha -CGRP-/-, respectively, and 6.9 ± 1.3 and 16.5 ± 1.5 mg/ml for OA-treated alpha -CGRP+/+ and alpha -CGRP-/-, respectively (P < 0.05, OA-treated alpha -CGRP+/+ vs. other groups). Although bronchial hyperresponsiveness to MCh was observed in the OA-challenged wild-type mice, responses in the OA-challenged alpha -CGRP-/- mice were significantly reduced compared with the OA-challenged wild-type group.

Assessment of the BALF. Antigen exposure increased protein amount in BALF, although there was no difference between the wild-type and mutant mice (Fig. 3). Total cell counts and cell fractions in BALF are shown in Table 1, indicating the increases in the total cell number in the OA-sensitized groups. OA challenge induced eosinophil infiltration, whereas no differences in the fraction and number of BALF eosinophils were observed between the wild-type and alpha -CGRP-/- mice.


View larger version (9K):
[in this window]
[in a new window]
 
Fig. 3.   Roles of CGRP in antigen-induced protein leakage. Protein leakage is assessed by total protein amount in bronchoalveolar lavage (BAL) fluid in wild-type and alpha -CGRP-deficient mice (n = 4-6). # P < 0.05 compared with saline groups.


                              
View this table:
[in this window]
[in a new window]
 
Table 1.   Total cell counts and cell fractions in BALF

The IgE levels were significantly greater in OA- than in saline-treated mice. However, there were no significant differences between the wild-type and alpha -CGRP-/- mice (Fig. 4).


View larger version (8K):
[in this window]
[in a new window]
 
Fig. 4.   IgE levels in BAL fluid in wild-type and alpha -CGRP-deficient mice (n = 4-6 ). # P < 0.05 compared with saline groups.

Measurements of thromboxane, leukotriene, and ET-1 in BALF. There were no significant differences in BALF TxB2 among the groups: 0.140 ± 0.047 and 0.135 ± 0.076 ng in saline-treated alpha -CGRP+/+ and alpha -CGRP-/-, respectively, and 0.423 ± 0.204 and 0.498 ± 0.262 ng in OA-treated alpha -CGRP+/+ and alpha -CGRP-/-, respectively.

BALF LTC4/D4/E4 was significantly greater in OA-treated CGRP+/+ mice than in any other group (Fig. 5). Meanwhile, LTC4/D4/E4 was reduced to the same level in antigen-treated CGRP-/- mice and the saline-treated groups.


View larger version (8K):
[in this window]
[in a new window]
 
Fig. 5.   Leukotriene (LT) C4/D4/E4 levels in BAL fluid in wild-type and alpha -CGRP-deficient mice (n = 4-6 ). * P < 0.05 compared with sensitized wild-type mice. # P < 0.05 compared with saline groups.

There were no differences in BALF ET-1 among the groups: 1.35 ± 0.32 and 1.01 ± 0.11 pg in saline-treated alpha -CGRP+/+ and alpha -CGRP-/-, respectively, and 2.03 ± 0.38 and 2.70 ± 0.60 pg in OA-treated alpha -CGRP+/+ and alpha -CGRP-/-, respectively.

Assessment of CGRP immunoreactivity. Figure 6 demonstrates the immunohistochemistry of CGRP in large airways. In the OA-sensitized wild-type mouse, significant immunoreactivity for CGRP was observed in the airway epithelium and submucosa, while the immunostaining was modest in the saline-treated wild-type animal. On the other hand, there was little CGRP immunoreactivity in saline- or OA-treated alpha -CGRP-/- mice. In small airways and lung parenchyma, little CGRP immunoreactivity was observed in each experimental group (Fig. 7). Table 2 summarizes the visual assessment of CGRP immunoreactivity in each group. In airway epithelium, submucosa, and smooth muscle of large airways, the scores were significantly higher in OA-treated CGRP+/+ mice than in any other group, although there were marked differences between alpha -CGRP+/+ and alpha -CGRP-/- mice. Meanwhile, the scores were much lower in peripheral airways and lung parenchyma than in large airways in the wild-type mice.


View larger version (107K):
[in this window]
[in a new window]
 
Fig. 6.   Photomicrographs of CGRP immunohistochemical staining in large airways from saline-treated wild-type (A), saline-treated alpha -CGRP-deficient (B), OA antigen-treated wild-type (C), and OA antigen-treated alpha -CGRP-deficient (D) mice. Arrowheads, CGRP immunoreactivity. Immunoreactivity for CGRP was increased in antigen-treated wild-type mice (C) compared with unsensitized control (A). There was little immunostaining for CGRP in alpha -CGRP-deficient mice (B and D). Specimens were counterstained with hematoxylin. Scale bar, 50 µm.



View larger version (120K):
[in this window]
[in a new window]
 
Fig. 7.   Photomicrographs of CGRP immunohistochemical staining in small airways and lung parenchyma from saline-treated wild-type (A), saline-treated alpha -CGRP-deficient (B), OA antigen-treated wild-type (C), and OA antigen-treated alpha -CGRP-deficient (D) mice. There was little immunostaining for CGRP in each group. Specimens were counterstained with hematoxylin. Scale bar, 50 µm.


                              
View this table:
[in this window]
[in a new window]
 
Table 2.   Visual assessment of airway CGRP immunoreactivity using immunoreactivity scores


    DISCUSSION
TOP
ABSTRACT
INTRODUCTION
METHODS
RESULTS
DISCUSSION
REFERENCES

The results of the present experiments show that antigen-induced bronchial hyperresponsiveness was significantly reduced in CGRP-deficient mice. Meanwhile, eosinophil infiltration elicited by antigen challenge was unaffected by disruption of the CGRP gene. Antigen-induced increases in BALF LTC4/D4/E4 were significantly attenuated in alpha -CGRP-disrupted mice. These findings suggest that CGRP could be involved in the antigen-induced airway hyperresponsiveness, but not eosinophil infiltration, in mice.

CGRP has pleiotropic and pathophysiological effects on various cells and organs (3, 44). CGRP exerts trophic effects on skeletal muscle and vascular smooth muscle (3). CGRP also modulates some macrophage functions, including antigen presentation (13, 36). In the respiratory system, CGRP is synthesized by sensory C-fibers in the respiratory tree (47). However, the pathophysiological roles of CGRP in the lung have not been determined. Palmer et al. (39) demonstrated that CGRP potently constricts human airway smooth muscle. On the other hand, recent studies have reported that CGRP acts as a potent inhibitor of responses elicited by bronchoconstrictive stimuli (4, 33). Regarding eosinophil chemotaxis, Numao and Agrawal (37) reported that neuropeptides, including CGRP, may play a significant role in eosinophil infiltration by priming cells in allergic inflammation. Meanwhile, Teixeira et al. (46) demonstrated that CGRP has little effect on eosinophil accumulation in guinea pig skin. In the present study, we hypothesized that CGRP could play a significant role in the underlying mechanism of asthma. To test this hypothesis, we studied the allergic pulmonary responses using alpha -CGRP gene-disrupted mice, which have been recently established by Oh-hashi et al. (38).

Allergen-induced airway hyperresponsiveness was significantly attenuated in the alpha -CGRP-deficient mice, suggesting that the existence of CGRP per se might be associated with bronchial hyperresponsiveness, which is a major trait of asthma (11, 12, 23). To our knowledge, this is the first report to use mutant mice to study whether the CGRP gene and endogenous CGRP could be involved in the airway hyperresponsiveness. Recently, using a pharmacological approach, Dakhama et al. (6) found that exogenous administration of CGRP to sensitized and challenged mice results in the normalization of airway responsiveness. However, the exact mechanism to explain the involvement of CGRP in airway hyperresponsiveness remains to be clarified. In the present study, the molecular and pathophysiological mechanisms underlying airway hyperresponsiveness were further examined using CGRP-mutant mice.

One of the possible mechanisms is that CGRP and CGRP gene expression might affect airway inflammation, including eosinophilia, after antigen challenge. Airway eosinophilia is one of the common features in asthmatic patients and could be involved in bronchial hyperresponsiveness (11, 23). In the present study, however, no significant difference in BALF eosinophil counts was observed between the wild-type and CGRP-deficient mice. These results suggest that disruption of the CGRP gene has little effect on antigen-induced airway eosinophilia in mice. Although the sequence of rodent CGRP contains the tetrapeptide eosinophil chemotactic factor, the effect of CGRP on airway eosinophil infiltration is not remarkable in the present model. Therefore, CGRP-dependent airway hyperresponsiveness might not be mediated by eosinophilia.

Possibly, immunization provoked by antigen challenge might be affected by modulation of the CGRP gene. However, increased IgE levels after antigen challenge were observed in both groups, whereas there were no significant differences in measured IgE levels between the wild-type and CGRP-deficient groups. Alveolar protein leakage or airway mucus secretion assessed by BALF protein was consistent with the results of IgE measurement in this study. These findings indicate that modulation of the CGRP gene might not affect the mechanism of IgE production.

Recently, it has been shown that bronchial asthma is related to the generation of various potent mediators, including thromboxane, leukotriene, and ET-1 (28, 45, 49). These mediators are reported to be involved in airway hyperresponsiveness (28). Cysteinyl leukotrienes (LTC4, LTD4, and LTE4) are reported to be among the most important targets for treating bronchial asthma. It has been shown that administration of cysteinyl leukotriene antagonist reduces antigen-induced airway hyperresponsiveness (10, 49) and the increases in airway smooth muscle after antigen exposure (49). Irvin et al. (14) demonstrated that antigen-induced airway hyperresponsiveness is significantly decreased in 5-lipoxygenase-deficient mice, suggesting the important role of leukotrienes in development of airway hyperresponsiveness. The potential sources of cysteinyl leukotrienes in the lung include alveolar macrophages, eosinophils, basophils, mast cells, and platelets (40). The proinflammatory activities of cysteinyl leukotrienes, including bronchoconstriction, mucus secretion, and plasma exudation, are mediated via the interaction with its receptor, the CysLT1 receptor (9). In humans, it has been recently demonstrated that the CysLT1 receptor is expressed in lung smooth muscle, lung macrophages, and peripheral blood leukocytes, while the identification of the CysLT1 receptor is consistent with the anti-inflammatory actions of CysLT1 receptor antagonists (9).

Potentially, genetic disruption of the CGRP gene may modulate the production levels of various potent mediators. We therefore measured possible mediators in the BALF and found that the production of cysteinyl leukotrienes was enhanced in the sensitized wild-type mice. In contrast, the level of cysteinyl leukotrienes was significantly reduced in the sensitized CGRP-deficient mice. There were no significant differences in thromboxane or ET-1 in each group. These observations indicate that CGRP gene disruption might inhibit the production of cysteinyl leukotrienes, which could be associated with reduced airway hyperresponsiveness. Meanwhile, after antigen challenge of wild-type and CGRP-deficient mice, there were no significant differences in the number of alveolar macrophages or eosinophils, i.e., potential sources of cysteinyl leukotrienes. One of the possible mechanisms to explain this observation is that CGRP might be involved in activation of the 5-lipoxygenase pathway.

In the present study, we used mutant mice deficient in alpha -CGRP but not beta -CGRP. Therefore, these mutant mice should express beta -CGRP. Because the alpha -CGRP antibody used in this study cross-reacts with beta -CGRP (79%), the CGRP immunoreactivity represents alpha -CGRP and, similarly, beta -CGRP. The very small amounts of CGRP immunoreactivity in the mutant mice may indicate beta -CGRP expression in the lung. It has been previously reported that alpha -CGRP concentrations are approximately four times greater than beta -CGRP concentrations in the rat lung, whereas in the intestine, beta -CGRP concentrations are up to seven times greater than alpha -CGRP concentrations (26). Presumably, it seems that beta -CGRP expression in the lung might not be affected by disruption of the alpha -CGRP gene in mice.

In the wild-type mice, we observed substantial CGRP immunoreactivity in the epithelium and submucosal tissues in large airways but not in small airways or parenchyma. Presumably, CGRP-immunoreactive cells and tissues include nerve fibers in submucosal tissues, whereas airway epithelium contains nerves and NEBs. Terada et al. (47) reported that nerve plexuses of CGRP-immunoreactive fibers are located in the basal part of the rat tracheal epithelium. These CGRP-immunoreactive intraepithelial nerves lack myelin and Schwann sheaths and run through the bases of the epithelial cells (47). In this study, the CGRP immunoreactivity in large airway epithelium was remarkable, and it was enhanced by antigen challenge. These observations suggest that the epithelium of central airways, including nerves and NEBs, may have a significant role in antigen-induced airway hyperresponsiveness. Meanwhile, it is assumed that the contribution of peripheral airways and parenchyma to CGRP-related airway physiology is small.

Recently, Dakhama et al. (6) showed that CGRP expression was diminished in airway epithelium and submucosal nerve plexuses only after the third OA challenge, although CGRP depletion did not occur after the single antigen exposure. In our study, however, the single antigen challenge enhanced CGRP immunoreactivity in large airways in the wild-type mice, whereas little CGRP immunoreactivity was observed in CGRP-deficient mice in the absence or presence of antigen challenge. The present findings suggest that endogenous CGRP per se may be related to the development of antigen-induced airway hyperresponsiveness.

Genetic features, including single-nucleotide polymorphism, are potentially associated with the etiology of asthma. On the basis of the inheritance pattern, a number of genes could have substantial roles in the pathogenesis of bronchial asthma (43). Murine models of asthma have been recently used to investigate individual genes associated with airway hyperresponsiveness (8, 15, 16, 18, 31, 48). Because CGRP is one of the potent mediators possibly involved in bronchial asthma (39), genes regulating the function of CGRP, calcitonin receptor-like receptor, and RAMP1 could be targets to study the pathogenesis of asthma. Consistently, the present observations suggest that alpha -CGRP and the alpha -CGRP gene play significant roles in the molecular mechanism underlying bronchial asthma, indicating that the alpha -CGRP gene could be a target for single-nucleotide polymorphism research. The alpha -CGRP-mutant mice used in this study may contribute to the study of the genetic roles of alpha -CGRP in bronchial asthma and may provide novel insights into the pathophysiological roles of alpha -CGRP and the alpha -CGRP gene in vivo.

In summary, reduction of antigen-induced airway hyperresponsiveness was detected in alpha -CGRP-deficient mice. Meanwhile, eosinophilic infiltration associated with antigen exposure was not altered by disruption of the alpha -CGRP gene. Antigen-induced increases in cysteinyl leukotriene production were significantly reduced in alpha -CGRP-disrupted mice. Disruption of the alpha -CGRP gene might inhibit production of cysteinyl leukotrienes, which could be associated with reduced airway hyperresponsiveness. Antigen challenge enhanced CGRP immunoreactivity in the wild-type mice, whereas little CGRP immunoreactivity in epithelium or submucosa was observed in alpha -CGRP-deficient mice. These findings suggest that endogenous CGRP may be involved in development of antigen-induced airway hyperresponsiveness. Taken together, CGRP and CGRP gene expression might be involved in the pathogenesis of bronchial asthma by acting as a mediator. The CGRP-mutant mice may provide appropriate models to study molecular and pathophysiological mechanisms underlying diseases related to CGRP.


    ACKNOWLEDGEMENTS

We thank Y. Tateno (University of Tokyo) for technical assistance.


    FOOTNOTES

This work was supported in part by a Grant-in-Aid for Scientific Research from the Ministry of Education, Science, Sports, and Culture, Japan, and an AstraZeneca research grant.

Address for reprint requests and other correspondence: T. Nagase, Dept. of Geriatric Medicine, Faculty of Medicine, University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan (E-mail: takahide-tky{at}umin.ac.jp).

The costs of publication of this article were defrayed in part by the payment of page charges. The article must therefore be hereby marked "advertisement" in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

June 21, 2002;10.1152/ajplung.00130.2002

Received 14 May 2002; accepted in final form 17 June 2002.


    REFERENCES
TOP
ABSTRACT
INTRODUCTION
METHODS
RESULTS
DISCUSSION
REFERENCES

1.   Amara, SG, Arriza JL, Leff SE, Swanson LW, Evans RM, and Rosenfeld MG. Expression in brain of a messenger RNA encoding a novel neuropeptide homologous to calcitonin gene-related peptide. Science 229: 1094-1097, 1985[ISI][Medline].

2.   Amara, SG, Jonas V, Rosenfeld MG, Ong ES, and Evans RM. Alternative RNA processing in calcitonin gene expression generates mRNAs encoding different polypeptide products. Nature 298: 240-244, 1982[ISI][Medline].

3.   Brain, SD, Williams TJ, Tippins JR, Morris HR, and MacIntyre I. Calcitonin gene-related peptide is a potent vasodilator. Nature 313: 54-56, 1985[ISI][Medline].

4.   Cadieux, A, Monast NP, Pomerleau F, Fournier A, and Lanoue C. Bronchoprotector properties of calcitonin-gene related peptide in guinea pig and human airways. Am J Respir Crit Care Med 159: 235-243, 1999[Abstract/Free Full Text].

5.   Chanez, P, Springall D, Vignola AM, Moradoghi-Hattvani A, Polak JM, Godard P, and Bousquet J. Bronchial mucosal immunoreactivity of sensory neuropeptides in severe airway diseases. Am J Respir Crit Care Med 158: 985-990, 1998[Abstract/Free Full Text].

6.   Dakhama, A, Kanehiro A, Makela MJ, Loader JE, Larsen GL, and Gelfand EW. Regulation of airway hyperresponsiveness by calcitonin gene-related peptide in allergen sensitized and challenged mice. Am J Respir Crit Care Med 165: 1137-1144, 2002[Abstract/Free Full Text].

7.   Davies, D, Medeiros MS, Keen J, Turner AJ, and Haynes LW. Endopeptidase-24.11 cleaves a chemotactic factor from alpha -calcitonin gene-related peptide. Biochem Pharmacol 43: 1753-1756, 1992[ISI][Medline].

8.   DeSanctis, GT, Merchant M, Beier DR, Dredge RD, Grobholz JK, Martin TR, Lander ES, and Drazen JM. Quantitative locus analysis of airway hyperresponsiveness in A/J and C57BL/6J mice. Nat Genet 11: 150-154, 1995[ISI][Medline].

9.   Figueroa, DJ, Breyer RM, Defoe SK, Kargman S, Daugherty BL, Waldburger K, Liu Q, Clements M, Zeng Z, O'Neill GP, Jones TR, Lynch KR, Austen CP, and Evans JF. Expression of the cysteinyl leukotriene 1 receptor in normal human lung and peripheral blood leukocytes. Am J Respir Crit Care Med 163: 226-233, 2001[Abstract/Free Full Text].

10.   Herd, CM, Donigi-Gale D, Shoupe TS, Burroughs DA, Yeadon M, and Page CP. Effect of a 5-lipoxygenase inhibitor and leukotriene antagonist (PF 5901) on antigen-induced airway responses in neonatally immunized rabbits. Br J Pharmacol 112: 292-298, 1994[Abstract].

11.   Hogg, JC. Pathology of asthma. J Allergy Clin Immunol 92: 1-5, 1993[ISI][Medline].

12.   Holgate, ST. The epidemic of allergy and asthma. Nature 402: B2-B4, 1999[ISI][Medline].

13.   Hosoi, J, Murphy GF, Egan CL, Lerner EA, Grabbe S, Asahina A, and Granstein RD. Regulation of Langerhans cell function by nerves containing calcitonin gene-related peptide. Nature 363: 159-163, 1993[ISI][Medline].

14.   Irvin, CG, Tu YP, Sheller JR, and Funk CD. 5-Lipoxygenase products are necessary for ovalbumin-induced airway responsiveness in mice. Am J Physiol Lung Cell Mol Physiol 272: L1053-L1058, 1997[Abstract/Free Full Text].

15.   Ishii, S, Kuwaki T, Nagase T, Maki K, Tashiro F, Sunaga S, Cao WH, Kume K, Fukuchi Y, Ikuta K, Miyazaki J, Kumada M, and Shimizu T. Impaired anaphylactic responses but intact sensitivity to endotoxin in mice lacking a platelet-activating factor receptor. J Exp Med 187: 1779-1788, 1998[Abstract/Free Full Text].

16.   Ishii, S, Nagase T, Tashiro F, Ikuta K, Sato S, Waga I, Kume K, Miyazaki J, and Shimizu T. Bronchial hyperreactivity, increased endotoxin lethality and melanocytic tumorigenesis in transgenic mice overexpressing platelet-activating factor receptor. EMBO J 16: 133-142, 1997[Abstract/Free Full Text].

17.   Janssen, PL, and Tucker A. Calcitonin gene-related peptide modulates pulmonary vascular reactivity in isolated rat lungs. J Appl Physiol 77: 142-146, 1994[Abstract/Free Full Text].

18.   Levitt, RC, and Mitzner W. Autosomal recessive inheritance of airway hyperreactivity to 5-hydroxytryptamine. J Appl Physiol 67: 1125-1132, 1989[Abstract/Free Full Text].

19.   Lu, B, Fu WM, Greengard P, and Poo MM. Calcitonin gene-related peptide potentiates synaptic responses at developing neuromuscular junction. Nature 363: 76-79, 1993[ISI][Medline].

20.   Lundberg, JM, Anders FC, Hua X, Hökfelt T, and Fischer JA. Coexistence of substance P and calcitonin gene-related peptide-like immunoreactivities in sensory nerves in relation to cardiovascular and bronchoconstrictor effects of capsaicin. Eur J Pharmacol 108: 315-319, 1985[ISI][Medline].

21.   Mapp, CE, Lucchini RE, Miotto D, Chitano P, Jovine L, Saetta M, Maestrelli P, Springall DR, Polak J, and Fabbri LM. Immunization and challenge with toluene diisocyanate decrease tachykinin and calcitonin gene-related peptide immunoreactivity in guinea pig central airways. Am J Respir Crit Care Med 158: 263-269, 1998[Abstract/Free Full Text].

22.   McCormack, D, Mak J, Coupe M, and Barnes PJ. Calcitonin gene-related peptide vasodilation of human pulmonary vessels. J Appl Physiol 67: 1265-1270, 1989[Abstract/Free Full Text].

23.   McFadden, ER, and Gilbert IA. Asthma. N Engl J Med 327: 1928-1937, 1992[Abstract].

24.   McLatchie, LM, Fraser NJ, Main MJ, Wise A, Brown J, Thompson N, Solary R, Lee MG, and Foord SM. RAMPs regulate the transport and ligand specificity of the calcitonin-receptor-like receptor. Nature 393: 333-339, 1998[ISI][Medline].

25.   Merighi, A, Polak JM, Gibson SJ, Gulbenkian S, Valentino KL, and Peirone SM. Ultrastructural studies on calcitonin gene-related peptide-, tachykinin-, and somatostatin-immunoreactive neurons in rat dorsal root ganglia: evidence for the colocalization of different peptides in single secretory granules. Cell Tissue Res 254: 101-109, 1988[ISI][Medline].

26.   Mulderry, PK, Ghatei MA, Spokes RA, Jones PM, Pierson AM, Hamid QA, Kanse S, Amara SG, Burrin JM, Legon S, Polak JM, and Bloom SR. Differential expression of alpha CGRP and beta CGRP by primary sensory neurons and enteric autonomic neurons of the rat. Neuroscience 25: 195-205, 1988[ISI][Medline].

27.   Nagase, T, Fukuchi Y, Matsuse T, Sudo E, Matsui H, and Orimo H. Antagonism of ICAM-1 attenuates airway and tissue responses to antigen in sensitized rats. Am J Respir Crit Care Med 151: 1244-1249, 1995[Abstract].

28.   Nagase, T, Ishii S, Katayama H, Fukuchi Y, Ouchi Y, and Shimizu T. Airway responsiveness in transgenic mice overexpressing platelet-activating factor receptor: roles of thromboxanes and leukotrienes. Am J Respir Crit Care Med 156: 1621-1627, 1997[Abstract/Free Full Text].

29.   Nagase, T, Ishii S, Kume K, Uozumi N, Izumi T, Ouchi Y, and Shimizu T. Platelet-activating factor mediates acid-induced lung injury in genetically engineered mice. J Clin Invest 104: 1071-1076, 1999[Abstract/Free Full Text].

30.   Nagase, T, Ishii S, Shindou H, Ouchi Y, and Shimizu T. Airway hyperresponsiveness in transgenic mice overexpressing platelet-activating factor receptor is mediated by an atropine-sensitive pathway. Am J Respir Crit Care Med 165: 200-205, 2002[Abstract/Free Full Text].

31.   Nagase, T, Kurihara H, Kurihara Y, Aoki T, Fukuchi Y, Yazaki Y, and Ouchi Y. Airway hyperresponsiveness to methacholine in mutant mice deficient in endothelin-1. Am J Respir Crit Care Med 157: 560-564, 1998[Abstract/Free Full Text].

32.   Nagase, T, Matsui H, Aoki T, Ouchi Y, and Fukuchi Y. Lung tissue behaviour in the mouse during constriction induced by methacholine and endothelin-1. J Appl Physiol 81: 2373-2378, 1996[Abstract/Free Full Text].

33.   Nagase, T, Ohga E, Katayama H, Sudo E, Aoki T, Matsuse T, Ouchi Y, and Fukuchi Y. Roles of calcitonin-gene related peptide (CGRP) in hyperpnea-induced constriction in guinea pigs. Am J Respir Crit Care Med 154: 1551-1556, 1996[Abstract].

34.   Nagase, T, Uozumi N, Ishii S, Kita Y, Yamamoto H, Ohga E, Ouchi Y, and Shimizu T. A pivotal role of cytosolic phospholipase A2 in bleomycin-induced pulmonary fibrosis. Nat Med 8: 480-484, 2002[ISI][Medline].

35.   Nagase, T, Uozumi N, Ishii S, Kume K, Izumi T, Ouchi Y, and Shimizu T. Acute lung injury by sepsis and acid aspiration: a key role for cytosolic phospholipase A2. Nat Immunol 1: 42-46, 2000[ISI][Medline].

36.   Nong, YH, Titus RG, Ribeiro JMC, and Remold HG. Peptides encoded by the calcitonin gene inhibit macrophage function. J Immunol 143: 45-49, 1989[Abstract/Free Full Text].

37.   Numao, T, and Agrawal DK. Neuropeptides modulate human eosinophil chemotaxis. J Immunol 149: 3309-3315, 1992[Abstract/Free Full Text].

38.   Oh-hashi, Y, Shindo T, Kurihara Y, Imai T, Wang Y, Morita H, Imai Y, Kayaba Y, Nishimatsu H, Suematsu Y, Hirata Y, Yazaki Y, Nagai R, Kuwaki T, and Kurihara H. Elevated sympathetic nervous activity in mice deficient in alpha CGRP. Circ Res 89: 983-990, 2001[Abstract/Free Full Text].

39.   Palmer, JBD, Cuss FMC, Mulderry PK, Ghatei MA, Springall DR, Cadieux A, Bloom SR, Polak JM, and Barnes PJ. Calcitonin gene-related peptide is localized to human airway nerves and potently constricts human airway smooth muscle. Br J Pharmacol 91: 95-101, 1987[Abstract].

40.   Penrose, JF, Spector J, Baldasaro M, Xu K, Boyce J, Arm JP, Austen KF, and Lam BK. Molecular cloning of the gene for human leukotriene C4 synthase: organization, nucleotide sequence, and chromosomal localization to 5q35. J Biol Chem 271: 11356-11361, 1996[Abstract/Free Full Text].

41.   Pinto, A, Sekizawa K, Yamaya M, Ohrui T, Jia YX, and Sasaki H. Effects of adrenomedullin and calcitonin gene-related peptide on airway and pulmonary vascular smooth muscle in guinea-pigs. Br J Pharmacol 119: 1477-1483, 1996[Abstract].

42.   Rosenfeld, MG, Mermod JJ, Amara SG, Swanson LW, Sawchenko PE, Rivier J, Vale WW, and Evans RM. Production of a novel neuropeptide encoded by the calcitonin gene via tissue-specific RNA processing. Nature 304: 129-135, 1983[ISI][Medline].

43.   Sandford, A, Weir T, and Paré P. The genetics of asthma. Am J Respir Crit Care Med 153: 1749-1765, 1996[Abstract].

44.   Solway, J, and Leff AR. Sensory neuropeptides and airway function. J Appl Physiol 71: 2077-2087, 1991[Abstract/Free Full Text].

45.   Springall, DR, Howarth PH, Counihan H, Djukanovic R, Holgate ST, and Polak JM. Endothelin immunoreactivity of airway epithelium in asthmatic patients. Lancet 337: 697-701, 1991[ISI][Medline].

46.   Teixeira, MM, Williams TJ, and Hellewell PG. E-type prostaglandins enhance local oedema formation and neutrophil accumulation but suppress eosinophil accumulation in guinea-pig skin. Br J Pharmacol 110: 416-422, 1993[Abstract].

47.   Terada, M, Iwanaga T, Iwanaga HT, and Adachi I. Calcitonin gene-related peptide (CGRP)-immunoreactive nerves in the tracheal epithelium of rats: an immunohistochemical study by means of whole mount preparations. Arch Histol Cytol 55: 219-233, 1992[ISI][Medline].

48.   Uozumi, N, Kume K, Nagase T, Nakatani N, Ishii S, Tashiro F, Komagata Y, Maki K, Ikuta K, Ouchi Y, Miyazaki J, and Shimizu T. Role of cytosolic phospholipase A2 in allergic response and parturition. Nature 390: 618-622, 1997[ISI][Medline].

49.   Wang, CG, Du T, Xu LJ, and Martin JG. Role of leukotriene D4 in allergen-induced increases in airway smooth muscle in the rat. Am Rev Respir Dis 148: 413-417, 1993[ISI][Medline].


Am J Physiol Lung Cell Mol Physiol 283(5):L963-L970
1040-0605/02 $5.00 Copyright © 2002 the American Physiological Society