Open Access

Effect of P2X4R on airway inflammation and airway remodeling in allergic airway challenge in mice

  • Authors:
    • Hongxia Chen
    • Qingqing Xia
    • Xiaoqian Feng
    • Fangyuan Cao
    • Hang Yu
    • Yinli Song
    • Xiuqin Ni
  • View Affiliations

  • Published online on: November 27, 2015     https://doi.org/10.3892/mmr.2015.4622
  • Pages: 697-704
  • Copyright: © Chen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

P2X4 receptor (P2X4R) is the most widely expressed subtype of the P2XRs in the purinergic receptor family. Adenosine triphosphate (ATP), a ligand for this receptor, has been implicated in the pathogenesis of asthma. ATP‑P2X4R signaling is involved in pulmonary vascular remodeling, and in the proliferation and differentiation of airway and alveolar epithelial cell lines. However, the role of P2X4R in asthma remains to be elucidated. This aim of the present study was to investigate the effects of P2X4R in a murine experimental asthma model. The asthmatic model was established by the inhalation of ovalbumin (OVA) in BALB/c mice. The mice were treated with P2X4R‑specific agonists and antagonists to investigate the role of this receptor in vivo. Pathological changes in the bronchi and lung tissues were examined using hematoxylin and eosin staining, Masson's trichrome staining and Alcian blue staining. The inflammatory cells in the bronchoalveolar lavage fluid were counted, and the expression levels of P2X4R, α‑smooth muscle actin (α‑SMA) and proliferating cell nuclear antigen (PCNA) were detected using western blotting. In the OVA‑challenged mice, inflammation, infiltration, collagen deposition, mucus production, and the expression levels of P2X4R and PCNA were all increased; however, the expression of α‑SMA was decreased, compared with the mice in the control group. Whereas treatment with the P2X4R agonist, ATP, enhanced the allergic reaction, treatment with the P2X4R antagonist, 5‑BDBD, attenuated the allergic reaction. The results suggested that ATP‑P2X4R signaling may not only contribute to airway inflammation, but it may also contribute to airway remodeling in allergic asthma in mice.

Introduction

Asthma is a chronic inflammatory airway disease involving several different types of cell. It is considered an allergic disease with characteristic airway inflammation, airway hyper-responsiveness (AHR) and airway remodeling (13). At present, anti-inflammatories are the predominant therapeutic option for patients with asthma (4,5). Previous studies (68) have shown that airway remodeling results in the progressive loss of lung function. Airway remodeling includes epithelial denudation, subepithelial fibrosis, mucus gland hypertrophy, myofibroblast and smooth muscle cell proliferation, and angiogenesis (911). The expression levels of α-smooth muscle actin (α-SMA) (12,13) and proliferating cell nuclear antigen (PCNA) (14,15) are indicative of asthmatic cell proliferation.

Adenosine triphosphate (ATP), a key mediator of acute and chronic inflammation, can be released in substantial quantities from various cell types following cellular stress or tissue injury (1619). Extracellular ATP exerts pro-inflammatory and immunomodulatory effects in the tumor microenvironment by binding to purinergic P2-receptors, which can be subdivided into two families: The G-protein-coupled P2YR (P2Y 1–14) and the ligand-gated ion channel, P2X receptors (P2XR), which include P2X1-7 (2022). Data from patient and animal models of allergic bronchial asthma indicate that ATP contributes to the pathophysiology of allergic airway inflammation (16). However, whether ATP-mediated P2X4R signaling is activated in allergic asthma remains to be elucidated.

Among the P2XR subtypes, P2X4R is the most widely expressed (23). P2X4 receptors are located in immune and non-immune cells, including alveolar epithelial cells, alveolar macrophages, platelets, lymphocytes and mast cells (2426). P2X4R can affect T cell activation via Ca2+ influx. ATP-P2X4R signaling is involved in peripheral nerve injury inflammation, which includes the promotion of the expression levels of interleukin (IL)-1β, IL-6 and tumor necrosis factor (TNF)-α via mitogen-activated protein kinase activation; indicating that P2X4R is involved in the immune response to nerve inflammation (27). Several studies have demonstrated that P2X4 receptors are involved in hypersensitivity by acting on prostaglandin E2 (2830). ATP-P2X4R signaling is also involved in pulmonary vascular remodeling, and in the proliferation and differentiation of airway and alveolar epithelial cell lines in pulmonary hypertension (31,32). However, whether P2X4R signaling is involved in allergic airway inflammation and airway remodeling requires further investigation.

The aim of the present study was to investigate the effects of P2X4R in a murine experimental asthma model and the effects of a P2X4R-specific antagonist, 5-BDBD, on airway inflammation and airway remodeling. Our findings may ultimately lead to the development of novel therapeutic targets for allergic asthma.

Materials and methods

Chemicals and reagents

Rabbit polyclonal anti-P2X4R primary antibody was purchased from Calbiochem (EMD Millipore, Billerica, MA, USA; cat. no. PC376) and used for western blotting. Mouse monoclonal anti-PCNA primary antibody (cat. no. BM0104) and anti-α-SMA primary antibody (cat. no. BM0002) were purchased from Boster Systems, Inc. (Wuhan, China). Mouse monoclonal anti-β-actin primary antibody (cat. no. TA-09) was purchased from ZSGB Biotechnology, Inc. (Beijing, China). Mouse anti-rabbit (cat. no. sc-2357) and rabbit anti-mouse (cat. no. sc-358914) IgG-horseradish peroxidase-conjugated secondary antibodies were purchased from Santa Cruz Biotechnology Inc. (Santa Cruz, CA, USA). The P2X4R-specific antagonist, 5-BDBD, was obtained from Tocris Bioscience (Bristol, UK). The Masson's trichromatic staining kit and Alcian blue-periodic acid-schiff (AB-PAS) mucosa staining kit were purchased from Fuzhou Maixin Biotechnology, Co., Ltd. (Fuzhou, China). ATP and the P2XR1/2/3/5/7 antagonist, phenyl-isopropyl-amine dopes (PPAD), were all purchased from Sigma-Aldrich (St. Louis, MO, USA). All other reagents were obtained from common commercial sources.

Experimental animals

Pathogen-free female BALB/c mice, aged 6–8 weeks and weighing ~18–20 g, were purchased from the Laboratory Animal Research Center (Beijing, China). Mice were maintained under 12 h light/dark cycles at room temperature, with free access to food and water. All animal experiments and manipulation procedures were approved by the Ethics Committee for Animal Use and Care of Harbin Medical University-Daqing (Daqing, China).

Animal groups and allergic sensitization protocol

A total of 48 BALB/c mice were randomized into the following five groups: Phosphate-buffered saline (PBS) control group (n=8), OVA group (n=10), ATP group (n=10), PPAD group (n=10) and 5-BDBD group (n=10). In the OVA group, the mice were sensitized to OVA (20 µg per injection), absorbed to 2.0 mg per injection of aluminum hydroxide (Tianjin Fuchen Chemical Reagents Factory, Tianjin, China), by intraperitoneal injection on day 1. On days 8, 15 and 22, the mice were sensitized again to OCA (10 µg per injection), absorbed to 1.0 mg per injection of aluminum hydroxide, by intraperitoneal injection. From day 23, the mice were exposed, using inhalation aerosols, of 4% OVA in PBS for 25–30 min, until the onset of bronchial obstruction, daily for 7 days consecutively, according to the methods described by Shen et al (31) and Vanacker et al (32) with modifications. The mice in the ATP, PPAD and 5-BDBD groups were also subjected to OVA sensitization and asthma induction, in the same manner. Intranasal application of 50 µl ATP (1 mM) (33), PPAD (0.1 mM) (34) and 5-BDBD (30 µmol) (35) were performed 3 h prior to each airway allergen challenge in the ATP, PPAD and 5-BDBD groups, respectively. All treatments were administered daily for 5 days consecutively. Mice in the control group were administered with PBS alone (0.5 ml) by injection, and were challenged with PBS (20 ml). All animals were humanely sacrificed within 24 h of the final OVA or PBS exposure.

Morphometric analysis

The lungs were fixed in 4% paraformaldehyde, embedded in paraffin and sectioned at a thickness of 5 µm. The sections from each animal were stained with hematoxylin and eosin for inflammation, Masson's trichrome staining for collagen-deposition and Alcian blue/periodic acid-Schiff (AB-PAS) staining for the analysis of goblet cell hyperplasia/mucus production. All staining procedures were performed in accordance with the manufacturer's protocol. Pathological changes in the lung and bronchial tissues were observed and images were captured using a BX-60 microscope (Olympus, Tokyo, Japan). The tissues were scored based on the presence of inflammatory cells, using the following scale: Absent (0), rare (1), mild (2), moderate (3) or severe (4). Scoring was performed by an observer in a blinded-manner. Positively stained areas of collagen and mucus in the lung and bronchial tissues were quantified using high-resolution images of individual vessels by image analysis, using a color-recognition algorithm, in Image Pro Plus 6.0 (Media Cybernetics, Inc., Rockville, MD, USA). The results were expressed as the percentage of collagen area or mucus area, compared to the total measured area.

Bronchoalveolar lavage fluid (BALF)

Samples of BALF were collected 24 h following the final challenge, according to previously described methods. The lungs were lavaged with 1.6 ml (0.8 ml per lavage) of ice-cold PBS via the tracheal cannula. The retrieved volume (~75–80%) of the instilled PBS was then centrifuged at 500 × g at 4°C for 5 min. The resultant pellet was washed twice with PBS and was resuspended in 1 ml PBS. The total numbers of cells were counted using a hemocytometer (Bio-Rad Laboratories, Inc., Hercules, CA, USA). Smears of the BALF cells were stained with Wright and Giemsa staining fluid (Beijing Solarbio Science & Technology Co., Ltd., Beijing, China) for differential cell counting. Using a BX-60 Olympus microscope, the numbers of cells were counted by two independent investigators, in a blinded-manner. A total of ~200 cells were counted in each of four randomly selected locations.

Western blotting

The lung tissues were homogenized with cold lysis buffer (Beyotime Institute of Biotechnology, Shanghai, China), containing 50 mM Tris (pH 7.4), 150 mM NaCl, 1% Triton X-100, 1 mM EDTA and 2 mM PMSF. Following ultrasonication on ice for 5 sec using an FA-25 (Fluko Equipment Shanghai Co., Ltd., Shanghai, China) and centrifugation at 12,500 × g at 4°C for 15 min, the supernatant was collected, and the protein concentration was determined using a Bicinchoninic Acid Protein Assay (Pierce Biotechnology, Inc., Rockford, IL, USA) with bovine serum albumin (Biosharp, Hefei, China) as a standard. Each sample, containing 20 µg protein, was separated on 8–10% SDS-PAGE gels and transferred onto nitrocellulose membranes (EMD Millipore). Following blocking with Tris-buffered saline, containing 20 mM Tris, 150 mM NaCl (pH 7.6) and 0.1% Tween 20, with 5% nonfat milk at room temperature, the membranes were incubated overnight at 4°C with specific antibodies against P2X4R (1:400 dilution), α-SMA (1:500 dilution), PCNA (1:500 dilution) or β-actin (1:1,000 dilution). This was followed by incubation for 2 h at room temperature with horseradish peroxidase-conjugated secondary antibodies (1:5,000 dilution). All protein blots were visualized using enhanced chemiluminescence reagents (GE Healthcare Life Sciences). Integrated density values were analyzed using a computerized image analysis system (Fluor Chen 2.0; Olympus Corporation) and were normalized to those of β-actin.

Statistical analysis

Data were analyzed using SPSS 15.0 software (SPSS, Inc., Chicago, IL, USA). Data are expressed as the mean ± standard error of the mean. Differences between groups were analyzed using Student's t-test and one-way analysis of variance, followed by Fisher's least significant difference test. P<0.05 was considered to indicate a statistically significant difference.

Results

OVA-challenge upregulates the protein expression of P2X4R in mice

The present study performed western blotting to compare the protein expression levels of P2X4R in the lung tissues of the treatment groups (Fig. 1A and B). As shown in Fig. 1, OVA-challenge significantly increased the expression of P2X4R, compared with the control mice.

P2X4R contributes to inflammation of the bronchi and lung tissues in allergic airway challenge in mice

Histological analyses of the tissues of the mice in the OVA group showed more extensive infiltration of inflammatory cells around the bronchioles, blood vessels and alveoli. The airway mucosa showed edema, with bronchiolar wall thickening causing luminal stenosis, suggesting successful establishment of the asthmatic model. In the OVA-challenged mice, treatment with ATP significantly increased OVA-induced inflammation, infiltration, and pathological changes of the bronchi and lung tissues. By contrast, treatment of OVA-challenged mice with PPAD (a P2X1, 2, 3, 5 and 7R antagonist) (36) and 5-BDBD (a P2X4R antagonist) (37) significantly reduced the extent of inflammation and cellular infiltration in the airway. In addition, the pathological changes of the bronchi and lung tissues were milder, compared with the changes found in the asthma group without treatment (Fig. 2A and B).

Cell numbers and differential cell percentages in BALF

The total cell numbers (Fig. 3A) and the percentages of eosinophils and lymphocytes (Fig. 3B) in the BALF from the asthmatic mice increased significantly, compared with those in the BALF from the control mice. The increased numbers of inflammatory cells, including eosinophils and lymphocytes, were significantly reduced by the administration of PPAD and 5-BDBD in the asthmatic mice, compared with the numbers found in the untreated asthmatic mice. The changes in the percentages of macrophages showed the opposite results.

P2X4R is required for collagen deposition in allergic airway challenge in mice

Collagen deposition in airway remodeling was assessed using Masson's trichrome staining of lung histological sections. Collagenous matrix was identified by its characteristic blue color. No obvious collagen fibers were present in the lungs of mice in the control group. However, deposition of collagen fibers was increased around the bronchial airways and vessels in the lungs of the OVA-challenged and ATP-treated mice. By contrast, intervention with PPAD and 5-BDBD significantly reduced collagen deposition (Fig. 4A and B).

P2X4R is required for goblet cell hyperplasia and mucus production in allergic airway challenge in mice

To assess mucus production and the hyperplasia of goblet cells, lung sections were stained with AB-PAS. In the OVA-challenged mice, bronchial epithelial mucus secretion and goblet cell hyperplasia were clearly observed, as a violet color in the bronchial airways, compared with the control group. The level of goblet cell hyperplasia in the OVA-challenged mice treated with ATP was significantly increased, compared with that in the OVA group. Intervention by PPAD or 5-BDBD significantly reduced goblet cell hyperplasia (Fig. 5A and B).

P2X4R regulates the expression of α-SMA and PCNA in the asthmatic mouse lung

Western blotting was performed to examine the protein levels of α-SMA (Fig. 6A and B) and PCNA (Fig. 7A and B) in each group. The expression levels of α-SMA in the OVA-challenged mice and ATP-treated mice were significantly lower, compared with those in the control mice. In addition, the mice treated with 5-BDBD showed significantly increased protein expression levels of α-SMA, compared with the mice in the OVA and ATP groups. However, the mice treated with PPAD showed no significant differences in protein expression, compared with the OVA group. The expression level of PCNA in the OVA-challenged mice and the ATP-treated mice were significantly higher than the level in the control mice. In addition, the mice treated with 5-BDBD showed a significant reduction in the protein expression levels of PCNA, compared with the mice in the OVA and ATP groups. Treatment with PPAD had no significant effects.

Discussion

Allergic asthma is a chronic airway inflammatory-driven disease, which is characterized by airway hyper-responsiveness, airway inflammation and varying degrees of smooth muscle cell proliferation, termed airway remodeling (3638). In the present study, airway inflammation and airway remodeling were examined in allergen-challenged mice. The pathophysiological mechanisms underlying airway remodeling remain to be fully elucidated. Compelling evidence indicates a pathophysiological relevance for ATP, acting via P2 purinergic receptors, in pain hypersensitivity and peripheral inflammation (3941). Certain studies have suggested that ATP contributes to the pathophysiology of allergic airway inflammation (14,16,42). In the present study, treatment of OVA-challenged mice with ATP significantly increased OVA-induced inflammation, infiltration, and pathological changes of the bronchi and lung tissues. Increased collagen deposition, goblet cell hyperplasia/mucus production, increased expression levels of PCNA expression, and decreased expression levels of α-SMA are indicative of airway remodeling (4346), and were observed in mice belonging to the OVA-challenged and ATP-treated groups. These results indicated that ATP may enhance airway inflammation and airway remodeling in allergic asthma. However, whether P2X4R is activated in allergic asthma by ATP-mediated signaling has not been determined.

Among the P2XR subtypes, P2X4R is the most widely expressed, which is expressed in blood vessels, the lungs, kidneys and immune cells (47,48). Additionally, P2X4R is involved in regulating blood pressure and vascular remodeling by ATP-induced Ca2+ influx and flow-mediated vasodilatation (49,50). P2X4Rs are also involved in modulating right ventricular hypertrophy, which occurs due to pulmonary hypertension in hypobaric hypoxia (51). Asthma and pulmonary arterial hypertension exhibit three major pathological features: Inflammation, smooth muscle constriction and smooth muscle cell proliferation (52). In the present study, the results demonstrated that treatment with the P2X4R antagonist, 5-BDBD, prevented the development of the airway and pulmonary inflammation, and airway remodeling induced by OVA challenge and OVA challenge coupled with ATP treatment. By contrast, treatment with PPAD showed no significant effect on the expression levels of α-SMA or PCNA. These data indicated that ATP-mediated P2X4R activation, but not P2X1/2/3/5/7R activation, was involved in airway inflammation and airway remodeling in allergic asthma. This suggests that P2X4R is involved in the pathogenesis of asthma via an ATP-mediated signaling pathway. To the best of our knowledge, the present study provides the first indication that P2X4R signaling is activated in allergic asthma, and contributes to airway inflammation and remodeling.

In conclusion, the results of the present study demonstrated the importance of P2X4R in allergic asthma. ATP-P2X4R signaling contributes to the pathogenesis of allergic asthma in mice. OVA-induced allergic asthma in mice was enhanced by the P2X4R agonist and attenuated by the P2X4R antagonist. The finding suggested that the ATP-P2X4R pathway may not only contribute to airway inflammation, but also to airway remodeling in models of allergic asthma in mice. The exact mechanisms of ATP-P2X4R signaling responsible for asthma pathogenesis requires further investigation.

Acknowledgments

This study was supported by the Natural Science Foundation of China (grant no. 81200011) and the Postdoctoral Science Foundation of Heilongjiang Province in China (grant no. LBH-Z12213).

References

1 

Benayoun L and Pretolani M: Airway remodeling in asthma: Mechanisms and therapeutic perspectives. Med Sci (Paris). 19:319–326. 2003. View Article : Google Scholar

2 

Uhlík J, Simůnková P, Žaloudíková M, Partlová S, Jarkovský J and Vajner L: Airway wall remodeling in young and adult rats with experimentally provoked bronchial asthma. Int Arch Allergy Immunol. 164:289–300. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Rhee CK, Kim JW, Park CK, Kim JS, Kang JY, Kim SJ, Kim SC, Kwon SS, Kim YK, Park SH and Lee SY: Effect of imatinib on airway smooth muscle thickening in a murine model of chronic asthma. Int Arch Allergy Immunol. 155:243–251. 2011. View Article : Google Scholar : PubMed/NCBI

4 

Cates C: Inhaled corticosteroids in COPD: Quantifying risks and benefits. Thorax. 68:499–500. 2013. View Article : Google Scholar

5 

Pascual RM and Peters SP: Airway remodeling contributes to the progressive loss of lung function in asthma: An overview. J Allergy Clin Immunol. 116:477–486. 2005. View Article : Google Scholar : PubMed/NCBI

6 

McParland BE, Macklem PT and Pare PD: Airway wall remodeling: Friend or foe? J Appl Physiol (1985). 95:426–434. 2003. View Article : Google Scholar

7 

James AL and Wenzel S: Clinical relevance of airway remodelling in airway diseases. Eur Respir J. 30:134–155. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Royce SG and Tang ML: The effects of current therapies on airway remodeling in asthma and new possibilities for treatment and prevention. Curr Mol Pharmacol. 2:169–181. 2009. View Article : Google Scholar : PubMed/NCBI

9 

Raissy HH, Kelly HW, Harkins M and Szefler SJ: Inhaled corticosteroids in lung diseases. Am J Respir Crit Care Med. 187:798–803. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Tagaya E and Tamaoki J: Mechanisms of airway remodeling in asthma. Allergol Int. 56:331–340. 2007. View Article : Google Scholar : PubMed/NCBI

11 

Warner SM and Knight DA: Airway modeling and remodeling in the pathogenesis of asthma. Curr Opin Allergy Clin Immunol. 8:44–48. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Roy SG, Nozaki Y and Phan SH: Regulation of alpha-smooth muscle actin gene expression in myofibroblast differentiation from rat lung fibroblasts. Int J Biochem Cell Biol. 33:723–734. 2001. View Article : Google Scholar : PubMed/NCBI

13 

Stumm CL, Halcsik E, Landgraf RG, Camara NO, Sogayar MC and Jancar S: Lung remodeling in a mouse model of asthma involves a balance between tgf-β1 and BMP-7. PloS One. 9:e959592014. View Article : Google Scholar

14 

Kesavan R, Potunuru UR, Nastasijević B, T A, Joksić G and Dixit M: Inhibition of vascular smooth muscle cell proliferation by gentiana lutea root extracts. PloS One. 8:e613932013. View Article : Google Scholar : PubMed/NCBI

15 

Zhao L, Wu J, Zhang X, Kuang H, Guo Y and Ma L: The effect of shenmai injection on the proliferation of rat airway smooth muscle cells in asthma and underlying mechanism. BMC Complement Altern Med. 13:2212013. View Article : Google Scholar : PubMed/NCBI

16 

Idzko M, Hammad H, van Nimwegen M, Kool M, Willart MA, Muskens F, Hoogsteden HC, Luttmann W, Ferrari D, Di Virgilio F, et al: Extracellular atp triggers and maintains asthmatic airway inflammation by activating dendritic cells. Nat Med. 13:913–919. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Khakh BS and North RA: P2x receptors as cell-surface ATP sensors in health and disease. Nature. 442:527–532. 2006. View Article : Google Scholar : PubMed/NCBI

18 

Lommatzsch M, Cicko S, Müller T, Lucattelli M, Bratke K, Stoll P, Grimm M, Dürk T, Zissel G, Ferrari D, et al: Extracellular adenosine triphosphate and chronic obstructive pulmonary disease. Am J Respir Crit Care Med. 181:928–934. 2010. View Article : Google Scholar : PubMed/NCBI

19 

Müller T, Vieira RP, Grimm M, Dürk T, Cicko S, Zeiser R, Jakob T, Martin SF, Blumenthal B, Sorichter S, et al: A potential role for p2×7r in allergic airway inflammation in mice and humans. Am J Respir Cell Mol Biol. 44:456–464. 2011. View Article : Google Scholar

20 

Di Virgilio F: Purines, purinergic receptors and cancer. Cancer Res. 72:5441–5447. 2012. View Article : Google Scholar : PubMed/NCBI

21 

Aymeric L, Apetoh L, Ghiringhelli F, Tesniere A, Martins I, Kroemer G, Smyth MJ and Zitvogel L: Tumor cell death and ATP release prime dendritic cells and efficient anticancer immunity. Cancer Res. 70:855–858. 2010. View Article : Google Scholar : PubMed/NCBI

22 

la Sala A, Ferrari D, Corinti S, Cavani A, Di Virgilio F and Girolomoni G: Extracellular ATP induces a distorted maturation of dendritic cells and inhibits their capacity to initiate Th1 responses. J Immunol. 166:1611–1617. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Tanaka J, Murate M, Wang CZ, Seino S and Iwanaga T: Cellular distribution of the P2X4 ATP receptor mRNA in the brain and non-neuronal organs of rats. Arch Histol Cytol. 59:485–490. 1996. View Article : Google Scholar : PubMed/NCBI

24 

Weinhold K, Krause-Buchholz U, Rödel G, Kasper M and Barth K: Interaction and interrelation of P2X7 and P2X4 receptor complexes in mouse lung epithelial cells. Cell Mol Life Sci. 67:2631–2642. 2010. View Article : Google Scholar : PubMed/NCBI

25 

Barth K and Kasper M: Membrane compartments and purinergic signalling: Occurrence and function of P2X receptors in lung. FEBS J. 276:341–353. 2009. View Article : Google Scholar

26 

Wareham K, Vial C, Wykes RC, Bradding P and Seward EP: Functional evidence for the expression of P2X1, P2X4 and P2X7 receptors in human lung mast cells. British journal of pharmacology. 157:1215–1224. 2009. View Article : Google Scholar : PubMed/NCBI

27 

Inoue K: The function of microglia through purinergic receptors: Neuropathic pain and cytokine release. Pharmacol Ther. 109:210–226. 2006. View Article : Google Scholar

28 

Ulmann L, Hirbec H and Rassendren F: P2X4 receptors mediate PGE2 release by tissue-resident macrophages and initiate inflammatory pain. EMBO J. 29:2290–2300. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Tsuda M, Kuboyama K, Inoue T, Nagata K, Tozaki-Saitoh H and Inoue K: Behavioral phenotypes of mice lacking purinergic P2X4 receptors in acute and chronic pain assays. Mol Pain. 5:282009. View Article : Google Scholar : PubMed/NCBI

30 

Jakobsson PJ: Pain: How macrophages mediate inflammatory pain via ATP signaling. Nat Rev Rheumatol. 6:679–681. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Shen HH, Xu F, Zhang GS, Wang SB and Xu WH: CCR3 monoclonal antibody inhibits airway eosinophilic inflammation and mucus overproduction in a mouse model of asthma. Acta Pharmacol Sin. 27:1594–1599. 2006. View Article : Google Scholar : PubMed/NCBI

32 

Vanacker NJ, Palmans E, Kips JC and Pauwels RA: Fluticasone inhibits but does not reverse allergen-induced structural airway changes. Am J Respir Crit Care Med. 163:674–679. 2001. View Article : Google Scholar : PubMed/NCBI

33 

Wu T, Dai M, Shi XR, Jiang ZG and Nuttall AL: Functional expression of P2X4 receptor in capillary endothelial cells of the cochlear spiral ligament and its role in regulating the capillary diameter. Am J Physiol Heart Circ Physiol. 301:H69–H78. 2011. View Article : Google Scholar : PubMed/NCBI

34 

Krugel U, Schraft T, Regenthal R, Illes P and Kittner H: Purinergic modulation of extracellular glutamate levels in the nucleus accumbens in vivo. Int J Dev Neurosci. 22:565–570. 2004. View Article : Google Scholar : PubMed/NCBI

35 

Chen K, Zhang J, Zhang W, Zhang J, Yang J, Li K and He Y: ATP-P2X4 signaling mediates NLRP3 inflammasome activation: A novel pathway of diabetic nephropathy. Int J Biochem Cell Biol. 45:932–943. 2013. View Article : Google Scholar : PubMed/NCBI

36 

Mauad T, Bel EH and Sterk PJ: Asthma therapy and airway remodeling. J Allergy Clin Immunol. 120:997–1009. 2007. View Article : Google Scholar : PubMed/NCBI

37 

Broide DH: Immunologic and inflammatory mechanisms that drive asthma progression to remodeling. J Allergy Clin Immunol. 121:560–570. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Bousquet J, Jeffery PK, Busse WW, Johnson M and Vignola AM: Asthma. From bronchoconstriction to airways inflammation and remodeling. Am J Respir Crit Care Med. 161:1720–1745. 2000. View Article : Google Scholar : PubMed/NCBI

39 

Di Virgilio F: Purinergic signalling between axons and microglia. Novartis Found Symp. 276:253–258. 2006. View Article : Google Scholar : PubMed/NCBI

40 

Donnelly-Roberts D, McGaraughty S, Shieh CC, Honore P and Jarvis MF: Painful purinergic receptors. J Pharmacol Exp Ther. 324:409–415. 2008. View Article : Google Scholar

41 

Jarvis MF: The neural-glial purinergic receptor ensemble in chronic pain states. Trends Neurosci. 33:48–57. 2010. View Article : Google Scholar

42 

Riteau N, Gasse P, Fauconnier L, Gombault A, Couegnat M, Fick L, Kanellopoulos J, Quesniaux VF, Marchand-Adam S, Crestani B, et al: Extracellular ATP is a danger signal activating P2X7 receptor in lung inflammation and fibrosis. Am J Respir Crit Care Med. 182:774–783. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Chiappara G, Gagliardo R, Siena A, Bonsignore MR, Bousquet J, Bonsignore G and Vignola AM: Airway remodelling in the pathogenesis of asthma. Curr Opin Allergy Clin Immunol. 1:85–93. 2001. View Article : Google Scholar

44 

Kudo M, Ishigatsubo Y and Aoki I: Pathology of asthma. Front Microbiol. 4:2632013. View Article : Google Scholar : PubMed/NCBI

45 

Moir LM, Leung SY, Eynott PR, McVicker CG, Ward JP, Chung KF and Hirst SJ: Repeated allergen inhalation induces phenotypic modulation of smooth muscle in bronchioles of sensitized rats. Am J Physiol Lung Cell Mol Physiol. 284:L148–L159. 2003. View Article : Google Scholar

46 

Labonté I, Hassan M, Risse PA, Tsuchiya K, Laviolette M, Lauzon AM and Martin JG: The effects of repeated allergen challenge on airway smooth muscle structural and molecular remodeling in a rat model of allergic asthma. Am J Physiol Lung Cell Mol Physiol. 297:L698–L705. 2009. View Article : Google Scholar : PubMed/NCBI

47 

Kim MJ, Turner CM, Hewitt R, Smith J, Bhangal G, Pusey CD, Unwin RJ and Tam FW: Exaggerated renal fibrosis in P2X4 receptor-deficient mice following unilateral ureteric obstruction. Nephrol Dial Transplant. 29:1350–1361. 2014. View Article : Google Scholar : PubMed/NCBI

48 

Soto F, Garcia-Guzman M, Gomez-Hernandez JM, Hollmann M, Karschin C and Stühmer W: P2X4: An ATP-activated ionotropic receptor cloned from rat brain. Proc Natl Acad Sci USA. 93:3684–3688. 1996. View Article : Google Scholar : PubMed/NCBI

49 

Kellner R, Matzkies F, Sailer D and Berg G: Potassium substitution in fasting. Z Ernahrungswiss. 16:73–76. 1977.In German. View Article : Google Scholar : PubMed/NCBI

50 

Yamamoto K, Sokabe T, Matsumoto T, Yoshimura K, Shibata M, Ohura N, Fukuda T, Sato T, Sekine K, Kato S, et al: Impaired flow-dependent control of vascular tone and remodeling in P2X4-deficient mice. Nat Med. 12:133–137. 2006. View Article : Google Scholar

51 

Ohata Y, Ogata S, Nakanishi K, Kanazawa F, Uenoyama M, Hiroi S, Tominaga S and Kawai T: Expression of P2X4R mRNA and protein in rats with hypobaric hypoxia-induced pulmonary hypertension. Circ J. 75:945–954. 2011. View Article : Google Scholar : PubMed/NCBI

52 

Said SI, Hamidi SA and Gonzalez Bosc L: Asthma and pulmonary arterial hypertension: Do they share a key mechanism of pathogenesis? Eur Respir J. 35:730–734. 2010. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2016
Volume 13 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Chen H, Xia Q, Feng X, Cao F, Yu H, Song Y and Ni X: Effect of P2X4R on airway inflammation and airway remodeling in allergic airway challenge in mice. Mol Med Rep 13: 697-704, 2016
APA
Chen, H., Xia, Q., Feng, X., Cao, F., Yu, H., Song, Y., & Ni, X. (2016). Effect of P2X4R on airway inflammation and airway remodeling in allergic airway challenge in mice. Molecular Medicine Reports, 13, 697-704. https://doi.org/10.3892/mmr.2015.4622
MLA
Chen, H., Xia, Q., Feng, X., Cao, F., Yu, H., Song, Y., Ni, X."Effect of P2X4R on airway inflammation and airway remodeling in allergic airway challenge in mice". Molecular Medicine Reports 13.1 (2016): 697-704.
Chicago
Chen, H., Xia, Q., Feng, X., Cao, F., Yu, H., Song, Y., Ni, X."Effect of P2X4R on airway inflammation and airway remodeling in allergic airway challenge in mice". Molecular Medicine Reports 13, no. 1 (2016): 697-704. https://doi.org/10.3892/mmr.2015.4622