Open Access

Characterization of the human mucin 5AC promoter and its regulation by the histone acetyltransferase P300

  • Authors:
    • Shiyao Xu
    • Yu Hui
    • Jin Shu
    • Jun Qian
    • Ling Li
  • View Affiliations

  • Published online on: January 9, 2019     https://doi.org/10.3892/ijmm.2019.4054
  • Pages: 1263-1270
  • Copyright: © Xu 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

Histone acetylation is important in the modification of gene transcription in asthma and is regulated by histone acetyltransferases (HATs). P300 (P300 HAT) is an enzyme that is able to acetylate a wide variety of proteins. The modification of core histones can further regulate gene transcription, cell proliferation and other cell processes. Airway mucus hypersecretion is one of the most serious pathophysiological symptoms of chronic airway inflammatory diseases, and the human mucin 5AC (MUC5AC) gene has been reported to be a major component of respiratory secretions related to asthma and chronic obstructive pulmonary disease. In the present study, the 5' sequence of the human MUC5AC gene with a 1,348‑bp DNA sequence was amplified from human A549 cells genomic DNA by polymerase chain reaction (PCR), and the product of the PCR was sequenced. By promoter deletion analysis, five promoter segments with different lengths were amplified by PCR. The products were identified by DNA sequencing and the six promoter segments were inserted into pGL3‑enhancer vectors. The core promoter area was identified with a series of 5' deletion promoter plasmids using luciferase reporter assays. MUC5AC promoter activity, and the mRNA and protein expression levels of MUC5AC were observed in P300 wild‑type, P300 mutant, P300 small interfering RNA and P300 control groups. The results showed that the core promoter area of MUC5AC was located within the ‑935/+48 region and that P300 reduced the expression of MUC5AC in A549 cells.

Introduction

Asthma is a chronic airway disease that affects >300,000,000 individuals worldwide (1). The prevalence of atopy and asthma has markedly increased in developing countries in recent years (2). The annual worldwide mortality rate of asthma has been reported to be ~250,000 and it is also a substantial economic burden (1).

Failure to clear bronchial secretions is the main pathological reason for asthmatic lung disease. The primary cause for asthma-associated mortality is intraluminal airway asphyxiation of mucus plugs (3,4). Defective mucociliary clearance is observed even in mild stable asthma (5-7). A decrease in clearance causes acute exacerbation (8).

In total, 21 genes are reported to encode mucins in the human genome. Mucin 5AC (MUC5AC) is expressed at high levels in the airway system (9,10). The expression of MUC5AC varies in airway diseases, including asthma, with an increase in the number of goblet cells. Mucus may alter the normal structure and status of goblet cells after failing to incorporate with MUC5AC. Without the normal reaction between MUC5AC and mucus, the airway viscoelasticity becomes vulnerable to plugging (11-13).

Current evidence shows that MUC5AC is regulated by several factors. Previous studies have found that interleukin-13 can increase the expression of MUC5AC in epithelial cell lines and in murine models (14-17). Epidermal growth factor receptor (EGFR) signaling increases the expression of MUC5AC and the expression of MUC5AC can be inhibited with an EGFR tyrosine kinase inhibitor (17). Previous studies have also shown that toll-like receptor 2 is related to the expression of MUC5AC in asthmatic models (18) and corticosteroids have been demonstrated to decrease mucus in airways and further decrease the expression of MUC5AC (19).

In the pathogenesis of asthma, epigenetic modifications are important environmental and genetic factors. In particular, the reversible process of histamine acetylation is mediated by histone acetyltransferases (HATs) and histone decarboxylases (HDACs); histone acetylation is closely associated with gene transcription and controlled by HATs.

P300 (EP300 or KAT3B) consists of 2,414 amino acids and its full length is 300 kDa. It was first cloned in 1994 (20,21) in studies aimed at identifying proteins that bind E1A, an adenoviral oncogenic transcription factor. P300 was found to have HAT activity and to acetylate a number of proteins (22). Studies have shown that the gain-of-function in P300 can be induced by mutation, the overexpression of P300 or several disease etiology conditions, such as diabetes, obesity, thrombocytopenia or hypertrophic heart diseases. Although, a P300 inhibitor has not been used in clinical trials (23), P300 inhibitors are considered potential therapeutic applications in several diseases.

In the present study, it was hypothesized that P300 may suppress the production of MUC5AC. The MUC5AC promoter was constructed and its activity was investigated with dual-luciferase assays. The study then observed the effects of the HAT protein P300 on the transcription of MUC5AC in adenocarcinomic human alveolar basal epithelial cells and examined the associated mechanisms.

Materials and methods

Cell culture

The A549 lung carcinoma cell line and Beas-2b cells were cultured in Dulbecco's modified Eagle's medium with 10% fetal bovine serum (both Thermo Fisher Scientific, Inc., Waltham, MA, USA) and antibiotics (50 U/ml penicillin and 50 µg/ml streptomycin). The cells were maintained at 37°C in a 5% CO2 humidified chamber.

Plasmids and small interfering RNA (siRNA)

The DNA sequence (-1,300 to +48) of the MUC5AC promoter region was amplified by polymerase chain reaction (PCR) and digested with KpnI and BglII (Thermo Fisher Scientific, Inc.). PCR was performed by using genomic DNA which was extracted from the A549 cells using a TIANamp Genomic DNA kit (Tiangen Biotech Co., Ltd., Beijing, China), DNA polymerase (LA Taq), dNTPs, GC Buffer (Thermo Fisher Scientific, Inc.) and forward and reverse primers. The forward primers contained a KpnI restriction site, and the reverse primer contained a BglII site. Sequences of primers were as follows: Forward, 5′-CGGGGTACCCTACCCATTCACATTTTCCCCATCC-3′ and reverse, 5′-GGAAGATCTGGGACCAAGCTGAGCTCTGC-3′. PCR was performed with the following thermocycling conditions: 94°C for 5 min followed by 30 cycles of amplification (94°C, 30 sec; 60°C, 30 sec; 72°C, 2 min) and followed by a final extension at 72°C for 10 min. Subsequently, it was subcloned into the promoter-less luciferase expression plasmid, pGL3-Basic (Promega Corporation, Madison, WI, USA). The resulting plasmid was termed pGL-1300/+48. Truncated plasmids of the MUC5AC promoter were constructed using pGL-1300/+48 as a template. KpnI and XhoI double digestions were performed on the MUC5AC promoter fragments of different lengths obtained from the above PCR reaction, and pGL3-enhancer vector. The vector and the target fragments were linked using a T4 ligase; competent DH5α cells (Tiangen Biotech Co., Ltd.) were transformed, positive clones were selected, and verification was conducted using restriction endonuclease analysis and DNA sequencing. The successfully constructed plasmids were named pGL-1300/+48, P2: pGL-935/+48, P3: pGL-583/+48, P4: pGL-205/+48, P5: pGL-116/+48, and P6: pGL-23/+48. The siRNAs were synthesized and purified (Shanghai GenePharma, Co. Ltd., Shanghai, China). The targeted sequence was designed to silence P300 gene transcription with the sequence 5′-GUCCUGGAUUAGGUUUGAUTT-3′. The control siRNA sequence was 5′-AUCAAACCUAAUCCAGGACTT-3′.

Transient transfection and luciferase assays

The p300 mutant (p300 mut; P300Δ1472-1522) and wild-type (p300 wt) plasmids were provided by Professor Zhou Guoping (Fourth School of Clinical Medicine, Nanjing Medical University, Nanjing, China) and were originally described by Boyes et al (24). siRNA transfection in the A549 cells was performed using Lipofectamine® 3000 (Invitrogen; Thermo Fisher Scientific, Inc.), according to the manufacturer's protocol. Transient transfection was performed following 24 h cell adaptation on 96-well plates (1.5×104 cells/well).

In the ectopic overexpression experiments, the cells were co-transfected with the P300 wt or P300 mut plasmids, and the pGL-935/+48 reporter plasmid. Following transfection for 24 h, the cells were collected for the luciferase assays. For the P300 siRNA and P300 overexpression experiments, reporter plasmids containing siRNA for P300 or the P300-overexpression plasmid were co-transfected into A549 cells and harvested after 24 h. Subsequently, the luciferase activity was measured with the Dual Reporter assay system (Promega Corporation). All experiments were conducted independently in triplicate.

RNA extraction and reverse transcription-quantitative (RT-q)PCR analysis

RT-qPCR analysis was performed to confirm the expression of MUC5AC in A549 cells. Following transfection for 24 h, the total R NA was extracted from the A549 cells using TRIzol®. The RT-qPCR analysis was performed with the SYBR PCR Master mix (Applied Biosystems; Thermo Fisher Scientific, Inc.) and ABI 7500 Fast system (Applied Biosystems; Thermo Fisher Scientific, Inc.). The primers were synthesized by Changzhou Bo Hong Biological Engineering Co., Ltd. (Changzhou, China). MUC5AC primer sequences were as follows: Forward, 5′-CTGTGAAGGTGGCTGACCAAGA3′ and reverse, 5′AAGGTGTAGTAGGTGCCGTCGAA-3′; GAPDH was selected as the control reference with the following primer sequences: Forward, 5′-TGGTATCGTGGAAGGACTCATGAC3′ and reverse, 5′TGCCAGTGAGCTTCCCGTTCAGC-3′. cDNA was reverse transcribed by primers using the PrimeScript RT reagent kit with gDNA Eraser (Perfect Real Time) according to the manufacturer's protocol. The reverse transcription cDNA products were stored at -20°C for PCR amplification. According to the instruction of the SYBR Premix Ex Taq II (Tli RnaseH Plus), the following reagents were added to the PCR reaction mixture: 2 µl cDNA template, 0.8 µl forward primer (10 µM), 0.8 µl reverse primer (10 µM), 10 µl SYBR® Premix EX Taq™ II (2X), 0.4 µl ROX Reference Dye II (50X) and 6 µl ddH2O to make a 20-µl total reaction system. Detection and quantification were performed as follows: Pre-denaturation, 95°C for 30 sec; 40 cycles of denaturation at 95°C for 5 sec; and extension, 60°C for 34 sec. Fluorescence data was collected at the extension step. The relative expression of the MUC5AC gene was analyzed with the 2−ΔΔCq method (25).

Immunofluorescence

The A549 cells were seeded on glass coverslips in 6-well plates (5×104 cells/well). When the cells reached ~50% confluence, the cells were washed twice with PBS and fixed with 4% paraformaldehyde. Prior to staining with antibodies for immunofluorescence, the cells were blocked with 0.2% Triton, 1% bovine serum albumin and 1% goat serum (Sangon Biotech, Co., Ltd., Shanghai, China) for 30 min at room temperature. Following blocking, the fixed cells were washed twice with PBS and incubated with MUC5AC primary antibody (EPR16904; cat. no. ab198294; 1:250) rabbit anti human monoclonal antibody (Abcam, Cambridge, UK) at 4°C in a humid chamber overnight. The following morning, the cells were incubated with fluorescein isothiocyanate-labeled goat anti-rabbit immunoglobulin G (cat. no. ab150077; 1:500; Abcam) for 1 h at room temperature and washed three times prior to observation under a confocal laser fluorescent microscope (Zeiss 710; Carl Zeiss AG, Oberkochen, Germany).

Statistical analysis

Statistical analysis was performed with SPSS software (version 12.0; SPSS, Inc., Chicago, IL, USA). All data are presented as the mean ± standard deviation. Student's t-test was used to analyze the statistical significance between groups. Differences between multiple groups were tested by one-way analysis of variance followed by Tukey's post hoc test. P<0.05 was considered to indicate a statistically significant difference.

Results

Identification of recombinant plasmid pGL-1300/+48

The pGL-1300/+48 gene fragment was amplified by PCR. The product and the pGL vector were digested using restriction endonucleases BglII and KpnI. The recombinant plasmid pGL-1300/+48 was constructed by connecting the digested products with DNA ligase. The recombinant plasmid was then identified by PCR. The endonuclease digestion result is presented in Fig. 1. The PCR product and the digested product had a size of ~1,350 bp, as expected. The results indicated that the recombinant plasmid pGL-1300/+48 was successfully constructed. Through agarose gel electrophoresis, the PCR products showed bands at ~983, 631, 253, 164 and 71 bp (Fig. 2). The electrophoresis results of the PCR products were as predicted.

Verification of the recombinant luciferase reporter plasmid

The electrophoresis results of the recombinant luciferase reporter plasmid containing the MUC5AC promoter fragments with restriction enzyme digestions are shown in Fig. 2. The results were as expected, and further sequencing was conducted. The sequencing results were consistent with the designed DNA fragment sequences, which further confirmed the successful construction of a recombinant plasmid of MUC5AC promoter fragments (Fig. 3).

Transcription activation activity of the expression of MUC5AC

The reporter gene plasmids containing regulatory sequences of different lengths of the human MUC5AC promoter region were co-transfected with the internal control plasmid -TK into A549 and Beas-2b cells, respectively. The transfected cells were stimulated for 30 min, and the specific luciferase activity was detected. The results showed that the luciferase activity of the recombinant pGL-MUC5AC-935/+48 plasmid was significantly induced (Fig. 4A and B).

mRNA expression of P300

To demonstrate transfection success, the empty vector control, the P300 expression plasmid (P300 wt) and the P300 siRNA, in addition to the control plasmids, were transfected into A549 cells respectively. The transfected cells were stimulated for 30 min and the mRNA expression of P300 was detected. The results showed that P300 wt markedly increased the mRNA expression of P300, whereas P300 siRNA inhibited the expression of P300 (Fig. 5A and B).

P300 inhibits MUC5AC promoter activity

In order to determine how P300 regulates MUC5AC promoter activity, the MUC5AC plasmid pGL-935/+48, the empty vector control, P300 wt, P300 siRNA, and the control plasmids were co-transfected into A549 cells, respectively (Fig. 6). By overexpressing P300, the promoter activity was significantly decreased in the A549 cells (Fig. 6A). However, P300 siRNA markedly increased the promoter activity of MUC5AC (Fig. 6B). Taken together, P300 inhibited the expression of MUC5AC by suppressing its promoter activity.

P300 decreases the mRNA level of MUC5AC

To determine whether P300 affects MUC5AC gene transcription, the co-transfected A549 cell RNA was isolated and the expression of MUC5AC was detected by RT-qPCR analysis (Fig. 7A and B). The mRNA expression of MUC5AC was significantly decreased following co-transfection with the P300 wt expression plasmid compared with the control. By contrast, P300 siRNA increased the mRNA levels of MUC5AC, which suggests that P300 decreased the mRNA expression of MUC5AC.

P300 decreases the protein expression level of MUC5AC

The empty vector, untransfected control, the P300 expression plasmid and P300 siRNA, together with their control plasmids, were transfected into A549 cells. Immunofluorescence was then performed to detect the protein expression of MUC5AC in each group. The A549 cells were stained with immunofluorescent MUC5AC (green) antibody, and the nuclei were stained with DAPI (blue). The results showed that the P300 expression plasmid downregulated the protein levels of MUC5AC, whereas P300 siRNA upregulated the protein levels of MUC5AC (Fig. 8).

Discussion

Mucosal airway hypersecretion is one of the most serious pathophysiological features of chronic airway inflammatory diseases. In cystic fibrosis, the thick and dehydrated airway mucus, which is difficult to clear, makes patients prone to chronic inflammation and bacterial infections. It is beneficial to reduce the impact of lung infections and improve lung function by clearing the airways of mucus (26). Excessive mucus production and secretion in airways are associated with disabling symptoms (cough and sputum), decline in lung function, exacerbations and mortality in patients with chronic obstructive pulmonary disease (COPD) (27). With an increasing number of individuals suffering from mucosal airway hypersecretion, it is becoming increasingly urgent to develop an effective treatment. It is well known that mucus plugging is the main cause of acute asthma (3). In patients with asthma, bronchial secretion deposits are frequently observed (28). At present, anticholinergics, corticosteroids or other medicines are applied to decrease mucus hypersecretion in the clinical setting. However, these treatments have a number of side effects. Therefore, attempts to identify novel efficient therapeutic methods for airway inflammatory diseases are ongoing (29).

Mucins are high-molecular-weight glycoproteins secreted by goblet cells, which cover the airways, the esophagus and other glandular organs (30). At present, 21 mucin-like genes have been identified and sorted into two categories; membrane-bound and secreted mucins (31). Secreted mucins have terminal cysteine-rich domains and form disulfide bonds for polymers that may be detected on gels (9,10,32).

MUC5AC and MUC5B are two typical polymers of airway mucins (9,10). MUC5B, a tenacious secretory mucin, is produced in the lung airway system in normal and pathological conditions. MUC5AC is also a gel-forming mucin and secreted at high levels in asthmatic inflammation or COPD. Increasing evidence has demonstrated that MUC5AC may have a less important role in human airway systems in normal conditions. Therefore, suppression of the production of MUC5AC may be a therapeutic method for secretory chronic airway inflammatory diseases (4). In our previous studies, mucosal airway hypersecretion was investigated in A549 lung adenocarcinoma cells, human NCI-H292 airway epithelial cells and normal human bronchial epithelial (NHBE) cells (33-35). In the present study, A549 alveolar only basal epithelial cells were used to investigate mucosal airway hypersecretion, owing to their high stability, medium culture conditions and low cost. Furthermore, this cell line has been investigated widely and it is known to express MUC5AC, MUC1 and MUC5B (36).

Histone acetylation, particularly by HATs, is essential for gene transcription and protein expression in asthmatic diseases. The decreased activity of HDACs is likely to cause increased secretion of inflammatory factors, thus inducing asthma (37). P300 has HAT activity, which can acetylate a number of proteins that regulate lung function and glandular secretion. Additionally, P300 exerts its effect through its binding domains with different proteins. P300 is able to interact with >411 proteins, to induce a signal response or influence gene expression. In addition, P300 and other acetyltransferases can influence protein-coupled structures by protein lysine acetylation. P300 is involved in the integration of protein complexes and regulating DNA element functions (3).

In the present study, the MUC5AC gene promoter was cloned and a luciferase reporter with different lengths was successfully constructed. The results demonstrated that the core promoter area was in the region of -935 to -583 bp upstream of the MUC5AC gene. In addition, the expression of MUC5AC was reduced by P300. Through a literature review and analyses using relevant software, it was demonstrated that the region of the core promoter contained nuclear factor (NF)-κB, and transcription factor Sp1 (Sp1) binding sites. Each binding site can bind with different transcription factors, which influence MUC5AC gene transcription (38). There are several studies showing that Sp1 and NF-κb (39,40) are important ubiquitous transcription factors, which regulate the transcription of MUC5AC and mucous metaplasia by interacting with gene promoters (19). Furthermore, studies have shown that glucocorticoids can influence gene expression and activity by integrating with the ligand-activated glucocorticoid receptor and activating glucocorticoid-responsive elements (GREs) in the promoter region. The MUC5AC gene also has GRE regions in its 5′ sequence and may be similarly influenced by glucocorticoids. All signals influencing P300 (activity, structure or recruitment), may have an impact on gene transcription to further affect the gene expression of MUC5AC. P300 is ubiquitously expressed in multiple tissues/organs and is involved in numerous physiological processes. Studies on P300 have mainly focused on viral infections, cancer and neurodegenerative diseases (41). Consistent with these diseases, P300 is important in mucosal airway hypersecretion. Differences in the mechanisms of P300 in these diseases remain to be elucidated. Therefore, the task of investigating the clinical applicability of targeting the expression of P300 as a potential treatment for mucosal airway hypersecretion involves understanding how the protein receives signals in cells, what induces its recruitment in a given signal transduction pathway, and what determines the final outcome of its individual activity. Therefore, further investigations are required focusing on the pathways that regulate the P300 pathway, and investigating the effects of MUC5AC in lung inflammatory secretion diseases.

In conclusion, the findings of the present study indicate that P300 inhibited the gene expression of MUC5AC in A549 cells, which may be a novel therapeutic target for chronic airway inflammatory diseases. However, further investigations are required to identify the signal pathways influencing P300.

Funding

The present study was supported by the Medical Innovation Team of Jiangsu Province (grant no. CXTDB2017016), the Major Program of Wuxi Health and Family Planning Commission (grant no. Z201606) and the General Program of Wuxi Municipal Health Bureau (grant no. MS201506).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

SX carried out the experiments, collected and analyzed the data and wrote the manuscript. YH designed the experimental study and assisted with revising the manuscript. JS was involved in experiments and analysis. LL and JQ evaluated the data, and revised and edited the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

The authors would like to thank Professor Joan Boyes (The Institute of Cancer Research, London, UK) for providing the P300 wt plasmid and P300 mut plasmid (P300mut-P300Δ1472-1522), and Professor Zhou Guoping (The Fourth School of Clinical Medicine, Nanjing Medical University) for providing the A549 cells.

References

1 

Masoli M, Fabian D, Holt S, Beasley R and Global Initiative; for Asthma (GINA) Program: The global burden of asthma: Executive summary of the GINA dissemination committee report. Allergy. 59:469–478. 2004. View Article : Google Scholar : PubMed/NCBI

2 

D'Amato G, Holgate ST, Pawankar R, Ledford DK, Cecchi L, Al-Ahmad M, Al-Enezi F, Al-Muhsen S, Ansotegui I, Baena-Cagnani CE, et al: Meteorological conditions, climate change, new emerging factors, and asthma and related allergic disorders. A statement of the world allergy organization. World Allergy Organ J. 8:252015.PubMed/NCBI

3 

Kuyper LM, Paré PD, Hogg JC, Lambert RK, Ionescu D, Woods R and Bai TR: Characterization of airway plugging in fatal asthma. Am J Med. 115:6–11. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Evans CM, Kim K, Tuvim MJ and Dickey BF: Mucus hypersecretion in asthma: Causes and effects. Curr Opin Pulm Med. 15:4–11. 2009. View Article : Google Scholar :

5 

Bateman JR, Pavia D, Sheahan NF, Agnew JE and Clarke SW: Impaired tracheobronchial clearance in patients with mild stable asthma. Thorax. 38:463–467. 1983. View Article : Google Scholar : PubMed/NCBI

6 

Agnew JE, Bateman JR, Pavia D and Clarke SW: Radionuclide demonstration of ventilatory abnormalities in mild asthma. Clin Sci (Lond). 66:525–531. 1984. View Article : Google Scholar

7 

O'Riordan TG, Zwang J and Smaldone GC: Mucociliary clearance in adult asthma. Am Rev Respir Dis. 146:598–603. 1992. View Article : Google Scholar : PubMed/NCBI

8 

Messina MS, O'riordan TG and Smaldone GC: Changes in mucociliary clearance during acute exacerbations of asthma. Am Rev Respir Dis. 143:993–997. 1991. View Article : Google Scholar : PubMed/NCBI

9 

Thornton DJ, Rousseau K and McGuckin MA: Structure and function of the polymeric mucins in airways mucus. Annu Rev Physiol. 70:459–486. 2008. View Article : Google Scholar

10 

Rose MC and Voynow JA: Respiratory tract mucin genes and mucin glycoproteins in health and disease. Physiol Rev. 86:245–278. 2006. View Article : Google Scholar

11 

Bonser LR and Erle DJ: Airway mucus and asthma: The role of MUC5AC and MUC5B. J Clin Med. 6:E1122017. View Article : Google Scholar : PubMed/NCBI

12 

Jeffery PK and Li D: Airway mucosa: Secretory cells, mucus and mucin genes. Eur Respir J. 10:1655–1662. 1997. View Article : Google Scholar : PubMed/NCBI

13 

Woodruff PG, Modrek B, Choy DF, Jia G, Abbas AR, Ellwanger A, Koth LL, Arron JR and Fahy JV: T-helper type 2-driven inflammation defines major subphenotypes of asthma. Am J Respir Crit Care Med. 180:388–395. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Bonser LR, Zlock L, Finkbeiner W and Erle DJ: Epithelial tethering of MUC5AC-rich mucus impairs mucociliary transport in asthma. J Clin Invest. 126:2367–2371. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Kuperman DA, Lewis CC, Woodruff PG, Rodriguez MW, Yang YH, Dolganov GM, Fahy JV and Erle DJ: Dissecting asthma using focused transgenic modeling and functional genomics. J Allergy Clin Immunol. 116:305–311. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Zhen G, Park SW, Nguyenvu LT, Rodriguez MW, Barbeau R, Paquet AC and Erle DJ: IL-13 and epidermal growth factor receptor have critical but distinct roles in epithelial cell mucin production. Am J Respir Cell Mol Biol. 36:244–253. 2007. View Article : Google Scholar

17 

Takeyama K, Dabbagh K, Lee HM, Agustí C, Lausier JA, Ueki IF, Grattan KM and Nadel JA: Epidermal growth factor system regulates mucin production in airways. Proc Natl Acad Sci USA. 96:3081–3086. 1999. View Article : Google Scholar : PubMed/NCBI

18 

Kraft M, Adler KB, Ingram JL, Crews AL, Atkinson TP, Cairns CB, Krause DC and Chu HW: Mycoplasma pneumoniae induces airway epithelial cell expression of MUC5AC in asthma. Eur Respir J. 31:43–46. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Chen Y, Nickola TJ, DiFronzo NL, Colberg-Poley AM and Rose MC: Dexamethasone-mediated repression of MUC5AC gene expression in human lung epithelial cells. Am J Respir Cell Mol Biol. 34:338–347. 2006. View Article : Google Scholar

20 

Bannister AJ and Kouzarides T: The CBP co-activator is a histone acetyltransferase. Nature. 384:641–643. 1996. View Article : Google Scholar : PubMed/NCBI

21 

Ogryzko VV, Schiltz RL, Russanova V, Howard BH and Nakatani Y: The transcriptional coactivators P300 and CBP are histone acetyltransferases. Cell. 87:953–959. 1996. View Article : Google Scholar : PubMed/NCBI

22 

Ito K, Caramori G, Lim S, Oates T, Chung KF, Barnes PJ and Adcock IM: Expression and activity of histone deacetylases (HDAC) in human asthmatic airways. Am J Respir Crit Care Med. 166:392–396. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Dancy BM and Cole PA: Protein lysine acetylation by P300/CBP. Chem Rev. 115:2419–2452. 2015. View Article : Google Scholar : PubMed/NCBI

24 

Boyes J, Byfield P, Nakatani Y and Ogryzko V: Regulation of activity of the transcription factor GATA-1 by acetylation. Nature. 396:594–598. 1998. View Article : Google Scholar : PubMed/NCBI

25 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar

26 

Martin C, Frija-Masson J and Burgel PR: Targeting mucus hyper-secretion: New therapeutic opportunities for COPD? Drugs. 74:1073–1089. 2014. View Article : Google Scholar : PubMed/NCBI

27 

Button BM and Button B: Structure and function of the mucus clearance system of the lung. Cold Spring Harb Perspect Med. 3:a0097202013. View Article : Google Scholar : PubMed/NCBI

28 

Dunnill MS: The pathology of asthma, with special reference to changes in the bronchial mucosa. J Clin Pathol. 13:27–33. 1960. View Article : Google Scholar : PubMed/NCBI

29 

Rogers DF and Barnes PJ: Treatment of airway mucus hypersecretion. Ann Med. 38:116–125. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Di Valentin E, Crahay C, Garbacki N, Hennuy B, Guéders M, Noël A, Foidart JM, Grooten J, Colige A, Piette J and Cataldo D: New asthma biomarkers: Lessons from murine models of acute and chronic asthma. Am J Physiol Lung Cell Mol Physiol. 296:L185–L197. 2009. View Article : Google Scholar

31 

Yu H, Li Q, Kolosov VP, Perelman JM and Zhou X: Interlenkin-13 induces mucin 5AC production involving STAT6/SPDEF in human airway epithelial cells. Cell Commun Adhes. 17:83–92. 2010. View Article : Google Scholar

32 

Hattrup CL and Gendler SJ: Structure and function of the cell surface (tethered) mucins. Annu Rev Physiol. 70:431–457. 2008. View Article : Google Scholar

33 

Lee JW, Kim YI, Im CN, Kim SW, Kim SJ, Min S, Joo YH, Yim SV and Chung N: Grape seed proanthocyanidin inhibits mucin synthesis and viral replication by suppression of AP-1 and NF-κB via p38 MAPKs/JNK signaling pathways in respiratory syncytial virus-infected A549 cells. J Agric Food Chem. 65:4472–4483. 2017. View Article : Google Scholar : PubMed/NCBI

34 

Al-Sawalha N, Pokkunuri I, Omoluabi O, Kim H, Thanawala VJ, Hernandez A, Bond RA and Knoll BJ: Epinephrine activation of the β2-adrenoceptor is required for IL-13-induced mucin production in human bronchial epithelial cells. PLoS One. 10:e01325592015. View Article : Google Scholar

35 

Song WY, Song YS, Ryu HW, Oh SR, Hong J and Yoon DY: Tilianin inhibits MUC5AC expression mediated via down-regulation of EGFR-MEK-ERK-Sp1 signaling pathway in NCI-H292 human airway cells. J Microbiol Biotechnol. 27:49–56. 2017. View Article : Google Scholar

36 

Berger JT, Horger T and Voynow JA: Respiratory carcinoma cell lines express MUC5/5AC mRNA and secrete MUC5/5AC mucins(C). In: 5th International Workshop on Carcinoma Associated Mucins; Cambridge, UK. 1998;

37 

Cui ZL, Gu W, Ding T, Peng XH, Chen X, Luan CY, Han RC, Xu WG and Guo XJ: Histone modifications of Notch1 promoter affect lung CD4+T cell differentiation in asthmatic rats. Int J Immunopathol Pharmacol. 26:371–381. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Hewson CA, Edbrooke MR and Johnston SL: PMA induces the MUC5AC respiratory mucin in human bronchial epithelial cells, via PKC, EGF/TGF-alpha, Ras/Raf, MEK, ERK and Sp1-dependent mechanisms. J Mol Biol. 344:683–695. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Perrais M, Pigny P, Copin MC, Aubert JP and Van Seuningen I: Induction of MUC2 and MUC5AC mucins by factors of the epidermal growth factor (EGF) family is mediated by EGF receptor/Ras/Raf/extracellular signal-regulated kinase cascade and Sp1. J Biol Chem. 277:32258–32267. 2002. View Article : Google Scholar : PubMed/NCBI

40 

Chen Y, Garvin LM, Nickola TJ, Watson AM, Colberg-Poley AM and Rose MC: IL-1β induction of MUC5AC gene expression is mediated by CREB and NF-κB and repressed by dexamethasone. Am J Physiol Lung Cell Mol Physiol. 306:L797–L807. 2014. View Article : Google Scholar : PubMed/NCBI

41 

Janknecht R: The versatile functions of the transcriptional coactivators P300 and CBP and their roles in disease. Histol Histopathol. 17:657–668. 2002.PubMed/NCBI

Related Articles

Journal Cover

March-2019
Volume 43 Issue 3

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xu S, Hui Y, Shu J, Qian J and Li L: Characterization of the human mucin 5AC promoter and its regulation by the histone acetyltransferase P300. Int J Mol Med 43: 1263-1270, 2019
APA
Xu, S., Hui, Y., Shu, J., Qian, J., & Li, L. (2019). Characterization of the human mucin 5AC promoter and its regulation by the histone acetyltransferase P300. International Journal of Molecular Medicine, 43, 1263-1270. https://doi.org/10.3892/ijmm.2019.4054
MLA
Xu, S., Hui, Y., Shu, J., Qian, J., Li, L."Characterization of the human mucin 5AC promoter and its regulation by the histone acetyltransferase P300". International Journal of Molecular Medicine 43.3 (2019): 1263-1270.
Chicago
Xu, S., Hui, Y., Shu, J., Qian, J., Li, L."Characterization of the human mucin 5AC promoter and its regulation by the histone acetyltransferase P300". International Journal of Molecular Medicine 43, no. 3 (2019): 1263-1270. https://doi.org/10.3892/ijmm.2019.4054