Curcumin inhibits the proliferation of airway smooth muscle cells in vitro and in vivo

  • Authors:
    • Xin Zeng
    • Ying Cheng
    • Yuejun Qu
    • Jide Xu
    • Zhiyuan Han
    • Tingting Zhang
  • View Affiliations

  • Published online on: June 27, 2013     https://doi.org/10.3892/ijmm.2013.1425
  • Pages: 629-636
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The inhibition of the proliferation of airway smooth muscle cells (ASMCs) is crucial for the prevention and treatment of asthma. Recent studies have revealed some important functions of curcumin; however, its effects on the proliferation of ASMCs in asthma remain unknown. Therefore, in this study, we performed in vitro and in vivo experiments to investigate the effects of curcumin on the proliferation of ASMCs in asthma. The thickness of the airway wall, the airway smooth muscle layer, the number of ASMCs and the expression of extracellular signal-regulated kinase (ERK) were significantly reduced in the curcumin-treated group as compared with the model group. Curcumin inhibited the cell proliferation induced by platelet-derived growth factor (PDGF) and decreased the PDGF-induced phosphorylation of ERK1/2 in the rat ASMCs. Moreover, the disruption of caveolae using methyl-β-cyclodextrin (MβCD) attenuated the anti-proliferative effects of curcumin in the ASMCs, which suggests that caveolin is involved in this process. Curcumin upregulated the mRNA and protein expression of caveolin-1. The data presented in this study demonstrate that the proliferation of ASMCs is inhibited by curcumin in vitro and in vivo; curcumin exerts these effects by upregulating the expression of caveolin-1 and blocking the activation of the ERK pathway.

Introduction

Asthma is a chronic inflammatory disorder of the airways in which many cells and cellular elements play an important role (1). Patients with severe asthma with an incidence rate of 5–10% account for the high morbidity and health costs; thus, novel treatment strategies for this disorder are urgently required (2). The airway obstruction in asthma is usually reversible; however, if left untreated, the obstruction may become irreversible due to airway remodeling (35). The process of airway remodeling in asthma includes subepithelial basement membrane fibrosis, epithelial goblet cell hyperplasia, an increase in the number of blood vessels and a proliferative state of the airway smooth muscle with increased mass due to hyperplasia and hypertrophy (5). Recent studies have indicated that the increased mass of airway smooth muscle cells (ASMCs) plays a critical role in the histopathological characteristics of airway remodeling (24). Thus, this may be a potential therapeutic target for asthma. Several classes of drugs that target the airway smooth muscle, including β-agonists, anti-cholinergics, antihistamines and anti-leukotrienes were developed decades ago (6). However, these life-long therapies only treat the symptoms, but have little or no effect on the structural alterations in asthma.

Curcumin [diferuloylmethane (C21H20O6)], a polyphenol, is derived from the roots (rhizomes) of the plant, Curcuma longa (7). Curcumin has been used as a traditional medicine for the treatment of asthma in China and India since 1900 BC (8,9). Several studies have demonstrated that curcumin has wide range of properties, including anti-inflammatory, anti-oxidant, anticancer, chemopreventive and chemotherapeutic activities (7,912), affecting multiple signaling pathways, such as the extracellular signal-regulated kinase (ERK)1/2 pathway (1216). The anti-proliferative effects of curcumin have been investigated in various cell lines (1720). Studies have shown that curcumin inhibits vascular smooth muscle cell (VSMC) proliferation by restoring the expression of caveolin-1, blocking the activation of ERK1/2 (13,15,2123). However, to our knowledge, whether curcumin inhibits ASMC proliferation in asthma has not been reported to date.

In this study, we investigated whether curcumin inhibits the proliferation of ASMCs in vitro and in vivo. Our results demonstrate that curcumin inhibits the proliferation of ASMCs by upregulating the expression of caveolin-1 and thus blocking the ERK signaling pathway; these results may promote the clinical application of curcumin in asthma.

Materials and methods

Animals

Eighty female BALB/c mice, weighing approximately 18–22 g, were purchased from the Guangdong Medical Laboratory Animal Center, Foshan, China. The mice were divided into 5 groups (n=16 per group): the control group (normal saline-challenged mice treated with normal saline), model group [ovalbumin (OVA)-challenged mice treated with normal saline] and the curcumin-treated groups (OVA-challenged mice treated with 50, 100 and 150 mg/kg curcumin). All mice were kept in well-controlled animal housing facilities, and had free access to tap water and food pellets throughout the experimental period. All experimental procedures involving animals were carried out in accordance with the Guide for the Care and Use of Laboratory Animals and the Institutional Ethical Guidelines for experiments with animals.

Sensitization and challenge with OVA

Mice underwent OVA sensitization and challenge as previously described (24,25) with slight modifications. Mice in the model and curcumin-treated groups were immunized by an intraperitoneal (i.p.) injection of 0.2 ml of 50 μg/ml OVA (Sigma-Aldrich Co., St. Louis, MO, USA) on days 0 and 12. The control group received the same volume of physiological saline at the same time. Mice in the model and curcumin-treated groups were then challenged once a day with 5% OVA (aerosolized for 30 min) via the airways between days 18 and 23, while normal saline was administered to the control group in a similar manner. Prolonged inflammation was induced by the subsequent exposure of mice to aerosolized 5% OVA 3 times a week for 30 min from day 26 onwards (chronic phase). Mice in the curcumin-treated groups were administered with an i.p. injection of curcumin 30 min prior to stimulation with OVA, while the others were administered normal saline. The mice were sacrificed on day 55.

Airway hyper-responsiveness (AHR)

The activity of all the mice was observed closely following challenge with OVA. Airway responsiveness was measured indirectly by whole body plethysmography to calculate enhanced pause (Penh: Buxco Technologies, Petersfield, UK). Response to inhaled methacholine (Sigma-Aldrich Co.) at concentrations of 0.78, 1.56, 3.12, 6.25, 12.5, 25 and 50 mg/ml were measured for 1 min, as described previously (26). Airway responsiveness activities (Penh% values) were calculated by the formula (P = m/s ×100%, in which P, m and s represent the Penh% value, methacholine-stimulated Penh value and the saline-stimulated Penh value, respectively).

Histology and immunohistochemistry

Paraffin-embedded lung sections from mice of the 5 groups were stained with hematoxylin and eosin (H&E) to observe the pathological changes in the airways and lung tissue under an optical microscope. We performed immunohistochemistry using the monoclonal mouse antibody to α-smooth muscle actin (α-SMA) (1:100, Sigma-Aldrich Co.) and the polyclonal rabbit antibodies to ERK (1:100, Cell Signaling Technology, Inc., Danver, MA, USA). The sections from mice in the 5 groups were deparaffinized, and a 3% hydrogen peroxide solution was applied for 15 min to inhibit endogenous peroxidase activity. Antigen retrieval was performed with citrate solution for 45 min. After the sections were blocked with 10% goat serum in phosphate-buffered saline (PBS) for 20 min, they were incubated with the primary antibody overnight at 4°C. The horseradish peroxidase (HRP)-conjugated rabbit-anti-mouse immunoglobulin (Sigma-Aldrich Co.) and HRP-conjugated goat-anti-rabbit immunoglobulin (Sigma-Aldrich Co.) were used as the secondary antibodies and 3,3-diaminobenzidine (DAB) (Sigma-Aldrich Co.) was used as the chromogen. Hematoxylin was used for the counterstaining of the sections. For the negative control, the primary antibody was replaced with PBS. The sections were observed under a microscope. The thickness of the airway wall, the thickness of the airway smooth muscle layer, the numbers of smooth muscle cells and the intensity of ERK staining were measured as an average optical density using Image-Pro Plus (IPP) software (Image-Pro Plus 6.0; Media Cybernetics, Inc., Rockville, MD, USA). A non-stained region was selected and set as the background.

Primary cell culture

The ASMCs were cultivated as an explant culture as previously described with slight modifications (27). Briefly, primary rat airway smooth muscle was isolated from the bronchial smooth muscle from 10–12-week-old specific-pathogen-free (SPF) Sprague-Dawley (SD) male rats and the smooth muscle was isolated, cut into sections of 1–2 mm in cubic size, and placed on culture flasks with Dulbecco's modified Eagle's medium/Ham's F-12 (DMEM/F-12) medium (Gibco, Carlsbad, CA, USA) supplemented with 10% fetal bovine serum (FBS) in an incubator at 37°C, 5% CO2. The ASMCs migrated from the tissue explants and approached confluence around the explants. The culture medium was changed every 3 days. When the cells reached 90–100% confluence, they were passaged with 0.25% trypsin-EDTA (Gibco). The cells at passage 3 to 6 were used in the following experiments.

Immunofluorescence in vitro

The primary ASMCs were characterized by microscopy and their purity was verified by the immunofluorescence staining of α-SMA. The location of caveolin-1 in the ASMCs was also detected by immunofluorescence. After the cells grew to approximately 50% confluence, they were fixed in the culture plate using 4% paraformaldehyde for 10 min, permeabilized with 0.3% Triton X-100, blocked with 5% bovine serum albumin (BSA) at 37°C for 1 h, incubated with monoclonal mouse antibodies against α-SMA (1:100, Sigma-Aldrich Co.) and rabbit anti-rat caveolin-1 (1:500, Abcam, Cambridge. UK) respectively overnight at 4°C, and then labeled with fluorescein isothiocynate (FITC) secondary anti-mouse or anti-rabbit antibody (1:100, Sigma-Aldrich Co.) at 37°C for 1 h in the dark. The nuclei were counterstained with 20 μg/ml propidium iodide (PI) and 1 μg/ml DAPI in methanol for 5 min. The culture plate was washed 3 times between each step for 5 min in 0.1 M PBS (pH 7.4). The cells were examined and images were acquired using a fluorescence microscope.

Cell proliferation assay

The ASMCs were dispensed in a 96-well culture plate at a cell density of 2×104/ml and incubated at 37°C, 5% CO2. The cells were divided into 5 groups: i) negative control group (DMEM/F-12 containing 2.5% FBS), ii) positive control group (DMEM/F-12 containing 10% FBS), iii) proliferation group [25 ng/ml platelet-derived growth factor (PDGF) in DMEM/F-12 supplemented with 2.5% FBS], iv) curcumin-treated group with caveolin-1 (25 μmol/l curcumin with 25 ng/ml PDGF in DMEM/F-12 2.5% FBS), and v) curcumin-treated group without caveolin-1 [the cells were pre-treated with 5 μmol/l methyl-β-cyclodextrin (MβCD) for 60 min, then 25 μmol/l curcumin were added with 25 ng/ml PDGF in DMEM/F-12 containing 2.5% FBS]. Cell Counting Kit-8 (CCK-8) assays (Dojindo Laboratories, Kumamoto Japan) were used to measure cell proliferation according to the manufacturer's instructions. After the cells were treated with the culture medium for 6, 12, 24 and 48 h, they were incubated in 10% CCK-8 diluted in normal culture medium at 37°C for 2 h and the absorbance at 450 nm (OD at 450 nm) was measured. Each group was analyzed in triplicate. The results were expressed as OD values at 450 nm.

Real time PCR

Total RNA was extracted from the ASMCs in each group at 5 time points (0, 4, 8, 12 and 24 h) using TRIzol reagent (Invitrogen, Carlsbad, CA, USA) according to the manufacturer's instructions. The concentration, purity and amount of total RNA were determined by ultraviolet spectrometry (ND-1000 spectrophotometer; NanoDrop Technologies, Wilmington, DE, USA). Reverse transcriptase PCR was carried out using the Prime Script™ RT reagent kit (Takara Bio Inc., Otsu, Japan). The sense and antisense primers for caveolin-1 (GenBank ID: NM_133651) and β-actin (GenBank ID: NM_031144.2) were synthesized by Takara Bio Inc. The primers used are listed in Table I.

Table I

Oligonucleotide sequences of primers used for real-time PCR.

Table I

Oligonucleotide sequences of primers used for real-time PCR.

Gene nameSense/antisense primers
Caveolin-1 5′-GGGCAACATCTAGAAGCCCAACAA-3′
5′-CTGATGCACTGAATTCCAATCAGGAA-3′
β-actin 5′-CTTCCTTCCTGGGTATGGAATC-3′
5′-GAGATGATCTGGGGGTCTGA-3′

Real-time PCR analyses were performed using SYBR Premix Ex Taq (Takara Bio Inc.). Amplification was performed in an ABI PRISM 7500 Real-Time PCR System with the following temperature profile: a pre-denaturation step of 30 sec at 95°C, extended at 60°C for 34 sec, and denaturion for 5 sec at 95°C for 40 cycles. The data were calculated using the 2−ΔΔCt method normalized to β-actin. Each experiment was performed 3 times.

Western blot analysis

The cells were lysed in cell lysis buffer (Cell Signaling Technology, Inc.). Total protein was extracted by sonication and centrifugation of the cell lysates at 12,000 × g for 10 min at 4°C. Whole-cell extracts were fractionated by 15% sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE). Proteins were transferred onto a nitrocellulose membrane (Millipore, Bedford, MA, USA) using a wet transfer apparatus. For immune detection, the membranes were washed and incubated with rabbit anti-rat ERK1/2 polyclonal antibody (1:4,000), rabbit-anti-rat phospho-ERK1/2 polyclonal antibody (1:4,000), anti-GADPH polyclonal antibody (1:2,000; all from Cell Signaling Technology, Inc., USA), rabbit-anti-rat caveolin-1 polyclonal antibody (1:500, Abcam) and anti-β-actin monoclonal antibody (1:2,000, Sigma-Aldrich Co.) overnight at 4°C. The signals were amplified using the appropriate HRP-conjugated secondary antibody, and visualized by enhanced chemiluminescence (Pierce Biotechnology, Inc., Rockford, IL, USA).

Statistical analysis

All statistical analyses were performed using SPSS for Windows v.13.0 (SPSS, Chicago, IL, USA). All data are expressed as the means ± SD. Statistical analyses were carried out using one-way ANOVA, the rank-sum test or least significant difference tests where appropriate. A P-value <0.05 was considered to indicate a statistically significant difference.

Results

Curcumin decreases the airway responsiveness in the mouse model of airway remodeling

Positive symptoms, such as dysphoria, tachypnea and abdominal spasms were observed in the mice in the model group 5 to 10 min following challenge with OVA, whereas less severe symptoms, including mild dysphoria and tachypnea were observed in the mice treated with curcumin; all the symptoms soon dissipated.

The mice with asthma (model group), the mice with asthma treated with curcumin (curcumin-treated groups) and the untreated normal mice (control group) began to suffer from breathing difficulties after inhaling methacholine at concentrations of 1.56, 6.25 and 25 mg/ml. The airway responsiveness (Penh%) of the model group and the curcumin-treated groups (treated with various concentrations of curcumin) significantly increased compared with the normal group when methacholine was inhaled at the concentrations of 3.12, 6.25, 12.5 and 25 mg/ml (P<0.05) and at the concentration of 50 mg/ml (P<0.01) (Fig. 1). The airway responsiveness (Penh%) of the curcumin-treated groups significantly decreased compared with the model group when methacholine was inhaled at the concentrations of 3.12, 6.25, 12.5, 25 mg/ml (P<0.05) and at the concentration of 50 mg/ml (P<0.01) (Fig. 1).

Effects of curcumin on OVA-induced histopathological changes in lungs

To assess the anti-remodeling effects of curcumin, histopathological experiments were performed. Using H&E staining, inflammatory cell infiltration in the peribronchial and perivascular areas was observed in OVA-challenged mice. Treatment with curcumin (50, 100 and 150 mg/kg) markedly reduced the degree of inflammatory cell infiltration in the peribronchial and perivascular areas (Fig. 2, upper panel). OVA induced the proliferation of ASMCs. In the curcumin-treated groups, curcumin inhibited the OVA-induced ASMC proliferation compared with the model group (Fig. 2, middle panel). The expression of α-SMA and ERK was detected by immunohistochemistry and the expression of ERK was increased in mice with asthma (Fig. 2, lower panel; ERK protein is stained brown). Curcumin decreased the expression of ERK in the curcumin-treated groups compared with the model group. The bronchial wall thickness (WAi/Pi), the thickness of the smooth muscle layer (WAm/Pi), the number of smooth muscle cells (N/Pi) and the ERK gray values in the ASMCs in each group were measured using IPP software (Table II). In the model group, the thickness of the airway wall, the thickness of the airway smooth muscle layer and the numbers of smooth muscle cells were significantly increased compared with the control group (P<0.01). In the curcumin-treated groups, the thickness of the airway wall, the thickness of the airway smooth muscle layer and the numbers of smooth muscle cells were significantly decreased compared with the model group (P<0.05).

Table II

Lung pathology of the mice in the 5 groups.

Table II

Lung pathology of the mice in the 5 groups.

GroupWAi/Pi (μm)WAm/Pi (μm)N/Pi (/mm)ERK gray-scale value
Control5.6±0.51.2±0.512.4±1.268.5±6.4
Model14.6±3.6b6.4±1.4b35.3±4.4b97.6±15.2b
50 mg/kg curcumin7.7±1.7a4.2±1.5a25.8±2.5a90.5±15.8a
100 mg/kg curcumin5.6±1.5a3.2±1.3a16.5±1.6a80.6±10.3a
150 mg/kg curcumin5.6±0.7a1.5±0.7a12.5±1.7a74.5±7.1a

{ label (or @symbol) needed for fn[@id='tfn1-ijmm-32-03-0629'] } The data are expressed as the the means ± SD (n=16).

a P<0.05, compared with the model group and

b P<0.01, compared with the control group.

{ label (or @symbol) needed for fn[@id='tfn4-ijmm-32-03-0629'] } Pi, internal perimeter; N, number of smooth muscle cells; WAi, inner wall area; WAm, smooth muscle area.

Correlation between ERK expression and ASMC proliferation

Semi-quantitative image analysis demonstrated that the expression level of ERK in the airways positively correlated with the thickness of the airway smooth muscle layer (r=0.745, P<0.05), and positively correlated with the number of ASMCs (r=0.821, P<0.05).

Rat primary ASMC identification and location of caveolin-1 in ASMCs

The ASMCs generally became adherent within 2 days, and assumed a stretched, spindle-shaped valley-like growth patterns after 4–5 days (Fig. 3A). The ASMCs grew to approximately 90% confluence after 8–10 days; they were then digested by trypsin and passaged for immunofluorescence staining (Fig. 3B). The cytoplasmic filaments of positive cells were stained green. The percentage of pure ASMCs was approximately 94.32% using Image Pro Plus 6.0 software. Caveolin-1 was detected in the ASMC plasma membrane under a fluorescence microscope (Fig. 3C–E).

Effect of curcumin on PDGF-induced ASMC proliferation

Cell proliferation was determined using the CCK-8 assay as shown in Fig. 4. At 12, 24 and 48 h, the absorbance at 450 nm was significantly higher in the PDGF + curcumin group compared with the negative control group (P<0.05). Of note, the ASMC proliferation induced by PDGF in group B peaked at 48 h, compared with the negative control group (P<0.01). Curcumin (25 μmol/l) significantly decreased the PDGF-induced the proliferation of ASMCs at 24 h (P<0.05) (group C compared with group B). After caveolae was disrupted by MβCD, the PDGF-induced proliferation of ASMCs in group D was markedly increased compared with group C. This suggests that the anti-proliferative effects of curcumin are weakened without caveolin-1; thus, caveolin-1 may play an important role in the anti-proliferative effects of curcumin on ASMCs.

Effect of curcumin on the expression of ERK1/2 protein in ASMCs

After the 4th passage ASMCs were treated with or without curcumin (25 μmol/l) for 30 and 60 min, total protein was extracted for western blot analysis to detect the expression of total ERK1/2 protein and the phosphorylation of ERK1/2 protein. A control group was set up. Treatment with PDGF increased ERK1/2 phosphorylation compared with the control group. The expression of PDGF-induced phosphorylated-ERK was significantly decreased following treatment with curcumin (Fig. 5), and phosphorylated ERK could not be detected at 60 min. Curcumin had no effect on the expression of total ERK1/2.

mRNA and protein expression of caveolin-1 following treatment with curcumin

After the rat ASMCs were treated with curcumin (25 μmol/l) for 0, 4, 8, 12 and 24 h, the mRNA expression of caveolin-1 in the ASMCs was measured by real-time PCR. The result revealed that following treatment with PDGF with or without crucumin for 4 h, the caveolin-1 mRNA expression was increased (compared with the control group) (P<0.01). The mRNA expression of caveolin-1 was significantly increased in the PDGF + curcumin-treated group compared with the PDGF group (P<0.05) (Fig. 6A). After the 4th passage ASMCs were treated with PDGF or curcumin and PDGF for 24 h, total protein was extracted for western blot analysis to detect the expression of caveolin-1 protein. The results are shown in Fig. 6B. Compared with the PDGF group, the protein expression of caveolin-1 in the cucumin + PDGF group was significantly increased (P<0.01).

Discussion

Asthma is traditionally defined as a chronic disease characterized by airway inflammation, AHR and airway remodeling. In a previous study, Chung examined patients with asthma using airway histopathology and lung function dynamic observation and indicated that a series of structural changes could be found in the airway wall in the patients with asthma (5). Among the histopathological characteristics of airway remodeling, previous studies have indicated that the increased mass of ASMCs plays a critical role (2,4,5,28,29). Due to their important role in airway remodeling, ASMCs may be a potential therapeutic target for the treatment of patients with asthma.

Curcumin has been used in indigenous medicine for the treatment of a variety of inflammatory conditions and chronic diseases (9,13,14,23) and studies have mainly focused on the effects of curcumin on inhibiting bronchial inflammation (3034). However, limited studies have investigated the effects of curcumin on structural changes in asthma, such as ASMC proliferation in airway remodeling. Therefore, in this study, we performed in vitro and in vivo experiments to examine the effects of curcumin on the proliferation of ASMCs and to elucidate the underlying mechanisms. Our findings demonstrate that curcumin inhibits the proliferation of ASMCs both in vitro and in vivo.

In in vivo experiments, we used OVA-challenged mice to establish a model of chronic asthma airway remodeling; the curcumin-treated groups were administered various doses of curcumin by an i.p. injection prior to challenge with OVA. The mouse model of airway remodeling induced by OVA has previously been used in a number of studiesl; however, the majority of these studies focused on the inflammatory infiltration in this model. Our results were identical with those from previous studies (3033). Our findings indicated that the symptoms of an asthma attack dissipated following treatment with curcumin and that curcumin significantly reduced the airway responsiveness. In addition, we paid particular attention to the changes in airway smooth muscle. We found that following treatment with curcumin, the thickness of the airway wall and the bronchial smooth muscle layer became thinner and the number of smooth muscle cells decreased; the most significant changes were observed in the high-dose curcumin-treated group. These results demonstrate that the administration of curcumin inhibits the proliferation of ASMCs in vivo.

In in vitro experiments, we stimulated the proliferation of primary rat ASMCs by PDGF and provided a proliferative experimental model for the study of asthma at the cellular and molecular level. Our results revealed that curcumin inhibited the PDGF-induced proliferation of ASMCs in vitro.

The mitogen-activated protein kinase (MAPK) signaling cascade has been shown to play an important role in the activation of various cells (3538). It is activated by the 3-tiered sequential phosphorylation of MAPK kinase, MAPK/ERK kinase (MEK) and MAPK. There are 3 major groups of MAPK in mammalian cells, including ERK, p38 MAPK and c-Jun N-terminal kinase. Accumulating evidence has shown that ERK plays a crucial role in human ASMC growth. A previous study demonstrated that ERK activity in the lungs of asthmatic mice was significantly higher compared with normal mice (39). ERK activation is necessary for the proliferation of ASMCs (4). In addition, PDGF and other growth factors are promoters of ERK1/2 (p42/44 MAPK) expression and the activation of phosphorylation, which are closely associated with the proliferation of smooth muscle cells (27,4044). Thus, the ERK signaling pathway plays a crucial role in the pathological process of airway remodeling in asthma. The results of our study are in accordance with these findings; we found that the phosphorylated-ERK1/2 protein levels were significantly decreased following the treatment of ASMCs with curcumin. This suggests that the inhibition of the activation of ERK1/2 by curcumin is involved in the inhibition of ASMC proliferation. We provide evidence that curcumin inhibits ASMC proliferation and that caveolin-1 plays a crucial role in this process; the involvement of caveolin-1 is partly due to the fact that it has the ability to regulate the ERK 1/2 pathway.

We found that curcumin significantly inhibited the PDGF-induced proliferation of ASMCs and that this effect was significantly attenuated by MβCD. The expression of caveolin-1 was significantly increased in the curcumin-treated group as compared with the PDGF group in our study. Caveolae are small vesicular invaginations of the cell membrane. It is within this organelle that cells perform transcytosis and signal transduction. Caveolae are composed of a mixture of lipids and proteins (4550). The chief structural proteins of caveolae are caveolins (caveolin-1, caveolin-2 and caveolin-3), and caveolin-1 appears to be an essential component of caveolae (5157). Evidence suggests that caveolin-1 regulates the ERK1/2 pathway during cell proliferation (55,5860). Buitrago and Boland found that when proliferating mouse skeletal myoblastic cells were pre-treated with MβCD, a caveolae-disrupting agent, the 1α,25(OH)2D3-dependent activation of ERK1/2, p38 MAPK and c-Src was suppressed (58). Furthermore, studies on human ASMCs have shown that caveolae and caveolin-1 coordinate PDGF receptor signaling, leading to myocyte proliferation, and inhibit the constitutive activity of p42/p44 MAPK, sustaining cell quiescence (55). These data are in accordance with those in the study by Peterson et al, who showed that the treatment of VSMCs with PDGF for 24 h resulted in a loss of caveolin-1 protein expression and plasma membrane-associated caveolae (21). The data mentioned above indicate that caveolin-1 is an important negative regulator of cell proliferation and PDGF signaling events. Our results therefore suggest that caveolin-1 plays a crucial role in the anti-proliferative effects of curcumin through the ERK1/2 pathway.

In conclusion, we demonstrate that curcumin inhibits the proliferation of ASMCs in vitro and in vivo; the possible mechanisms behind the inhibitory effects of curcumin may be the upregulation of the expression of caveolin-1 and the blocking of the ERK pathway, thereby inhibiting the proliferation of ASMCs. Our findings provide new insight into the application of curcumin in the prevention and treatment of asthma, particularly airway remodeling in severe asthma.

Acknowledgements

This study was supported by grants from the Guangzhou Science and Technology Committee (no. 2008J1-C261) and the Guangzhou University Science and Technology Project (no. 10A149), China.

References

1 

Bateman ED: Global strategy for asthma management and prevention 2009 (update). Report of Global Initiative for Asthma. 2009:22009.

2 

Girodet PO, Ozier A, Bara I, Tunon de Lara JM, Marthan R and Berger P: Airway remodeling in asthma: new mechanisms and potential for pharmacological intervention. Pharmacol Ther. 130:325–337. 2011. View Article : Google Scholar : PubMed/NCBI

3 

Delacourt C: Bronchial changes in untreated asthma. Arch Pediatr. 11(Suppl 2): 71s–73s. 2004.(In French).

4 

Black JL, Roth M, Lee J, Carlin S and Johnson PR: Mechanisms of airway remodeling. Airway smooth muscle. Am J Respir Crit Care Med. 164:S63–S66. 2001. View Article : Google Scholar : PubMed/NCBI

5 

Chung KF: The role of airway smooth muscle in the pathogenesis of airway wall remodeling in chronic obstructive pulmonary disease. Proc Am Thorac Soc. 2:347–354. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Siddiqui S, Redhu NS, Ojo OO, et al: Emerging airway smooth muscle targets to treat asthma. Pulm Pharmacol Ther. 26:132–144. 2012. View Article : Google Scholar

7 

Bar-Sela G, Epelbaum R and Schaffer M: Curcumin as an anti-cancer agent: review of the gap between basic and clinical applications. Curr Med Chem. 17:190–197. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Anand P, Sundaram C, Jhurani S, Kunnumakkara AB and Aggarwal BB: Curcumin and cancer: an ‘old-age’ disease with an ‘age-old’ solution. Cancer Lett. 267:133–164. 2008.

9 

Schaffer M, Schaffer PM, Zidan J and Bar Sela G: Curcuma as a functional food in the control of cancer and inflammation. Curr Opin Clin Nutr Metab Care. 14:588–597. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Ammon HP and Wahl MA: Pharmacology of Curcuma longa. Planta Med. 57:1–7. 1991.

11 

Hatcher H, Planalp R, Cho J, Torti FM and Torti SV: Curcumin: from ancient medicine to current clinical trials. Cell Mol Life Sci. 65:1631–1652. 2008. View Article : Google Scholar : PubMed/NCBI

12 

Aggarwal BB and Harikumar KB: Potential therapeutic effects of curcumin, the anti-inflammatory agent, against neurodegenerative, cardiovascular, pulmonary, metabolic, autoimmune and neoplastic diseases. Int J Biochem Cell Biol. 41:40–59. 2009. View Article : Google Scholar

13 

Yang X, Thomas DP, Zhang X, et al: Curcumin inhibits platelet-derived growth factor-stimulated vascular smooth muscle cell function and injury-induced neointima formation. Arterioscler Thromb Vasc Biol. 26:85–90. 2006. View Article : Google Scholar : PubMed/NCBI

14 

Wong TF, Takeda T, Li B, et al: Curcumin disrupts uterine leiomyosarcoma cells through AKT-mTOR pathway inhibition. Gynecol Oncol. 122:141–148. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Laporte JC, Moore PE, Baraldo S, et al: Direct effects of interleukin-13 on signaling pathways for physiological responses in cultured human airway smooth muscle cells. Am J Respir Crit Care Med. 164:141–148. 2001. View Article : Google Scholar : PubMed/NCBI

16 

Weng SH, Tsai MS, Chiu YF, Kuo YH, Chen HJ and Lin YW: Enhancement of mitomycin C-induced cytotoxicity by curcumin results from down-regulation of MKK1/2-ERK1/2-mediated thymidine phosphorylase expression. Basic Clin Pharmacol Toxicol. 110:298–306. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Saab MB, Bec N, Martin M, et al: Differential effect of curcumin on the nanomechanics of normal and cancerous mammalian epithelial cells. Cell Biochem Biophys. 65:399–411. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Dhandapani KM, Mahesh VB and Brann DW: Curcumin suppresses growth and chemoresistance of human glioblastoma cells via AP-1 and NFkappaB transcription factors. J Neurochem. 102:522–538. 2007. View Article : Google Scholar : PubMed/NCBI

19 

Yang CL, Liu YY, Ma YG, et al: Curcumin blocks small cell lung cancer cells migration, invasion, angiogenesis, cell cycle and neoplasia through Janus kinase-STAT3 signalling pathway. PLoS One. 7:e379602012. View Article : Google Scholar : PubMed/NCBI

20 

Chen JW, Tang YL, Liu H, et al: Anti-proliferative and anti-metastatic effects of curcumin on oral cancer cells. Hua Xi Kou Qiang Yi Xue Za Zhi. 29:83–86. 2011.(In Chinese).

21 

Peterson TE, Guicciardi ME, Gulati R, et al: Caveolin-1 can regulate vascular smooth muscle cell fate by switching platelet-derived growth factor signaling from a proliferative to an apoptotic pathway. Arterioscler Thromb Vasc Biol. 23:1521–1527. 2003. View Article : Google Scholar

22 

Luo DX, Cheng J, Xiong Y, et al: Static pressure drives proliferation of vascular smooth muscle cells via caveolin-1/ERK1/2 pathway. Biochem Biophys Res Commun. 391:1693–1697. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Qin L, Yang YB, Tuo QH, et al: Effects and underlying mechanisms of curcumin on the proliferation of vascular smooth muscle cells induced by Chol:MbetaCD. Biochem Biophys Res Commun. 379:277–282. 2009. View Article : Google Scholar : PubMed/NCBI

24 

McMillan SJ and Lloyd CM: Prolonged allergen challenge in mice leads to persistent airway remodelling. Clin Exp Allergy. 34:497–507. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Lloyd CM, Gonzalo JA, Nguyen T, et al: Resolution of bronchial hyperresponsiveness and pulmonary inflammation is associated with IL-3 and tissue leukocyte apoptosis. J Immunol. 166:2033–2040. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Hamelmann E, Schwarze J, Takeda K, et al: Noninvasive measurement of airway responsiveness in allergic mice using barometric plethysmography. Am J Respir Crit Care Med. 156:766–775. 1997. View Article : Google Scholar : PubMed/NCBI

27 

Chen G and Khalil N: TGF-beta1 increases proliferation of airway smooth muscle cells by phosphorylation of map kinases. Respir Res. 7:22006. View Article : Google Scholar : PubMed/NCBI

28 

Tang ML, Wilson JW, Stewart AG and Royce SG: Airway remodelling in asthma: current understanding and implications for future therapies. Pharmacol Ther. 112:474–488. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Nath P, Leung SY, Williams A, et al: Importance of p38 mitogen-activated protein kinase pathway in allergic airway remodelling and bronchial hyperresponsiveness. Eur J Pharmacol. 544:160–167. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Oh SW, Cha JY, Jung JE, et al: Curcumin attenuates allergic airway inflammation and hyper-responsiveness in mice through NF-κB inhibition. J Ethnopharmacol. 136:414–421. 2011.PubMed/NCBI

31 

Moon DO, Kim MO, Lee HJ, et al: Curcumin attenuates ovalbumin-induced airway inflammation by regulating nitric oxide. Biochem Biophys Res Commun. 375:275–279. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Karaman M, Firinci F, Cilaker S, et al: Anti-inflammatory effects of curcumin in a murine model of chronic asthma. Allergol Immunopathol (Madr). 40:210–214. 2012. View Article : Google Scholar : PubMed/NCBI

33 

Venkatesan N, Punithavathi D and Babu M: Protection from acute and chronic lung diseases by curcumin. Adv Exp Med Biol. 595:379–405. 2007. View Article : Google Scholar : PubMed/NCBI

34 

Sharafkhaneh A, Velamuri S, Badmaev V, Lan C and Hanania N: The potential role of natural agents in treatment of airway inflammation. Ther Adv Respir Dis. 1:105–120. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Nel AE: T-cell activation through the antigen receptor. Part 1: signaling components, signaling pathways, and signal integration at the T-cell antigen receptor synapse. J Allergy Clin Immunol. 109:758–770. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Jacob A, Cooney D, Pradhan M and Coggeshall KM: Convergence of signaling pathways on the activation of ERK in B cells. J Biol Chem. 277:23420–23426. 2002. View Article : Google Scholar : PubMed/NCBI

37 

Gauld SB, Dal Porto JM and Cambier JC: B cell antigen receptor signaling: roles in cell development and disease. Science. 296:1641–1642. 2002. View Article : Google Scholar : PubMed/NCBI

38 

Nadler MJ, Matthews SA, Turner H and Kinet JP: Signal transduction by the high-affinity immunoglobulin E receptor Fc epsilon RI: coupling form to function. Adv Immunol. 76:325–355. 2000. View Article : Google Scholar : PubMed/NCBI

39 

Duan W, Chan JH, Wong CH, Leung BP and Wong WS: Anti-inflammatory effects of mitogen-activated protein kinase kinase inhibitor U0126 in an asthma mouse model. J Immunol. 172:7053–7059. 2004. View Article : Google Scholar : PubMed/NCBI

40 

De S, Zelazny ET, Souhrada JF and Souhrada M: Role of phospholipase C and tyrosine kinase systems in growth response of human airway smooth muscle cells. Am J Physiol. 270:L795–L802. 1996.PubMed/NCBI

41 

Walker TR, Moore SM, Lawson MF, Panettieri RA Jr and Chilvers ER: Platelet-derived growth factor-BB and thrombin activate phosphoinositide 3-kinase and protein kinase B: role in mediating airway smooth muscle proliferation. Mol Pharmacol. 54:1007–1015. 1998.

42 

Kumar A, Lnu S, Malya R, et al: Mechanical stretch activates nuclear factor-kappaB, activator protein-1, and mitogen-activated protein kinases in lung parenchyma: implications in asthma. FASEB J. 17:1800–1811. 2003. View Article : Google Scholar

43 

Chiou YL, Shieh JJ and Lin CY: Blocking of Akt/NF-kappaB signaling by pentoxifylline inhibits platelet-derived growth factor-stimulated proliferation in Brown Norway rat airway smooth muscle cells. Pediatr Res. 60:657–662. 2006. View Article : Google Scholar

44 

Bai J, Liu XS, Xu YJ, Zhang ZX, Xie M and Ni W: The effect of ERK signaling pathway on cell apoptosis in airway smooth muscle cells of chronic asthmatic rats. Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi. 26:738–741. 2010.(In Chinese).

45 

Schlegel A, Volonte D, Engelman JA, et al: Crowded little caves: structure and function of caveolae. Cell Signal. 10:457–463. 1998. View Article : Google Scholar : PubMed/NCBI

46 

Shaul PW and Anderson RG: Role of plasmalemmal caveolae in signal transduction. Am J Physiol. 275:L843–L851. 1998.PubMed/NCBI

47 

Anderson RG: The caveolae membrane system. Annu Rev Biochem. 67:199–225. 1998. View Article : Google Scholar : PubMed/NCBI

48 

Okamoto T, Schlegel A, Scherer PE and Lisanti MP: Caveolins, a family of scaffolding proteins for organizing ‘preassembled signaling complexes’ at the plasma membrane. J Biol Chem. 273:5419–5422. 1998.

49 

Couet J, Belanger MM, Roussel E and Drolet MC: Cell biology of caveolae and caveolin. Adv Drug Deliv Rev. 49:223–235. 2001. View Article : Google Scholar : PubMed/NCBI

50 

Razani B, Woodman SE and Lisanti MP: Caveolae: from cell biology to animal physiology. Pharmacol Rev. 54:431–467. 2002. View Article : Google Scholar : PubMed/NCBI

51 

Schubert W, Frank PG, Woodman SE, et al: Microvascular hyperpermeability in caveolin-1 (−/−) knock-out mice. Treatment with a specific nitric-oxide synthase inhibitor, L-NAME, restores normal microvascular permeability in Cav-1 null mice. J Biol Chem. 277:40091–40098. 2002.

52 

Ramirez MI, Pollack L, Millien G, Cao YX, Hinds A and Williams MC: The alpha-isoform of caveolin-1 is a marker of vasculogenesis in early lung development. J Histochem Cytochem. 50:33–42. 2002. View Article : Google Scholar : PubMed/NCBI

53 

Thyberg J: Caveolin-1 and caveolae act as regulators of mitogenic signaling in vascular smooth muscle cells. Arterioscler Thromb Vasc Biol. 23:1481–1483. 2003. View Article : Google Scholar : PubMed/NCBI

54 

Miyawaki-Shimizu K, Predescu D, Shimizu J, Broman M, Predescu S and Malik AB: siRNA-induced caveolin-1 knockdown in mice increases lung vascular permeability via the junctional pathway. Am J Physiol Lung Cell Mol Physiol. 290:L405–L413. 2006. View Article : Google Scholar : PubMed/NCBI

55 

Gosens R, Stelmack GL, Dueck G, et al: Role of caveolin-1 in p42/p44 MAP kinase activation and proliferation of human airway smooth muscle. Am J Physiol Lung Cell Mol Physiol. 291:L523–L534. 2006. View Article : Google Scholar : PubMed/NCBI

56 

Sun Y, Hu G, Zhang X and Minshall RD: Phosphorylation of caveolin-1 regulates oxidant-induced pulmonary vascular permeability via paracellular and transcellular pathways. Circ Res. 105:676–685, 615 p following 685. 2009. View Article : Google Scholar : PubMed/NCBI

57 

Feng H, Guo L, Song Z, et al: Caveolin-1 protects against sepsis by modulating inflammatory response, alleviating bacterial burden, and suppressing thymocyte apoptosis. J Biol Chem. 285:25154–25160. 2010. View Article : Google Scholar

58 

Buitrago C and Boland R: Caveolae and caveolin-1 are implicated in 1α,25(OH)2-vitamin D3-dependent modulation of Src, MAPK cascades and VDR localization in skeletal muscle cells. J Steroid Biochem Mol Biol. 121:169–175. 2010.

59 

Park JH, Ryu JM and Han HJ: Involvement of caveolin-1 in fibronectin-induced mouse embryonic stem cell proliferation: role of FAK, RhoA, PI3K/Akt, and ERK 1/2 pathways. J Cell Physiol. 226:267–275. 2011. View Article : Google Scholar : PubMed/NCBI

60 

Watson CS, Jeng YJ, Hu G, Wozniak A, Bulayeva N and Guptarak J: Estrogen- and xenoestrogen-induced ERK signaling in pituitary tumor cells involves estrogen receptor-α interactions with G protein-αi and caveolin I. Steroids. 77:424–432. 2012.PubMed/NCBI

Related Articles

Journal Cover

September 2013
Volume 32 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
Zeng X, Cheng Y, Qu Y, Xu J, Han Z and Zhang T: Curcumin inhibits the proliferation of airway smooth muscle cells in vitro and in vivo. Int J Mol Med 32: 629-636, 2013
APA
Zeng, X., Cheng, Y., Qu, Y., Xu, J., Han, Z., & Zhang, T. (2013). Curcumin inhibits the proliferation of airway smooth muscle cells in vitro and in vivo. International Journal of Molecular Medicine, 32, 629-636. https://doi.org/10.3892/ijmm.2013.1425
MLA
Zeng, X., Cheng, Y., Qu, Y., Xu, J., Han, Z., Zhang, T."Curcumin inhibits the proliferation of airway smooth muscle cells in vitro and in vivo". International Journal of Molecular Medicine 32.3 (2013): 629-636.
Chicago
Zeng, X., Cheng, Y., Qu, Y., Xu, J., Han, Z., Zhang, T."Curcumin inhibits the proliferation of airway smooth muscle cells in vitro and in vivo". International Journal of Molecular Medicine 32, no. 3 (2013): 629-636. https://doi.org/10.3892/ijmm.2013.1425