Open Access

IL‑10/IL‑10 receptor 1 pathway promotes the viability and collagen synthesis of pulmonary fibroblasts originated from interstitial pneumonia tissues

  • Authors:
    • Hong Ye
    • Jiongwei Pan
    • Xiaoping Cai
    • Zhangyong Yin
    • Lu Li
    • Enhui Gong
    • Cunlai Xu
    • Hao Zheng
    • Zhuo Cao
    • Enguo Chen
    • Junfeng Qian
  • View Affiliations

  • Published online on: June 15, 2022     https://doi.org/10.3892/etm.2022.11445
  • Article Number: 518
  • Copyright: © Ye 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

Interstitial pneumonia is a pulmonary interstitial inflammatory and fibrosis disease with a variety of causes that causes respiratory disorders and threatens the lives of patients. The present study aimed to investigate the expression of interleukin (IL)‑10 in peripheral blood of patients with interstitial pneumonia and its biological functions in pulmonary fibroblasts. A total of 42 patients with idiopathic pulmonary fibrosis (IPF) and 20 healthy subjects were included. ELISA was used to determine IL‑10 concentration in serum from the patients and healthy subjects. Primary fibroblasts were isolated from lung tissue successfully and determined by morphology. The CCK‑8 assay was performed to determine the effect of IL‑10 expression on cell viability. Western blotting was used to determine COL1a1, COL1a2 and IL‑10R1 protein expression. Flow cytometry was used for cell cycle analysis and to determine the number of IL‑10+ cells. Expression of IL‑10 in serum from IPF patients was higher compared to that from healthy subjects. IL‑10 promoted the viability and collagen synthesis and secretion of MRC‑5 cells and primary pulmonary fibroblasts. IL‑10 and IL‑10 receptor (R) 1 served regulatory roles in the viability and collagen synthesis of MRC‑5 cells. The ratio of peripheral mononuclear lymphocytes with positive expression of IL‑10 was elevated in peripheral blood from patients with IPF. The present study demonstrated that IL‑10 expression in peripheral blood of patients with IPF is increased significantly compared with healthy subjects. Activation of the IL‑10/IL‑10R1 signaling pathway promoted the viability and collagen synthesis and secretion of pulmonary fibroblasts, leading to pulmonary fibrosis. The present study provided experimental basis for further understanding the development mechanism of pulmonary fibrosis.

Introduction

Interstitial pulmonary diseases are a group of diseases caused mainly by pathological changes in the alveolar wall (1). The basic pathological changes of interstitial pulmonary diseases include diffuse lung parenchyma, alveolitis and interstitial fibrosis, and their clinical manifestations comprise active dyspnea, diffuse shadow in X-ray chest film, restrictive ventilation disorder, decreased diffusion function and hypoxemia (1,2). Interstitial pulmonary diseases have a variety of subtypes, among which idiopathic pulmonary fibrosis (IPF) is the most harmful one (3). IPF is a chronic, progressive and fatal interstitial pulmonary disease, and there is no effective treatment due to its unknown pathogenesis (4,5). The incidence rate of IPF varies according to different regions, different populations and different occupations (6). In developed countries, the incidence rate is as low as three in 100,000(6). In developing countries, its incidence is slightly higher. Overall, the incidence rate of males is higher than that of females (6). It is believed that repeated pulmonary epithelial cell injury and myofibroblast activation are the direct causes of pulmonary fibrosis (7,8). A large number of fibrosis-promoting cytokines produced in the process of injury repair can induce fibroblasts to differentiate, proliferate, migrate and invade through the autocrine or paracrine tissues (9). This results in continuous deposition of extracellular matrix (ECM), hinders normal injury repair and ultimately leads to the destruction of lung structure and the occurrence of fibrosis (9,10).

Abnormal proliferation of fibroblasts and increase of ECM are the basic pathological features of interstitial pulmonary diseases, indicating that fibroblasts serve. Important biological roles in the diseases (11,12). Fibroblasts are the main cell components in lung connective tissues, differentiated from mesenchymal cells with a spindle flat star shape and protuberances (13,14). Fibroblasts have large cell bodies and their cytoplasm has weak basophilia (15). Under normal conditions, the main functions of fibroblasts include sustaining normal lung morphology, synthesizing and releasing ECM, participating in gas exchange in lung tissues and maintaining normal physiological functions of pulmonary epithelial and endothelial cells such as secretion (16). However, fibroblasts demonstrate abnormal proliferation and differentiation and secrete a large amount of ECM in interstitial pneumonia (17).

Cytokines serve important roles in the occurrence and development of interstitial pneumonia (18). For example, TGF-β is the strongest fibrogenic cytokine that can stimulate the proliferation of pulmonary fibroblasts and make them transform into myofibroblasts, hence it is an important target for the inhibition of pulmonary fibrosis (19). Studies on cytokines that are involved in pulmonary fibrosis have important clinical implications (20). Interleukin (IL)-10 is a type of inflammatory cytokine that is involved in tumor development and progression, interstitial pulmonary diseases and autoimmune diseases (21,22). IL-10 is secreted mainly by lymphocytes, macrophages and mast cells and it can inhibit macrophages and Th1 cells and enhance the biological function of B cells (23). In addition, IL-10 reduces the production of inflammatory cytokines, such as IL-2 and IFN-γ, thus inhibiting inflammatory responses (24). It has also been reported that IL-10 participates in the occurrence and development of pulmonary fibrosis (25). However, the role of IL-10 in pulmonary fibroblasts remains unclear. The present study aimed to investigate the regulatory relationship between IL-10 and lung fibroblasts at a cellular and molecular level and to provide experimental basis for clinical targeted intervention in interstitial pneumonia.

Materials and methods

Subjects

A total of 42 patients with IPF (31 males and 11 females; age range, 53-71 years; mean age, 62.4±7.3 years) were diagnosed at Lishui People's Hospital (Lishui, China) between July 012 and July 2018 according to IPF non-traumatic diagnostic criteria formulated by ATS/ERS/JRS/ALAT in 2011(26). The exclusion criteria for the patients were: i) Not having chronic obstructive pulmonary disease; ii) having collagen angiopathy complicated with interstitial pulmonary diseases; iii) having unstable angina pectoris; iv) having interstitial pulmonary diseases caused by occupational and environmental exposures; and v) having interstitial pulmonary diseases and other serious diseases caused by drugs or known causes. The inclusion criterion was IPF patients diagnosed in Lishui People's Hospital (Lishui, China) who had no history of cancer, diabetes, hypertension, autoimmune diseases or chronic medication. Among the 42 patients, 38 were smokers and 4 were non-smokers. Twenty healthy subjects were included as control group (age range, 33-45 years; 15 males and 5 females).

From all subjects, 10 ml blood was drawn from the elbow vein. Of the 10 ml blood, 8 ml was used for separating serum after centrifugation at 1,500 x g and 4˚C for 10 min, and 2 ml was used to obtain peripheral mononuclear lymphocytes for flow cytometry. The serum and peripheral mononuclear lymphocytes were stored at -80˚C until use. All procedures performed in the current study were approved by the Ethics Committee of Wenzhou Medical University (Lishui, China). Written informed consent was obtained from all patients or their families.

Cells and transfection

Lung tissues were collected during lobectomy and cut into 1 mm3 size pieces. Then, the tissues were cultured in DMEM supplemented with 20% fetal bovine serum (Thermo Fisher Scientific, Inc.) at 37˚C and 5% CO2. Every 2 days, the tissues were observed and the medium was replenished. Subsequently, ~3 weeks later, primary fibroblasts crawled out of tissue mass and continued growing. On reaching 70-80% confluency, the fibroblasts were passaged at a volume ratio of 1:3 (cell suspension vs. medium) and cultured in DMEM medium supplemented with 10% fetal bovine serum at 37˚C and 5% CO2.

MRC-5 cells (2x105; Cell Bank of the Chinese Academy of Sciences, Shanghai, China) in logarithmic growth phase were seeded into 24-well plates, and cultured in RPMI-1640 medium supplemented with 10% fetal bovine serum (Thermo Fisher Scientific, Inc.) at 37˚C and 5% CO2. When cells reached 70% confluency, they were transfected with the small-interfering RNA of IL-10RA1 (siR-IL-10RA1; 5'-GGTCTACAGCATCGAGTAT-3'; Hanbio Biotechnology Co., Ltd.) or siR-NC (non-targeting siRNA sequences; Hanbio Biotechnology Co., Ltd.) using Lipofectamine 3000® (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. Briefly, 1.5 µl siR-IL-10RA1 or siR-NC (50 pmol/µl; Hanbio Biotechnology Co., Ltd.) was mixed with 50 µl Opti Mem medium (Thermo Fisher Scientific, Inc.) in a vial. In another vial, 1 µl Lipofectamine 3000® (Invitrogen; Thermo Fisher Scientific, Inc.) was mixed with 50 µl Opti Mem medium. After standing for 5 min, the two vials were combined for further incubation at room temperature for another 20 min. Then, the mixtures were added onto cells in thesiR-IL-10RA1 and siR-NC groups. Next, 6 h later, the medium was replaced with RPMI-1640 medium containing 10% fetal bovine serum. After culturing at 37˚C for 48 h, the cells were collected for subsequent experimentation. To examine the effect of IL-10 on fibroblasts, MRC-5 cells were cultured with the serum of IPF patients (serum group) or the serum of IPF patients containing IL-10 antibody (serum+IL-10 antibody group) at 37˚C and 5% CO2 for 24 h. Untreated MRC-5 cells were used as negative control (NC) group.

The present study included eight cases of primary cell culture and five cases were successful. Cellular experiments were performed on the five successful cases.

Enzyme-linked immunosorbent assay (ELISA)

HumanIL-10 ELISA kit (abs510005-96T; Absin Bioscience Inc.) was used to determine the concentration of IL-10 in serum. In microplates, standards (100 µl) and samples (100 µl serum) were added into predefined wells, while blank wells were left empty. In the wells for standards and samples, horseradish peroxidase-labelled conjugates (100 µl) were added before sealing the plates for incubation at 37˚C for 1 h. After washing the plates 5 times, substrates A (50 µl) and B (50 µl) were added into each well. After incubation at 37˚C for 15 min, stop solution (50 µl) was added into each well, and absorbance of each well was measured at 450 nm within 15 min.

Reverse transcription-quantitative (RT-q) PCR

Transfected cells (1x106) were directly lysed with 1 ml TRIzol® reagent (Invitrogen; Thermo Fisher Scientific Inc.) at room temperature for 30 min. Total RNA was extracted using the phenol chloroform method. The concentration and quality of RNA was measured using ultraviolet spectrophotometry (NanoDrop ND2000; Thermo Fisher Scientific Inc.). The acceptable concentration was 50-150 ng/µl and the acceptable ratio of A260/A280 was between 1.8 and 2.0. Next, cDNA was obtained by reverse transcription from 1 µg RNA and stored at -20˚C. Reverse transcription of mRNA was performed at 50˚C for 45 min using TIANScript II cDNA First Strand Synthesis kit (Tiangen Biotech Co., Ltd.) according to the manufacturer's manual. SuperReal PreMix (SYBR-Green) RT-qPCR kit (Tiangen Biotech Co., Ltd.) was used to detect mRNA expression of IL-10R1, using GAPDH as an internal reference gene. The sequences of IL-10R1 were: forward, 5'-TGAAAACAAGAGCAAGGCCG-3' and reverse, 5'-ATCCCTCCGAGACACTGGAA-3'. The sequences of GAPDH used were: forward, 5'-GGAGCGAGATCCCTCCAAAAT-3' and reverse, 5'-GGCTGTTGTCATACTTCTCATGG-3'. The reaction system (20 µl) was composed of 10 µl SYBR Premix EXTaq, 0.5 µl upstream primer, 0.5 µl downstream primer, 2 µl cDNA and 7 µl ddH2O. The thermocycling conditions used were as follows: Initial denaturation at 95˚C for 10 min; denaturation at 95˚C for 1 min and annealing at 60˚C for 30 sec (40 cycles); elongation at 72˚C for 30 sec and were performed using the iQ5 instrument (Bio-Rad Laboratories Inc.). The 2-ΔΔCq method (27) was used to calculate the relative expression of IL-10R1 mRNA against GAPDH. Each sample was tested in triplicate.

CCK-8 assay

MRC-5 cells were trypsinized and seeded into 96-well plates at a density of 2x103/well. At 0, 24, 48 and 72 h, the medium was discarded, and the cells were washed with phosphate-buffered saline (PBS) twice, followed by addition of DMEM medium and 10 µl CCK-8 reaction reagent (5 g/l; Beyotime Institute of Biotechnology). After incubation at 37˚C and 5% CO2 for 2 h, the absorbance of each well was measured at 490 nm for plotting cell viability curves.

Flow cytometry

Flow cytometry was used for cell cycle analysis. At 24 h after transfection or treatments with serum, 1x106 MRC-5 cells of NC, serum and serum + IL-10 antibody groups or siR-NC and siR-IL-10R1 groups were washed twice with precooled PBS. The centrifugation was at 2,000 x g for 10 min and 4˚C. BD Cycletest Plus DNA Reagent kit (catalogue no. 340242; BD Biosciences) was used to perform the cell cycle analysis according to the manufacturer's instructions. The cells were permeabilized using the liquid provided in BD Cycletest Plus DNA Reagent kit (catalogue no. 340242; BD Biosciences). The staining buffer contained RNAse. The cells were incubated with 200 µl liquid A for 10 min and 150 µl liquid B for another 10 min. Then, the cells were incubated with 120 µl liquid C in the dark for 10 min before flow cytometry (FACSCalibur; BD Biosciences). The result was analyzed using ModFit software version 3.2 (Verity Software House Inc.).

For separating peripheral mononuclear lymphocytes, 3 ml sterile mononuclear cell separation solution was added into 15 ml tube before gently adding 2 ml peripheral blood on top of the solution. After centrifugation at 100 x g at room temperature for 20 min, the layer of peripheral mononuclear lymphocytes was gently aspirated and mixed with 10 ml sterile PBS, before centrifugation at 100 x g at room temperature for 5 min. Then, the supernatant was discarded and 5 ml PBS was added to resuspend the cells. After centrifugation at 100 x g at room temperature for 5 min, 250 µl BD Cytofix/Cytoperm reagent (BD Biosciences) was used to resuspend the cells, which were incubated at 4˚C for 20 min and permeabilized. Then, 1 ml PBS was added to stop perforation. After centrifugation at 100 x g at room temperature for 5 min, the cells were collected and stained with IL-10 antibody (1:3,000; cat.no. PI528; Beyotime Institute of Biotechnology) in the dark at room temperature for 30 min. Finally, the cells were examined by flow cytometry (FACSCalibur; BD Biosciences). The result was analyzed using ModFit software version 3.2 (Verity Software House Inc.).

Western blotting

After treatment for 48 h, the cells in NC, serum and serum+IL-10 antibody groups or siR-NC and siR-IL-10R1 groups were collected and washed with PBS twice. Then, the cells were lysed with 600 µl precooled Radio-Immunoprecipitation Assay (RIPA) lysis buffer (Beyotime Institute of Biotechnology) for 5 min on ice. The mixture was centrifuged at 13,000 x g and 4˚C for 10 min. The supernatant was used to determine protein concentration using the bicinchoninic acid (BCA) protein concentration determination kit (RTP7102; Real-Times Biotechnology Co., Ltd.). Subsequently, the samples were mixed with 5X sodium dodecyl sulfate loading buffer before denaturation in a boiling water bath for 10 min. Subsequently, the protein samples (20 µg/lane) were subjected to 10% sodium dodecyl sulfate-polyacrylamide gel electrophoresis at 100 V. The resolved proteins were transferred to polyvinylidene difluoride membranes on ice (250 mA; 1 h) and blocked with 5% skimmed milk at room temperature for 1 h. Then, the membranes were incubated with rabbit anti-human IL-10R1 (1:1,000; abs136163), IL-10R2 (1:1,000; abs138477), collagen type I α1 chain (COL1a1; 1:1,000; abs118788), collagen type I α2 chain (COL1a2; 1:1,000; abs101119) or GAPDH (1:4,000; abs132004) polyclonal primary antibodies (all from Absin Bioscience Inc.) at 4˚C overnight. After extensive washing with PBS with 0.1% Tween-20 3 times (each wash for 15 min), the membranes were incubated with goat anti-rabbit IgG-horseradish peroxidase-conjugated secondary antibody (1:4,000; abs20040; Absin, Shanghai, China) for 1 h at room temperature before washing with PBS with Tween 20 3 times (each wash for 15 min). Subsequently, membrane development was performed using the enhanced chemiluminescence detection kit (Abcam) for imaging. Image lab v.3.0 software (Bio-Rad Laboratories Inc.) was used to acquire and analyze imaging signals. GAPDH was used as the loading control.

Immunohistochemistry

Sterilized cover glasses were placed in a 90 mm culture dish, and then the cells were seeded in the culture dish at a density of 2x104/ml for cell climbing. On the following day, the cells attached on the cover slip were washed with PBS for three times of 2 min. Then, the cells were fixed with 4% paraformaldehyde at room temperature for 15 min and dried under room temperature for 5 min. After washing with PBS for 3 times of 2 min, the cells were incubated with 0.5% Triton X-100 (dissolved in DPBS) at room temperature for 20 min before washing again with PBS for 3 times of 2 min. After addition of 3% H2O2, the cells were incubated for 15 min at room temperature. Following washing with PBS for 3 times of 2 min, the cells were incubated with blocking serum at room temperature for 20 min. Then, primary vimentin antibody (abs131996; Absin, Shanghai, China) was added before incubation at 40˚C overnight. Subsequently, the cells were incubated with secondary antibody pv6001 at 37˚C for 30 min. After washing with PBS for 5 times of 2 min, 1 drop of diaminobenzidine (DAB) liquid was added to the section was dripped with. After 10 min, the section was washed with water for 5 min. After staining with hematoxylin at room temperature for 10 min, the section was washed with water for 5 min before being sealed in gum.

Statistical analysis

Results were analyzed using SPSS 18.0 (IBM Corp.). The data were expressed as means ± standard deviations. The number of biological replicates was three. Comparison between two groups was carried out using paired Student's t-tests. Multiple group comparisons were performed by one-way analysis of variance followed by a post hoc Student-Newman-Keuls test. P<0.05 was considered to indicate a statistically significant difference.

Results

IL-10 promotes the viability and collagen synthesis of MRC-5 cells and primary pulmonary fibroblasts

To measure the levels of IL-10 in serum samples from 42 IPF patients and 20 healthy subjects, ELISA was performed. The data demonstrated that the level of IL-10 in serum from patients with (135.7±12.5 ng/µl) was significantly higher compared with that in normal subjects (21.6±5.8 ng/µl) (P<0.05; Fig. 1A). To examine the effect of IL-10 on fibroblasts, MRC-5 cells were cultured with the serum of patients with IPF for 24 h in the absence or presence of IL-10 antibody. The CCK-8 assay demonstrated that the absorbance of MRC-5 cells stimulated with serum from patients with IPF was significantly higher compared with that of MRC-5 cells treated with serum from healthy subjects at 24, 48 and 72 h (P<0.05 for all time points; Fig. 1B), while co-incubation with IL-10 antibody reduced the absorbance of MRC-5 cells to a level similar to that of MRC-5 cells treated with serum from healthy subjects (P>0.05; Fig. 1B). Flow cytometry demonstrated that, compared with MRC-5 cells that were not treated with serum (NC group), the transition from G1 to S phase in MRC-5 cells treated with serum from IPF patients was accelerated (P<0.05; Fig. 1C), while treatment with IL-10 antibody reversed this to a level similar to NC group (P>0.05; Fig. 1C). Western blotting demonstrated that COL1a1 and COL1a2 protein expression in MRC-5 cells treated with serum from IPF patients was significantly higher compared with that in the MRC-5 cells of the negative control group (P<0.05; Fig. 1D), while treatment with IL-10 antibody reversed this to a level similar to the negative control group (P>0.05; Fig. 1D). To further confirm the effect of IL-10 on fibroblasts, primary fibroblasts were isolated and identified. Microscopy demonstrated that primary pulmonary fibroblasts were spindle-shaped (yellow-brown indicated positive signals), and immunohistochemistry detected the marker protein vimentin in the fibroblasts (Fig. 2A). The present study included 8 cases of primary cell culture and 5 cases were successful. Cellular experiments were performed on the 5 successful cases. Similar results were observed in primary fibroblasts compared to those from MRC-5 cells (Fig. 2B-D). The aforementioned results suggested that IL-10 promotes the viability and collagen synthesis of MRC-5 cells and primary pulmonary fibroblasts.

IL-10 and IL-10R1 play regulatory roles in the viability and collagen synthesis of MRC-5 cells

To investigate the mechanism by which IL-10 regulates MRC-5 cells, the mRNA and protein expression of IL-10 receptor IL-10R1. RT-qPCR and western blotting demonstrated that IL-10R1 mRNA and protein expression in MRC-5 cells treated with serum from patients with IPF was significantly higher compared with that in the negative control group (P<0.05; Fig. 3A and B), while addition of IL-10 antibody did not decrease the level of IL-10R1 mRNA and protein compared with serum group (P>0.05; Fig. 3A and B). After transfecting MRC-5 cells with siR-IL-10R1, the expression of IL-10R1 protein was decreased compared with the siR-NC group (P<0.05; Fig. 3C). The CCK-8 assay demonstrated that the viability of MRC-5 cells in the siR-IL-10R1 group was decreased compared with the siR-NC group (P<0.05 at all time points; Fig. 3D). Flow cytometry demonstrated that siR-IL-10R1 slowed down the transition from G1 phase to S phase compared with that of siR-NC group (P<0.05; Fig. 3E). In addition, expression of COL1a1 and COL1a2 in siR-IL-10R1 group was lower compared with that in the siR-NC group (Fig. 3F). The aforementioned results suggested that IL-10 and IL-10R1 serve regulatory roles in the viability and collagen synthesis of MRC-5 cells.

The ratio of peripheral mononuclear lymphocytes with positive expression of IL-10 is elevated in peripheral blood from patients with IPF compared with healthy subjects

To detect the expression of IL-10 in peripheral mononuclear lymphocytes, flow cytometry was performed. The data demonstrated that the ratio of IL-10+ peripheral mononuclear lymphocytes in patients with IPF was significantly higher compared with that in normal subjects (P<0.05; Fig. 4A and B). Peripheral mononuclear lymphocyte population itself looked much smaller in the normal subjects compared with the patients with IPF, possibly because of the proliferation of inflammatory cells caused by IPF inflammation. The aforementioned result indicated that the ratio of peripheral mononuclear lymphocytes with positive expression of IL-10 is elevated in peripheral blood from patients with IPF.

Discussion

Chronic airway inflammation, myofibroblast proliferation and ECM synthesis are the main mechanisms of pulmonary subepithelial fibrosis in patients with interstitial pneumonia (28,29). In recent years, pulmonary fibroblasts have become important targets in improving airway remodeling (30). Hence, the role of pulmonary fibroblasts in airway remodeling in asthma has attracted increasing attention. IL-10 is a type of cytokine that has attracted a lot of attention in recent years (31). Numerous tissues and cells, such as T cells, B cells, macrophages, bronchial epithelial cells and tumor cells produce IL-10 (32,33). IL-10 is a multidirectional immunoregulatory factor and its main activity is immunosuppression (34). Studies have demonstrated that IL-10 has different biological functions in a number of other cells. For example, IL-10 induced the expression of E-selectin on the surface of vascular endothelial cells and regulated the barrier function of endothelial cells (35). Treatment targeting IL-10 inhibited the apoptosis of CD8+ cells induced by dendritic cells (36). In addition, IL-10 serves important roles in lung injury and pneumonia. For example, the expression of IL-10 and TGF-β in biopsy tissues of patients with IPF was significantly increased compared with that from healthy subjects and involved in pulmonary fibrosis, indicating that they are important therapeutic targets (37). In addition, a study demonstrated that IL-10 delivered by hydrogel improved treatment of bleomycin-induced lung fibrosis in mice (25). Consistent with a previous report (38), the present study revealed that expression of IL-10 in peripheral blood of patients with IPF was significantly increased compared with normal subjects, suggesting that IL-10 may serve a role in pulmonary fibrosis. Pulmonary fibroblasts serve important roles in pulmonary fibrosis and secrete cytokines that are involved in the local inflammatory response and collagen secretion inducing matrix microenvironment remodeling (37). During the occurrence and development of interstitial pneumonia, abnormal proliferation of pulmonary fibroblasts occurs and the ability of collagen synthesis increases significantly (39). In the present study, primary pulmonary fibroblasts were cultured with the serum of patients with IPF which stimulated the viability and collagen synthesis of MRC-5 cells and primary pulmonary fibroblasts. Following addition of IL-10 antibody, the stimulation effect by IPF serum was reduced which suggested that IL-10 promoted the viability and collagen synthesis of lung fibroblasts.

IL-10 is a secretory cytokine that transmits extracellular signals mainly by binding to its membrane surface receptor IL-10R (40). IL-10R mainly consists of IL-10R1 and IL-10R2 subunits, and IL-10 first binds to IL-10R1 and then binds to IL-10R2 to activate downstream signaling pathways (41). IL-10/IL-10R pathway is involved in numerous pathological processes (42). For example, detection of IL-10 expression in serum may help predict the prognosis of patients with rheumatoid arthritis (43). In addition, IL-10 promoted drug resistance in non-small cell lung cancer (44). The present study demonstrated that the expression of IL-10R1, but not IL-10R2, in primary fibroblasts was increased significantly compared with untreated primary fibroblasts after treatment with serum from IPF patients. Serum from IPF patients could promote collagen secretion of fibroblasts, and IL-10 antibody was able to block this effect. Following interference with the expression of IL-10R1, the treatment effect of IPF serum on primary fibroblasts was significantly weakened in the present study. This suggested that IL-10/IL-10R1 promoted the viability and collagen secretion of primary fibroblasts. IL-10 can be secreted by a variety of cells, of which monocytes are the main cells (45). Using flow cytometry, the present study demonstrated that the number of IL-10-positive mononuclear lymphocytes in peripheral blood of patients with IPF was significantly higher compared with that in normal subjects, which suggested that mononuclear lymphocytes secreted IL-10 in patients with IPF and IL-10 promoted the viability and collagen secretion of lung fibroblasts leading to pulmonary fibrosis.

The present study had several limitations. In the current study, the heterogeneity of primary fibroblasts was very large and even the generational differences between fibroblasts from the same patient were very large. If cell sorting was used to select monoclonal cells, the heterogeneity could have been avoided to some extent, but diversity would have been lost at the same time. In addition, the present study only suggested that monocytes secrete IL-10 that acts on fibroblasts as there is infiltration by a lot of monocytes in interstitial pneumonia. In order to prove that IL-10 is derived from monocytes, more evidences required from future studies.

In conclusion, the present study demonstrated that the IL-10 level in peripheral blood of patients with IPF is increased significantly compared with normal subjects. Activation of the IL-10/IL-10R1 signaling pathway promotes the viability and collagen synthesis of pulmonary fibroblasts leading to pulmonary fibrosis. The present study explored the mechanism of IL-10 in the occurrence and development of interstitial pneumonia, and provided experimental basis for understanding the role of inflammatory factors in this disease.

Acknowledgements

Not applicable.

Funding

Funding: This work was supported by the Subject Fund of Zhejiang Provincial Science and Technology Hall (grant no. LGF19H010006) and Lishui Science and Technology Bureau Project (grant no. 2016GYX29).

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

HY, JP, XC, ZC, EC and JQ contributed to the design of the study. HY, JP, XC, ZY, LL, EG, CX and HZ performed the experiments. HY and JP confirm the authenticity of all the raw data. HY, JP and XC analyzed the data. HY, JP, XC, ZC, EC and JQ interpreted results and drafted the manuscript. All authors have read and approved the manuscript.

Ethics approval and consent to participate

All procedures performed in the current study were approved by the Ethics Committee of Wenzhou Medical University (Lishui, China) with approval no. IACUC-20180901-28. Written informed consent was obtained from all subjects or their families.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Hobbs BD, Putman RK, Araki T, Nishino M, Gudmundsson G, Gudnason V, Eiriksdottir G, Zilhao Nogueira NR, Dupuis J, Xu H, et al: Overlap of genetic risk between interstitial lung abnormalities and idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 200:1402–1413. 2019.PubMed/NCBI View Article : Google Scholar

2 

Pereira CAC, Soares MR, Boaventura R, Castro MDC, Gomes PS, Gimenez A, Fukuda C, Cerezoli M and Missrie I: Squawks in interstitial lung disease prevalence and causes in a cohort of one thousand patients. Medicine (Baltimore). 98(e16419)2019.PubMed/NCBI View Article : Google Scholar

3 

Singh P, Thakur B, Mohapatra AK and Padhan P: Clinical features and outcome of acute exacerbation in connective tissue disease-associated interstitial lung disease: A single-center study from India. Int J Rheum Dis. 22:1741–1745. 2019.PubMed/NCBI View Article : Google Scholar

4 

Gasperini ML, Gigante A, Iacolare A, Pellicano C, Lucci S and Rosato E: The predictive role of lung ultrasound in progression of scleroderma interstitial lung disease. Clin Rheumatol. 39:119–123. 2020.PubMed/NCBI View Article : Google Scholar

5 

Willemsen AECAB, Tol J, van Erp NP, Jonker MA, de Boer M, Meek B, de Jong PC, van Moorsel C, Gerritsen WR, Grutters JC and van*Herpen CML: Prospective study of drug-induced interstitial lung disease in advanced breast cancer patients receiving everolimus plus exemestane. Target Oncol. 14:441–451. 2019.PubMed/NCBI View Article : Google Scholar

6 

Saito S, Alkhatib A, Kolls JK, Kondoh Y and Lasky JA: Pharmacotherapy and adjunctive treatment for idiopathic pulmonary fibrosis (IPF). J Thorac Dis. 11 (Suppl 14):S1740–S1754. 2019.PubMed/NCBI View Article : Google Scholar

7 

Ussavarungsi K, Nugent K, Gerke AK, Krasowski MD, Tuetken RS and Lenert PS: Interstitial lung disease associated with anti-PM-Scl antibody: A single center experience. Autoimmun Rev. 18(102355)2019.PubMed/NCBI View Article : Google Scholar

8 

Chai GT, Tan TC, Lee YS, Kaw GJ, Chuah KL, Lim YJ, Abisheganaden JA and Thong BY: Impact of an interstitial lung disease service in the diagnosis and management of interstitial lung disease in Singapore. Singapore Med J. 61:302–307. 2019.PubMed/NCBI View Article : Google Scholar

9 

Chen Z, Wang X and Ye S: Tofacitinib in Amyopathic Dermatomyositis-associated interstitial lung disease. N Engl J Med. 381:291–293. 2019.PubMed/NCBI View Article : Google Scholar

10 

Hoffmann-Vold AM, Fretheim H, Halse AK, Seip M, Bitter H, Wallenius M, Garen T, Salberg A, Brunborg C, Midtvedt Ø, et al: Tracking impact of interstitial lung disease in systemic sclerosis in a complete nationwide cohort. Am J Respir Crit Care Med. 200:1258–1266. 2019.PubMed/NCBI View Article : Google Scholar

11 

Ye Y, Fu Q, Wang R, Guo Q and Bao C: Serum KL-6 level is a prognostic marker in patients with anti-MDA5 antibody-positive dermatomyositis associated with interstitial lung disease. J Clin Lab Anal. 33(e22978)2019.PubMed/NCBI View Article : Google Scholar

12 

Lee JU, Son JH, Shim EY, Cheong HS, Shin SW, Shin HD, Baek AR, Ryu S, Park CS, Chang HS and Park JS: Global DNA methylation pattern of fibroblasts in idiopathic pulmonary fibrosis. DNA Cell Biol. 38:905–914. 2019.PubMed/NCBI View Article : Google Scholar

13 

Kheirollahi V, Wasnick RM, Biasin V, Vazquez-Armendariz AI, Chu X, Moiseenko A, Weiss A, Wilhelm J, Zhang JS, Kwapiszewska G, et al: Metformin induces lipogenic differentiation in myofibroblasts to reverse lung fibrosis. Nat Commun. 10(2987)2019.PubMed/NCBI View Article : Google Scholar

14 

Caporarello N, Meridew JA, Jones DL, Tan Q, Haak AJ, Choi KM, Manlove LJ, Prakash YS, Tschumperlin DJ and Ligresti G: PGC1α repression in IPF fibroblasts drives a pathologic metabolic, secretory and fibrogenic state. Thorax. 74:749–760. 2019.PubMed/NCBI View Article : Google Scholar

15 

Woodley DT: Distinct fibroblasts in the papillary and reticular dermis: Implications for wound healing. Dermatol Clin. 35:95–100. 2017.PubMed/NCBI View Article : Google Scholar

16 

Schruf E, Schroeder V, Kuttruff CA, Weigle S, Krell M, Benz M, Bretschneider T, Holweg A, Schuler M, Frick M, et al: Human lung fibroblast-to-myofibroblast transformation is not driven by an LDH5-dependent metabolic shift towards aerobic glycolysis. Respir Res. 20(87)2019.PubMed/NCBI View Article : Google Scholar

17 

Woodcock HV, Eley JD, Guillotin D, Platé M, Nanthakumar CB, Martufi M, Peace S, Joberty G, Poeckel D, Good RB, et al: The mTORC1/4E-BP1 axis represents a critical signaling node during fibrogenesis. Nat Commun. 10(6)2019.PubMed/NCBI View Article : Google Scholar

18 

Ebrahimpour A, Shrestha S, Bonnen MD, Eissa NT, Raghu G and Ghebre YT: Nicotine modulates growth factors and MicroRNA to promote inflammatory and fibrotic processes. J Pharmacol Exp Ther. 368:169–178. 2019.PubMed/NCBI View Article : Google Scholar

19 

Nie Y, Zhang D, Qian F and Wu Y: Baccatin III ameliorates bleomycin-induced pulmonary fibrosis via suppression of TGF-β1 production and TGF-β1-induced fibroblast differentiation. Int Immunopharmacol. 74(105696)2019.PubMed/NCBI View Article : Google Scholar

20 

Wynn TA: Integrating mechanisms of pulmonary fibrosis. J Exp Med. 208:1339–1350. 2011.PubMed/NCBI View Article : Google Scholar

21 

Nenov MN, Konakov MV, Teplov IY and Levin SG: Interleukin-10 facilitates glutamatergic synaptic transmission and homeostatic plasticity in cultured hippocampal neurons. Int J Mol Sci. 20(3375)2019.PubMed/NCBI View Article : Google Scholar

22 

Krakow S, Crescimone ML, Bartels C, Wiegering V, Eyrich M, Schlegel PG and Wölfl M: Re-expression of CD14 in response to a combined IL-10/TLR stimulus defines monocyte-derived cells with an immunoregulatory phenotype. Front Immunol. 10(1484)2019.PubMed/NCBI View Article : Google Scholar

23 

Xiao S, Huang G, Wei Z, Nie K, Liu Z, Deng C and Wang D: IL-10 Gene-modified human amniotic mesenchymal stem cells augment regenerative wound healing by multiple synergistic effects. Stem Cells Int. 2019(9158016)2019.PubMed/NCBI View Article : Google Scholar

24 

Sheridan MP, Browne JA, Doyle MB, Fitzsimons T, McGill K and Gormley E: IL-10 suppression of IFN-γ responses in tuberculin-stimulated whole blood from Mycobacterium bovis infected cattle. Vet Immunol Immunopathol. 189:36–42. 2017.PubMed/NCBI View Article : Google Scholar

25 

Shamskhou EA, Kratochvil MJ, Orcholski ME, Nagy N, Kaber G, Steen E, Balaji S, Yuan K, Keswani S, Danielson B, et al: Hydrogel-based delivery of Il-10 improves treatment of bleomycin-induced lung fibrosis in mice. Biomaterials. 203:52–62. 2019.PubMed/NCBI View Article : Google Scholar

26 

Raghu G, Collard HR, Egan JJ, Martinez FJ, Behr J, Brown KK, Colby TV, Cordier JF, Flaherty KR, Lasky JA, et al: An official ATS/ERS/JRS/ALAT statement: Idiopathic pulmonary fibrosis: Evidence-based guidelines for diagnosis and management. Am J Respir Crit Care Med. 183:788–824. 2011.PubMed/NCBI View Article : Google Scholar

27 

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.PubMed/NCBI View Article : Google Scholar

28 

Deterding RR, DeBoer EM, Cidon MJ, Robinson TE, Warburton D, Deutsch GH and Young LR: Approaching clinical trials in childhood interstitial lung disease and pediatric pulmonary fibrosis. Am J Respir Crit Care Med. 200:1219–1227. 2019.PubMed/NCBI View Article : Google Scholar

29 

Algamdi M, Sadatsafavi M, Fisher JH, Morisset J, Johannson KA, Fell CD, Kolb M, Manganas H, Cox G, Gershon AS, et al: Costs of workplace productivity loss in patients with fibrotic interstitial lung disease. Chest. 156:887–895. 2019.PubMed/NCBI View Article : Google Scholar

30 

Wen J, Lin X, Gao W, Qu B, Zuo Y, Liu R and Yu M: Inhibition of LPA1 Signaling impedes conversion of human Tenon's fibroblasts into myofibroblasts via suppressing TGF-β/Smad2/3 signaling. J Ocul Pharmacol Ther. 35:331–340. 2019.PubMed/NCBI View Article : Google Scholar

31 

Saraiva M and O'Garra A: The regulation of IL-10 production by immune cells. Nat Rev Immunol. 10:170–181. 2010.PubMed/NCBI View Article : Google Scholar

32 

Wang Y, Sun SN, Liu Q, Yu YY, Guo J, Wang K, Xing BC, Zheng QF, Campa MJ, Patz EF Jr, et al: Autocrine complement inhibits IL10-dependent T-cell-mediated antitumor immunity to promote tumor progression. Cancer Discov. 6:1022–1035. 2016.PubMed/NCBI View Article : Google Scholar

33 

Yang S, Tang X, Sheng X, Xing J and Zhan W: Analysis of the role of IL-10 in the phagocytosis of mIgM+ B lymphocytes in flounder (Paralichthys olivaceus). Fish Shellfish Immunol. 92:813–820. 2019.PubMed/NCBI View Article : Google Scholar

34 

Ramakrishna C, Kujawski M, Chu H, Li L, Mazmanian SK and Cantin EM: Bacteroides fragilis polysaccharide A induces IL-10 secreting B and T cells that prevent viral encephalitis. Nat Commun. 10(2153)2019.PubMed/NCBI View Article : Google Scholar

35 

Vora M, Romero LI and Karasek MA: Interleukin-10 induces E-selectin on small and large blood vessel endothelial cells. J Exp Med. 184:821–829. 1996.PubMed/NCBI View Article : Google Scholar

36 

Qiao J, Liu Z, Dong C, Luan Y, Zhang A, Moore C, Fu K, Peng J, Wang Y, Ren Z, et al: Targeting Tumors with IL-10 prevents dendritic cell-mediated CD8+ T cell apoptosis. Cancer Cell. 35:901–915.e4. 2019.PubMed/NCBI View Article : Google Scholar

37 

Bergeron A, Soler P, Kambouchner M, Loiseau P, Milleron B, Valeyre D, Hance AJ and Tazi A: Cytokine profiles in idiopathic pulmonary fibrosis suggest an important role for TGF-beta and IL-10. Eur Respir J. 22:69–76. 2003.PubMed/NCBI View Article : Google Scholar

38 

Papiris SA, Tomos IP, Karakatsani A, Spathis A, Korbila I, Analitis A, Kolilekas L, Kagouridis K, Loukides S, Karakitsos P and Manali ED: High levels of IL-6 and IL-8 characterize early-on idiopathic pulmonary fibrosis acute exacerbations. Cytokine. 102:168–172. 2018.PubMed/NCBI View Article : Google Scholar

39 

Wollin L, Distler JH, Redente EF, Riches DW, Stowasser S, Schlenker-Herceg R, Maher TM and Kolb M: Potential of nintedanib in treatment of progressive fibrosing interstitial lung diseases. Eur Respir J. 54(1900161)2019.PubMed/NCBI View Article : Google Scholar

40 

Kotlarz D, Beier R, Murugan D, Diestelhorst J, Jensen O, Boztug K, Pfeifer D, Kreipe H, Pfister ED, Baumann U, et al: Loss of interleukin-10 signaling and infantile inflammatory bowel disease: Implications for diagnosis and therapy. Gastroenterology. 143:347–355. 2012.PubMed/NCBI View Article : Google Scholar

41 

Glocker EO, Kotlarz D, Boztug K, Gertz EM, Schäffer AA, Noyan F, Perro M, Diestelhorst J, Allroth A, Murugan D, et al: Inflammatory bowel disease and mutations affecting the interleukin-10 receptor. N Engl J Med. 361:2033–2045. 2009.PubMed/NCBI View Article : Google Scholar

42 

Engelhardt KR, Shah N, Faizura-Yeop I, Kocacik Uygun DF, Frede N, Muise AM, Shteyer E, Filiz S, Chee R, Elawad M, et al: Clinical outcome in IL-10- and IL-10 receptor-deficient patients with or without hematopoietic stem cell transplantation. J Allergy Clin Immunol. 131:825–830. 2013.PubMed/NCBI View Article : Google Scholar

43 

McDonald S, Reed R and Baricevic-Jones I: Biologics in Rheumatoid Arthritis Genetics and Genomics Study Syndicate. Ling S, Plant D and Barton A: Can serum interleukin-17 and interleukin-10 levels predict response to biologic treatments in patients with rheumatoid arthritis? Rheumatology (Oxford). 58:1872–1873. 2019.PubMed/NCBI View Article : Google Scholar

44 

Vahl JM, Friedrich J, Mittler S, Trump S, Heim L, Kachler K, Balabko L, Fuhrich N, Geppert CI, Trufa DI, et al: Interleukin-10-regulated tumour tolerance in non-small cell lung cancer. Br J Cancer. 117:1644–1655. 2017.PubMed/NCBI View Article : Google Scholar

45 

Lopes RL, Borges TJ, Zanin RF and Bonorino C: IL-10 is required for polarization of macrophages to M2-like phenotype by mycobacterial DnaK (heat shock protein 70). Cytokine. 85:123–129. 2016.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

August-2022
Volume 24 Issue 2

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ye H, Pan J, Cai X, Yin Z, Li L, Gong E, Xu C, Zheng H, Cao Z, Chen E, Chen E, et al: IL‑10/IL‑10 receptor 1 pathway promotes the viability and collagen synthesis of pulmonary fibroblasts originated from interstitial pneumonia tissues. Exp Ther Med 24: 518, 2022
APA
Ye, H., Pan, J., Cai, X., Yin, Z., Li, L., Gong, E. ... Qian, J. (2022). IL‑10/IL‑10 receptor 1 pathway promotes the viability and collagen synthesis of pulmonary fibroblasts originated from interstitial pneumonia tissues. Experimental and Therapeutic Medicine, 24, 518. https://doi.org/10.3892/etm.2022.11445
MLA
Ye, H., Pan, J., Cai, X., Yin, Z., Li, L., Gong, E., Xu, C., Zheng, H., Cao, Z., Chen, E., Qian, J."IL‑10/IL‑10 receptor 1 pathway promotes the viability and collagen synthesis of pulmonary fibroblasts originated from interstitial pneumonia tissues". Experimental and Therapeutic Medicine 24.2 (2022): 518.
Chicago
Ye, H., Pan, J., Cai, X., Yin, Z., Li, L., Gong, E., Xu, C., Zheng, H., Cao, Z., Chen, E., Qian, J."IL‑10/IL‑10 receptor 1 pathway promotes the viability and collagen synthesis of pulmonary fibroblasts originated from interstitial pneumonia tissues". Experimental and Therapeutic Medicine 24, no. 2 (2022): 518. https://doi.org/10.3892/etm.2022.11445