Effect of transplanted adipose‑derived stem cells in mice exhibiting idiopathic pulmonary fibrosis

  • Authors:
    • Hongbin Jiang
    • Jun Zhang
    • Zhemin Zhang
    • Shengxiang Ren
    • Chuansen Zhang
  • View Affiliations

  • Published online on: August 5, 2015     https://doi.org/10.3892/mmr.2015.4178
  • Pages: 5933-5938
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Stem cell‑based cell therapy has provided a promising method for the treatment of pulmonary diseases, including idiopathic pulmonary fibrosis (IPF). Furthermore, adipose‑derived stem cells (ADSCs) have been reported to be effective in lung repair and regeneration. In the current study, IPF was induced in mice by intratracheal instillation of bleomycin (BLM), and ADSCs were delivered systemically into the mice via the tail vein to evaluate the effects of ADSC transplantation. The ADSC engraftment rate and morphometric changes in lung tissue samples in vivo were investigated by histochemistry and immunohistochemistry, as well as by western blotting. The results indicated that ADSCs may relieve IPF and provide a significant contribution to lung repair when administered at an early stage.

Introduction

Idiopathic pulmonary fibrosis (IPF) is a refractory and lethal form of progressive fibrotic lung disorder that is characterized by fibroblast proliferation, extracellular matrix deposition and progressive lung scarring. IPF exhibits the histopathologic pattern of usual interstitial pneumonia and the incidence of IPF worldwide has gradually increased annually (1). Mesenchymal stem cell (MSC) therapy has previously been reported as a promising method for lung tissue regeneration and protection (2). Zhang et al revealed that MSCs are important in lung repair and promote collagen fiber deposition following induction of secondary damage in alveolar type (AT) II cells using rAAV-SPA-TK, which involves hypoxia-inducible factor-1α and stromal cell-derived factor 1 signaling (3). Moodley et al reported that human umbilical cord MSCs reduce fibrosis of bleomycin (BLM)-induced lung injury (4), and Gupta et al reported that intrapulmonary delivery of bone marrow-derived MSCs (BMSCs) improves survival and attenuates endotoxin-induced acute lung injury in mice (5).

However, reports on the effect of transplanted adipose-derived stem cells (ADSCs) in mice with IPF were rare. When compared with BMSCs, ADSCs are more abundant, widely available and possess similar properties to BMSCs, such as multiple differentiation potential, high proliferation and immunoregulatory ability (6,7). Therefore, investigating the effect of ADSCs in mice with IPF was required.

In the present study, it was hypothesized that transplanted ADSCs could promote the repair of lung injury and relieve IPF. To test the hypothesis, ADSCs were delivered systemically into IPF mice via the tail vein to assess the effects.

Materials and methods

Animals

Sixty C57BL/6 mice (weight, 12–16 g) were procured from the Center of Experimental Animals, Shanghai Medical College, Fudan University (Shanghai, China). Mice were given free access to water and a standard rodent diet (no. 2920; Harlan Laboratories, Indianapolis, IN, USA).

To induce fibrotic changes, the animals were intratracheally injected with BLM (Blenoxane; Mead Johnson, Princeton, NJ, USA) at a dose of 2.5 U/kg mouse body weight, as previously described (8). All of the animal studies were reviewed and approved by the Ethics Committee of Tongji University School of Medicine (Shanghai, China).

Isolation and expansion of mouse ADSCs

ADSCs were harvested from the inguinal groove of the mice. The adipose tissue samples were isolated and incubated in alpha-minimal essential medium (MEM) containing 10% fetal bovine serum (FBS; GE Healthcare Life Sciences, Logan, UT, USA) and 0.1% collagenase type IV solution at 37°C for 45–60 min. The digested tissues were filtered through 45-mm nylon filter mesh (BD Biosciences, Franklin Lakes, NJ, USA) and centrifuged at 157 × g for 5 min. Following removal of the supernatant, pellets was resuspended in alpha-MEM supplemented with 10% FBS, and the cells were plated in 10-cm tissue culture dishes and cultured at 37°C in a 5% CO2 incubator. The culture medium was changed every two days, and the cells were passaged after reaching 80–90% confluence (9).

Experimental design

The mice were randomly divided into four groups: i) A, normal control; ii) B, IPF; iii) C, IPF + phosphate-buffered saline (PBS); and iv) D, IPF + ADSC transplantation. ADSCs (1×106 cells/ml) were systemically administered to the mice in ~250 μl PBS via the tail vein, as described previously (3). The control group received the same volume of PBS only. The mice were sacrificed on day 7, and the lung tissues were sampled for morphometric analysis and immunohistochemical staining.

TUNEL assay for apoptosis detection

Paraffin-embedded samples (thickness, 4 μm) was rehydrated and then incubated with Proteinase K solution (Life Technologies, Carlsbad, CA, USA) for 30 min at room temperature. The samples were incubated with TUNEL reaction solution (BD Pharmingen, San Diego, CA, USA) at 37°C for 60 min after two washes with PBS. As previously described (10), the transforming solution was added and the samples were incubated at 37°C for 30 min. Staining was developed with 3,3′-diaminobenzidine tetrahydrochloride for 15 min. The samples were subsequently counterstained with hematoxylin for 15 min, dehydrated in graded alcohol and covered with resin. Positive staining was indicated by a nuclei that was pale brown.

Morphometric analysis and immunohistochemical staining

Cryosections (thickness, 5 μm) were stained with hematoxylin and eosin for morphometry or collagen fiber detection. Additional cryosections were used for the immunostaining of the matrix metalloproteinase (MMP)-2 antibody (cat. no. ab124292; Abcam, Cambridge, UK) and the MMP-9 antibody (cat. no. ab38898; Abcam) as previously described (11).

Hydroxyproline (Hyp) detection

The Hyp level in the lungs of mice was used to quantify the lung collagen content, as described previously (10). At the time of sacrifice, the mouse lungs were removed and any extra pulmonary airways and blood vessels were excised and discarded. The lung parenchyma was homogenized in 1.0 ml PBS and 1.0 ml 12N HCl was added. The samples were subsequently hydrolyzed at 110°C for 24 h and 5 μl of each sample was mixed with 5 μl citrate/acetate buffer (5% citric acid, 1.2% glacial acetic acid, 7.25% sodium acetate and 3.4% sodium hydroxide). Chloramine-T solution (100 μl; 1.4% chloramine-T, 10% N-propanol and 80% citrate/acetate buffer) was added and the mixture was incubated for 20 min at room temperature. Ehrlich's solution (1 ml) was added and the samples were incubated at 65°C for 18 min. The absorbance was measured at 550 nm on a spectrophotometer (Lambda 20 UV VIS; Perkin Elmer, Norwalk, CT, USA).

Measurement of bronchoalveolar lavage fluid (BALF)

An enzyme-linked immunosorbent assay kit (Abcam) was used to detect the levels of BALF cytokines [interleukin-1 (IL-1), platelet-derived growth factor (PDGF), and tumor necrosis factor (TNF)-α] in accordance with the manufacturer's instructions.

Western blotting

Tissues were harvested on ice in a radioimmunoprecipitation assay buffer (50 mmol/l Tris-HCl, pH 7.4, 0.25% Na-deoxycholate, 1 mmol/l EDTA, 1 mmol/l phenylmethylsulfonyl fluoride, 150 mmol/l NaCl, 1 mg/ml aprotinin, 1 mg/ml pepstatin, 1 mg/ml leupeptin and 1 mmol/l sodium fluoride) and centrifuged at 12,000 × g for 18 min at 4°C. Protein concentrations were determined using a bicinchoninic acid protein assay (Abcam) with bovine serum albumin (Sigma-Aldrich, St. Louis, MO, USA) as the standard. The samples were boiled in sample buffer at 100°C for 15 min. Equal quantities of protein (110 μg per sample) were separated by electrophoresis on 7.6–11% Tris-glycine sodium dodecyl sulfate polyacrylamide gels. Following electrophoresis, the proteins were transferred onto nitrocellulose membranes that were blocked and incubated with anti-MMP-2 (1:300; Abcam) and anti-MMP-9 (1:500; Abcam) at 4°C overnight. After washing, the membranes were incubated with anti-rabbit secondary antibodies (1:2000; Life Technologies) and reacted with enhanced chemiluminescence horseradish peroxidase detection reagents (GE Healthcare Life Sciences). The western blots were scanned and analyzed on a Storm 860 Phosphorimager (GE Healthcare).

Statistical analysis

Values are expressed as means ± standard deviation and were analyzed by one-way analysis of variance. P<0.05 was considered to indicate a statistically significant difference.

Results

ADSC intervention alleviated IPF

Mice in the IPF and IPF + PBS treatment groups demonstrated severe alveolar destruction compared with that of the normal control group and the ADSC intervention group. Pathological changes were observed, such as a significant increase in collagen fibers around the bronchi and vessels of the IPF and IPF + PBS treatment groups, and light blue collagen fibers were identified in the interstitial lung. These data indicate that ADSC intervention alleviates IPF (Fig. 1).

ADSC transplantation decreases the apoptosis of alveolar epithelial cells

As shown in Fig. 2, the cell nuclei of the apoptotic alveolar epithelial cells were stained yellow. The number of apoptotic cells in the ADSC transplantation group was less than that of groups B and C, indicating that the transplantation of ADSCs contributes to a decrease in apoptotic lung cells.

Immunohistochemical staining of MMP-2 and MMP-9

A large number of cells with a yellow-stained cytoplasm were identified in the IPF and the IPF + PBS treatment groups, although not in the normal control group, indicating increased collagen deposition in these two groups. However, no significant difference was observed between the IPF and the IPF + PBS treatment groups (P>0.05; Figs. 3 and 4).

Hyp content in lung tissue samples

When compared with that of groups A and D, an increase in the Hyp content was observed in groups B and C, indicating that collagen deposition increased following lung IPF, and ADSC transplantation decreased the occurrence of lung fibrosis (P<0.05; Fig. 5).

Increased IL-1, PDGF and TNF-α levels in the BALF of each IPF group

The concentrations of IL-1, PDGF, and TNF-α in the BALF of groups B, C and D were observed to be significantly increased when compared with those observed in the control group (A). The expression of IL-1, PDGF,and TNF-α was not significantly different between groups B and C; however, the cytokine level was significantly decreased in the ADSC transplantation group, when compared with that of groups B and C. These results indicate that transplantation of ADSCs decreases the production of these particular proinflammatory factors within the mouse lung (Fig. 6).

Discussion

It has been reported that MSCs may be used for the treatment of lung disease, such as chronic obstructive pulmonary disease (COPD) and IPF (1215). Furthermore, BMSCs have been identified as effective in the therapy of pulmonary fibrosis (1618). As the harvesting of BMSCs is considered to be difficult and low yielding, ADSCs may potentially be an ideal cell source, owing to the biological features that are comparable with BMSCs. Therefore, in the current study, the effect of transplanted ADSCs in mice with IPF was evaluated.

The IPF mouse model was established by injecting mice with BLM (a glycopeptide antibiotic that is produced by the bacterium, Streptomyces verticillus), which causes pulmonary fibrosis and impairs lung function. The epithelium of the mouse airway was subjected to BLM exposure, which resulted in the development IPF and cystic fibrosis (19).

Previous studies have identified that AT II cells may be involved in the repair and regeneration of damaged alveoli in cases of IPF; however, this type of cell is rare (20,21). Therefore, the transplantation of exogenous stem cells to enhance the repair capacity of the lungs may provide an effective alternative. In the current study, the effect of ADSC transplantation into mice with IPF was investigated. Histological staining revealed thickening of the pulmonary interstitium, narrowing alveolar width, and pulmonary fibrosis, which were observed in the IPF and the IPF + PBS groups, whereas attenuating responses were observed in the ADSC transplantation group. The collagen fiber deposition was also apparent in the IPF and the IPF + PBS groups. This finding indicates that injection of ADSCs decreases pulmonary fibrosis; however, the specific underlying mechanism remains unclear. Recent studies have indicated that lung MSCs are triggered to differentiate into myofibroblasts in a local microenvironment, which induces pulmonary fibrosis (22,23). In the present study, the ADSCs were injected into mice 1 h after pulmonary fibrosis induction, and it was hypothesized that ADSCs could not differentiate into myofibroblasts at this early stage.

MMP is a type of proteinase that is capable of degrading the extracellular matrix, which has been reported as vital in lung fibrosis (24). MMP-2 and MMP-9 levels were considered to be directly proportional to the occurrence of pulmonary fibrosis, and overactivation of MMP-2 may be the cause of type IV collagen degradation, which is involved in the initiation of lung fibrosis. In the present study, immunohistochemical staining of MMP-2 and MMP-9 revealed that the expression of MMPs in the lungs of mice increased following BLM treatment. The same results were also observed during western blot analysis. Kim et al reported that the MMP-2/MMP-9 activity was upregulated in the early stage of lung fibrosis, while the expression of tissue inhibitor of metalloproteinases, TIMP1/2 increased and MMP-2/MMP-9 activity decreased in the later time period (25). In addition, it has been reported that the level of MMPs in the serum and sputum of COPD patients increased, although the specific association between MMPs and COPD requires further investigation. Notably, the expression of MMPs decreased significantly following ADSC transplantation, indicating that ADSC transplantation may be involved in the process of lung repair. It was hypothesized in the current study that ADSCs migrate into the injured lung and proliferate, thus inhibiting fibroblast proliferation and collagen deposition.

Additionally, Hyp has been recognized as a key marker of collagen deposition in pulmonary fibrosis. Therefore, in the present study, the expression of Hyp was detected and the result revealed increased collagen fiber deposition in the IPF and IPF + PBS groups. Wang et al also reported that immediate MSC transplantation following fibrosis induction decreased the Hyp content (26), however, the MSC transplantation at a later stage of fibrosis induction in mice demonstrated no significant improvement to lung fibrosis, indicating that stem cells transplantation at an early stage may be critical in lung repair.

In conclusion, ADSC transplantation in the lungs may enhance lung injury repair and extenuate lung fibrosis when administered at an early stage, thus providing an effective option for the treatment of lung diseases, such as IPF and COPD.

Acknowledgments

The present study was supported by the Shanghai Municipal Natural Science Foundation (no. 14ZR1434500).

References

1 

Tzouvelekis A, Koliakos G, Ntolios P, Baira I, Bouros E, Oikonomou A, Zissimopoulos A, Kolios G, Kakagia D, Paspaliaris V, et al: Stem cell therapy for idiopathic pulmonary fibrosis: a protocol proposal. J Transl Med. 9:1822011. View Article : Google Scholar : PubMed/NCBI

2 

D'Agostino B, Sullo N, Siniscalco D, De Angelis A and Rossi F: Mesenchymal stem cell therapy for the treatment of chronic obstructive pulmonary disease. Expert Opin Biol Ther. 10:681–687. 2010. View Article : Google Scholar : PubMed/NCBI

3 

Zhang WG, He L, Shi XM, Wu SS, Zhang B, Mei L, Xu YJ, Zhang ZX, Zhao JP and Zhang HL: Regulation of transplanted mesenchymal stem cells by the lung progenitor niche in rats with chronic obstructive pulmonary disease. Respir Res. 15:332014. View Article : Google Scholar : PubMed/NCBI

4 

Moodley Y, Atienza D, Manuelpillai U, Samuel CS, Tchongue J, Ilancheran S, Boyd R and Trounson A: Human umbilical cord mesenchymal stem cells reduce fibrosis of bleomycin-induced lung injury. Am J Pathol. 175:303–313. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Gupta N, Su X, Popov B, Lee JW, Serikov V and Matthay MA: Intrapulmonary delivery of bone marrow-derived mesenchymal stem cells improves survival and attenuates endotoxin-induced acute lung injury in mice. J Immunol. 179:1855–1863. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Taghi GM, Ghasem Kashani Maryam H, Taghi L, Leili H and Leyla M: Characterization of in vitro cultured bone marrow and adipose tissue-derived mesenchymal stem cells and their ability to express neurotrophic factors. Cell Biol Int. 36:1239–1249. 2012. View Article : Google Scholar : PubMed/NCBI

7 

Nakao N, Nakayama T, Yahata T, Muguruma Y, Saito S, Miyata Y, Yamamoto K and Naoe T: Adipose tissue-derived mesenchymal stem cells facilitate hematopoiesis in vitro and in vivo: advantages over bone marrow-derived mesenchymal stem cells. Am J Pathol. 177:547–554. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Ding L, Dolgachev V, Wu Z, Liu T, Nakashima T, Wu Z, Ullenbruch M, Lukacs NW, Chen Z and Phan SH: Essential role of stem cell factor-c-Kit signalling pathway in bleomycin-induced pulmonary fibrosis. J Pathol. 230:205–214. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Liu HY, Chiou JF, Wu AT, Tsai CY, Leu JD, Ting LL, Wang MF, Chen HY, Lin CT, Williams DF, et al: The effect of diminished osteogenic signals on reduced osteoporosis recovery in aged mice and the potential therapeutic use of adipose-derived stem cells. Biomaterials. 33:6105–6112. 2012. View Article : Google Scholar : PubMed/NCBI

10 

Hattori N, Degen JL, Sisson TH, Liu H, Moore BB, Pandrangi RG, Simon RH and Drew AF: Bleomycin-induced pulmonary fibrosis in fibrinogen-null mice. J Clin Invest. 106:1341–1350. 2000. View Article : Google Scholar : PubMed/NCBI

11 

Monteiro-Amado F, Castro-Silva II, Lima CJ, Soares FA, Kowalski LP and Granjeiro JM: Immunohistochemical evaluation of MMP-2, MMP-9 and CD31/microvascular density in squamous cell carcinomas of the floor of the mouth. Braz Dent J. 24:3–9. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Inamdar AC and Inamdar AA: Mesenchymal stem cell therapy in lung disorders: pathogenesis of lung diseases and mechanism of action of mesenchymal stem cell. Exp Lung Res. 39:315–327. 2013. View Article : Google Scholar : PubMed/NCBI

13 

Moodley Y, Manuelpillai U and Weiss DJ: Cellular therapies for lung disease: a distant horizon. Respirology. 16:223–237. 2011. View Article : Google Scholar : PubMed/NCBI

14 

Sdrimas K and Kourembanas S: MSC microvesicles for the treatment of lung disease: a new paradigm for cell-free therapy. Antioxid Redox Signal. 21:1905–1915. 2014. View Article : Google Scholar : PubMed/NCBI

15 

Huleihel L, Levine M and Rojas M: The potential of cell-based therapy in lung diseases. Expert Opin Biol Ther. 13:1429–1440. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Rhieu BH, Epperly MW, Cao S, Franicola D, Shields D, Goff J, Wang H and Greenberger JS: Increased hematopoiesis in long-term bone marrow cultures and reduced irradiation-induced pulmonary fibrosis in Von Willebrand factor homologous deletion recombinant mice. In Vivo. 28:449–456. 2014.PubMed/NCBI

17 

Gilpin SE, Lung K, de Couto GT, Cypel M, Sato M, Singer LG, Keshavjee S and Waddell TK: Bone marrow-derived progenitor cells in end-stage lung disease patients. BMC Pulm Med. 13:482013. View Article : Google Scholar : PubMed/NCBI

18 

Aguilar S, Scotton CJ, McNulty K, Nye E, Stamp G, Laurent G, Bonnet D and Janes SM: Bone marrow stem cells expressing keratinocyte growth factor via an inducible lentivirus protects against bleomycin-induced pulmonary fibrosis. PLoS One. 4:e80132009. View Article : Google Scholar : PubMed/NCBI

19 

Kalayarasan S, Sriram N, Soumyakrishnan S and Sudhandiran G: Diallylsulfide attenuates excessive collagen production and apoptosis in a rat model of bleomycin induced pulmonary fibrosis through the involvement of protease activated receptor-2. Toxicol Appl Pharmacol. 271:184–195. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Rawlins EL: Lung epithelial progenitor cells: lessons from development. Proc Am Thorac Soc. 5:675–681. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Reynolds SD and Malkinson AM: Clara cell: progenitor for the bronchiolar epithelium. Int J Biochem Cell Biol. 42:1–4. 2010. View Article : Google Scholar

22 

Popova AP, Bozyk PD, Goldsmith AM, Linn MJ, Lei J, Bentley JK and Hershenson MB: Autocrine production of TGF-beta1 promotes myofibroblastic differentiation of neonatal lung mesenchymal stem cells. Am J Physiol Lung Cell Mol Physiol. 298:L735–L743. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Sun Z, Wang C, Shi C, Sun F, Xu X, Qian W, Nie S and Han X: Activated Wnt signaling induces myofibroblast differentiation of mesenchymal stem cells, contributing to pulmonary fibrosis. Int J Mol Med. 33:1097–1109. 2014.PubMed/NCBI

24 

Lagente V, Manoury B, Nénan S, Le Quément S, Martin-Chouly C and Boichot E: Role of matrix metalloproteinases in the development of airway inflammation and remodeling. J Med Biol Res. 38:1521–1530. 2005. View Article : Google Scholar

25 

Kim JY, Choeng HC, Ahn C and Cho SH: Early and late changes of MMP-2 and MMP-9 in bleomycin-induced pulmonary fibrosis. Yonsei Med J. 50:68–77. 2009. View Article : Google Scholar : PubMed/NCBI

26 

Wang Q, Zhu H, Zhou WG, Guo XC, Wu MJ, Xu ZY, Jiang JF, Shen C and Liu HQ: N-acetylcysteine-pretreated human embryonic mesenchymal stem cell administration protects against bleomycin-induced lung injury. Am J Med Sci. 346:113–122. 2013. View Article : Google Scholar

Related Articles

Journal Cover

October-2015
Volume 12 Issue 4

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

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Jiang H, Zhang J, Zhang Z, Ren S and Zhang C: Effect of transplanted adipose‑derived stem cells in mice exhibiting idiopathic pulmonary fibrosis. Mol Med Rep 12: 5933-5938, 2015
APA
Jiang, H., Zhang, J., Zhang, Z., Ren, S., & Zhang, C. (2015). Effect of transplanted adipose‑derived stem cells in mice exhibiting idiopathic pulmonary fibrosis. Molecular Medicine Reports, 12, 5933-5938. https://doi.org/10.3892/mmr.2015.4178
MLA
Jiang, H., Zhang, J., Zhang, Z., Ren, S., Zhang, C."Effect of transplanted adipose‑derived stem cells in mice exhibiting idiopathic pulmonary fibrosis". Molecular Medicine Reports 12.4 (2015): 5933-5938.
Chicago
Jiang, H., Zhang, J., Zhang, Z., Ren, S., Zhang, C."Effect of transplanted adipose‑derived stem cells in mice exhibiting idiopathic pulmonary fibrosis". Molecular Medicine Reports 12, no. 4 (2015): 5933-5938. https://doi.org/10.3892/mmr.2015.4178