Open Access

Prostaglandin E1 protects bone marrow‑derived mesenchymal stem cells against serum deprivation‑induced apoptosis

  • Authors:
    • Kuan Zeng
    • Bao Ping Deng
    • Hui‑Qi Jiang
    • Meng Wang
    • Ping Hua
    • Hong‑Wu Zhang
    • Yu‑Bin Deng
    • Yan‑Qi Yang
  • View Affiliations

  • Published online on: August 5, 2015     https://doi.org/10.3892/mmr.2015.4176
  • Pages: 5723-5729
  • Copyright: © Zeng 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

Mesenchymal stem cells (MSCs) have become a recent focus of experimental and clinical research regarding myocardial regeneration. However, the therapeutic potential of these cells is limited by poor survival. Prostaglandin E1 (PGE1) is known to have anti‑inflammatory and anti‑apoptotic effects on the myocardium. The aim of the present study was to determine whether PGE1 could protect MSCs against serum deprivation (SD)‑induced apoptosis. An SD model was used to induce apoptosis in MSCs in vitro. Apoptotic morphological changes were detected by Hoechst 33258 fluorescent nuclear staining; and Annexin V‑fluorescein isothiocyanate/propidium iodide (PI) double staining and flow cytometry was used to quantify the rate of apoptosis. Western blot analysis was used to detect the expression levels of the apoptosis‑associated proteins Bcl‑2, Bax and caspase‑3. The results of the present study demonstrated that SD induced apoptosis of MSCs, and that treatment with PGE1 attenuated the morphological changes characteristic of apoptosis. Annexin V/PI staining showed that the rate of apoptosis gradually increased with the duration of ischemia. Furthermore, treatment with PGE1 significantly reduced SD‑induced apoptosis, decreased the protein expression levels of Bax and caspase‑3, and increased the expression levels of Bcl‑2. These data suggest that PGE1 is able to influence the survival of MSCs under certain conditions. These results may aid in improving the therapeutic efficacy of MSC transplantation used to treat chronic ischemic heart disease.

Introduction

Ischemic heart disease is one of the most common diseases worldwide. The traditional treatment of ischemic heart disease includes the prevention of atherosclerosis, and revascularization of the coronary arteries; however, these strategies cannot reverse or repair myocardial necrosis. Heart transplantation is an effective treatment for patients with late-stage heart failure, however due to the insufficient supply of organs there are limits to its clinical application (13). Recently, the rapid development of stem cell technology has led to novel treatment methods, including the transplantation of mesenchymal stem cells (MSCs) to repair or regenerate damaged myocardium (47). Previous studies have demonstrated that MSCs transplanted into areas of myocardial ischemia may differentiate into myocardial cells and repair necrotic myocardial tissue. However, the effects of MSCs are insufficient, since the majority of transplanted MSCs die shortly after transplantation in the ischemic microenvironment (7,8). Therefore, a key focus of research is to improve the survival of MSCs following transplantation into ischemic tissue.

Prostaglandin E1 (PGE1), also termed alprostadil, is an endogenous substance, which has numerous effects, including vasodilation, protection of endothelial cells, and inhibition of the activation and aggregation of neutrophils and thrombocytes (9). Furthermore, PGE1 is widely used in the treatment of ischemic heart disease. Clinical research has previously demonstrated the potential of PGE1 for improving myocardial microcirculation, and counteracting the effects of ischemia-reperfusion injury and apoptosis in the myocardium (1013). These findings indicate that PGE1 may have a general cytoprotective action; however, there are currently no studies investigating whether PGE1 may prevent apoptosis of MSCs.

Apoptosis is a type of physiological cell death, for which it is considered difficult to generate a comprehensive in vitro model. Serum deprivation (SD) injury in vitro is widely used to mimic the ischemic environment (14,15). The mitochondrial pathway is the major underlying mechanism of physiological cell death in apoptosis (16,17), and the Bcl-2 family proteins have an important role in the apoptotic response (18,19). To the best of our knowledge, the molecular mechanism by which PGE1 may inhibit apoptosis of MSCs is currently unknown.

The present study established an in vitro model of SD-induced apoptosis, in order to explore the potential mechanisms by which PGE1 may improve the survival of MSCs in the myocardial microenvironment following transplantation.

Materials and methods

Animals

Sprague-Dawley rats (specific pathogen free; weight, 80–100 g) of either sex were purchased from the Animal Center of Sun Yat-sen University (Guangzhou, China). All procedures of the present study were approved by the Institutional Animal Care and Use Committee of Sun Yat-sen University. The rats were maintained in 12 h light/dark cycles at a temperature of 26–26°C and with a humidity of 40–70%, with free access to a standard laboratory diet and water.

Reagents and instruments

PGE1 was purchased from Zhuhai Schwarz Pharma Co., Ltd. (Zhuhai, China). Dulbecco's modified Eagle's medium (DMEM) and fetal bovine serum (FBS) were obtained from Gibco Life Technologies (Carlsbad, CA, USA). Penicillin-streptomycin was purchased from Solarbio Science & Technology Co., Ltd. (Beijing, China). A Hoechst 33258 Staining kit and propidium iodide (PI) were obtained from Sigma-Aldrich (St. Louis, MO, USA). Antibodies targeting Bcl-2 (cat. no. 2870), Bax (cat. no. 2772), and caspase-3 (cat. no. 9662), were purchased from Cell Signaling Technology Inc. (Danvers, MA, USA) and were used at a dilution of 1:1,000. A Bicinchoninic Acid (BCA) Protein Assay kit was purchased from Kangchen Bio-tech (Shanghai, China). Enhanced Chemiluminescence (ECL) solution was purchased from KeyGen Biotech Co., Ltd. (Nanjing, China). Fluorescein isothiocyanate (FITC)-labeled Annexin V and anti-rat CD90 (cat. no. 11-0900-81), CD45 (cat. no. 17-0461-80), CD11b/c (cat. no. 12-0110-80), and CD29 (cat. no. 46-0291-80), antibodies were purchased from BD Biosciences (San Jose, CA, USA).

Cell preparation and culture

MSCs were isolated from the femora and tibiae of Sprague Dawley rats, which had were sacrificed by cervical dislocation. The cells were cultured in DMEM supplemented with 10% FBS and penicillin-streptomycin (50 U/ml), and were incubated at 37°C in a humidified atmosphere containing 5% CO2. The culture medium was refreshed every 2–3 days. Each primary culture was passaged 1:2 once the MSCs had grown to 80% confluence. MSCs at passage 4, which were positive for CD90 and CD29, and negative for CD45 and CD11b/c, were collected and used for subsequent experiments.

SD-induced apoptosis

The MSCs were randomly divided into three groups and cultured for 24 h. The control group was cultured with complete medium supplemented with 10% FBS; the SD group was cultured with SD medium; and the PGE1+SD group was cultured with SD medium plus 10 ng/ml PGE1.

Hoechst 33342 staining

The culture medium was discarded, and the cells were washed three times with phosphate-buffered saline (PBS), and fixed in 4% paraformaldehyde for 15 min at room temperature. The fixing solution was then discarded and the cells were washed with PBS for 5 min and incubated in Hoechst 33342 in the dark for 15 min at room temperature. Following the 15 min incubation, the Hoechst 33342 solution was discarded, and the cells were washed for a further 5 min with PBS. The cells were then supplemented with PBS and observed under an inverted fluorescence microscope (Olympus, Tokyo, Japan). The apoptotic cells exhibited morphological changes, including shrinkage and condensed nuclei.

Flow cytometric (FCM) analysis of apoptosis

The MSCs from passage 4 were digested with 0.05% Trypsin-EDTA (Gibco Life Technologies) for 5 min at 37°C, resuspended at a concentration of 10×105 cells/ml and centrifuged at 300 x g for 5 min. Following centrifugation, the cells were collected in PBS at 4°C, washed twice, and resuspended in 100 ml binding buffer (BD Biosciences). In a 5 ml dry flow tube the cells were added to 5 µl Annexin V-FITC (BD Biosciences) and 5 µl PI (BD Biosciences), lightly vortexed, and incubated in the dark at room temperature for 15 min. Following incubation, 400 µl binding buffer was added, and the flow tube was placed on ice. PI and Annexin V-FITC fluorescence was measured using a flow cytometer (BD FACSVerse; BD Biosciences; excitation, 488 nm; emission, 615 nm). The research software (BD FACSuite softwared; BD Bioscience) matched with FCM was used to analyze the data. Positive Annexin V staining indicated apoptosis, and positive PI staining indicated necrosis. The experiment was repeated three times.

Western blot analysis of Bcl-2, Bax and caspase-3

The MSCs were seeded into a 60 mm petri dish, at a density of 10×105 cells/dish. The total protein was extracted using radioimmunoprecipitation buffer (EMD Millipore), supplemented with PMSF. The cells were sonicated briefly and centrifuged at 10,000 x g at 4°C. The protein concentration was measured using the BCA Protein Assay kit, according to the manufacturer's instructions. Equal samples of protein (20 µg) were separated by 12% SDS-PAGE and then transferred onto polyvinylidene difluoride membranes (EMD Millipore, Billerica, MA, USA). The membranes were incubated with blocking solution (Beyotime Institute of Biotechnology, Jiangsu, China) at room temperature for 2 h, and then incubated with the following primary antibodies: Bcl-2, Bax, caspase-3 and GAPDH (cat. no. KC-5G5; KangChen Biotech, Shanghai, China; dilution, 1:10,000) at 4°C overnight. The membranes were then incubated for 1 h at room temperature with the appropriate horseradish peroxidase conjugated-secondary antibodies. The blots were visualized using an enhanced chemiluminescence solution (EMD Millipore) and were exposed to X-ray film (Kodak, Tokyo, Japan). The density of the protein bands was analyzed using ImageJ 1.41 software (National Institutes of Health, Bethesda, MD, USA). The expression levels of the target proteins were normalized to those of GAPDH.

Statistical analysis

The data are expressed as the mean ± standard deviation. Comparisons between the groups were analyzed by one-way analysis of variance or Student's t-test. Statistical analyses were performed using SPSS 16.0 (SPSS, Inc., Chicago, IL, USA). P<0.05 was considered to indicate a statistically significant difference.

Results

MSC morphology and identification

MSCs were isolated and expanded from Sprague Dawley rats. The control MSCs were shown to sparsely attach to the culture flasks, and the majority of cells displayed a spindle-like shape (Fig. 1A). In the MSCs cultured with SD medium cells did not display the typical spindle-like shape, and a large number of floating dead cells were observed (Fig. 1B). Following treatment with PGE1, the morphology of the MSCs cultured with SD changed, and the number of floating dead cells was reduced, as compared with the cells cultured with SD alone (Fig. 1C). The MSCs at passage 4 were stably positive for CD90 and CD29 markers, and negative for CD45 and CD11b/c markers.

PGE1 inhibits SD-induced apoptosis

Hoechst 33342 staining showed that the majority of the MSCs in the control group were round or oval-shaped, and had light-blue regular nuclei (Fig. 2A). In the PGE1+SD group, the apoptotic cells were round or oval-shaped, with bright-blue irregular nuclei, which indicated chromosome condensation. The number of apoptotic cells in the SD group was significantly increased, as compared with the control group. Treatment with PGE1 significantly reduced the rate of MSC apoptosis, as compared with the SD group. Furthermore, flow cytometry of Annexin V/PI-stained cells was used to quantify the apoptotic rate of the MSCs. The percentage of apoptotic cells was significantly higher in the SD (P<0.01) and SD+PGE1 (P<0.05) groups, as compared with the control group (Fig. 2B and C). In addition, the rate of apoptosis was lower in the PGE1-treated MSCs, as compared with the untreated MSCs cultured with SD (P<0.05).

SD influences the expression levels of apoptosis-associated proteins Bax, Bcl-2 and caspase-3

The present study determined whether SD could affect the expression levels of the proapoptotic Bcl-2 family members, Bax and Bcl-2. Western blot analysis showed that the protein expression levels of Bax were significantly increased and the protein expression levels of Bcl-2 gradually decreased, following exposure to SD for 12–24 h (Fig. 3A and B). Cleaved caspase-3 expression was not detected in the control MSCs, however its expression was significantly increased in the MSCs cultured in SD medium (Fig. 3C).

Effects of PGE1 on the protein expression levels of Bax, Bcl-2 and caspase-3

Treatment with PGE1 significantly reduced SD-induced Bax protein expression levels and increased the protein expression levels of Bcl-2 (Fig. 4A). PGE1 also reduced the protein expression levels of cleaved caspase-3 in the MSCs (Figure 4B). These results indicate that PGE1 was able to attenuate SD-induced apoptosis though activation of Bax and deactivation of Bcl-2, thus reducing the expression of cleaved caspase-3.

Discussion

The present study reported the protective effects of PGE1 on SD-induced apoptosis in MSCs, and this effect was shown to be mediated through the mitochondrial caspase-3 pathway. The results of the present study demonstrated that treatment with PGE1 (10 ng/ml) decreased SD-induced apoptosis in MSCs, as shown by Hoechst 33342 staining, flow cytometry and measurement of caspase-3 protein expression levels. Furthermore, PGE1 protected MSCs against SD-induced apoptosis by downregulating Bax expression and upregulating Bcl-2 expression.

MSCs are nonhematogenic stem cells, which are present in the bone marrow. Due to their availability, potential for differentiation (20) and amplification, and association with fewer ethical issues (21), MSCs have become a focus of attention in the field of experimental and clinical research regarding myocardial regeneration. However, the ischemic cardiac microenvironment reduces the survival rate of transplanted cells, and limits their therapeutic effects (22). More than 90% of MSCs have previously been shown to die within 24 h of transplantation (23). Another study demonstrated that only ~21% of MSCs survive after 4 h of transplantation, and only 3.6% survive seven days (24). To address the problem of poor survival, research has focused on strategies that inhibit apoptosis of MSCs and improve their therapeutic effects in the ischemic myocardium. Such strategies include the use of genetically modified stem cells, preconditioning of stem cells, and combination drug therapy prior to transplantation of the cells into the damaged myocardium (25). Genetic modification (26,27) and preconditioning (28) of MSCs may improve the survival rate of stem cells; however, these are difficult to perform clinically. Whereas, combination drug therapy improves the viability of MSCs and is convenient for clinical application. Zhang et al (29) previously used rosuvastatin as a combination therapy to improve the therapeutic efficacy of MSCs for treating myocardial infarction. Furthermore, Dong et al (30) used combination therapy with atorvastatin, which was shown to activate AMP-activated protein kinase (AMPK); phosphorylation of AMPK resulted in activation of endothelial nitric oxide synthase. This mechanism may also be associated with the protection of MSCs against SD-induced apoptosis, through the mitochondrial apoptosis signaling pathway.

PGE1 is widely used in the treatment of ischemic heart disease. Previous research has identified the ability of PGE1 to improve myocardial microcirculation, reduce ischemicreperfusion injury, and exert anti-inflammatory and antiapoptotic effects on the myocardium (1013,31). PGE1 also exhibits general cytoprotective effects and anti-apoptotic activity (32). It has previously been reported that PGE1 is able to significantly upregulate antiapoptotic proteins, such as Bcl-2 (33); and downregulate Bax and caspase-3 (13). The present study established an in vitro SD-induced apoptosis model, in order to explore the potential mechanisms for the protective effects of PGE1 on MSCs. Apoptosis was detected by Hoechst 33258 and Annexin V-FITC/PI double staining. PGE1 was shown to protect the MSCs against SD-induced apoptosis. However, apoptosis involves a series of gene activation, expression and regulation; therefore, further investigation is required to explore the underlying molecular mechanisms by which PGE1 inhibits apoptosis of MSCs.

The Bcl-2 family is an important apoptosis-regulating family, which includes the antiapoptotic molecule Bcl-2 and proapoptotic molecule Bax (34). Numerous studies have demonstrated that Bcl-2 and Bax are associated with the mitochondrial membrane (3539). Bcl-2 is predominantly localized to endoplasmic reticulum and mitochondrial membranes, where it prevents the release of cytochrome c from the mitochondria and inhibits glutathione leakage, thus blocking programmed cell death (40,41). Bcl-2 can inhibit the activation of caspases, including caspase-9 and caspase-3, and thereby acts as an antiapoptotic agent (42). Bax is predominantly localized to the cytosol, or may be loosely attached to the mitochondrial membrane in an inactive form in healthy cells. Apoptotic stimuli result in structural changes to Bax, which may facilitate the translocation of Bax from the cytosol to the mitochondria, leading to apoptosis (43). Bax exhibits extensive amino acid homology with Bcl-2, and can form homodimers and heterodimers with Bcl-2 in vivo (44). The overexpression of Bax counteracts the death repressor activity of Bcl-2, and the activation of caspase-3 is dependent on the ratio of Bcl-2 to Bax, which controls cell survival and death following an apoptotic stimulus (44,45). Caspase-3 is a critical mediator of mitochondrial apoptosis (46), which can be activated by SD in MSCs (47).

The effects of PGE1 on the inhibition of caspase-3 and regulation of Bcl-2 and Bax have previously been reported (13,33,48); however, the present study is the first, to the best of our knowledge, to report such effects in MSCs. To confirm these findings, western blot analysis was used to detect the protein expression levels of Bax, Bcl-2 and caspase-3. SD downregulated the protein expression levels of Bcl-2, and upregulated the expression levels of Bax in MSCs, resulting in overexpression of caspase-3, which caused an increased rate of MSC apoptosis. Furthermore, treatment with PGE1 significantly increased the expression levels of Bcl-2 and inhibited the expression levels of Bax and caspase-3, thereby attenuating apoptosis in MSCs. Apoptosis is a complex process, and the SD model used in the present study attempted to simulate the myocardial microenvironment in vitro. However, further investigation is required to confirm these findings in vivo.

In conclusion, the results of the present study demonstrated that PGE1 exerts protective effects against SD-induced MSC apoptosis. PGE1 downregulated the protein expression levels of Bax and caspase-3, and upregulated the protein expression levels of Bcl-2 in the SD in vitro model. These findings may be useful in the clinical application of PGE1 alongside MSC transplantation into ischemic tissue, and may enhance the efficacy of cell therapy.

Acknowledgments

The authors of the present study would like to thank Dr Run-min Guo, Professor Hong-fu Wu and Dr Qing-feng Xiang for technical assistance. This study was supported by the program of 'One Hundred Talented Scholars' of Sun Yat-sen University (grant no. F002009011).

References

1 

Yu Q, Fan W and Cao F: Mechanistic molecular imaging of cardiac cell therapy for ischemic heart disease. Am J Physiol Heart Circ Physiol. 305:H947–H959. 2013. View Article : Google Scholar : PubMed/NCBI

2 

Toyoda Y, Guy TS and Kashem A: Present status and future perspectives of heart transplantation. Circ J. 77:1097–1110. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Burchill LJ and Ross HJ: Heart transplantation in adults with end-stage congenital heart disease. Future Cardiol. 8:329–342. 2012. View Article : Google Scholar : PubMed/NCBI

4 

Schächinger V, Erbs S, Elsässer A, et al: Improved clinical outcome after intracoronary administration of bone-marrow-derived progenitor cells in acute myocardial infarction: final 1-year results of the REPAIR-AMI trial. Eur Heart J. 27:2775–2783. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Tse HF, Thambar S, Kwong YL, et al: Prospective randomized trial of direct endomyocardial implantation of bone marrow cells for treatment of severe coronary artery diseases (PROTECT-CAD trial). Eur Heart J. 28:2998–3005. 2007. View Article : Google Scholar : PubMed/NCBI

6 

Strauer BE and Steinhoff G: 10 years of intracoronary and intra-myocardial bone marrow stem cell therapy of the heart: from the methodological origin to clinical practice. J Am Coll Cardiol. 58:1095–1104. 2011. View Article : Google Scholar : PubMed/NCBI

7 

Tang YL, Tang Y, Zhang YC, Qian K, Shen L and Phillips MI: Improved graft mesenchymal stem cell survival in ischemic heart with a hypoxia-regulated heme oxygenase-1 vector. J Am Coll Cardiol. 46:1339–1350. 2005. View Article : Google Scholar : PubMed/NCBI

8 

Menasché P: Current status and future prospects for cell transplantation to prevent congestive heart failure. Semin Thorac Cardiovasc Surg. 20:131–137. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Zhao XS, Pan W, Bekeredjian R and Shohet RV: Endogenous endothelin-1 is required for cardiomyocyte survival in vivo. Circulation. 114:830–837. 2006. View Article : Google Scholar : PubMed/NCBI

10 

Fang WT, Li HJ and Zhou LS: Protective effects of prostaglandin E1 on human umbilical vein endothelial cell injury induced by hydrogen peroxide. Acta Pharmacol Sin. 31:485–492. 2010. View Article : Google Scholar : PubMed/NCBI

11 

Takikawa M, Sumi Y, Tanaka Y, et al: Protective effect of pros-taglandin E1 on radiation-induced proliferative inhibition and apoptosis in keratinocytes and healing of radiation-induced skin injury in rats. J Radiat Res. 53:385–394. 2012. View Article : Google Scholar

12 

Li JH, Yang P, Li AL, Wang Y, Ke YN and Li XL: Cardioprotective effect of liposomal prostaglandin E1 on a porcine model of myocardial infarction reperfusion no-reflow. J Zhejiang Univ Sci B. 12:638–643. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Jia C, Dai C, Bu X, et al: Co-administration of prostaglandin E1 with somatostatin attenuates acute liver damage after massive hepatectomy in rats via inhibition of inflammatory responses, apoptosis and endoplasmic reticulum stress. Int J Mol Med. 31:416–422. 2013.

14 

Zeng X, Yu SP, Taylor T, Ogle M and Wei L: Protective effect of apelin on cultured rat bone marrow mesenchymal stem cells against apoptosis. Stem Cell Res. 8:357–367. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Chauvier D, Lecoeur H, Langonné A, et al: Upstream control of apoptosis by caspase-2 in serum-deprived primary neurons. Apoptosis. 10:1243–1259. 2005. View Article : Google Scholar : PubMed/NCBI

16 

Chalah A and Khosravi-Far R: The mitochondrial death pathway. Adv Exp Med Biol. 615:25–45. 2008. View Article : Google Scholar : PubMed/NCBI

17 

Suen DF, Norris KL and Youle RJ: Mitochondrial dynamics and apoptosis. Genes Dev. 22:1577–1590. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Martinou JC and Youle RJ: Mitochondria in apoptosis: Bcl-2 family members and mitochondrial dynamics. Dev Cell. 21:92–101. 2011. View Article : Google Scholar : PubMed/NCBI

19 

Gross A, McDonnell JM and Korsmeyer SJ: BCL-2 family members and the mitochondria in apoptosis. Genes Dev. 13:1899–1911. 1999. View Article : Google Scholar : PubMed/NCBI

20 

Zhang X, Wang Y, Gao Y, et al: Maintenance of high proliferation and multipotent potential of human hair follicle-derived mesenchymal stem cells by growth factors. Int J Mol Med. 31:913–921. 2013.PubMed/NCBI

21 

Ding DC, Shyu WC and Lin SZ: Mesenchymal stem cells. Cell Transplant. 20:5–14. 2011. View Article : Google Scholar : PubMed/NCBI

22 

Hale SL, Dai W, Dow JS and Kloner RA: Mesenchymal stem cell administration at coronary artery reperfusion in the rat by two delivery routes: a quantitative assessment. Life Sci. 83:511–515. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Hodgetts SI, Beilharz MW, Scalzo AA and Grounds MD: Why do cultured transplanted myoblasts die in vivo? DNA quantification shows enhanced survival of donor male myoblasts in host mice depleted of CD4+ and CD8+ cells or Nk1.1+ cells. Cell Transplant. 9:489–502. 2000.PubMed/NCBI

24 

Tang YL, Tang Y, Zhang YC, Qian K, Shen L and Phillips MI: Improved graft mesenchymal stem cell survival in ischemic heart with a hypoxia-regulated heme oxygenase-1 vector. J Am Coll Cardiol. 46:1339–1350. 2005. View Article : Google Scholar : PubMed/NCBI

25 

Mingliang R, Bo Z and Zhengguo W: Stem cells for cardiac repair: status, mechanisms and new strategies. Stem Cells Int. 2011:3109282011. View Article : Google Scholar

26 

Huang J, Zhang Z, Guo J, et al: Genetic modification of mesenchymal stem cells overexpressing CCR1 increases cell viability, migration, engraftment, and capillary density in the injured myocardium. Circ Res. 106:1753–1762. 2010. View Article : Google Scholar : PubMed/NCBI

27 

Huang F, Zhu X, Hu XQ, et al: Mesenchymal stem cells modified with miR-126 release angiogenic factors and activate Notch ligand Delta-like-4, enhancing ischemic angiogenesis and cell survival. Int J Mol Med. 31:484–492. 2013.

28 

Liu XB, Chen H, Chen HQ, et al: Angiopoietin-1 preconditioning enhances survival and functional recovery of mesenchymal stem cell transplantation. J Zhejiang Univ Sci B. 13:616–623. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Zhang Z, Li S, Cui M, et al: Rosuvastatin enhances the therapeutic efficacy of adipose-derived mesenchymal stem cells for myocardial infarction via PI3K/Akt and MEK/ERK pathways. Basic Res Cardiol. 108:3332013. View Article : Google Scholar : PubMed/NCBI

30 

Dong Q, Yang Y, Song L, Qian H and Xu Z: Atorvastatin prevents mesenchymal stem cells from hypoxia and serum-free injury through activating AMP-activated protein kinase. Int J Cardiol. 153:311–316. 2011. View Article : Google Scholar

31 

Huang CL, Wu YW, Wang SS, et al: Continuous intravenous infusion of prostaglandin E1 improves myocardial perfusion reserve in patients with ischemic heart disease assessed by positron emission tomography: a pilot study. Ann Nucl Med. 25:462–468. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Hara Y, Akamatsu Y, Maida K, et al: A new liver graft preparation method for uncontrolled non-heart-beating donors, combining short oxygenated warm perfusion and prostaglandin E1. J Surg Res. 184:1134–1142. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Ma XQ, Fu RF, Feng GQ, Wang ZJ, Ma SG and Weng SA: Hypoxia-reoxygenation-induced apoptosis in cultured neonatal rat cardiomyocyets and the protective effect of prostaglandin E. Clin Exp Pharmacol Physiol. 32:1124–1130. 2005. View Article : Google Scholar

34 

Dietrich JB: Apoptosis and anti-apoptosis genes in the Bcl-2 family. Arch Physiol Biochem. 105:125–135. 1997.In French. View Article : Google Scholar : PubMed/NCBI

35 

Renault TT, Teijido O, Antonsson B, Dejean LM and Manon S: Regulation of Bax mitochondrial localization by Bcl-2 and Bcl-x(L): keep your friends close but your enemies closer. Int J Biochem Cell Biol. 45:64–67. 2013. View Article : Google Scholar

36 

Degli EM and Dive C: Mitochondrial membrane permeabilisation by Bax/Bak. Biochem Biophys Res Commun. 304:455–461. 2003. View Article : Google Scholar

37 

Nemec KN and Khaled AR: Therapeutic modulation of apoptosis: targeting the BCL-2 family at the interface of the mitochondrial membrane. Yonsei Med J. 49:689–697. 2008. View Article : Google Scholar : PubMed/NCBI

38 

Autret A and Martin SJ: Emerging role for members of the Bcl-2 family in mitochondrial morphogenesis. Mol Cell. 36:355–363. 2009. View Article : Google Scholar : PubMed/NCBI

39 

Garcia-Saez AJ, Fuertes G, Suckale J and Salgado J: Permeabilization of the outer mitochondrial membrane by Bcl-2 proteins. Adv Exp Med Biol. 677:91–105. 2010. View Article : Google Scholar : PubMed/NCBI

40 

Hockenbery D, Nuñez G, Milliman C, Schreiber RD and Korsmeyer SJ: Bcl-2 is an inner mitochondrial membrane protein that blocks programmed cell death. Nature. 348:334–336. 1990. View Article : Google Scholar : PubMed/NCBI

41 

Haeberlein SL: Mitochondrial function in apoptotic neuronal cell death. Neurochem Res. 29:521–530. 2004. View Article : Google Scholar : PubMed/NCBI

42 

Mukhopadhyay A, Shishodia S, Suttles J, et al: Ectopic expression of protein-tyrosine kinase Bcr-Abl suppresses tumor necrosis factor (TNF)-induced NF-kappa B activation and IkappaBalpha phosphorylation. Relationship with down-regulation of TNF receptors. J Biol Chem. 277:30622–30628. 2002. View Article : Google Scholar : PubMed/NCBI

43 

Tan KO, Fu NY, Sukumaran SK, et al: MAP-1 is a mitochondrial effector of Bax. Proc Natl Acad Sci USA. 102:14623–14628. 2005. View Article : Google Scholar : PubMed/NCBI

44 

Oltvai ZN, Milliman CL and Korsmeyer SJ: Bcl-2 heterodimerizes in vivo with a conserved homolog, Bax, that accelerates programmed cell death. Cell. 74:609–619. 1993. View Article : Google Scholar : PubMed/NCBI

45 

Heon Seo K, Ko HM, Kim HA, et al: Platelet-activating factor induces up-regulation of antiapoptotic factors in a melanoma cell line through nuclear factor-kappaB activation. Cancer Res. 66:4681–4686. 2006. View Article : Google Scholar : PubMed/NCBI

46 

Lakhani SA, Masud A, Kuida K, et al: Caspases 3 and 7: key mediators of mitochondrial events of apoptosis. Science. 311:847–851. 2006. View Article : Google Scholar : PubMed/NCBI

47 

Zhu W, Chen J, Cong X, Hu S and Chen X: Hypoxia and serum deprivation-induced apoptosis in mesenchymal stem cells. Stem Cells. 24:416–425. 2006. View Article : Google Scholar

48 

Liu HJ, Ma JW, Qiao ZY and Xu B: Study of molecular mechanism of Prostaglandin E1 in inhibiting coronary heart disease. Mol Biol Rep. 40:6701–6708. 2013. View Article : Google Scholar : PubMed/NCBI

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
Zeng K, Deng BP, Jiang HQ, Wang M, Hua P, Zhang HW, Deng YB and Yang YQ: Prostaglandin E1 protects bone marrow‑derived mesenchymal stem cells against serum deprivation‑induced apoptosis. Mol Med Rep 12: 5723-5729, 2015
APA
Zeng, K., Deng, B.P., Jiang, H., Wang, M., Hua, P., Zhang, H. ... Yang, Y. (2015). Prostaglandin E1 protects bone marrow‑derived mesenchymal stem cells against serum deprivation‑induced apoptosis. Molecular Medicine Reports, 12, 5723-5729. https://doi.org/10.3892/mmr.2015.4176
MLA
Zeng, K., Deng, B. P., Jiang, H., Wang, M., Hua, P., Zhang, H., Deng, Y., Yang, Y."Prostaglandin E1 protects bone marrow‑derived mesenchymal stem cells against serum deprivation‑induced apoptosis". Molecular Medicine Reports 12.4 (2015): 5723-5729.
Chicago
Zeng, K., Deng, B. P., Jiang, H., Wang, M., Hua, P., Zhang, H., Deng, Y., Yang, Y."Prostaglandin E1 protects bone marrow‑derived mesenchymal stem cells against serum deprivation‑induced apoptosis". Molecular Medicine Reports 12, no. 4 (2015): 5723-5729. https://doi.org/10.3892/mmr.2015.4176