Lysophosphatidic acid increases the proliferation and migration of adipose‑derived stem cells via the generation of reactive oxygen species

  • Authors:
    • Sangjin Kang
    • Juhee Han
    • Seung Yong Song
    • Won‑Serk Kim
    • Soyoung Shin
    • Ji Hye Kim
    • Hyosun Ahn
    • Jin‑Hyun Jeong
    • Sung‑Joo Hwang
    • Jong‑Hyuk Sung
  • View Affiliations

  • Published online on: July 2, 2015     https://doi.org/10.3892/mmr.2015.4023
  • Pages: 5203-5210
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Phospholipid derivatives, such as lysophosphatidic acid (LPA), exhibit mitogenic effects on mesenchymal stem cells; however, the molecular mechanism underlying this stimulation has yet to be identified. The aims of the present study were as follows: To evaluate the stimulatory effects of LPA on the proliferation and migration of adipose‑derived stem cells (ASCs); to study the association between reactive oxygen species (ROS) and LPA signaling in ASCs; and to investigate the microRNAs upregulated by LPA treatment in ASCs. The results of the present study demonstrated that LPA increased the proliferation and migration of ASCs, and acted as a mitogenic signal via extracellular signal‑regulated kinases 1/2 and the phosphoinositide 3‑kinase/Akt signaling pathways. The LPA1 receptor is highly expressed in ASCs, and pharmacological inhibition of it by Ki16425 significantly attenuated the proliferation and migration of ASCs. In addition, LPA treatment generated ROS via NADPH oxidase 4, and ROS were able to function as signaling molecules to increase the proliferation and migration of ASCs. The induction of ROS by LPA treatment also upregulated the expression of miR‑210. A polymerase chain reaction array assay demonstrated that the expression levels of adrenomedullin and Serpine1 were increased following treatment with LPA. Furthermore, transfection with Serpine1‑specific small interfering RNA attenuated the migration of ASCs. In conclusion, the present study is the first, to the best of our knowledge, to report that ROS generation and miR‑210 expression are associated with the LPA‑induced stimulation of ASCs, and that Serpine1 mediates the LPA‑induced migration of ASCs. These results further suggest that LPA may be used for ASC stimulation during stem cell expansion.

Introduction

Lysophosphatidic acid (LPA) is a phospholipid derivative that is synthesized by the removal of the choline group from lysophosphatidylcholine. LPA is an intermediate in the synthesis of phosphatidic acid and is able to act as a signaling molecule in various cell types (14). In particular, LPA acts as a potent mitogen via activation of high-affinity G protein-coupled receptors, such as EDG2 and EDG4 (5,6). Due to its ability to stimulate cell proliferation, aberrant LPA signaling has been linked to cancer progression, through numerous mechanisms. Dysregulation of the LPA receptors may lead to cellular hyperproliferation, which may in turn contribute to oncogenesis and metastasis (79). Signaling associated with LPA has also been reported to regulate the growth of fibroblasts, vascular smooth muscle cells, endothelial cells, and keratinocytes (1013). In addition, the small GTPase Rho, a downstream molecule of the LPA receptor signal cascade, induces the formation of stress fibers and enhances cell migration (4). Therefore, LPA has been identified as a strong mitogenic factor. Recently, the molecular mechanisms underlying the mitogenic effects of LPA have been reported. Saunders et al (14) reported that LPA increases cell growth and inhibits apoptosis via the redox-dependent activation of extracellular signal-regulated kinases (ERK)-, Akt-, and nuclear factor-κB-dependent signaling pathways in ovarian cancer cells (14). Furthermore, the existence of an association between LPA signaling and reactive oxygen species (ROS)-mediated signaling has been suggested in other cell types (1517).

In addition to cancer cells, numerous studies have reported that LPA exerts stimulatory effects on mesenchymal stem cells (MSCs). LPA protects MSCs against hypoxia and serum deprivation-induced apoptosis via the ERK12 and phosphoinositide 3-kinase (PI3K)/Akt signaling pathways, and induces the migration of human lung-resident MSCs via the β-catenin pathway (18,19). LPA also rescues MSCs from endoplasmic reticulum stress-associated apoptosis, induced by hypoxia and serum deprivation through the p38 pathway (20). In addition, LPA induces the osteoblastic differentiation of MSCs by increasing cyclic adenosine mono-phosphate and Ca2+ levels (21). LPA also affects the paracrine activity of MSCs, and stimulates secretion of vascular endothelial growth factor (VEGF) and stem cell-derived factor 1 (22). LPA promotes VEGF secretion by increasing the expression of 150 kDa oxygen-related proteins in MSCs (23). However, there are few studies that specifically address the molecular mechanisms underlying LPA-induced ASC proliferation and migration. Our previous studies demonstrated that ROS generation exerts stimulatory effects on ASC proliferation and migration (2427); however, the association between ROS and LPA signaling in ASCs remains poorly understood. Therefore, the present study aimed to investigate the stimulatory effects of LPA on ASC proliferation and migration, and the association between ROS and LPA signaling in ASCs, as well as the microRNA (miR) expression in ASCs following LPA treatment.

Materials and methods

Cell culture and chemical inhibition

The sampling of human subcutaneous adipose tissue, isolation of ASCs, and characterization of ASCs with ASC-specific surface markers using flow cytometry were performed as previously described (28,29). Liposuction aspirates were obtained from a healthy female donor following the attainment of informed consent and approval from Bundang CHA Hospital (Seongnam, Korea; BD2011-152D). Human ASCs were cultured in α-minimum essential medium (Gibco Life Technologies, Carlsbad, CA, USA) supplemented with 10% fetal bovine serum (Gibco Life Technologies) and 1% penicillin-streptomycin (Gibco Life Technologies) at 37°C in a humidified atmosphere containing 5% CO2.

For chemical inhibition studies, the ASCs were co-treated with 5 mM U0126 (ERK12 inhibitor; EMD Millipore, Billerica, MA, USA), 5 mM LY294002 (PI3KAkt inhibitor; EMD Millipore), 0.1–1 mM Ki16425 (Sigma-Aldrich, St. Louis, MO, USA) or oleoyl-L-α-lysophosphatidic acid sodium salt (Sigma-Aldrich). For the inhibition of ROS generation, the ASCs were treated with 100 µM NADPH oxidase (Nox) inhibitor diphenyleneiodonium chloride (DPI; Sigma-Aldrich).

Cell proliferation assay

ASCs were seeded at 7×103 cells/well in 48-well plates. Following incubation with the LPA (1–50 µM) for 48 or 72 h, cell numbers were measured using the Cell Counting kit (CCK)-8 Assay (Dojindo Molecular Technologies, Inc., Rockville, MD, USA). Briefly, the medium in each well was replaced with medium containing the water-soluble tetrazolium salt WST-8 (10% vv). Following incubation for 2 h at 37°C, absorbance was measured at 450 nm using a Sunrise™ Microplate Absorbance Reader (Tecan Group Ltd., Männedorf, Switzerland).

Cell migration assay

Cell migration was measured using a transwell chamber. The transwell inserts with an 8 µm 3422 pore polycarbonate membrane (Corning Life Sciences, Cambridge, MA, USA), were coated with 0.1% gelatin (Sigma-Aldrich) in phosphate-buffered saline (PBS) for 2 h at 37°C. The ASCs were seeded at 1×104 cells per 200 µl in the upper chamber and incubated for 2 h at 37°C in an atmosphere containing 5% CO2. The ASCs were allowed to migrate towards 500 µl medium containing 10 µM LPA in the lower chamber. Migration induced by serum-free medium was used as a negative control. Following 16 h incubation at 37°C, the transwell inserts were fixed with ice-cold 100% methanol for 20 min and stained with 0.23% crystal violet (Sigma-Aldrich) in 2% ethanol for 30 min. Following further washing with distilled water, the non-migratory cells were removed by gently wiping the upper face of the transwell inserts with a cotton swab. Images of the stained cells were captured using a light microscope (CKX41; Olympus, Tokyo, Japan) and were analyzed using Image J 1.47 software (National Institutes of Health, Bethesda, MD, USA), in order to determine the number of cells that had migrated to the lower side of membrane.

Intracellular ROS staining

Intracellular ROS production was measured by flow cytometry using the ROS-sensitive dye 2′,7′-dichlorofluorescin diacetate (DCF-DA; Molecular Probes Life Technologies, Carlsbad, CA, USA), as previously described (25,26,30). Briefly, the ASCs were seeded at 2.5×105 cells/60 mm dish. The following day, the cells were incubated with 20 µM DCF-DA for 10 min at 37°C, and treated with LPA for 20 min at 37°C. Following incubation, the cells were trypsinized and resuspended in PBS. Green fluorescence intensity was measured using a BD FACSCalibur flow cytometer and analyzed using CellQuest Pro software (BD Biosciences, Franklin Lakes, NJ, USA).

Polymerase chain reaction (PCR) array and reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was isolated using the RNeasy kit (Qiagen, Inc., Valencia, CA, USA), and was reverse transcribed using a cDNA Synthesis kit (Promega Corporation, Madison, WI, USA), according to the manufacturer's instructions.

The synthesized cDNA was used in a PCR Array analysis using a CFX96 Real-Time PCR Detection system (Bio-Rad Laboratories, Inc., Hercules, CA, USA), according to the manufacturer's instructions. A PAHS-014ZD Human Signal Transduction Pathway Finder RT2 Profiler PCR Array (Qiagen, Inc.), which was composed of 84 transcripts representing 10 distinct signaling pathways, was used to identify the genes associated with signaling pathways that may be affected by LPA treatment. Differential gene expression and statistical analysis of the data were performed using PCR Array Data Analysis software (Qiagen, Inc.).

RT-qPCR was performed using a StepOne Plus Real-Time PCR system (Applied Biosystems Life Technologies, Foster City, CA, USA) using the following primers: Adrenomedullin (ADM) forward, CTT ATT CGG CCC CAG GACAT, and reverse, ACT GGT AGA TCT GGT GTGCC; Serpine1 forward, CCG CCT CTT CCA CAA ATCAG, and reverse, AAT GTT GGT GAG GGC AGAGA; LPA receptor 1 (LPAR1) forward, TCA TCT GGA CTA TGG CCATC, and reverse, CAA GTT GAA AAT GGC CCAGA; LPAR2 forward, CAA TGC TGC TGT GTA CTCTT, and reverse, TGG GCA GAG GAT GTA TAGTG; LPAR3 forward, CTA CAA GGA CGA GGA CATGT, and reverse, ATC CTC TAT GTA CTG GCTGC; LPAR4 forward, TCT GCA AGA TCT CTG GAACT, and reverse, ACA CAA TGG CAG AAT TCCTC; LPAR5 forward, TTC TCC CGT GTC CTG ACTA, and reverse, ACA TGT ACA CGC TCA CCAC; LPAR6 forward, TTG TAT GGG TGC ATG TTCAG, and reverse, CAA GTC TGA CAT TGC CAAGT; and GAPDH forward, CGA GAT CCC TCC AAA ATCAA, and reverse, TGT GGT CAT GAG TCC TTCCA. GAPDH served as a loading control. Thermal cycling over 40 cycles consisted of an initial denaturation at 95°C for 10 min, then 95°C for 15 sec, 60°C for 1 min and 95°C for 15 sec, and was terminated by a final extension at 60°C for 1 min.

Measurement of the expression levels of miR-210

Total RNA was extracted using TRIzol® reagent (Invitrogen Life Technologies, Carlsbad, CA, USA), and 0.5 µg total RNA was reverse transcribed using a Mir-X miRNA First-Strand Synthesis kit (Clontech Laboratories, Inc., Mountain View, CA, USA) according to the manufacturer's instructions. The expression levels of miR-210 were measured by RT-qPCR using the miR-210 primer (CTG TGC GTG TGA CAG CGG CTGA; Genolution Pharmaceuticals, Inc., Seoul, Korea) as previously described (24). U6 small nuclear RNA served as a loading control (Clontech Laboratories, Inc.).

Transfection with small interfering (si)RNA and miR-210 inhibitor

The Stealth RNAi™ siRNA specific to human Nox4 (sense, UUA UCC AAC AAU CUC CUG GUU CUCC; and antisense, GGA GAA CCA GGA GAU UGU UGG AUAA), the Silencer Select siRNA for human Serpine1 (ID# s10013), and the non-targeting negative control (scramble) siRNA (Silencer Select Negative Control#1 siRNA) were purchased from Invitrogen Life Technologies. The miR-210 inhibitor (hsa-miR210) and the miRNA inhibitor negative control were purchased from Genolution Pharmaceuticals, Inc. ASCs were seeded in 60 mm dishes at a density of 2×106 with complete medium. The following day, confluent ASCs were maintained in serum-free medium without antibiotics for 2 h. Subsequently, the ASCs were transfected with 20 nM of each siRNA or miR-210 inhibitor using Lipofectamine® 2000 transfection reagent (Invitrogen Life Technologies), according to the manufacturer's instructions in 24-well plates or 60 mm dishes.

Western blotting

Proteins of ASCs were isolated using SDS sample buffer. The soluble protein concentration was determined using the Quick start Bradford 1X dye reagent (Bio-Rad Laboratories, Inc.). The western blot analysis was performed as previously described (25) using antibodies targeting p-ERK12 (1:2,000; cat. no. 9106; in 5% milk), ERK12 (1:4,000; cat. no. 9102; in 5% milk), p-Akt (1:1,000; cat. no. 9271; in 5% milk), and Akt (1:4,000; cat. no. 9272; in 5% milk). All antibodies were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA). Horseradish-peroxidase (HRP)-conjugated secondary mouse antibody (1:10,000) was purchased from Vector Laboratories, Inc. (Burlingame, CA, USA) and HRP-conjugated secondary rabbit antibody was purchased from Cell Signaling Technology, Inc. (1:10,000; cat. no. 7074).

Statistical analysis

All data are presented as the mean ± standard deviation. Statistical analysis of the data was performed using Microsoft Excel software (Microsoft Corporation, Redmond, WA, USA). A Student's t-test was used to analyze the data. P<0.05 was considered to indicate a statistically significant difference.

Results

LPA increases the proliferation and migration of ASCs

The present study initially investigated whether LPA treatment increased the proliferation and migration of ASCs. As expected, 1–50 mM LPA significantly increased the proliferation (Fig. 1A) and migration (Fig. 1B) of ASCs in a dose-dependent manner. Since PI3K/Akt and mitogen-activated protein kinase (MAPK) signaling pathways have previously been reported to be associated with the mitogenic effects of LPA, the phosphorylation levels of Akt and ERK12 were measured following LPA treatment, and phosphorylation of both proteins increased in a time-dependent manner (Fig. 1C). In the inhibition study, pharmacological inhibition of PI3K/Akt by LY294002 significantly reduced the LPA-induced proliferation of ASCs (Fig. 1D). In addition, pharmacological inhibition of PI3K/Akt by LY294002, and of ERK by U0126 significantly reduced the LPA-induced migration of ASCs (Fig. 1E). These results suggest that LPA acts as a strong mitogenic factor in ASCs.

LPA mediates the mitogenic effects of ASCs via the LPA receptor

The expression levels of LPAR were measured in the ASCs (Table I), indicating that LPAR1 is highly expressed in ASCs, as compared with the other isoforms. To measure the involvement of LPAR in the mitogenic function of LPA, pharmacological inhibition of LPAR by Ki16425 was performed. Co-treatment with Ki16425 (0.1 and 1 mM) significantly attenuated LPA-induced proliferation (Fig. 2A) and migration (Fig. 2B) of ASCs.

Table I

LPAR expression in adipose-derived stem cells.

Table I

LPAR expression in adipose-derived stem cells.

GeneCt-value
LPAR124.44±0.50
LPAR229.82±0.78
LPAR330.85±0.94
LPAR430.84±0.90
LPAR531.44±0.54
LPAR629.60±0.57
GAPDH18.84±0.09

[i] Ct, cycle threshold; LPAR, lysophophatidic acid receptor.

LPA treatment induces ROS generation

The present study also investigated whether the LPA-induced proliferation and migration of ASCs was mediated by ROS generation. As expected, LPA treatment significantly increased the fluorescence intensity of DCF-DA, as determined by flow cytometric analysis (Fig. 3A); and pharmacological inhibition of Nox following treatment with DPI significantly attenuated the fluorescence intensity of DCF-DA (Fig. 3B). In addition, DPI treatment significantly reduced LPA-induced proliferation (Fig. 3C) and migration (Fig. 3D) of ASCs. As Nox4 is highly expressed in ASCs and primarily mediates ROS generation (26), downregulation of Nox4 expression in ASCs by siRNA transfection and Nox4 silencing significantly decreased LPA-induced cell proliferation (Fig. 3E) and migration (Fig. 3F). These results suggest that Nox4 may be associated with LPA-mediated ROS generation and downstream mitogenic effects in ASCs.

LPA increases miR-210 expression

Our previous study demonstrated that ROS generation induced by hypoxia, or chemicals, induced miR-210 expression and increased the proliferation and migration of ASCs (24); therefore, the present study investigated the involvement of miR-210 in the LPA-induced proliferation and migration of ASCs. As expected, LPA treatment increased miR-210 expression (Fig. 4A); however, the extent of upregulation was not as great as that induced by hypoxia and chemical ROS donors. In the inhibition study, miR-210 inhibitors significantly attenuated LPA-induced proliferation (Fig. 4B) and migration (Fig. 4C) of ASCs. These results suggest that miR-210 is involved in the LPA-induced proliferation and migration of ASCs.

LPA upregulates the expression of ADM and Serpine1

The human Signal Transduction Pathway Finder RT2 Profiler PCR Array was used to identify the LPA target genes that mediated the proliferation and migration of ASCs. A total of 4 h following LPA treatment, the expression levels of ADM increased 5.12-fold, as demonstrated by the PCR array (Fig. 5A). RT-qPCR analysis confirmed that the expression levels of ADM were upregulated ~6-fold (Fig. 5B). A total of 24 h following LPA treatment, the expression levels of Serpine1 increased 8.28-fold, as determined by the PCR array (Fig. 5C), and ~18-fold, as determined by the RT-qPCR (Fig. 5D). Therefore, the present study investigated whether ADM and/or Serpine1 mediated LPA-induced proliferation and migration of ASCs using siRNAs specific for ADM and Serpine1. Although silencing of ADM did not affect proliferation or migration (data not shown), transfection with Serpine1 siRNA significantly attenuated the LPA-induced migration of ASCs (Fig. 5E).

Discussion

The present study investigated the signal pathways and molecular mechanisms underlying LPA-induced proliferation and migration of ASCs. Previous studies have demonstrated that LPA is able to mediate the proliferation and migration of various cells, primarily via the ERK12 and PI3K/Akt signaling pathways (19,31,32). The results of the present study demonstrated that LPA may function as a mitogenic signal via LPARs, and the ERK12 and PI3K/Akt signaling pathways in ASCs. In addition to these signaling pathways, the present study demonstrated that LPA treatment induced ROS generation via Nox4, and that these ROS act as signaling molecules to increase the proliferation and migration of ASCs. Furthermore, upregulation of miR-210 expression by ROS may contribute to the increased proliferation and migration of ASCs, results which are concordant with those of a previous study (24). LPA also upregulated Serpine1 expression, which increased ASC migration.

As determined by PCR array, the expression levels of ADM and Serpine1 increased 4 and 24 h following LPA treatment, respectively. In addition, the results of the present study demonstrated that Serpine1 mediated LPA-induced migration of ASCs; however, no proliferation-associated genes were identified in the present study. Although the involvement of ADM in ASC proliferation was hypothesized, siRNA silencing of ADM did not inhibit the proliferation of ASCs. A previous study also reported that ADM did not increase, but rather inhibited, LPA-induced proliferation, and attenuated the stimulation of the MAPK pathway by LPA treatment in adventitial cells (33). Therefore, the mediator(s) of LPA-induced proliferation of ASCs have yet to be identified.

miRNAs exert their actions primarily at the post-transcriptional level, and their expression is known to be regulated by redox signaling (24,3436). In our previous study, the expression levels of miR-210 were significantly increased via various ROS generators (hypoxia, antimycin, rotenone, and platelet-derived growth factor subunit B), and regulated the proliferation and migration of ASCs (24). The present study demonstrated that LPA-induced ROS generation upregulated miR-210 expression in ASCs, and increased the proliferation and migration of ASCs. However, the extent of miR-210 LPA-induced upregulation was lower, as compared with other ROS generators such as hypoxia. In addition, ROS generation by S1P did not induce miR-210 expression in ASCs (data not shown). Therefore, it is reasonable to assume that ROS generation is not a prerequisite for the upregulation of miR-210 expression in ASCs.

In conclusion, the present study demonstrated that LPA increases the proliferation and migration of ASCs via Nox4-induced ROS generation. The present study is the first, to the best of our knowledge, to demonstrate that miR-210 expression is associated with LPA-induced stimulation, and that Serpine1 mediates the LPA-induced migration of ASCs. Therefore, phospholipid derivatives, such as LPA and S1P, may be used to stimulate ASCs during stem cell expansion.

Acknowledgments

The present study was supported by a grant from the Yonsei University Research Fund (grant no. 2014-12-0040).

References

1 

Glazier AT, Blackmore PF, Nolan RD and Wasilenko WJ: Attenuation of LPA-mediated calcium signaling and inositol poly-phosphate production in rat-1 fibroblasts transformed by the v-src oncogene. Biochem Biophys Res Commun. 245:607–612. 1998. View Article : Google Scholar : PubMed/NCBI

2 

Jonkers J and Moolenaar WH: Mammary tumorigenesis through LPA receptor signaling. Cancer Cell. 15:457–459. 2009. View Article : Google Scholar : PubMed/NCBI

3 

Sorensen SD, Nicole O, Peavy RD, Montoya LM, Lee CJ, Murphy TJ, Traynelis SF and Hepler JR: Common signaling pathways link activation of murine PAR-1, LPA, and S1P receptors to proliferation of astrocytes. Mol Pharmacol. 64:1199–1209. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Tangkijvanich P, Melton AC, Santiskulvong C and Yee HF Jr: Rho and p38 MAP kinase signaling pathways mediate LPA-stimulated hepatic myofibroblast migration. J Biomed Sci. 10:352–358. 2003. View Article : Google Scholar : PubMed/NCBI

5 

Bandoh K, Aoki J, Taira A, Tsujimoto M, Arai H and Inoue K: Lysophosphatidic acid (LPA) receptors of the EDG family are differentially activated by LPA species. Structure-activity relationship of cloned LPA receptors. FEBS Lett. 478:159–165. 2000. View Article : Google Scholar : PubMed/NCBI

6 

Yanagida K, Kurikawa Y, Shimizu T and Ishii S: Current progress in non-Edg family LPA receptor research. Biochim Biophys Acta. 1831:33–41. 2013. View Article : Google Scholar

7 

Hao F, Tan M, Xu X, Han J, Miller DD, Tigyi G and Cui MZ: Lysophosphatidic acid induces prostate cancer PC3 cell migration via activation of LPA(1), p42 and p38alpha. Biochim Biophys Acta. 1771:883–892. 2007. View Article : Google Scholar : PubMed/NCBI

8 

Sengupta S, Xiao YJ and Xu Y: A novel laminin-induced LPA autocrine loop in the migration of ovarian cancer cells. FASEB J. 17:1570–1572. 2003.PubMed/NCBI

9 

Shida D, Watanabe T, Aoki J, Hama K, Kitayama J, Sonoda H, Kishi Y, Yamaguchi H, Sasaki S, Sako A, et al: Aberrant expression of lysophosphatidic acid (LPA) receptors in human colorectal cancer. Lab Invest. 84:1352–1362. 2004. View Article : Google Scholar : PubMed/NCBI

10 

Kwon YJ, Sun Y, Kim NH and Huh SO: Phosphorylation of CREB, a cyclic AMP responsive element binding protein, contributes partially to lysophosphatidic acid-induced fibroblast cell proliferation. Biochem Biophys Res Commun. 380:655–659. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Lee H, Goetzl EJ and An S: Lysophosphatidic acid and sphingosine 1-phosphate stimulate endothelial cell wound healing. Am J Physiol Cell Physiol. 278:C612–C618. 2000.PubMed/NCBI

12 

Sauer B, Vogler R, Zimmermann K, Fujii M, Anzano MB, Schäfer-Korting M, Roberts AB and Kleuser B: Lysophosphatidic acid interacts with transforming growth factor-beta signaling to mediate keratinocyte growth arrest and chemotaxis. J Invest Dermatol. 123:840–849. 2004. View Article : Google Scholar : PubMed/NCBI

13 

Tokumura A, Iimori M, Nishioka Y, Kitahara M, Sakashita M and Tanaka S: Lysophosphatidic acids induce proliferation of cultured vascular smooth muscle cells from rat aorta. Am J Physiol. 267:C204–C210. 1994.PubMed/NCBI

14 

Saunders JA, Rogers LC, Klomsiri C, Poole LB and Daniel LW: Reactive oxygen species mediate lysophosphatidic acid induced signaling in ovarian cancer cells. Free Radic Biol Med. 49:2058–2067. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Chen Q, Olashaw N and Wu J: Participation of reactive oxygen species in the lysophosphatidic acid-stimulated mitogen-activated protein kinase kinase activation pathway. J Biol Chem. 270:28499–28502. 1995. View Article : Google Scholar : PubMed/NCBI

16 

Idzko M, Laut M, Panther E, Sorichter S, Dürk T, Fluhr JW, Herouy Y, Mockenhaupt M, Myrtek D, Elsner P and Norgauer J: Lysophosphatidic acid induces chemotaxis, oxygen radical production, CD11b up-regulation, Ca2+ mobilization, and actin reorganization in human eosinophils via pertussis toxin-sensitive G proteins. J Immunol. 172:4480–4485. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Lin CC, Lin CE, Lin YC, Ju TK, Huang YL, Lee MS, Chen JH and Lee H: Lysophosphatidic acid induces reactive oxygen species generation by activating protein kinase C in PC-3 human prostate cancer cells. Biochem Biophys Res Commun. 440:564–569. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Badri L and Lama VN: Lysophosphatidic acid induces migration of human lung-resident mesenchymal stem cells through the β-catenin pathway. Stem Cells. 30:2010–2019. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Chen J, Baydoun AR, Xu R, Deng L, Liu X, Zhu W, Shi L, Cong X, Hu S and Chen X: Lysophosphatidic acid protects mesenchymal stem cells against hypoxia and serum deprivation-induced apoptosis. Stem Cells. 26:135–145. 2008. View Article : Google Scholar

20 

Li Z, Wei H, Liu X, Hu S, Cong X and Chen X: LPA rescues ER stress-associated apoptosis in hypoxia and serum deprivation-stimulated mesenchymal stem cells. J Cell Biochem. 111:811–820. 2010. View Article : Google Scholar : PubMed/NCBI

21 

Liu YB, Kharode Y, Bodine PV, Yaworsky PJ, Robinson JA and Billiard J: LPA induces osteoblast differentiation through interplay of two receptors: LPA1 and LPA4. J Cell Biochem. 109:794–800. 2010.PubMed/NCBI

22 

Jeon ES, Heo SC, Lee IH, Choi YJ, Park JH, Choi KU, Park do Y, Suh DS, Yoon MS and Kim JH: Ovarian cancer-derived lysophosphatidic acid stimulates secretion of VEGF and stromal cell-derived factor-1 alpha from human mesenchymal stem cells. Exp Mol Med. 42:280–293. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Wei H, Wang F, Wang X, Yang J, Li Z, Cong X and Chen X: Lysophosphatidic acid promotes secretion of VEGF by increasing expression of 150-kD Oxygen-regulated protein (ORP150) in mesenchymal stem cells. Biochim Biophys Acta. 1831:1426–1434. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Kim JH, Park SG, Song SY, Kim JK and Sung JH: Reactive oxygen species-responsive miR-210 regulates proliferation and migration of adipose-derived stem cells via PTPN2. Cell Death Dis. 4:e5882013. View Article : Google Scholar : PubMed/NCBI

25 

Kim JH, Park SH, Park SG, Choi JS, Xia Y and Sung JH: The pivotal role of reactive oxygen species generation in the hypoxia-induced stimulation of adipose-derived stem cells. Stem Cells Dev. 20:1753–1761. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Kim JH, Song SY, Park SG, Song SU, Xia Y and Sung JH: Primary involvement of NADPH oxidase 4 in hypoxia-induced generation of reactive oxygen species in adipose-derived stem cells. Stem Cells Dev. 21:2212–2221. 2012. View Article : Google Scholar :

27 

Park SG, Kim JH, Xia Y and Sung JH: Generation of reactive oxygen species in adipose-derived stem cells: Friend or foe? Expert Opin Ther Targets. 15:1297–1306. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Kim WS, Park BS, Park SH, Kim HK and Sung JH: Antiwrinkle effect of adipose-derived stem cell: Activation of dermal fibroblast by secretory factors. J Dermatol Sci. 53:96–102. 2009. View Article : Google Scholar

29 

Kim WS, Park BS, Sung JH, Yang JM, Park SB, Kwak SJ and Park JS: Wound healing effect of adipose-derived stem cells: A critical role of secretory factors on human dermal fibroblasts. J Dermatol Sci. 48:15–24. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Jeong YM, Sung YK, Kim WK, Kim JH, Kwack MH, Yoon I, Kim DD and Sung JH: Ultraviolet B preconditioning enhances the hair growth-promoting effects of adipose-derived stem cells via generation of reactive oxygen species. Stem Cells Dev. 22:158–168. 2013. View Article : Google Scholar :

31 

Kue PF, Taub JS, Harrington LB, Polakiewicz RD, Ullrich A and Daaka Y: Lysophosphatidic acid-regulated mitogenic ERK signaling in androgen-insensitive prostate cancer PC-3 cells. Int J Cancer. 102:572–579. 2002. View Article : Google Scholar : PubMed/NCBI

32 

Hu X, Haney N, Kropp D, Kabore AF, Johnston JB and Gibson SB: Lysophosphatidic acid (LPA) protects primary chronic lymphocytic leukemia cells from apoptosis through LPA receptor activation of the anti-apoptotic protein AKT/PKB. J Biol Chem. 280:9498–9508. 2005. View Article : Google Scholar

33 

Yang JH, Jiang W, Pan CS, Qi YF, Wu QZ, Pang YZ and Tang CS: Effects of adrenomedullin on cell proliferation in rat adventitia induced by lysophosphatidic acid. Regul Pept. 121:49–56. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Boesch-Saadatmandi C, Wagner AE, Wolffram S and Rimbach G: Effect of quercetin on inflammatory gene expression in mice liver in vivo - role of redox factor 1, miRNA-122 and miRNA-125b. Pharmacol Res. 65:523–530. 2012. View Article : Google Scholar : PubMed/NCBI

35 

Ishimoto T, Sugihara H, Watanabe M, Sawayama H, Iwatsuki M, Baba Y, Okabe H, Hidaka K, Yokoyama N, Miyake K, et al: Macrophage-derived reactive oxygen species suppress miR-328 targeting CD44 in cancer cells and promote redox adaptation. Carcinogenesis. 35:1003–1011. 2014. View Article : Google Scholar

36 

Marin T, Gongol B, Chen Z, Woo B, Subramaniam S, Chien S and Shyy JY: Mechanosensitive microRNAs-role in endothelial responses to shear stress and redox state. Free Radic Biol Med. 64:61–68. 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
Kang S, Han J, Song SY, Kim WS, Shin S, Kim JH, Ahn H, Jeong JH, Hwang SJ, Sung JH, Sung JH, et al: Lysophosphatidic acid increases the proliferation and migration of adipose‑derived stem cells via the generation of reactive oxygen species. Mol Med Rep 12: 5203-5210, 2015
APA
Kang, S., Han, J., Song, S.Y., Kim, W., Shin, S., Kim, J.H. ... Sung, J. (2015). Lysophosphatidic acid increases the proliferation and migration of adipose‑derived stem cells via the generation of reactive oxygen species. Molecular Medicine Reports, 12, 5203-5210. https://doi.org/10.3892/mmr.2015.4023
MLA
Kang, S., Han, J., Song, S. Y., Kim, W., Shin, S., Kim, J. H., Ahn, H., Jeong, J., Hwang, S., Sung, J."Lysophosphatidic acid increases the proliferation and migration of adipose‑derived stem cells via the generation of reactive oxygen species". Molecular Medicine Reports 12.4 (2015): 5203-5210.
Chicago
Kang, S., Han, J., Song, S. Y., Kim, W., Shin, S., Kim, J. H., Ahn, H., Jeong, J., Hwang, S., Sung, J."Lysophosphatidic acid increases the proliferation and migration of adipose‑derived stem cells via the generation of reactive oxygen species". Molecular Medicine Reports 12, no. 4 (2015): 5203-5210. https://doi.org/10.3892/mmr.2015.4023