Open Access

Changes in mitochondrial function during EMT induced by TGFβ-1 in pancreatic cancer

  • Authors:
    • Qingqu Guo
  • View Affiliations

  • Published online on: January 18, 2017     https://doi.org/10.3892/ol.2017.5613
  • Pages: 1575-1580
  • Copyright: © Guo . 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

Mitochondrial dysfunction is linked to cancer. Differences in the number, morphology and function of mitochondria have been observed between normal cells and cancer cells. However, changes in mitochondrial function during epithelial-mesenchymal transition (EMT) in pancreatic cancer are less known. In the present study, the cultured human pancreatic cancer cell line Panc‑1 was treated with transforming growth factor (TGF)β‑1. Mitochondrial functions following TGFβ‑1 exposure in pancreatic cancer were investigated. It was noticed that TGFβ‑1 treatment induces morphologic changes and a shift from epithelial to mesenchymal phenotype in pancreatic cancer. Furthermore, increased mitochondrial mass was detected in pancreatic cancer following TGFβ‑1 treatment. Besides, the production of reactive oxygen species in TGFβ-1-treated pancreatic cancer cells significantly increased compared with the control cells. Our results indicate that the phenomenon of EMT in pancreatic cancer has an association with mitochondrial dysfunction. Mitochondrial dysfunction may be a cause of EMT in pancreatic cancer, which leads to heterogeneity in pancreatic cancer, and may be a potential therapeutic target in the future.

Introduction

Mitochondria, which are multifunctional organelles, are important in cellular proliferation and physiology, including cellular energy (adenosine triphosphate) production via oxidative phosphorylation and apoptosis via cytochrome c release (1). Mitochondrial dysfunction is linked to several human diseases, including premature aging, diabetes mellitus and cancer (2). Differences in the number, morphology and function of mitochondria have been detected between normal cells and cancer cells (3). Previous studies have revealed that mitochondrial dysfunction contributes to the development and progression of cancer (4).

The epithelial-mesenchymal transition (EMT) is a process by which cells undergo a morphological switch from the epithelial polarized phenotype to the mesenchymal fibroblastoid phenotype. During EMT, the function and expression of the epithelial cell-cell adhesion molecule E-cadherin is lost, whereas the expression of the mesenchymal cell-cell adhesion molecule N-cadherin is induced (5). Numerous studies have previously demonstrated that EMT is often activated during cancer cell migration, invasion, metastatic dissemination and chemoresistance (6). However, changes in mitochondrial function during EMT are largely unknown. In the present study, the mitochondrial function was examined during EMT induced by transforming growth factor (TGF)β-1 exposure in pancreatic cancer. Mitochondrial DNA (mtDNA), mitochondrial mass and reactive oxygen species (ROS) increased, while the mitochondrial membrane potential (ΔΨm) decreased, following TGFβ-1 treatment in pancreatic cancer.

Materials and methods

Cell culture and EMT induction

The pancreatic cell line Panc-1 was purchased from the Type Culture Collection of Chinese Academy of Sciences (Shanghai, China), and was cultured in RPMI 1640 medium (Jinuo, Hangzhou, China) containing 10% fetal bovine serum (HyClone; GE Healthcare Life Sciences, Logan, UT, USA) 1 µg/ml streptomycin (Jinuo), 100 IU/ml penicillin (Jinuo), 2 mM glutamine (Jinuo) and 1 mM sodium pyruvate (Jinuo) at 37°C in a 5% CO2 atmosphere. EMT induction by TGF-β1 (PeproTech, Inc., Rocky Hill, NJ, USA) was performed according to a previous protocol (7). Briefly, when cells grew to ~80% confluence, Panc-1 cells were trypsinized (Jinuo) and seeded into 6-well plates in duplicate (4×105 cells/well). Following 24–48 h, EMT-inducing medium (serum free, containing 1 ng/ml TGF-β1 and 50 ng/ml epidermal growth factor) was used to replace the common medium of Panc-1 cells, and the cells were incubated for additional 24–48 h. Next, the optical density was determined.

Western blot analysis

EMT-induced Panc-1 cells were washed twice using ice-cold PBS and lysed on ice using radioimmunoprecipitation assay buffer (20 mM Tris-HCl, 150 mM NaCl, 1% NP-40, 0.1% SDS, 0.5% sodium deoxycholate, 2 mM sodium fluoride, 2 mM Na3VO4·2H2O, 1 mM EDTA and protease inhibitor phenylmethane sulfonyl fluoride) (Beyotime Institute of Biotechnology, Haimen, China). The protein concentration of the lysates was determined using a BCA Protein Assay kit (P0009; Beyotime Institute of Biotechnology), and then marker proteins of EMT were detected using 10% SDS-PAGE and western blotting. Cell lysates were heated at 100°C for 10 min and electrophoresed though 10% SDS-PAGE. Then, proteins were transferred to a polyvinylidene difluoride membrane (EMD Millipore, Billerica, MA, USA), which were next incubated with 5% bovine serum albumin (BSA) (ST023; Beyotime Institute of Biotechnology) for 1 h at room temperature. Subsequently, membranes were incubated with the corresponding antibodies in a 5% BSA solution at 4°C overnight. Antibodies against N-cadherin (14215; 1:1,000 dilution), E-cadherin (5296; 1:1,000 dilution) and vimentin (3390; 1:1,000 dilution) (8) were purchased from Cell Signaling Technology, Inc. (Danvers, MA, USA). Upon washing three times with PBS containing Tween 20 [0.1% (v/v) Tween 20, 137 mM NaCl, 2.7 mM KCl, 10 mM Na2HPO4 and 1.8 mM KH2PO4), the membranes were incubated with horseradish peroxidase-conjugated rabbit anti-mouse immunoglobulin G (58802; Cell Signaling Technology, Inc.; 1:1,000 dilution) for 2 h at room temperature. Finally, protein bands were detected with an enhanced chemiluminescence detection kit (P0018A; Beyotime Institute of Biotechnology).

Mitochondrial density of EMT-Panc-1 cells determined by MitoTracker Green FM staining

Panc-1 cells were seeded in 6-well plates and cultured in EMT-inducing medium. After 24–48 h, Panc-1 cells in plates were stained with MitoTracker Green FM kit (Beyotime Institute of Biotechnology) according to the manufacturer's protocol to determine mitochondria density (9). Briefly, cells were firstly washed using PBS twice and then incubated at 37°C with 50 nM MitoTracker Green FM probe for 30 min. Next, the staining buffer was removed and replaced with fresh complete medium. Fluorescence was detected with a fluorescent microscope (Olympus Corporation, Tokyo, Japan), and the cells were detached for analysis using flow cytometry at 490 nm.

Genome DNA extraction and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) analysis of mtDNA

The mtDNA copy number of EMT-Panc-1 cells was determined by a standard protocol (10). Total genomic DNA was isolated with the Gentra Puregene Cell kit (Qiagen GmbH, Hilden, Germany) according to manufacturer's protocol. The mtDNA content was determined in cells utilizing RT-qPCR via SYBR Green assay (RR820A; Takara Biotechnology Co., Ltd., Dalian, China). The method for mtDNA copy number detection based on qPCR used a 107-bp sized amplicon of mtDNA transfer RNA (tRNA)Leu (UUR) (forward primer, 5′-CACCCAAGAACAGGGTTTGT-3′ and reverse primer, 5′-TGGCCATGGGTATGTTGTTA-3′) to determine the mtDNA copy number, and an 86-bp sized amplicon of β2-microglobulin (forward primer, 5′-TGCTGTCTCCATGTTTGATGTATCT-3′ and reverse primer, 5′-TCTCTGCTCCCCACCTCTAAGT-3′) to determine nuclear DNA (nDNA) as an internal control of the experiment (10). The PCR process consisted of 1 cycle at 95°C for 10 min followed by 40 cycles (95°C for 15 sec and 62°C for 30 sec) and melting curve (50–95°C with a 0.5°C interval). qPCR was conducted in a StepOnePlus Real-Time PCR System (Applied Biosystems; Thermo Fisher Scientific, Inc., Waltham, MA, USA). PCR assays were performed in triplicate for each DNA sample. The expression of mtDNA copy number relative to that of nDNA was determined using the formula 2×2ΔCq, where ΔCq is the difference of the Cq values between the β2-microglobulin gene and the tRNALeu (UUR) gene.

Detection of ROS by flow cytometry and fluorescence spectroscopy

Panc-1 cells were pretreated with EMT-inducing buffer for 24–48 h and stained with the ROS detection probe 2′,7′-dichlorofluorescin diacetate at a final concentration of 10 µM for 20–30 min at 37°C. The level of ROS was detected using fluorescence spectroscopy. Subsequently, the cells were harvested and washed twice with PBS, resuspended in PBS, and analyzed by flow cytometry.

ΔΨm determination

ΔΨm determination of EMT-Panc-1 cells was conducted using the lipophilic cationic fluorescent dye 5,5′,6,6′-tetrachloro-1,1′,3,3′-tetraethylbenzimidazolcarbocyanine iodide (JC-1) (Beyotime Institute of Biotechnology). The cells were plated and treated according to the manufacturer's protocol, and 1 µM JC-1 was added 30 min prior to harvesting the cells. JC-1-stained cells were firstly counted using fluorescence spectroscopy (Leica Microsystems GmbH, Wetzlar, Germany), and then the cells were collected by trypsinization and washed with PBS. The red (aggregated JC-1; R2 region) and green (monomeric JC-1; R1 region) fluorescence signals were analyzed by flow cytometry.

Statistical analysis

Data were analyzed with SPSS 19.0 software (IBM SPSS, Armonk, NY, USA) and expressed as means ± standard deviation. The statistical significance of differences was evaluated using an unpaired, non-parametric Student's t test. P<0.05 was considered to indicate a statistically significant difference.

Results

TGFβ-1 treatment induces morphological changes and a shift from epithelial to mesenchymal phenotype in pancreatic cancer

For EMT induction in cancer cells, TGFβ-1, which is a major factor during EMT (11), was used. As demonstrated by Ikenaga et al (11), pancreatic cancer cells treated with TGFβ-1 exhibited a spindle-shaped fibroblastic morphology and cell scattering compared with untreated cancer cells (Fig. 1). Lower E-cadherin expression, and higher N-cadherin and vimentin expression, were detected in Panc-1 cells following treatment with TGFβ-1 by western blotting (Fig. 1C). These results indicate that the pancreatic cancer Panc-1 cells acquired a mesenchymal phenotype, suggesting that EMT was induced by TGFβ-1 in these cells.

Increased mitochondrial mass following TGFβ-1 treatment in pancreatic cancer

To examine the mitochondrial mass upon TGFβ-1 treatment, pancreatic cancer cells were stained with MitoTracker Green FM dye. As shown in Fig. 2, there were significant differences in mitochondrial mass between treated and control (untreated) cells (P=0.0004). Cells that underwent EMT had more mitochondria than control cells. To determine the mitochondrial mass more precisely, the fluorescence intensity of the cells was quantified immediately by flow cytometric scanning. The results of flow cytometry revealed that the mitochondrial mass of the treated group was higher than that of the control cells (Fig. 2B and E). In addition, the mtDNA content was also significantly increased in treated cells relative to that in control cells (P=0.0005) (Fig. 2C).

Effect of TGFβ-1 treatment on the production of ROS in pancreatic cancer cells

Mitochondria are considered the main source of ROS, and ROS production is generally associated with impairments of the respiratory chain and mitochondrial function (12,13). Therefore, in order to ascertain whether alterations in the generation of ROS also occur in association with EMT, the intracellular ROS levels were measured in pancreatic cancer cells with or without TGFβ-1 treatment. The results are shown in Fig. 3. The production of ROS in TGFβ-1-treated tumor cells was significantly increased compared with that in control cells (P=0.004).

Decreased ΔΨm upon TGFβ-1 treatment in pancreatic cancer

The present study further investigated whether TGFβ-1 treatment affected the ΔΨm, since this is important for mitochondrial functions (14). A fluorescent probe, JC-1, was used to stain polarized mitochondria. As represented in Fig. 4A and C, incubation with TGFβ-1 decreased the number of pancreatic cancer cells displaying a high ΔΨm. Flow cytometry revealed the same results than fluorescence microscopy (Fig. 4B and D).

Discussion

The EMT is a basic physiological process in which epithelial cells lose their polarity and undergo a transition to a mesenchymal phenotype (6). Hallmarks of EMT include loss of cell-cell adhesion, re-organization of cytoskeletal actin and acquisition of migratory characteristics (15).

Emerging evidence suggests that EMT is essential in promoting tumor invasion, metastasis, recurrence and drug resistance in various types of cancer, including pancreatic cancer (16,17). TGFβ-1 promotes EMT by transcriptional and post-transcriptional regulation of a group of transcription factors that suppress epithelial features and enhance mesenchymal features (18,19). In agreement with previous reports (20,21), the present study demonstrated that TGFβ-1 induces EMT in pancreatic cancer cells by acquisition of mesenchymal morphology, increased expression of the mesenchymal markers vimentin and N-cadherin, and decreased expression of the epithelial marker E-cadherin.

Mitochondria are the primary energy producers of the cell, and regulate intracellular energy metabolism, cell death and free radical (ROS) production (22). In this context, the aim of the present study is to demonstrate the change in mitochondrial function during EMT induced by TGFβ-1 in pancreatic cancer. First, mitochondrial mass and mtDNA were investigated. Human mitochondria contain a small quantity of their own DNA (mtDNA), which is essential for normal mitochondrial function (23). Changes in mtDNA content have been described in a wide variety of cancers, with both increases and decreases being reported in either tumor tissue, circulating cells or metastatic cancer cells (24,25). However, in the present study, the mitochondrial mass and mtDNA content were significantly increased in TGFβ-1-treated pancreatic cancer cells relative to those detected in the control cells. Furthermore, a recent study revealed that high mtDNA content is associated with tumor invasion and EMT characteristics in esophageal squamous cell carcinoma (ESCC) cells, and suggested that a relatively high mtDNA copy number may confer an advantage for tumor invasion in ESCC (26). Other studies have associated mitochondrial dysfunction with mtDNA copy number, and have proposed that the mtDNA content may be a potential biomarker of mitochondrial dysfunction (23,27). Therefore, the increase in mitochondrial mass and mtDNA in pancreatic cancer cells with EMT phenotype observed in the present study suggests that mitochondrial dysfunction happened during the process of EMT, and may promote pancreatic cancer cell migration.

Mitochondrial dysfunction has been regarded as a hallmark of malignancy in human gastric cancer (28). Several studies have implied that mitochondrial dysfunction is important in cancer metastasis, in which induction of ROS was a key element. Thus, increased generation of ROS has an association with mitochondrial damage and dysfunction (2729). Additionally, accumulating evidence suggests that cancer cells exhibit increased intrinsic ROS stress partly due to mitochondrial malfunction (30). In turn, excessive production of ROS in cancer cells may contribute to mitochondrial dysfunction and further lead to the stimulation of cellular proliferation, cell migration and invasion, thus contributing to carcinogenesis (31). Therefore, the current study next examined whether EMT induced by TGFβ-1 resulted in excessive production of ROS in pancreatic cancer cells (32). As expected, an increase in ROS was observed in TGFβ-1-treated cancer cells. This is consistent with the results of Zhou et al (33), who noticed that hypoxia-induced EMT requires the generation of mitochondrial ROS, which participate in hypoxia-induced TGF-β1 production and results in EMT. Furthermore, a previous study reported that EMT-promoted mitochondrial dysfunction was accompanied by increased ROS generation and decreased ΔΨm (27). Thus, determination of ΔΨm following TGF-β1 treatment was conducted in the present study. As previously reported, the ΔΨm was markedly increased in the present study, suggesting that mitochondrial dysfunction occurred during the process of EMT (27).

In conclusion, the present study investigated the change in mitochondrial function that occurs during EMT induced by TGFβ-1 exposure in pancreatic cancer. Our results indicate that the phenomenon of EMT is associated with mitochondrial dysfunction. Mitochondrial dysfunction may be a cause of EMT in pancreatic cancer, which leads to heterogeneity in pancreatic cancer, and may be a potential therapeutic target in the future.

Acknowledgements

The present study was supported by grants from the National Natural Science Foundation of China (Beijing, China; grant no. 81071960) and the New Teacher Foundation of Ministry of Education (Beijing, China; grant no. 20100101120129).

References

1 

Kim AJ, Jee HJ, Song N, Kim M, Jeong SY and Yun J: p21 (WAF1/C1P1) deficiency induces mitochondrial dysfunction in HCT116 colon cancer cells. Biochem Biophys Res Commun. 430:653–658. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Kang D and Hamasaki N: Alterations of mitochondrial DNA in common diseases and disease states: Aging, neurodegeneration, heart failure, diabetes, and cancer. Curr Med Chem. 12:429–441. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Jose C and Rossignol R: Rationale for mitochondria-targeting strategies in cancer bioenergetic therapies. Int J Biochem Cell Biol. 45:123–129. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Gottfried E, Kreutz M and Mackensen A: Tumor metabolism as modulator of immune response and tumor progression. Semin Cancer Biol. 22:335–341. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Yilmaz M and Christofori G: EMT, the cytoskeleton, and cancer cell invasion. Cancer Metastasis Rev. 28:15–33. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Pirozzi G, Tirino V, Camerlingo R, Franco R, La Rocca A, Liguori E, Martucci N, Paino F, Normanno N and Rocco G: Epithelial to mesenchymal transition by TGFβ-1 induction increases stemness characteristics in primary non small cell lung cancer cell line. PLoS One. 6:e215482011. View Article : Google Scholar : PubMed/NCBI

7 

Moreno-Bueno G, Peinado H, Molina P, Olmeda D, Cubillo E, Santos V, Palacios J, Portillo F and Cano A: The morphological and molecular features of the epithelial-to-mesenchymal transition. Nat Protoc. 4:1591–1613. 2009. View Article : Google Scholar : PubMed/NCBI

8 

Mani SA, Guo W, Liao MJ, Eaton EN, Ayyanan A, Zhou AY, Brooks M, Reinhard F, Zhang CC, Shipitsin M, et al: The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell. 133:704–715. 2008. View Article : Google Scholar : PubMed/NCBI

9 

Fetisova EK, Avetisyan AV, Izyumov DS, Korotetskaya MV, Chernyak BV and Skulachev VP: Mitochondria-targeted antioxidant SkQR1 selectively protects MDR (Pgp 170)-negative cells against oxidative stress. FEBS Lett. 584:562–566. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Venegas V, Wang J, Dimmock D and Wong LJ: Real-time quantitative PCR analysis of mitochondrial DNA content. Curr Protoc Hum Genet Chapter. 19:Unit 19.7. 2011. View Article : Google Scholar

11 

Ikenaga N, Ohuchida K, Mizumoto K, Akagawa S, Fujiwara K, Eguchi D, Kozono S, Ohtsuka T, Takahata S and Tanaka M: Pancreatic cancer cells enhance the ability of collagen internalization during epithelial-mesenchymal transition. PLoS One. 7:e404342012. View Article : Google Scholar : PubMed/NCBI

12 

Rigoulet M, Yoboue ED and Devin A: Mitochondrial ROS generation and its regulation: Mechanisms involved in H(2)O(2) signaling. Antioxid Redox Signal. 14:459–468. 2011. View Article : Google Scholar : PubMed/NCBI

13 

Figueira TR, Barros MH, Camargo AA, Castilho RF, Ferreira JC, Kowaltowski AJ, Sluse FE, Souza-Pinto NC and Vercesi AE: Mitochondria as a source of reactive oxygen and nitrogen species: From molecular mechanisms to human health. Antioxid Redox Signal. 18:2029–2074. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Brand MD and Nicholls DG: Assessing mitochondrial dysfunction in cells. Biochem J. 435:297–312. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Chaffer CL and Weinberg RA: A perspective on cancer cell metastasis. Science. 331:1559–1564. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Bao B, Wang Z, Ali S, Kong D, Li Y, Ahmad A, Banerjee S, Azmi AS, Miele L and Sarkar FH: Notch-1 induces epithelial-mesenchymal transition consistent with cancer stem cell phenotype in pancreatic cancer cells. Cancer Lett. 307:26–36. 2011. View Article : Google Scholar : PubMed/NCBI

17 

Wang Z, Li Y, Ahmad A, Azmi AS, Kong D, Banerjee S and Sarkar FH: Targeting miRNAs involved in cancer stem cell and EMT regulation: An emerging concept in overcoming drug resistance. Drug Resist Updat. 13:109–118. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Heldin CH, Vanlandewijck M and Moustakas A: Regulation of EMT by TGFβ in cancer. FEBS Lett. 586:1959–1970. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Horiguchi K, Sakamoto K, Koinuma D, Semba K, Inoue A, Inoue S, Fujii H, Yamaguchi A, Miyazawa K, Miyazono K and Saitoh M: TGF-β drives epithelial-mesenchymal transition through δEF1-mediated downregulation of ESRP. Oncogene. 31:3190–3201. 2012. View Article : Google Scholar : PubMed/NCBI

20 

Boreddy SR and Srivastava SK: Deguelin suppresses pancreatic tumor growth and metastasis by inhibiting epithelial-to-mesenchymal transition in an orthotopic model. Oncogene. 32:3980–3991. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Lou C, Zhang F, Yang M, Zhao J, Zeng W, Fang X, Zhang Y, Zhang C and Liang W: Naringenin decreases invasiveness and metastasis by inhibiting TGF-β-induced epithelial to mesenchymal transition in pancreatic cancer cells. PLoS One. 7:e509562012. View Article : Google Scholar : PubMed/NCBI

22 

Chen EI: Mitochondrial dysfunction and cancer metastasis. J Bioenerg Biomembr. 44:619–622. 2012. View Article : Google Scholar : PubMed/NCBI

23 

Malik AN and Czajka A: Is mitochondrial DNA content a potential biomarker of mitochondrial dysfunction? Mitochondrion. 13:481–492. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Yu M: Generation, function and diagnostic value of mitochondrial DNA copy number alterations in human cancers. Life Sci. 89:65–71. 2011. View Article : Google Scholar : PubMed/NCBI

25 

Sotgia F, Whitaker-Menezes D, Martinez-Outschoorn UE, Flomenberg N, Birbe RC, Witkiewicz AK, Howell A, Philp NJ, Pestell RG and Lisanti MP: Mitochondrial metabolism in cancer metastasis: Visualizing tumor cell mitochondria and the ‘reverse Warburg effect’ in positive lymph node tissue. Cell Cycle. 11:1445–1454. 2012. View Article : Google Scholar : PubMed/NCBI

26 

Lin CS, Lee HT, Lee SY, Shen YA, Wang LS, Chen YJ and Wei YH: High mitochondrial DNA copy number and bioenergetic function are associated with tumor invasion of esophageal squamous cell carcinoma cell lines. Int J Mol Sci. 13:11228–11246. 2012. View Article : Google Scholar : PubMed/NCBI

27 

Yuan Y, Chen Y, Zhang P, Huang S, Zhu C, Ding G, Liu B, Yang T and Zhang A: Mitochondrial dysfunction accounts for aldosterone-induced epithelial-to-mesenchymal transition of renal proximal tubular epithelial cells. Free Radic Biol Med. 53:30–43. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Hung WY, Huang KH, Wu CW, Chi CW, Kao HL, Li AF, Yin PH and Lee HC: Mitochondrial dysfunction promotes cell migration via reactive oxygen species-enhanced β5-integrin expression in human gastric cancer SC-M1 cells. Biochim Biophys Acta. 1820:1102–1110. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Ma J, Zhang Q, Chen S, Fang B, Yang Q, Chen C, Miele L, Sarkar FH, Xia J and Wang Z: Mitochondrial dysfunction promotes breast cancer cell migration and invasion through HIF1α accumulation via increased production of reactive oxygen species. PLoS One. 8:e694852013. View Article : Google Scholar : PubMed/NCBI

30 

Moreno-Sánchez R, Rodríguez-Enríquez S, Marín-Hernández A and Saavedra E: Energy metabolism in tumor cells. FEBS J. 274:1393–1418. 2007. View Article : Google Scholar : PubMed/NCBI

31 

Ishikawa K, Takenaga K, Akimoto M, Koshikawa N, Yamaguchi A, Imanishi H, Nakada K, Honma Y and Hayashi J: ROS-generating mitochondrial DNA mutations can regulate tumor cell metastasis. Science. 320:661–664. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Bonnard C, Durand A, Peyrol S, Chanseaume E, Chauvin MA, Morio B, Vidal H and Rieusset J: Mitochondrial dysfunction results from oxidative stress in the skeletal muscle of diet-induced insulin-resistant mice. J Clin Invest. 118:789–800. 2008.PubMed/NCBI

33 

Zhou G, Dada LA, Wu M, Kelly A, Trejo H, Zhou Q, Varga J and Sznajder JI: Hypoxia-induced alveolar epithelial-mesenchymal transition requires mitochondrial ROS and hypoxia-inducible factor 1. Am J Physiol Lung Cell Mol Physiol. 297:L1120–L1130. 2009. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2017
Volume 13 Issue 3

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Guo Q: Changes in mitochondrial function during EMT induced by TGFβ-1 in pancreatic cancer. Oncol Lett 13: 1575-1580, 2017
APA
Guo, Q. (2017). Changes in mitochondrial function during EMT induced by TGFβ-1 in pancreatic cancer. Oncology Letters, 13, 1575-1580. https://doi.org/10.3892/ol.2017.5613
MLA
Guo, Q."Changes in mitochondrial function during EMT induced by TGFβ-1 in pancreatic cancer". Oncology Letters 13.3 (2017): 1575-1580.
Chicago
Guo, Q."Changes in mitochondrial function during EMT induced by TGFβ-1 in pancreatic cancer". Oncology Letters 13, no. 3 (2017): 1575-1580. https://doi.org/10.3892/ol.2017.5613