Skip to main content
Top
Published in: BMC Cancer 1/2018

Open Access 01-12-2018 | Research article

Hypoxia promotes acquisition of aggressive phenotypes in human malignant mesothelioma

Authors: Myung-Chul Kim, Sung-Hyun Hwang, Na-Yon Kim, Hong-Seok Lee, Sumin Ji, Yeseul Yang, Yongbaek Kim

Published in: BMC Cancer | Issue 1/2018

Login to get access

Abstract

Background

Hypoxia is a hallmark of the solid tumor microenvironment and is associated with poor outcomes in cancer patients. The present study was performed to investigate mechanisms underlying the hypoxia-induced phenotypic changes using human malignant mesothelioma (HMM) cells.

Methods

Hypoxic conditions were achieved by incubating HMM cells in the air chamber. The effect of hypoxia on phenotype changes in HMM cells was investigated by performing in vitro clonogenicity, drug resistance, migration, and invasion assays. Signaling pathways and molecules involved in the more aggressive behaviors of HMM cells under hypoxia were investigated. A two-tailed unpaired Student’s t-test or one-way ANOVA with Bonferroni post-test correction was used in this study.

Results

Hypoxic conditions upregulated hypoxia-inducible factor 1 alpha (HIF-1α) and HIF-2α in parallel with the upregulation of its target, Glut-1, in HMM cells. In vitro clonogenicity of HMM cells was significantly increased in hypoxic conditions, but the proliferation of cells at a high density in hypoxia was lower than that in normoxic conditions. The expression levels of HIF-2α and Oct4 were increased in hypoxic HMM cells. The percentage of cells with high CD44 expression was significantly higher in HMM cells cultured in hypoxia than those cultured in normoxia. Hypoxia significantly enhanced the resistance of HMM cells to cisplatin, which occurred through cytoprotection against cisplatin-induced apoptosis. While cisplatin treatment decreased the ratio of Bcl-2 to Bax in normoxic condition, hypoxia conversely increased the ratio in HMM cells treated with cisplatin. Hypoxia increased the mobility and invasiveness of HMM cells. Epithelial to mesenchymal transition was promoted, which was indicated by the repression of E-cadherin and the concomitant increase of vimentin in HMM cells.

Conclusions

The data illustrated that hypoxic conditions augmented the aggressive phenotypes of HMM cells at the biological and molecular levels. The present study provides valuable background information beginning to understand aggressiveness of HMM in tumor microenvironments, suggesting that a control measure for tumor hypoxia may be an effective therapeutic strategy to reduce the aggressiveness of cancer cells in HMM patients.
Appendix
Available only for authorised users
Literature
1.
go back to reference Ruan K, Song G, Ouyang G. Role of hypoxia in the hallmarks of human cancer. J Cell Biochem. 2009;107(6):1053–62.CrossRefPubMed Ruan K, Song G, Ouyang G. Role of hypoxia in the hallmarks of human cancer. J Cell Biochem. 2009;107(6):1053–62.CrossRefPubMed
2.
go back to reference Fukumura D, Jain RK. Tumor microenvironment abnormalities: causes, consequences, and strategies to normalize. J Cell Biochem. 2007;101(4):937–49.CrossRefPubMed Fukumura D, Jain RK. Tumor microenvironment abnormalities: causes, consequences, and strategies to normalize. J Cell Biochem. 2007;101(4):937–49.CrossRefPubMed
3.
go back to reference Cosse J-P, Michiels C. Tumour hypoxia affects the responsiveness of cancer cells to chemotherapy and promotes cancer progression. Anti-Cancer Agent Me. 2008;8(7):790–7.CrossRef Cosse J-P, Michiels C. Tumour hypoxia affects the responsiveness of cancer cells to chemotherapy and promotes cancer progression. Anti-Cancer Agent Me. 2008;8(7):790–7.CrossRef
4.
go back to reference Vaupel P, Mayer A. Hypoxia in cancer: significance and impact on clinical outcome. Cancer Metast Rev. 2007;26(2):225–39.CrossRef Vaupel P, Mayer A. Hypoxia in cancer: significance and impact on clinical outcome. Cancer Metast Rev. 2007;26(2):225–39.CrossRef
5.
go back to reference Zhou J, Schmid T, Schnitzer S, Brüne B. Tumor hypoxia and cancer progression. Cancer Lett. 2006;237(1):10–21.CrossRefPubMed Zhou J, Schmid T, Schnitzer S, Brüne B. Tumor hypoxia and cancer progression. Cancer Lett. 2006;237(1):10–21.CrossRefPubMed
6.
go back to reference Lee J-W, Bae S-H, Jeong J-W, Kim S-H, Kim K-W. Hypoxia-inducible factor (HIF-1) α: its protein stability and biological functions. Exp Mol Med. 2004;36(1):1–12.CrossRefPubMed Lee J-W, Bae S-H, Jeong J-W, Kim S-H, Kim K-W. Hypoxia-inducible factor (HIF-1) α: its protein stability and biological functions. Exp Mol Med. 2004;36(1):1–12.CrossRefPubMed
7.
go back to reference Keith B, Johnson RS, Simon MC. HIF1α and HIF2α: sibling rivalry in hypoxic tumour growth and progression. Nat Rev Cancer. 2012;12(1):9–22.CrossRef Keith B, Johnson RS, Simon MC. HIF1α and HIF2α: sibling rivalry in hypoxic tumour growth and progression. Nat Rev Cancer. 2012;12(1):9–22.CrossRef
10.
go back to reference Covello KL, Kehler J, Yu H, Gordan JD, Arsham AM, Hu C-J, Labosky PA, Simon MC, Keith B. HIF-2α regulates Oct-4: effects of hypoxia on stem cell function, embryonic development, and tumor growth. Genes Dev. 2006;20(5):557–70.CrossRefPubMedPubMedCentral Covello KL, Kehler J, Yu H, Gordan JD, Arsham AM, Hu C-J, Labosky PA, Simon MC, Keith B. HIF-2α regulates Oct-4: effects of hypoxia on stem cell function, embryonic development, and tumor growth. Genes Dev. 2006;20(5):557–70.CrossRefPubMedPubMedCentral
11.
12.
go back to reference Bianchi C, Bianchi T. Malignant mesothelioma: global incidence and relationship with asbestos. Ind Health. 2007;45(3):379–87.CrossRefPubMed Bianchi C, Bianchi T. Malignant mesothelioma: global incidence and relationship with asbestos. Ind Health. 2007;45(3):379–87.CrossRefPubMed
13.
go back to reference Mujoomdar AA, Tilleman TR, Richards WG, Bueno R, Sugarbaker DJ. Prevalence of in vitro chemotherapeutic drug resistance in primary malignant pleural mesothelioma: result in a cohort of 203 resection specimens. J Thorac Cardiov Sur. 2010;140(2):352–5.CrossRef Mujoomdar AA, Tilleman TR, Richards WG, Bueno R, Sugarbaker DJ. Prevalence of in vitro chemotherapeutic drug resistance in primary malignant pleural mesothelioma: result in a cohort of 203 resection specimens. J Thorac Cardiov Sur. 2010;140(2):352–5.CrossRef
14.
go back to reference Fennell DA, Rudd RM. Defective core-apoptosis signalling in diffuse malignant pleural mesothelioma: opportunities for effective drug development. Lancet Oncol. 2004;5(6):354–62.CrossRefPubMed Fennell DA, Rudd RM. Defective core-apoptosis signalling in diffuse malignant pleural mesothelioma: opportunities for effective drug development. Lancet Oncol. 2004;5(6):354–62.CrossRefPubMed
15.
go back to reference Blomberg C, Nilsson J, Holgersson G, Edlund P, Bergqvist M, Adwall L, Ekman S, Brattström D, Bergström S. Randomized trials of systemic medically-treated malignant mesothelioma: a systematic review. Anticancer Res. 2015;35(5):2493–501.PubMed Blomberg C, Nilsson J, Holgersson G, Edlund P, Bergqvist M, Adwall L, Ekman S, Brattström D, Bergström S. Randomized trials of systemic medically-treated malignant mesothelioma: a systematic review. Anticancer Res. 2015;35(5):2493–501.PubMed
16.
go back to reference Fennell DA, Gaudino G, O'Byrne KJ, Mutti L, Van Meerbeeck J. Advances in the systemic therapy of malignant pleural mesothelioma. Nat Clin Pract Oncol. 2008;5(3):136–47.CrossRefPubMed Fennell DA, Gaudino G, O'Byrne KJ, Mutti L, Van Meerbeeck J. Advances in the systemic therapy of malignant pleural mesothelioma. Nat Clin Pract Oncol. 2008;5(3):136–47.CrossRefPubMed
17.
go back to reference Klabatsa A, Sheaff MT, Steele JP, Evans MT, Rudd RM, Fennell DA. Expression and prognostic significance of hypoxia-inducible factor 1alpha (HIF-1alpha) in malignant pleural mesothelioma (MPM). Lung Cancer. 2006;51(1):53–9.CrossRefPubMed Klabatsa A, Sheaff MT, Steele JP, Evans MT, Rudd RM, Fennell DA. Expression and prognostic significance of hypoxia-inducible factor 1alpha (HIF-1alpha) in malignant pleural mesothelioma (MPM). Lung Cancer. 2006;51(1):53–9.CrossRefPubMed
18.
go back to reference Francis RJ, Segard T, Morandeau L, Lee YG, Millward MJ, Segal A, Nowak AK. Characterization of hypoxia in malignant pleural mesothelioma with FMISO PET-CT. Lung Cancer. 2015;90(1):55–60.CrossRefPubMed Francis RJ, Segard T, Morandeau L, Lee YG, Millward MJ, Segal A, Nowak AK. Characterization of hypoxia in malignant pleural mesothelioma with FMISO PET-CT. Lung Cancer. 2015;90(1):55–60.CrossRefPubMed
19.
go back to reference Franken NA, Rodermond HM, Stap J, Haveman J, Van Bree C. Clonogenic assay of cells in vitro. Nat Proto. 2006;1(5):2315.CrossRef Franken NA, Rodermond HM, Stap J, Haveman J, Van Bree C. Clonogenic assay of cells in vitro. Nat Proto. 2006;1(5):2315.CrossRef
20.
go back to reference Jampel HD. Effect of brief exposure to mitomycin C on viability and proliferation of cultured human Tenon's capsule fibroblasts. Ophthalmology. 1992;99(9):1471–6.CrossRefPubMed Jampel HD. Effect of brief exposure to mitomycin C on viability and proliferation of cultured human Tenon's capsule fibroblasts. Ophthalmology. 1992;99(9):1471–6.CrossRefPubMed
21.
go back to reference Kim M-C, Cui F-J, Kim Y. Hydrogen peroxide promotes epithelial to mesenchymal transition and stemness in human malignant mesothelioma cells. Asian Pac J Cancer P. 2013;14(6):3625–30.CrossRef Kim M-C, Cui F-J, Kim Y. Hydrogen peroxide promotes epithelial to mesenchymal transition and stemness in human malignant mesothelioma cells. Asian Pac J Cancer P. 2013;14(6):3625–30.CrossRef
22.
go back to reference Ghani FI, Yamazaki H, Iwata S, Okamoto T, Aoe K, Okabe K, Mimura Y, Fujimoto N, Kishimoto T, Yamada T. Identification of cancer stem cell markers in human malignant mesothelioma cells. Biochem Biophys Res Commun. 2011;404(2):735–42.CrossRefPubMed Ghani FI, Yamazaki H, Iwata S, Okamoto T, Aoe K, Okabe K, Mimura Y, Fujimoto N, Kishimoto T, Yamada T. Identification of cancer stem cell markers in human malignant mesothelioma cells. Biochem Biophys Res Commun. 2011;404(2):735–42.CrossRefPubMed
23.
go back to reference Cortes-Dericks L, Froment L, Boesch R, Schmid RA, Karoubi G. Cisplatin-resistant cells in malignant pleural mesothelioma cell lines show ALDH high CD44+ phenotype and sphere-forming capacity. BMC Cancer. 2014;14(1):304.CrossRefPubMedPubMedCentral Cortes-Dericks L, Froment L, Boesch R, Schmid RA, Karoubi G. Cisplatin-resistant cells in malignant pleural mesothelioma cell lines show ALDH high CD44+ phenotype and sphere-forming capacity. BMC Cancer. 2014;14(1):304.CrossRefPubMedPubMedCentral
24.
go back to reference Cao W, Yacoub S, Shiverick KT, Namiki K, Sakai Y, Porvasnik S, Urbanek C, Rosser CJ. Dichloroacetate (DCA) sensitizes both wild-type and over expressing Bcl-2 prostate cancer cells in vitro to radiation. Prostate. 2008;68(11):1223–31.CrossRefPubMed Cao W, Yacoub S, Shiverick KT, Namiki K, Sakai Y, Porvasnik S, Urbanek C, Rosser CJ. Dichloroacetate (DCA) sensitizes both wild-type and over expressing Bcl-2 prostate cancer cells in vitro to radiation. Prostate. 2008;68(11):1223–31.CrossRefPubMed
25.
go back to reference Campos L, Sabido O, Rouault J-P, Guyotat D. Effects of BCL-2 antisense oligodeoxynucleotides on in vitro proliferation and survival of normal marrow progenitors and leukemic cells. Blood. 1994;84(2):595–600.PubMed Campos L, Sabido O, Rouault J-P, Guyotat D. Effects of BCL-2 antisense oligodeoxynucleotides on in vitro proliferation and survival of normal marrow progenitors and leukemic cells. Blood. 1994;84(2):595–600.PubMed
26.
go back to reference Goudarzi H, Hida Y, Takano H, Teramae H, Iizasa H. Hamada J-i. Hypoxia affects in vitro growth of newly established cell lines from patients with malignant pleural mesothelioma. Biomedl Res. 2013;34(1):13–21.CrossRef Goudarzi H, Hida Y, Takano H, Teramae H, Iizasa H. Hamada J-i. Hypoxia affects in vitro growth of newly established cell lines from patients with malignant pleural mesothelioma. Biomedl Res. 2013;34(1):13–21.CrossRef
27.
go back to reference Mimeault M, Batra SK. Hypoxia-inducing factors as master regulators of stemness properties and altered metabolism of cancer-and metastasis-initiating cells. J Cell Mol Med. 2013;17(1):30–54.CrossRefPubMedPubMedCentral Mimeault M, Batra SK. Hypoxia-inducing factors as master regulators of stemness properties and altered metabolism of cancer-and metastasis-initiating cells. J Cell Mol Med. 2013;17(1):30–54.CrossRefPubMedPubMedCentral
28.
go back to reference Ma Y, Liang D, Liu J, Axcrona K, Kvalheim G, Stokke T, Nesland JM, Suo Z. Prostate cancer cell lines under hypoxia exhibit greater stem-like properties. PLoS One. 2011;6(12):e29170.CrossRefPubMedPubMedCentral Ma Y, Liang D, Liu J, Axcrona K, Kvalheim G, Stokke T, Nesland JM, Suo Z. Prostate cancer cell lines under hypoxia exhibit greater stem-like properties. PLoS One. 2011;6(12):e29170.CrossRefPubMedPubMedCentral
29.
30.
go back to reference Yeung TM, Gandhi SC, Bodmer WF. Hypoxia and lineage specification of cell line-derived colorectal cancer stem cells. P Natl Acad Sci. 2011;108(11):4382–7.CrossRef Yeung TM, Gandhi SC, Bodmer WF. Hypoxia and lineage specification of cell line-derived colorectal cancer stem cells. P Natl Acad Sci. 2011;108(11):4382–7.CrossRef
31.
go back to reference Heddleston JM, Li Z, McLendon RE, Hjelmeland AB, Rich JN. The hypoxic microenvironment maintains glioblastoma stem cells and promotes reprogramming towards a cancer stem cell phenotype. Cell Cycle. 2009;8(20):3274–84.CrossRefPubMedPubMedCentral Heddleston JM, Li Z, McLendon RE, Hjelmeland AB, Rich JN. The hypoxic microenvironment maintains glioblastoma stem cells and promotes reprogramming towards a cancer stem cell phenotype. Cell Cycle. 2009;8(20):3274–84.CrossRefPubMedPubMedCentral
32.
33.
go back to reference Gordan JD, Bertout JA, Hu C-J, Diehl JA, Simon MC. HIF-2α promotes hypoxic cell proliferation by enhancing c-myc transcriptional activity. Cancer Cell. 2007;11(4):335–47.CrossRefPubMedPubMedCentral Gordan JD, Bertout JA, Hu C-J, Diehl JA, Simon MC. HIF-2α promotes hypoxic cell proliferation by enhancing c-myc transcriptional activity. Cancer Cell. 2007;11(4):335–47.CrossRefPubMedPubMedCentral
34.
go back to reference Hu Y-Y, Fu L-A, Li S-Z, Chen Y, Li J-C, Han J, Liang L, Li L, Ji C-C, Zheng M-H. Hif-1α and Hif-2α differentially regulate notch signaling through competitive interaction with the intracellular domain of notch receptors in glioma stem cells. Cancer Lett. 2014;349(1):67–76.CrossRefPubMed Hu Y-Y, Fu L-A, Li S-Z, Chen Y, Li J-C, Han J, Liang L, Li L, Ji C-C, Zheng M-H. Hif-1α and Hif-2α differentially regulate notch signaling through competitive interaction with the intracellular domain of notch receptors in glioma stem cells. Cancer Lett. 2014;349(1):67–76.CrossRefPubMed
35.
go back to reference Rohwer N, Cramer T. Hypoxia-mediated drug resistance: novel insights on the functional interaction of HIFs and cell death pathways. Drug Resist Update. 2011;14(3):191–201.CrossRef Rohwer N, Cramer T. Hypoxia-mediated drug resistance: novel insights on the functional interaction of HIFs and cell death pathways. Drug Resist Update. 2011;14(3):191–201.CrossRef
36.
go back to reference Gross A, McDonnell JM, Korsmeyer SJ. BCL-2 family members and the mitochondria in apoptosis. Genes Dev. 1999;13(15):1899–911.CrossRefPubMed Gross A, McDonnell JM, Korsmeyer SJ. BCL-2 family members and the mitochondria in apoptosis. Genes Dev. 1999;13(15):1899–911.CrossRefPubMed
38.
go back to reference Toshiyuki M, Reed JC. Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell. 1995;80(2):293–9.CrossRef Toshiyuki M, Reed JC. Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell. 1995;80(2):293–9.CrossRef
39.
go back to reference Reddy RM, Yeow W-S, Chua A, Nguyen DM, Baras A, Ziauddin MF, Shamimi-Noori SM, Maxhimer JB, Schrump DS, Nguyen DM. Rapid and profound potentiation of Apo2L/TRAIL-mediated cytotoxicity and apoptosis in thoracic cancer cells by the histone deacetylase inhibitor Trichostatin a: the essential role of the mitochondria-mediated caspase activation cascade. Apoptosis. 2007;12(1):55–71.CrossRefPubMed Reddy RM, Yeow W-S, Chua A, Nguyen DM, Baras A, Ziauddin MF, Shamimi-Noori SM, Maxhimer JB, Schrump DS, Nguyen DM. Rapid and profound potentiation of Apo2L/TRAIL-mediated cytotoxicity and apoptosis in thoracic cancer cells by the histone deacetylase inhibitor Trichostatin a: the essential role of the mitochondria-mediated caspase activation cascade. Apoptosis. 2007;12(1):55–71.CrossRefPubMed
40.
go back to reference Goudarzi H, Iizasa H, Furuhashi M, Nakazawa S, Nakane R, Liang S, Hida Y, Yanagihara K, Kubo T, Nakagawa K. Enhancement of in vitro cell motility and invasiveness of human malignant pleural mesothelioma cells through the HIF-1α-MUC1 pathway. Cancer Lett. 2013;339(1):82–92.CrossRefPubMed Goudarzi H, Iizasa H, Furuhashi M, Nakazawa S, Nakane R, Liang S, Hida Y, Yanagihara K, Kubo T, Nakagawa K. Enhancement of in vitro cell motility and invasiveness of human malignant pleural mesothelioma cells through the HIF-1α-MUC1 pathway. Cancer Lett. 2013;339(1):82–92.CrossRefPubMed
41.
go back to reference Fassina A, Cappellesso R, Guzzardo V, Dalla Via L, Piccolo S, Ventura L, Fassan M. Epithelial–mesenchymal transition in malignant mesothelioma. Modern Pathol. 2012;25(1):86–99.CrossRef Fassina A, Cappellesso R, Guzzardo V, Dalla Via L, Piccolo S, Ventura L, Fassan M. Epithelial–mesenchymal transition in malignant mesothelioma. Modern Pathol. 2012;25(1):86–99.CrossRef
42.
go back to reference Jiang J, Tang Y-L, Liang X-H. EMT: a new vision of hypoxia promoting cancer progression. Cancer Biol Ther. 2011;11(8):714–23.CrossRefPubMed Jiang J, Tang Y-L, Liang X-H. EMT: a new vision of hypoxia promoting cancer progression. Cancer Biol Ther. 2011;11(8):714–23.CrossRefPubMed
43.
go back to reference Casarsa C, Bassani N, Ambrogi F, Zabucchi G, Boracchi P, Biganzoli E, Coradini D. Epithelial-to-mesenchymal transition, cell polarity and stemness-associated features in malignant pleural mesothelioma. Cancer Lett. 2011;302(2):136–43.CrossRefPubMed Casarsa C, Bassani N, Ambrogi F, Zabucchi G, Boracchi P, Biganzoli E, Coradini D. Epithelial-to-mesenchymal transition, cell polarity and stemness-associated features in malignant pleural mesothelioma. Cancer Lett. 2011;302(2):136–43.CrossRefPubMed
44.
go back to reference Klabatsa A, Sheaff M, Steele J, Evans M, Rudd R, Fennell D. Expression and prognostic significance of hypoxia-inducible factor 1α (HIF-1α) in malignant pleural mesothelioma (MPM). Lung Cancer. 2006;51(1):53–9.CrossRefPubMed Klabatsa A, Sheaff M, Steele J, Evans M, Rudd R, Fennell D. Expression and prognostic significance of hypoxia-inducible factor 1α (HIF-1α) in malignant pleural mesothelioma (MPM). Lung Cancer. 2006;51(1):53–9.CrossRefPubMed
45.
go back to reference Schramm A, Opitz I, Thies S, Seifert B, Moch H, Weder W, Soltermann A. Prognostic significance of epithelial–mesenchymal transition in malignant pleural mesothelioma. Eur J Cardio-Thorac. 2010;37(3):566–72.CrossRef Schramm A, Opitz I, Thies S, Seifert B, Moch H, Weder W, Soltermann A. Prognostic significance of epithelial–mesenchymal transition in malignant pleural mesothelioma. Eur J Cardio-Thorac. 2010;37(3):566–72.CrossRef
46.
go back to reference Manente AG, Pinton G, Zonca S, Tavian D, Habib T, Jithesh PV, Fennell D, Nilsson S, Moro L. KDM6B histone demethylase is an epigenetic regulator of estrogen receptor β expression in human pleural mesothelioma. Epigenomics. 2016;8(9):1227–38.CrossRefPubMed Manente AG, Pinton G, Zonca S, Tavian D, Habib T, Jithesh PV, Fennell D, Nilsson S, Moro L. KDM6B histone demethylase is an epigenetic regulator of estrogen receptor β expression in human pleural mesothelioma. Epigenomics. 2016;8(9):1227–38.CrossRefPubMed
Metadata
Title
Hypoxia promotes acquisition of aggressive phenotypes in human malignant mesothelioma
Authors
Myung-Chul Kim
Sung-Hyun Hwang
Na-Yon Kim
Hong-Seok Lee
Sumin Ji
Yeseul Yang
Yongbaek Kim
Publication date
01-12-2018
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2018
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-018-4720-z

Other articles of this Issue 1/2018

BMC Cancer 1/2018 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine