Skip to main content
Top
Published in: International Journal of Clinical Oncology 3/2013

01-06-2013 | Original Article

Epigallocatechin-3-gallate potentiates curcumin’s ability to suppress uterine leiomyosarcoma cell growth and induce apoptosis

Authors: Akiko Kondo, Takashi Takeda, Bin Li, Kenji Tsuiji, Mari Kitamura, Tze Fang Wong, Nobuo Yaegashi

Published in: International Journal of Clinical Oncology | Issue 3/2013

Login to get access

Abstract

Background

Uterine leiomyosarcoma (LMS) has an unfavorable response to standard chemotherapeutic regimens. Two natural occurring compounds, curcumin and epigallocatechin gallate (EGCG), are reported to have anti-cancer activity. We previously reported that curcumin reduced uterine LMS cell proliferation by targeting the AKT–mTOR pathway. However, challenges remain in overcoming curcumin’s low bioavailability.

Methods

The human LMS cell line SKN was used. The effect of EGCG, curcumin or their combination on cell growth was detected by MTS assay. Their effect on AKT, mTOR, and S6 was detected by Western blotting. The induction of apoptosis was determined by Western blotting using cleaved-PARP specific antibody, caspase-3 activity and TUNEL assay. Intracellular curcumin level was determined by a spectrophotometric method. Antibody against EGCG cell surface receptor, 67-kDa laminin receptor (67LR), was used to investigate the role of the receptor in curcumin’s increased potency by EGCG.

Results

In this study, we showed that the combination of EGCG and curcumin significantly reduced SKN cell proliferation more than either drug alone. The combination inhibited AKT, mTOR, and S6 phosphorylation, and induced apoptosis at a much lower curcumin concentration than previously reported. EGCG enhanced the incorporation of curcumin. 67LR antibody partially rescued cell proliferation suppression by the combination treatment, but was not involved in the EGCG-enhanced intracellular incorporation of curcumin.

Conclusions

EGCG significantly lowered the concentration of curcumin required to inhibit the AKT–mTOR pathway, reduce cell proliferation and induce apoptosis in uterine LMS cells by enhancing intracellular incorporation of curcumin, but the process was independent of 67LR.
Literature
1.
go back to reference Leitao MM, Sonoda Y, Brennan MF et al (2003) Incidence of lymph node and ovarian metastases in leiomyosarcoma of the uterus. Gynecol Oncol 91:209–212PubMedCrossRef Leitao MM, Sonoda Y, Brennan MF et al (2003) Incidence of lymph node and ovarian metastases in leiomyosarcoma of the uterus. Gynecol Oncol 91:209–212PubMedCrossRef
2.
go back to reference Naaman Y, Shveiky D, Ben-Shachar I et al (2011) Uterine sarcoma: prognostic factors and treatment evaluation. Isr Med Assoc J 13:76–79PubMed Naaman Y, Shveiky D, Ben-Shachar I et al (2011) Uterine sarcoma: prognostic factors and treatment evaluation. Isr Med Assoc J 13:76–79PubMed
3.
go back to reference Muss HB, Bundy B, DiSaia PJ et al (1985) Treatment of recurrent or advanced uterine sarcoma: a randomized trial of doxorubicin versus doxorubicin and cyclophosphamide (a phase III trial of the Gynecologic Oncology Group). Cancer 55:1648–1653PubMedCrossRef Muss HB, Bundy B, DiSaia PJ et al (1985) Treatment of recurrent or advanced uterine sarcoma: a randomized trial of doxorubicin versus doxorubicin and cyclophosphamide (a phase III trial of the Gynecologic Oncology Group). Cancer 55:1648–1653PubMedCrossRef
4.
go back to reference Omura GA, Major FJ, Blessing JA et al (1983) A randomized study of adriamycin with and without dimethyl triazenoimidazole carboxamide in advanced uterine sarcomas. Cancer 52:626–632PubMedCrossRef Omura GA, Major FJ, Blessing JA et al (1983) A randomized study of adriamycin with and without dimethyl triazenoimidazole carboxamide in advanced uterine sarcomas. Cancer 52:626–632PubMedCrossRef
5.
go back to reference Sutton G, Blessing JA, Malfetano JH (1996) Ifosfamide and doxorubicin in the treatment of advanced leiomyosarcomas of the uterus: a Gynecologic Oncology Group Study. Gynecol Oncol 62:226–229PubMedCrossRef Sutton G, Blessing JA, Malfetano JH (1996) Ifosfamide and doxorubicin in the treatment of advanced leiomyosarcomas of the uterus: a Gynecologic Oncology Group Study. Gynecol Oncol 62:226–229PubMedCrossRef
6.
go back to reference Piver MS, Lele SB, Marchetti DL et al (1988) Effect of adjuvant chemotherapy on time to recurrence and survival of stage I uterine sarcomas. J Surg Oncol 38:233–239PubMedCrossRef Piver MS, Lele SB, Marchetti DL et al (1988) Effect of adjuvant chemotherapy on time to recurrence and survival of stage I uterine sarcomas. J Surg Oncol 38:233–239PubMedCrossRef
7.
go back to reference Omura GA, Blessing JA, Major F et al (1985) A randomized clinical trial of adjuvant adriamycin in uterine sarcomas: a Gynecologic Oncology Group study. J Clin Oncol 3:1240–1245PubMed Omura GA, Blessing JA, Major F et al (1985) A randomized clinical trial of adjuvant adriamycin in uterine sarcomas: a Gynecologic Oncology Group study. J Clin Oncol 3:1240–1245PubMed
8.
go back to reference Hernando E, Charytonowicz E, Dudas ME et al (2007) The AKT–mTOR pathway plays a critical role in the development of leiomyosarcomas. Nat Med 13:748–753PubMedCrossRef Hernando E, Charytonowicz E, Dudas ME et al (2007) The AKT–mTOR pathway plays a critical role in the development of leiomyosarcomas. Nat Med 13:748–753PubMedCrossRef
9.
go back to reference Amant F, Coosemans A, Debiec-Rychter M et al (2009) Clinical management of uterine sarcomas. Lancet Oncol 10:1188–1198PubMedCrossRef Amant F, Coosemans A, Debiec-Rychter M et al (2009) Clinical management of uterine sarcomas. Lancet Oncol 10:1188–1198PubMedCrossRef
10.
go back to reference Sarbassov DD, Ali SM, Kim DH et al (2004) Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton. Curr Biol 14:1296–1302PubMedCrossRef Sarbassov DD, Ali SM, Kim DH et al (2004) Rictor, a novel binding partner of mTOR, defines a rapamycin-insensitive and raptor-independent pathway that regulates the cytoskeleton. Curr Biol 14:1296–1302PubMedCrossRef
11.
go back to reference Breuleux M, Klopfenstein M, Stephan C et al (2009) Increased AKT S473 phosphorylation after mTORC1 inhibition is rictor dependent and does not predict tumor cell response to PI3K/mTOR inhibition. Mol Cancer Ther 8:742–753PubMedCrossRef Breuleux M, Klopfenstein M, Stephan C et al (2009) Increased AKT S473 phosphorylation after mTORC1 inhibition is rictor dependent and does not predict tumor cell response to PI3K/mTOR inhibition. Mol Cancer Ther 8:742–753PubMedCrossRef
12.
go back to reference Sarbassov DD, Guertin DA, Ali SM et al (2005) Phosphorylation and regulation of Akt/PKB by the rictor–mTOR complex. Science 307:1098–1101PubMedCrossRef Sarbassov DD, Guertin DA, Ali SM et al (2005) Phosphorylation and regulation of Akt/PKB by the rictor–mTOR complex. Science 307:1098–1101PubMedCrossRef
13.
go back to reference Lambert JD, Yang CS (2003) Mechanisms of cancer prevention by tea constituents. J Nutr 133:3262S–3267SPubMed Lambert JD, Yang CS (2003) Mechanisms of cancer prevention by tea constituents. J Nutr 133:3262S–3267SPubMed
14.
go back to reference Yang CS, Sang S, Lambert JD et al (2006) Possible mechanisms of the cancer-preventive activities of green tea. Mol Nutr Food Res 50:170–175PubMedCrossRef Yang CS, Sang S, Lambert JD et al (2006) Possible mechanisms of the cancer-preventive activities of green tea. Mol Nutr Food Res 50:170–175PubMedCrossRef
15.
go back to reference Brown MD (1999) Green tea (Camellia sinensis) extract and its possible role in the prevention of cancer. Altern Med Rev 4:360–370PubMed Brown MD (1999) Green tea (Camellia sinensis) extract and its possible role in the prevention of cancer. Altern Med Rev 4:360–370PubMed
16.
go back to reference Bettuzzi S, Brausi M, Rizzi F et al (2006) Chemoprevention of human prostate cancer by oral administration of green tea catechins in volunteers with high-grade prostate intraepithelial neoplasia: a preliminary report from a one-year proof-of-principle study. Cancer Res 66:1234–1240PubMedCrossRef Bettuzzi S, Brausi M, Rizzi F et al (2006) Chemoprevention of human prostate cancer by oral administration of green tea catechins in volunteers with high-grade prostate intraepithelial neoplasia: a preliminary report from a one-year proof-of-principle study. Cancer Res 66:1234–1240PubMedCrossRef
17.
go back to reference Toda M, Okubo S, Ikigai H et al (1990) Antibacterial and anti-hemolysin activities of tea catechins and their structural relatives. Nippon Saikingaku Zassi 45:561–566CrossRef Toda M, Okubo S, Ikigai H et al (1990) Antibacterial and anti-hemolysin activities of tea catechins and their structural relatives. Nippon Saikingaku Zassi 45:561–566CrossRef
18.
go back to reference Lin YL, Lin JK (1997) (−)-Epigallocatechin-3-gallate blocks the induction of nitric oxide synthase by down-regulating lipopolysaccharide-induced activity of transcription factor nuclear factor-κB. Mol Pharmacol 52:465–472PubMed Lin YL, Lin JK (1997) (−)-Epigallocatechin-3-gallate blocks the induction of nitric oxide synthase by down-regulating lipopolysaccharide-induced activity of transcription factor nuclear factor-κB. Mol Pharmacol 52:465–472PubMed
19.
go back to reference Maeda-Yamamoto M, Inagaki N, Kitaura J et al (2004) O-Methylated catechins from tea leaves inhibit multiple protein kinases in mast cells. J Immunol 172:4486–4492PubMed Maeda-Yamamoto M, Inagaki N, Kitaura J et al (2004) O-Methylated catechins from tea leaves inhibit multiple protein kinases in mast cells. J Immunol 172:4486–4492PubMed
20.
go back to reference Wheeler DS, Catravas JD, Odoms K et al (2004) Epigallocatechin-3-gallate, a green tea-derived polyphenol, inhibits IL-1β-dependent proinflammatory signal transduction in cultured respiratory epithelial cells. J Nutr 134:1039–1044PubMed Wheeler DS, Catravas JD, Odoms K et al (2004) Epigallocatechin-3-gallate, a green tea-derived polyphenol, inhibits IL-1β-dependent proinflammatory signal transduction in cultured respiratory epithelial cells. J Nutr 134:1039–1044PubMed
21.
go back to reference Yang F, Oz HS, Barve S et al (2001) The green tea polyphenol (−)-epigallocatechin-3-gallate blocks nuclear factor-κB activation by inhibiting IκB kinase activity in the intestinal epithelial cell line IEC-6. Mol Pharmacol 60:528–533PubMed Yang F, Oz HS, Barve S et al (2001) The green tea polyphenol (−)-epigallocatechin-3-gallate blocks nuclear factor-κB activation by inhibiting IκB kinase activity in the intestinal epithelial cell line IEC-6. Mol Pharmacol 60:528–533PubMed
22.
go back to reference Van Aller GS, Carson JD, Tang W et al (2011) Epigallocatechin gallate (EGCG), a major component of green tea, is a dual phosphoinositide-3-kinase/mTOR inhibitor. Biochem Biophys Res Co 406:194–199CrossRef Van Aller GS, Carson JD, Tang W et al (2011) Epigallocatechin gallate (EGCG), a major component of green tea, is a dual phosphoinositide-3-kinase/mTOR inhibitor. Biochem Biophys Res Co 406:194–199CrossRef
23.
go back to reference Anand P, Sundaram C, Jhurani S et al (2008) Curcumin and cancer: an “old-age” disease with an “age-old” solution. Cancer Lett 267:133–164PubMedCrossRef Anand P, Sundaram C, Jhurani S et al (2008) Curcumin and cancer: an “old-age” disease with an “age-old” solution. Cancer Lett 267:133–164PubMedCrossRef
24.
go back to reference Wong TF, Takeda T, Li B et al (2011) Curcumin disrupts uterine leiomyosarcoma cells through AKT–mTOR pathway inhibition. Gynecol Oncol 122:141–148PubMedCrossRef Wong TF, Takeda T, Li B et al (2011) Curcumin disrupts uterine leiomyosarcoma cells through AKT–mTOR pathway inhibition. Gynecol Oncol 122:141–148PubMedCrossRef
25.
go back to reference Somers-Edgar TJ, Scandlyn MJ, Stuart EC et al (2008) The combination of epigallocatechin gallate and curcumin suppresses ERα-breast cancer cell growth in vitro and in vivo. Int J Cancer 122:1966–1971PubMedCrossRef Somers-Edgar TJ, Scandlyn MJ, Stuart EC et al (2008) The combination of epigallocatechin gallate and curcumin suppresses ERα-breast cancer cell growth in vitro and in vivo. Int J Cancer 122:1966–1971PubMedCrossRef
26.
go back to reference Ghosh AK, Kay NE, Secreto CR et al (2009) Curcumin inhibits prosurvival pathways in chronic lymphocytic leukemia B cells and may overcome their stromal protection in combination with EGCG. Clin Cancer Res 15:1250–1258PubMedCrossRef Ghosh AK, Kay NE, Secreto CR et al (2009) Curcumin inhibits prosurvival pathways in chronic lymphocytic leukemia B cells and may overcome their stromal protection in combination with EGCG. Clin Cancer Res 15:1250–1258PubMedCrossRef
27.
go back to reference Saha A, Kuzuhara T, Echigo N et al (2010) New role of (−)-epicatechin in enhancing the induction of growth inhibition and apoptosis in human lung cancer cells by curcumin. Cancer Prev Res 3:953–962CrossRef Saha A, Kuzuhara T, Echigo N et al (2010) New role of (−)-epicatechin in enhancing the induction of growth inhibition and apoptosis in human lung cancer cells by curcumin. Cancer Prev Res 3:953–962CrossRef
28.
go back to reference Tachibana H, Koga K, Fujimura Y et al (2004) A receptor of green tea polyphenol EGCG. Nat Struct Mol Biol 11:380–381PubMedCrossRef Tachibana H, Koga K, Fujimura Y et al (2004) A receptor of green tea polyphenol EGCG. Nat Struct Mol Biol 11:380–381PubMedCrossRef
29.
go back to reference Ishiwata I, Nozawa S, Nagal S et al (1977) Establishment of a human leiomyosarcoma cell line. Cancer Res 37:658–664PubMed Ishiwata I, Nozawa S, Nagal S et al (1977) Establishment of a human leiomyosarcoma cell line. Cancer Res 37:658–664PubMed
30.
go back to reference Byun EH, Fujimura Y, Yamada K et al (2010) TLR4 signaling inhibitory pathway induced by green tea polyphenol epigallocatechin-3-gallate through 67-kDa laminin receptor. J Immunol 185:33–45CrossRef Byun EH, Fujimura Y, Yamada K et al (2010) TLR4 signaling inhibitory pathway induced by green tea polyphenol epigallocatechin-3-gallate through 67-kDa laminin receptor. J Immunol 185:33–45CrossRef
31.
go back to reference Holy EW, Stänpfli SF, Akhmedov A et al (2010) Laminin receptor activation inhibits endothelial tissue factor expression. J Mol Cell Cardiol 48:1138–1145PubMedCrossRef Holy EW, Stänpfli SF, Akhmedov A et al (2010) Laminin receptor activation inhibits endothelial tissue factor expression. J Mol Cell Cardiol 48:1138–1145PubMedCrossRef
32.
go back to reference Kuesap J, Li B, Satarug S et al (2008) Prostaglandin D2 induces heme oxygenase-1 in human retinal pigment epithelial cells. Biochem Biophys Res Commun 367:413–419PubMedCrossRef Kuesap J, Li B, Satarug S et al (2008) Prostaglandin D2 induces heme oxygenase-1 in human retinal pigment epithelial cells. Biochem Biophys Res Commun 367:413–419PubMedCrossRef
33.
go back to reference Scott DW, Loo G (2007) Curcumin-induced GADD153 upregulation: modulation by glutathione. J Cell Biochem 101:307–320PubMedCrossRef Scott DW, Loo G (2007) Curcumin-induced GADD153 upregulation: modulation by glutathione. J Cell Biochem 101:307–320PubMedCrossRef
34.
go back to reference Shoba G, Joy D, Joseph T et al (1998) Influence of piperine on the pharmacokinetics of curcumin in animals and human volunteers. Planta Med 64:353–356PubMedCrossRef Shoba G, Joy D, Joseph T et al (1998) Influence of piperine on the pharmacokinetics of curcumin in animals and human volunteers. Planta Med 64:353–356PubMedCrossRef
35.
go back to reference Telang N, Katdare M (2007) Combinatorial prevention of carcinogenic risk in a model for familial colon cancer. Oncol Rep 17:909–914PubMed Telang N, Katdare M (2007) Combinatorial prevention of carcinogenic risk in a model for familial colon cancer. Oncol Rep 17:909–914PubMed
36.
go back to reference Wan X, Harkavy B, Shen N et al (2007) Rapamycin induces feedback activation of Akt signaling through an IGF-1R-dependent mechanism. Oncogene 26:1932–1940PubMedCrossRef Wan X, Harkavy B, Shen N et al (2007) Rapamycin induces feedback activation of Akt signaling through an IGF-1R-dependent mechanism. Oncogene 26:1932–1940PubMedCrossRef
37.
go back to reference Tewari M, Quan LT, O’Rourke K et al (1995) Yama/CPP32β, a mammalian homolog of CED-3, is a CrmA-inhibitable protease that cleaves the death substrate poly(ADP-ribose)polymerase. Cell 81:801–809PubMedCrossRef Tewari M, Quan LT, O’Rourke K et al (1995) Yama/CPP32β, a mammalian homolog of CED-3, is a CrmA-inhibitable protease that cleaves the death substrate poly(ADP-ribose)polymerase. Cell 81:801–809PubMedCrossRef
Metadata
Title
Epigallocatechin-3-gallate potentiates curcumin’s ability to suppress uterine leiomyosarcoma cell growth and induce apoptosis
Authors
Akiko Kondo
Takashi Takeda
Bin Li
Kenji Tsuiji
Mari Kitamura
Tze Fang Wong
Nobuo Yaegashi
Publication date
01-06-2013
Publisher
Springer Japan
Published in
International Journal of Clinical Oncology / Issue 3/2013
Print ISSN: 1341-9625
Electronic ISSN: 1437-7772
DOI
https://doi.org/10.1007/s10147-012-0387-7

Other articles of this Issue 3/2013

International Journal of Clinical Oncology 3/2013 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