Skip to main content
Top
Published in: Cellular Oncology 1/2016

01-02-2016 | Original Paper

Treatment of LS174T colorectal cancer stem-like cells with n-3 PUFAs induces growth suppression through inhibition of survivin expression and induction of caspase-3 activation

Published in: Cellular Oncology | Issue 1/2016

Login to get access

Abstract

Purpose

Colorectal cancer stem cells (CCSCs) are thought to contribute to tumor initiation, progression, metastasis, chemo-resistance and therapy failure. Therefore, assessment of the effectiveness of agents with anti-proliferative activities against CCSCs is warranted. Several studies have shown that different tumorigenic steps, ranging from initiation to metastasis, can be affected by n-3 polyunsaturated fatty acids (PUFAs). Here, we evaluated the effects of the PUFA components docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA), alone or in combination, on LS174T cells that serve as a model for colorectal cancer initiating cells with stem cell-like properties.

Methods

LS174T cells were treated with 50, 100 and 150 μM DHA and EPA, or equal mixtures of DHA/EPA (i.e., 25/25, 50/50 and 75/75 μM), after which cell number, viability, growth inhibition, survivin expression, caspase-3 activation and apoptotic rate were evaluated.

Results

We found that treatment of LS174T cells with increasing PUFA concentrations significantly increased growth inhibition in a dose- and time-dependent manner. After a 72 h treatment with 150 μM DHA and EPA, or their combination (75/75 μM), growth rates were inhibited by 80.3 ± 5.5 %, 79.3 ± 5 % and 71.1 ± 1 %, respectively, compared to untreated cells. We also found that treatment for 48 h with 100 μM DHA and EPA, or their combination (50/50 μM), resulted in 2.9-, 3- and 2.6-fold increases in caspase-3 activation, as well as 54, 62.4 and 100 % decreases in survivin mRNA expression levels, respectively, compared to untreated cells. Low survivin mRNA levels combined with high caspase-3 activity levels were found to correlate with a higher growth inhibition in PUFA-treated cells. DHA appears to be a more potent growth inhibitor than EPA and the DHA/EPA combination. An increase in the number of apoptotic cells (early + late), ranging from 12.9 to 44.7 %, was observed with increasing DHA doses.

Conclusion

From our data we conclude that PUFAs induce growth inhibition via targeting survivin expression in LS174T cells, which serve as a model for CCSCs.
Literature
1.
go back to reference R. Siegel, C. Desantis, A. Jemal, Colorectal cancer statistics. CA Cancer J. Clin. 64, 104–117 (2014)CrossRefPubMed R. Siegel, C. Desantis, A. Jemal, Colorectal cancer statistics. CA Cancer J. Clin. 64, 104–117 (2014)CrossRefPubMed
2.
go back to reference T. Cačev, G. Aralica, B. Lončar, S. Kapitanović, Loss of NF2/Merlin expression in advanced sporadic colorectal cancer. Cell. Oncol. 37, 69–77 (2014)CrossRef T. Cačev, G. Aralica, B. Lončar, S. Kapitanović, Loss of NF2/Merlin expression in advanced sporadic colorectal cancer. Cell. Oncol. 37, 69–77 (2014)CrossRef
3.
go back to reference B.J. Wilson, T. Schatton, M.H. Frank, N.Y. Frank, Colorectal cancer stem cells: biology and therapeutic implications. Curr. Colorectal Cancer Rep. 7, 128–135 (2011)PubMedCentralCrossRefPubMed B.J. Wilson, T. Schatton, M.H. Frank, N.Y. Frank, Colorectal cancer stem cells: biology and therapeutic implications. Curr. Colorectal Cancer Rep. 7, 128–135 (2011)PubMedCentralCrossRefPubMed
4.
go back to reference K. Vazquez-Santillan, J. Melendez-Zajgla, L. Jimenez-Hernandez, G. Martínez-Ruiz, V. Maldonado, NF-κB signaling in cancer stem cells: a promising therapeutic target? Cell. Oncol. 38, 327–339 (2015)CrossRef K. Vazquez-Santillan, J. Melendez-Zajgla, L. Jimenez-Hernandez, G. Martínez-Ruiz, V. Maldonado, NF-κB signaling in cancer stem cells: a promising therapeutic target? Cell. Oncol. 38, 327–339 (2015)CrossRef
5.
go back to reference V. Sulzyc-Bielicka, P. Domagala, D. Bielicki, K. Safranow, W. Domagala, Thymidylate synthase expression and p21 (WAF1)/p53 phenotype of colon cancers identify patients who may benefit from 5-fluorouracil based therapy. Cell. Oncol. 37, 17–28 (2014)CrossRef V. Sulzyc-Bielicka, P. Domagala, D. Bielicki, K. Safranow, W. Domagala, Thymidylate synthase expression and p21 (WAF1)/p53 phenotype of colon cancers identify patients who may benefit from 5-fluorouracil based therapy. Cell. Oncol. 37, 17–28 (2014)CrossRef
6.
go back to reference M.N. Hall, J.E. Chavarro, I.M. Lee, W.C. Willett, J. Ma, A 22-year prospective study of fish, n-3 fatty acid intake, and colorectal cancer risk in men. Cancer Epidemiol. Biomarkers Prev. 17, 1136–1143 (2008)PubMedCentralCrossRefPubMed M.N. Hall, J.E. Chavarro, I.M. Lee, W.C. Willett, J. Ma, A 22-year prospective study of fish, n-3 fatty acid intake, and colorectal cancer risk in men. Cancer Epidemiol. Biomarkers Prev. 17, 1136–1143 (2008)PubMedCentralCrossRefPubMed
7.
go back to reference N. Habermann, B. Christian, B. Luckas, B.L. Pool-Zobel, E.K. Lund, M. Glei, Effects of fatty acids on metabolism and cell growth of human colon cell lines of different transformation state. Biofactors 35, 460–467 (2009)CrossRefPubMed N. Habermann, B. Christian, B. Luckas, B.L. Pool-Zobel, E.K. Lund, M. Glei, Effects of fatty acids on metabolism and cell growth of human colon cell lines of different transformation state. Biofactors 35, 460–467 (2009)CrossRefPubMed
8.
go back to reference M. Anti, G. Marra, F. Armelao, G.M. Bartoli, R. Ficarelli, A. Percesepe, I. De Vitis, G. Maria, L. Sofo, G.L. Rapaccini, N. Gentiloni, E. Piccioni, G. Miggiano, Effect of omega-3 fatty acids on rectal mucosal cell proliferation in subjects at risk for colon cancer. Gastroenterology 103, 883–891 (1992)PubMed M. Anti, G. Marra, F. Armelao, G.M. Bartoli, R. Ficarelli, A. Percesepe, I. De Vitis, G. Maria, L. Sofo, G.L. Rapaccini, N. Gentiloni, E. Piccioni, G. Miggiano, Effect of omega-3 fatty acids on rectal mucosal cell proliferation in subjects at risk for colon cancer. Gastroenterology 103, 883–891 (1992)PubMed
9.
go back to reference M. Anti, F. Armelao, G. Giancarlo Marra, A. Percesepe, G.M. Bartoli, P. Palozza, P. Parrella, C. Cantelta, N. Gentiloni, I.D. Vitis, G. Gasbarri, Effects of different doses of fish oil on rectal cell proliferation in patients with sporadic colonic adenomas. Gastroenterology 107, 1709–1718 (1994)PubMed M. Anti, F. Armelao, G. Giancarlo Marra, A. Percesepe, G.M. Bartoli, P. Palozza, P. Parrella, C. Cantelta, N. Gentiloni, I.D. Vitis, G. Gasbarri, Effects of different doses of fish oil on rectal cell proliferation in patients with sporadic colonic adenomas. Gastroenterology 107, 1709–1718 (1994)PubMed
10.
go back to reference G. Calviello, F. Nicuolo, S. Gragnoli, E. Piccioni, S. Serini, N. Maggiano, G. Tringali, P. Navarra, F.O. Ranelletti, P. Palozza, n-3 PUFAs reduce VEGF expression in human colon cancer cells modulating the COX-2/PGE2 induced ERK-1 and −2 and HIF-1 alpha induction pathway. Carcinogenesis 25, 2303–2310 (2004)CrossRefPubMed G. Calviello, F. Nicuolo, S. Gragnoli, E. Piccioni, S. Serini, N. Maggiano, G. Tringali, P. Navarra, F.O. Ranelletti, P. Palozza, n-3 PUFAs reduce VEGF expression in human colon cancer cells modulating the COX-2/PGE2 induced ERK-1 and −2 and HIF-1 alpha induction pathway. Carcinogenesis 25, 2303–2310 (2004)CrossRefPubMed
11.
go back to reference B.A. Narayanan, N.K. Narayanan, D. Desai, B. Pittman, B.S. Reddy, Effects of a combination of docosahexaenoic acid and 1,4-phenylene bis (methylene) selenocyanate on cyclooxygenase 2, inducible nitric oxide synthase and beta-catenin pathways in colon cancer cells. Carcinogenesis 25, 2443–2449 (2004)CrossRefPubMed B.A. Narayanan, N.K. Narayanan, D. Desai, B. Pittman, B.S. Reddy, Effects of a combination of docosahexaenoic acid and 1,4-phenylene bis (methylene) selenocyanate on cyclooxygenase 2, inducible nitric oxide synthase and beta-catenin pathways in colon cancer cells. Carcinogenesis 25, 2443–2449 (2004)CrossRefPubMed
12.
go back to reference J. Hofmanová, A. Vaculová, A. Kozubík, Polyunsaturated fatty acids sensitize human colon adenocarcinoma HT-29 cells to death receptor-mediated apoptosis. Cancer Lett. 218, 33–41 (2005)CrossRefPubMed J. Hofmanová, A. Vaculová, A. Kozubík, Polyunsaturated fatty acids sensitize human colon adenocarcinoma HT-29 cells to death receptor-mediated apoptosis. Cancer Lett. 218, 33–41 (2005)CrossRefPubMed
13.
go back to reference C.Y. Kuan, T.H. Walker, P.G. Luo, C.F. Chen, Long-chain polyunsaturated fatty acids promote paclitaxel cytotoxicity via inhibition of the MDR1 gene in the human colon cancer Caco-2 cell line. J. Am. Coll. Nutr. 30, 265–273 (2011)CrossRefPubMed C.Y. Kuan, T.H. Walker, P.G. Luo, C.F. Chen, Long-chain polyunsaturated fatty acids promote paclitaxel cytotoxicity via inhibition of the MDR1 gene in the human colon cancer Caco-2 cell line. J. Am. Coll. Nutr. 30, 265–273 (2011)CrossRefPubMed
14.
go back to reference B. Wen, E. Deutsch, P. Opolon, A. Auperin, V. Frascogna, E. Connault, J. Bourhis, n-3 polyunsaturated fatty acids decrease mucosal/epidermal reactions and enhance antitumour effect of ionising radiation with inhibition of tumour angiogenesis. Br. J. Cancer 89, 1102–1107 (2003)PubMedCentralCrossRefPubMed B. Wen, E. Deutsch, P. Opolon, A. Auperin, V. Frascogna, E. Connault, J. Bourhis, n-3 polyunsaturated fatty acids decrease mucosal/epidermal reactions and enhance antitumour effect of ionising radiation with inhibition of tumour angiogenesis. Br. J. Cancer 89, 1102–1107 (2003)PubMedCentralCrossRefPubMed
15.
go back to reference T. Kato, N. Kolenic, R.S. Pardini, Docosahexaenoic acid (DHA), a primary tumor suppressive omega-3 fatty acid, inhibits growth of colorectal cancer independent of p53 mutational status. Nutr. Cancer 58, 178–187 (2007)CrossRefPubMed T. Kato, N. Kolenic, R.S. Pardini, Docosahexaenoic acid (DHA), a primary tumor suppressive omega-3 fatty acid, inhibits growth of colorectal cancer independent of p53 mutational status. Nutr. Cancer 58, 178–187 (2007)CrossRefPubMed
16.
go back to reference D.P. Rose, J.M. Connolly, C.L. Meschter, Effect of dietary fat on human breast cancer growth and lung metastasis in nude mice. J. Natl. Cancer Inst. 83, 1491–1495 (1991)CrossRefPubMed D.P. Rose, J.M. Connolly, C.L. Meschter, Effect of dietary fat on human breast cancer growth and lung metastasis in nude mice. J. Natl. Cancer Inst. 83, 1491–1495 (1991)CrossRefPubMed
17.
go back to reference T. Kato, R.L. Hancock, H. Mohammadpour, B. McGregor, P. Manalo, S. Khaiboullina, M.R. Hall, L. Pardini, R.S. Pardini, Influence of omega-3 fatty acids on the growth of human colon carcinoma in nude mice. Cancer Lett. 187, 169–177 (2002)CrossRefPubMed T. Kato, R.L. Hancock, H. Mohammadpour, B. McGregor, P. Manalo, S. Khaiboullina, M.R. Hall, L. Pardini, R.S. Pardini, Influence of omega-3 fatty acids on the growth of human colon carcinoma in nude mice. Cancer Lett. 187, 169–177 (2002)CrossRefPubMed
18.
go back to reference U.P. Kelavkar, J. Hutzley, R. Dhir, P. Kim, K.G. Allen, K. McHugh, Prostate tumor growth and recurrence can be modulated by the omega-6:omega-3 ratio in diet: athymic mouse xenograft model simulating radical prostatectomy. Neoplasia 8, 112–124 (2006)PubMedCentralCrossRefPubMed U.P. Kelavkar, J. Hutzley, R. Dhir, P. Kim, K.G. Allen, K. McHugh, Prostate tumor growth and recurrence can be modulated by the omega-6:omega-3 ratio in diet: athymic mouse xenograft model simulating radical prostatectomy. Neoplasia 8, 112–124 (2006)PubMedCentralCrossRefPubMed
19.
go back to reference P. Astorg, Dietary N-6 and N-3 polyunsaturated fatty acids and prostate cancer risk: a review of epidemiological and experimental evidence. Cancer Causes Control 15, 367–386 (2004)CrossRefPubMed P. Astorg, Dietary N-6 and N-3 polyunsaturated fatty acids and prostate cancer risk: a review of epidemiological and experimental evidence. Cancer Causes Control 15, 367–386 (2004)CrossRefPubMed
20.
go back to reference B.A. Narayanan, N.K. Narayanan, B.S. Reddy, Docosahexaenoic acid regulated genes and transcription factors inducing apoptosis in human colon cancer cells. Int. J. Oncol. 119, 1255–1262 (2001) B.A. Narayanan, N.K. Narayanan, B.S. Reddy, Docosahexaenoic acid regulated genes and transcription factors inducing apoptosis in human colon cancer cells. Int. J. Oncol. 119, 1255–1262 (2001)
21.
go back to reference H. Chamras, A. Ardashian, D. Heber, J.A. Glaspy, Fatty acid modulation of MCF-7 human breast cancer cell proliferation, apoptosis and differentiation. J. Nutr. Biochem. 13, 711–716 (2002)CrossRefPubMed H. Chamras, A. Ardashian, D. Heber, J.A. Glaspy, Fatty acid modulation of MCF-7 human breast cancer cell proliferation, apoptosis and differentiation. J. Nutr. Biochem. 13, 711–716 (2002)CrossRefPubMed
22.
go back to reference T. Yamagami, C.D. Porada, R.S. Pardini, E.D. Zanjani, G. Almeida-Porada, Docosahexaenoic acid induces dose dependent cell death in an early undifferentiated subtype of acute myeloid leukemia cell line. Cancer Biol. Ther. 8, 331–337 (2009)PubMedCentralCrossRefPubMed T. Yamagami, C.D. Porada, R.S. Pardini, E.D. Zanjani, G. Almeida-Porada, Docosahexaenoic acid induces dose dependent cell death in an early undifferentiated subtype of acute myeloid leukemia cell line. Cancer Biol. Ther. 8, 331–337 (2009)PubMedCentralCrossRefPubMed
23.
go back to reference P. Gazzaniga, A. Gradilone, A. Petracca, C. Nicolazzo, C. Raimondi, R. Iacovelli, G. Naso, E. Cortesi, Molecular markers in circulating tumour cells from metastatic colorectal cancer patients. J. Cell. Mol. Med. 14, 2073–2077 (2010)PubMedCentralCrossRefPubMed P. Gazzaniga, A. Gradilone, A. Petracca, C. Nicolazzo, C. Raimondi, R. Iacovelli, G. Naso, E. Cortesi, Molecular markers in circulating tumour cells from metastatic colorectal cancer patients. J. Cell. Mol. Med. 14, 2073–2077 (2010)PubMedCentralCrossRefPubMed
24.
go back to reference A.I. Sarela, R.C. Macadam, S.M. Farmery, A.F. Markham, P.J. Guillou, Expression of the antiapoptosis gene, survivin, predicts death from recurrent colorectal carcinoma. Gut 46, 645–650 (2000)PubMedCentralCrossRefPubMed A.I. Sarela, R.C. Macadam, S.M. Farmery, A.F. Markham, P.J. Guillou, Expression of the antiapoptosis gene, survivin, predicts death from recurrent colorectal carcinoma. Gut 46, 645–650 (2000)PubMedCentralCrossRefPubMed
25.
go back to reference P. Habbel, K.H. Weylandt, K. Lichopoj, J. Nowak, M. Purschke, J.D. Wang, C.W. He, D.C. Baumgart, J.X. Kang, Docosahexaenoic acid suppresses arachidonic acid-induced proliferation of LS-174T human colon carcinoma cells. World J. Gastroenterol. 15, 1079–1084 (2009) P. Habbel, K.H. Weylandt, K. Lichopoj, J. Nowak, M. Purschke, J.D. Wang, C.W. He, D.C. Baumgart, J.X. Kang, Docosahexaenoic acid suppresses arachidonic acid-induced proliferation of LS-174T human colon carcinoma cells. World J. Gastroenterol. 15, 1079–1084 (2009)
26.
go back to reference A. Kreso, P.V. Galen, N.M. Pedley, E. Lima-Fernandes, C. Frelin, T. Davis, L. Cao, R. Baiazitov, W. Du, N. Sydorenko, Y.C. Moon, L. Gibson, T. Wang, C. Leung, N.N. Iscove, C.H. Arrowsmith, E. Szentgyorgyi, S. Gallinger, J.E. Dick, C.A. O’Brien, Self-renewal as a therapeutic target in human colorectal cancer. Nat. Med. 20, 29–36 (2013)CrossRefPubMed A. Kreso, P.V. Galen, N.M. Pedley, E. Lima-Fernandes, C. Frelin, T. Davis, L. Cao, R. Baiazitov, W. Du, N. Sydorenko, Y.C. Moon, L. Gibson, T. Wang, C. Leung, N.N. Iscove, C.H. Arrowsmith, E. Szentgyorgyi, S. Gallinger, J.E. Dick, C.A. O’Brien, Self-renewal as a therapeutic target in human colorectal cancer. Nat. Med. 20, 29–36 (2013)CrossRefPubMed
27.
go back to reference S.L. Volchenboum, C. Li, S. Li, E.F. Attiyeh, C.P. Reynolds, J.M. Maris, A.T. Look, R.E. George, Comparison of primary neuroblastoma tumors and derivative early-passage cell lines using genome-wide single nucleotide polymorphism array analysis. Cancer Res. 69, 4143–4149 (2009)PubMedCentralCrossRefPubMed S.L. Volchenboum, C. Li, S. Li, E.F. Attiyeh, C.P. Reynolds, J.M. Maris, A.T. Look, R.E. George, Comparison of primary neuroblastoma tumors and derivative early-passage cell lines using genome-wide single nucleotide polymorphism array analysis. Cancer Res. 69, 4143–4149 (2009)PubMedCentralCrossRefPubMed
28.
go back to reference E.J. Douglas, H. Fiegler, A. Rowan, S. Halford, D.C. Bicknell, W. Bodmer, I.P. Tomlinson, N.P. Carter, Array comparative genomic hybridization analysis of colorectal cancer cell lines and primary carcinomas. Cancer Res. 64, 4817–4825 (2004)CrossRefPubMed E.J. Douglas, H. Fiegler, A. Rowan, S. Halford, D.C. Bicknell, W. Bodmer, I.P. Tomlinson, N.P. Carter, Array comparative genomic hybridization analysis of colorectal cancer cell lines and primary carcinomas. Cancer Res. 64, 4817–4825 (2004)CrossRefPubMed
29.
go back to reference J.K. Willson, G.N. Bittner, T.D. Oberley, L.F. Meisner, J.L. Weese, Cell culture of human colon adenomas and carcinomas. Cancer Res. 47, 2704–2713 (1987)PubMed J.K. Willson, G.N. Bittner, T.D. Oberley, L.F. Meisner, J.L. Weese, Cell culture of human colon adenomas and carcinomas. Cancer Res. 47, 2704–2713 (1987)PubMed
30.
go back to reference T. Yang, S. Fang, H.X. Zhang, L.X. Xu, Z.Q. Zhang, K.T. Yuan, C.L. Xue, H.L. Yu, S. Zhang, Y.F. Li, H.P. Shi, Y. Zhang, N-3 PUFAs have antiproliferative and apoptotic effects on human colorectal cancer stem-like cells in vitro. J. Nutr. Biochem. 24, 744–753 (2013) T. Yang, S. Fang, H.X. Zhang, L.X. Xu, Z.Q. Zhang, K.T. Yuan, C.L. Xue, H.L. Yu, S. Zhang, Y.F. Li, H.P. Shi, Y. Zhang, N-3 PUFAs have antiproliferative and apoptotic effects on human colorectal cancer stem-like cells in vitro. J. Nutr. Biochem. 24, 744–753 (2013)
31.
go back to reference G. Calviello, F. Resci, S. Serini, E. Piccioni, A. Toesca, A. Boninsegna, G. Monego, F.O. Ranelletti, P. Palozza, Docosahexaenoic acid induces proteasome-dependent degradation of beta-catenin, down-regulation of survivin and apoptosis in human colorectal cancer cells not expressing COX-2. Carcinogenesis 28, 1202–1209 (2007)CrossRefPubMed G. Calviello, F. Resci, S. Serini, E. Piccioni, A. Toesca, A. Boninsegna, G. Monego, F.O. Ranelletti, P. Palozza, Docosahexaenoic acid induces proteasome-dependent degradation of beta-catenin, down-regulation of survivin and apoptosis in human colorectal cancer cells not expressing COX-2. Carcinogenesis 28, 1202–1209 (2007)CrossRefPubMed
32.
go back to reference D.C. Altieri, Validating survivin as a cancer therapeutic target. Nat. Rev. Cancer 3, 46–54 (2003)CrossRefPubMed D.C. Altieri, Validating survivin as a cancer therapeutic target. Nat. Rev. Cancer 3, 46–54 (2003)CrossRefPubMed
34.
go back to reference M. Pennati, M. Folini, N. Zaffaroni, Targeting survivin in cancer therapy: fulfilled promises and open questions. Carcinogenesis 28, 1133–1139 (2007)CrossRefPubMed M. Pennati, M. Folini, N. Zaffaroni, Targeting survivin in cancer therapy: fulfilled promises and open questions. Carcinogenesis 28, 1133–1139 (2007)CrossRefPubMed
35.
go back to reference H. Yamamoto, C.Y. Ngan, M. Monden, Cancer cells survive with survivin. Cancer Sci. 99, 1709–1714 (2008)CrossRefPubMed H. Yamamoto, C.Y. Ngan, M. Monden, Cancer cells survive with survivin. Cancer Sci. 99, 1709–1714 (2008)CrossRefPubMed
36.
go back to reference P.J. Morin, A.B. Sparks, V. Korinek, N. Barker, H. Clevers, B. Vogelstein, K.W. Kinzler, Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science 275, 1787–1790 (1997)CrossRefPubMed P.J. Morin, A.B. Sparks, V. Korinek, N. Barker, H. Clevers, B. Vogelstein, K.W. Kinzler, Activation of beta-catenin-Tcf signaling in colon cancer by mutations in beta-catenin or APC. Science 275, 1787–1790 (1997)CrossRefPubMed
37.
go back to reference R. Rosin-Arbesfeld, A. Cliffe, T. Brabletz, M. Bienz, Nuclear export of the APC tumour suppressor controls beta-catenin function in transcription. EMBO J. 22, 1101–1113 (2003)PubMedCentralCrossRefPubMed R. Rosin-Arbesfeld, A. Cliffe, T. Brabletz, M. Bienz, Nuclear export of the APC tumour suppressor controls beta-catenin function in transcription. EMBO J. 22, 1101–1113 (2003)PubMedCentralCrossRefPubMed
38.
go back to reference P.J. Kim, J. Plescia, H. Clevers, E.R. Fearon, D.C. Altieri, Survivin and molecular pathogenesis of colorectal cancer. Lancet 362, 205–209 (2003)CrossRefPubMed P.J. Kim, J. Plescia, H. Clevers, E.R. Fearon, D.C. Altieri, Survivin and molecular pathogenesis of colorectal cancer. Lancet 362, 205–209 (2003)CrossRefPubMed
39.
go back to reference A.B. Di Stefano, F. Iovino, Y. Lombardo, V. Eterno, T. Höger, F. Dieli, G. Stassi, M. Todaro, Survivin is regulated by interleukin-4 in colon cancer stem cells. J. Cell. Physiol. 225, 555–561 (2010)CrossRefPubMed A.B. Di Stefano, F. Iovino, Y. Lombardo, V. Eterno, T. Höger, F. Dieli, G. Stassi, M. Todaro, Survivin is regulated by interleukin-4 in colon cancer stem cells. J. Cell. Physiol. 225, 555–561 (2010)CrossRefPubMed
40.
go back to reference V. Catalano, M. Gaggianesi, V. Spina, F. Iovino, F. Dieli, G. Stassi, M. Todaro, Colorectal cancer stem cells and cell death. Cancers (Basel) 3, 1929–1946 (2011) V. Catalano, M. Gaggianesi, V. Spina, F. Iovino, F. Dieli, G. Stassi, M. Todaro, Colorectal cancer stem cells and cell death. Cancers (Basel) 3, 1929–1946 (2011)
41.
go back to reference L.J. Lin, C.Q. Zheng, Y. Jin, Y. Ma, W.G. Jiang, T. Ma, Expression of survivin protein in human colorectal carcinogenesis. World J. Gastroenterol. 9, 974–977 (2003)PubMedCentralCrossRefPubMed L.J. Lin, C.Q. Zheng, Y. Jin, Y. Ma, W.G. Jiang, T. Ma, Expression of survivin protein in human colorectal carcinogenesis. World J. Gastroenterol. 9, 974–977 (2003)PubMedCentralCrossRefPubMed
Metadata
Title
Treatment of LS174T colorectal cancer stem-like cells with n-3 PUFAs induces growth suppression through inhibition of survivin expression and induction of caspase-3 activation
Publication date
01-02-2016
Published in
Cellular Oncology / Issue 1/2016
Print ISSN: 2211-3428
Electronic ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-015-0254-4

Other articles of this Issue 1/2016

Cellular Oncology 1/2016 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