Skip to main content
Erschienen in: Cellular Oncology 2/2017

03.02.2017 | Original Paper

Histone deacetylase inhibitors VPA and TSA induce apoptosis and autophagy in pancreatic cancer cells

verfasst von: Maria Saveria Gilardini Montani, Marisa Granato, Claudio Santoni, Paola Del Porto, Nicolò Merendino, Gabriella D’Orazi, Alberto Faggioni, Mara Cirone

Erschienen in: Cellular Oncology | Ausgabe 2/2017

Einloggen, um Zugang zu erhalten

Abstract

Purpose

Histone deacetylase inhibitors (HDACi) are anti-neoplastic agents that are known to affect the growth of different cancer types, but their underlying mechanisms are still incompletely understood. Here, we compared the effects of two HDACi, i.e., Trichostatin A (TSA) and Valproic Acid (VPA), on the induction of cell death and autophagy in pancreatic cancer-derived cells that exhibit a high metastatic capacity and carry KRAS/p53 double mutations.

Methods

Cell viability and proliferation tests were carried out using Trypan blue dye exclusion, MTT and BrdU assays. FACS analyses were carried out to assess cell cycle progression, apoptosis, reactive oxygen species (ROS) production and mitochondrial depolarization, while Western blot and immunoprecipitation analyses were employed to detect proteins involved in apoptosis and autophagy.

Results

We found that both VPA and TSA can induce apoptosis in Panc1 and PaCa44 pancreatic cancer-derived cells by triggering mitochondrial membrane depolarization, Cytochrome c release and Caspase 3 activation, although VPA was more effective than TSA, especially in Panc1 cells. As underlying molecular events, we found that ERK1/2 was de-phosphorylated and that the c-Myc and mutant p53 protein levels were reduced after VPA and, to a lesser extent, after TSA treatment. Up-regulation of p21 and Puma was also observed, concomitantly with mutant p53 degradation. In addition, we found that in both cell lines VPA increased the pro-apoptotic Bim level, reduced the anti-apoptotic Mcl-1 level and increased ROS production and autophagy, while TSA was able to induce these effects only in PaCA44 cells.

Conclusions

From our results we conclude that both VPA and TSA can induce pancreatic cancer cell apoptosis and autophagy. VPA appears have a stronger and broader cytotoxic effect than TSA and, thus, may represent a better choice for anti-pancreatic cancer therapy.
Literatur
1.
Zurück zum Zitat R. Siegel, E. Ward, O. Brawley, A. Jemal, Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin 61, 212–236 (2011)CrossRefPubMed R. Siegel, E. Ward, O. Brawley, A. Jemal, Cancer statistics, 2011: the impact of eliminating socioeconomic and racial disparities on premature cancer deaths. CA Cancer J Clin 61, 212–236 (2011)CrossRefPubMed
2.
Zurück zum Zitat M. Giulietti, G. Occhipinti, G. Principato, F. Piva, Weighted gene co-expression network analysis reveals key genes involved in pancreatic ductal adenocarcinoma development. Cell Oncol 39, 379–388 (2016) M. Giulietti, G. Occhipinti, G. Principato, F. Piva, Weighted gene co-expression network analysis reveals key genes involved in pancreatic ductal adenocarcinoma development. Cell Oncol 39, 379–388 (2016)
3.
Zurück zum Zitat X. Shi, S. Liu, J. Kleeff, H. Friess, M.W. Buchler, Acquired resistance of pancreatic cancer cells towards 5-fluorouracil and gemcitabine is associated with altered expression of apoptosis-regulating genes. Oncology 62, 354–362 (2002)CrossRefPubMed X. Shi, S. Liu, J. Kleeff, H. Friess, M.W. Buchler, Acquired resistance of pancreatic cancer cells towards 5-fluorouracil and gemcitabine is associated with altered expression of apoptosis-regulating genes. Oncology 62, 354–362 (2002)CrossRefPubMed
4.
Zurück zum Zitat H.H. Wong, N.R. Lemoine, Pancreatic cancer: molecular pathogenesis and new therapeutic targets. Nat Rev Gastroenterol Hepatol 6, 412–422 (2009)CrossRefPubMedPubMedCentral H.H. Wong, N.R. Lemoine, Pancreatic cancer: molecular pathogenesis and new therapeutic targets. Nat Rev Gastroenterol Hepatol 6, 412–422 (2009)CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat H. Lehrmann, L.L. Pritchard, A. Harel-Bellan, Histone acetyltransferases and deacetylases in the control of cell proliferation and differentiation. Adv Cancer Res 86, 41–65 (2002)CrossRefPubMed H. Lehrmann, L.L. Pritchard, A. Harel-Bellan, Histone acetyltransferases and deacetylases in the control of cell proliferation and differentiation. Adv Cancer Res 86, 41–65 (2002)CrossRefPubMed
6.
Zurück zum Zitat C. Das, T.K. Kundu, Transcriptional regulation by the acetylation of nonhistone proteins in humans -- a new target for therapeutics. IUBMB Life 57, 137–149 (2005)CrossRefPubMed C. Das, T.K. Kundu, Transcriptional regulation by the acetylation of nonhistone proteins in humans -- a new target for therapeutics. IUBMB Life 57, 137–149 (2005)CrossRefPubMed
7.
Zurück zum Zitat S. Spange, T. Wagner, T. Heinzel, O.H. Kramer, Acetylation of non-histone proteins modulates cellular signalling at multiple levels. Int J Biochem Cell Biol 41, 185–198 (2009)CrossRefPubMed S. Spange, T. Wagner, T. Heinzel, O.H. Kramer, Acetylation of non-histone proteins modulates cellular signalling at multiple levels. Int J Biochem Cell Biol 41, 185–198 (2009)CrossRefPubMed
8.
Zurück zum Zitat P.A. Jones, S.B. Baylin, The fundamental role of epigenetic events in cancer. Nat Rev Genet 3, 415–428 (2002)CrossRefPubMed P.A. Jones, S.B. Baylin, The fundamental role of epigenetic events in cancer. Nat Rev Genet 3, 415–428 (2002)CrossRefPubMed
9.
Zurück zum Zitat S. Minucci, P.G. Pelicci, Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer 6, 38–51 (2006)CrossRefPubMed S. Minucci, P.G. Pelicci, Histone deacetylase inhibitors and the promise of epigenetic (and more) treatments for cancer. Nat Rev Cancer 6, 38–51 (2006)CrossRefPubMed
10.
Zurück zum Zitat N. Carey, N.B. La Thangue, Histone deacetylase inhibitors: gathering pace. Curr Opin Pharmacol 6, 369–375 (2006)CrossRefPubMed N. Carey, N.B. La Thangue, Histone deacetylase inhibitors: gathering pace. Curr Opin Pharmacol 6, 369–375 (2006)CrossRefPubMed
12.
Zurück zum Zitat C. Cortes, S.C. Kozma, A. Tauler, S. Ambrosio, MYCN concurrence with SAHA-induced cell death in human neuroblastoma cells. Cell Oncol 38, 341–352 (2015)CrossRef C. Cortes, S.C. Kozma, A. Tauler, S. Ambrosio, MYCN concurrence with SAHA-induced cell death in human neuroblastoma cells. Cell Oncol 38, 341–352 (2015)CrossRef
13.
Zurück zum Zitat B. Jang, J.A. Shin, Y.S. Kim, J.Y. Kim, H.K. Yi, I.S. Park, N.P. Cho, S.D. Cho, Growth-suppressive effect of suberoylanilide hydroxamic acid (SAHA) on human oral cancer cells. Cell Oncol 39, 79–87 (2016)CrossRef B. Jang, J.A. Shin, Y.S. Kim, J.Y. Kim, H.K. Yi, I.S. Park, N.P. Cho, S.D. Cho, Growth-suppressive effect of suberoylanilide hydroxamic acid (SAHA) on human oral cancer cells. Cell Oncol 39, 79–87 (2016)CrossRef
14.
Zurück zum Zitat Z. Chen, Y. Yang, B. Liu, B. Wang, M. Sun, L. Zhang, B. Chen, H. You, M. Zhou, Promotion of metastasis-associated Gene expression in survived PANC-1 cells following trichostatin a treatment. Anti Cancer Agents Med Chem 15, 1317–1325 (2015)CrossRef Z. Chen, Y. Yang, B. Liu, B. Wang, M. Sun, L. Zhang, B. Chen, H. You, M. Zhou, Promotion of metastasis-associated Gene expression in survived PANC-1 cells following trichostatin a treatment. Anti Cancer Agents Med Chem 15, 1317–1325 (2015)CrossRef
15.
Zurück zum Zitat J. Jones, W. Bentas, R.A. Blaheta, J. Makarevic, L. Hudak, S. Wedel, M. Probst, D. Jonas, E. Juengel, Modulation of adhesion and growth of colon and pancreatic cancer cells by the histone deacetylase inhibitor valproic acid. Int J Mol Med 22, 293–299 (2008)PubMed J. Jones, W. Bentas, R.A. Blaheta, J. Makarevic, L. Hudak, S. Wedel, M. Probst, D. Jonas, E. Juengel, Modulation of adhesion and growth of colon and pancreatic cancer cells by the histone deacetylase inhibitor valproic acid. Int J Mol Med 22, 293–299 (2008)PubMed
16.
Zurück zum Zitat S. Schuler, P. Fritsche, S. Diersch, A. Arlt, R.M. Schmid, D. Saur, G. Schneider, HDAC2 attenuates TRAIL-induced apoptosis of pancreatic cancer cells. Mol Cancer 9, 80 (2010)CrossRefPubMedPubMedCentral S. Schuler, P. Fritsche, S. Diersch, A. Arlt, R.M. Schmid, D. Saur, G. Schneider, HDAC2 attenuates TRAIL-induced apoptosis of pancreatic cancer cells. Mol Cancer 9, 80 (2010)CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat J.E. Bolden, M.J. Peart, R.W. Johnstone, Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov 5, 769–784 (2006)CrossRefPubMed J.E. Bolden, M.J. Peart, R.W. Johnstone, Anticancer activities of histone deacetylase inhibitors. Nat Rev Drug Discov 5, 769–784 (2006)CrossRefPubMed
18.
Zurück zum Zitat M. Ouaissi, U. Giger, I. Sielezneff, N. Pirro, B. Sastre, A. Ouaissi, Rationale for possible targeting of histone deacetylase signaling in cancer diseases with a special reference to pancreatic cancer. J Biomed Biotechnol 2011, 315939 (2011)CrossRefPubMed M. Ouaissi, U. Giger, I. Sielezneff, N. Pirro, B. Sastre, A. Ouaissi, Rationale for possible targeting of histone deacetylase signaling in cancer diseases with a special reference to pancreatic cancer. J Biomed Biotechnol 2011, 315939 (2011)CrossRefPubMed
20.
Zurück zum Zitat G. Pistritto, D. Trisciuoglio, C. Ceci, A. Garufi, G. D'Orazi, Apoptosis as anticancer mechanism: function and dysfunction of its modulators and targeted therapeutic strategies. Aging (Albany NY) 8, 603–619 (2016)CrossRef G. Pistritto, D. Trisciuoglio, C. Ceci, A. Garufi, G. D'Orazi, Apoptosis as anticancer mechanism: function and dysfunction of its modulators and targeted therapeutic strategies. Aging (Albany NY) 8, 603–619 (2016)CrossRef
21.
Zurück zum Zitat F. Natoni, L. Diolordi, C. Santoni, M.S. Gilardini Montani, Sodium butyrate sensitises human pancreatic cancer cells to both the intrinsic and the extrinsic apoptotic pathways. Biochim Biophys Acta 1745, 318–329 (2005)CrossRefPubMed F. Natoni, L. Diolordi, C. Santoni, M.S. Gilardini Montani, Sodium butyrate sensitises human pancreatic cancer cells to both the intrinsic and the extrinsic apoptotic pathways. Biochim Biophys Acta 1745, 318–329 (2005)CrossRefPubMed
22.
Zurück zum Zitat O.H. Kramer, P. Zhu, H.P. Ostendorff, M. Golebiewski, J. Tiefenbach, M.A. Peters, B. Brill, B. Groner, I. Bach, T. Heinzel, M. Gottlicher, The histone deacetylase inhibitor valproic acid selectively induces proteasomal degradation of HDAC2. EMBO J 22, 3411–3420 (2003)CrossRefPubMedPubMedCentral O.H. Kramer, P. Zhu, H.P. Ostendorff, M. Golebiewski, J. Tiefenbach, M.A. Peters, B. Brill, B. Groner, I. Bach, T. Heinzel, M. Gottlicher, The histone deacetylase inhibitor valproic acid selectively induces proteasomal degradation of HDAC2. EMBO J 22, 3411–3420 (2003)CrossRefPubMedPubMedCentral
23.
Zurück zum Zitat M.G. Catalano, N. Fortunati, M. Pugliese, L. Costantino, R. Poli, O. Bosco, G. Boccuzzi, Valproic acid induces apoptosis and cell cycle arrest in poorly differentiated thyroid cancer cells. J Clin Endocrinol Metab 90, 1383–1389 (2005)CrossRefPubMed M.G. Catalano, N. Fortunati, M. Pugliese, L. Costantino, R. Poli, O. Bosco, G. Boccuzzi, Valproic acid induces apoptosis and cell cycle arrest in poorly differentiated thyroid cancer cells. J Clin Endocrinol Metab 90, 1383–1389 (2005)CrossRefPubMed
24.
Zurück zum Zitat M. Gottlicher, S. Minucci, P. Zhu, O.H. Kramer, A. Schimpf, S. Giavara, J.P. Sleeman, F. Lo Coco, C. Nervi, P.G. Pelicci, T. Heinzel, Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells. EMBO J 20, 6969–6978 (2001)CrossRefPubMedPubMedCentral M. Gottlicher, S. Minucci, P. Zhu, O.H. Kramer, A. Schimpf, S. Giavara, J.P. Sleeman, F. Lo Coco, C. Nervi, P.G. Pelicci, T. Heinzel, Valproic acid defines a novel class of HDAC inhibitors inducing differentiation of transformed cells. EMBO J 20, 6969–6978 (2001)CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat B.E. Schultz, S. Misialek, J. Wu, J. Tang, M.T. Conn, R. Tahilramani, L. Wong, Kinetics and comparative reactivity of human class I and class IIb histone deacetylases. Biochemistry 43, 11083–11091 (2004)CrossRefPubMed B.E. Schultz, S. Misialek, J. Wu, J. Tang, M.T. Conn, R. Tahilramani, L. Wong, Kinetics and comparative reactivity of human class I and class IIb histone deacetylases. Biochemistry 43, 11083–11091 (2004)CrossRefPubMed
26.
Zurück zum Zitat M.S. Gilardini Montani, A. Prodosmo, V. Stagni, D. Merli, L. Monteonofrio, V. Gatti, M.P. Gentileschi, D. Barila, S. Soddu, ATM-depletion in breast cancer cells confers sensitivity to PARP inhibition. J Exp Clin Cancer Res 32, 95 (2013)CrossRefPubMedPubMedCentral M.S. Gilardini Montani, A. Prodosmo, V. Stagni, D. Merli, L. Monteonofrio, V. Gatti, M.P. Gentileschi, D. Barila, S. Soddu, ATM-depletion in breast cancer cells confers sensitivity to PARP inhibition. J Exp Clin Cancer Res 32, 95 (2013)CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat M. Granato, B. Chiozzi, M.R. Filardi, L.V. Lotti, L. Di Renzo, A. Faggioni, M. Cirone, Tyrosine kinase inhibitor tyrphostin AG490 triggers both apoptosis and autophagy by reducing HSF1 and mcl-1 in PEL cells. Cancer Lett 366, 191–197 (2015)CrossRefPubMed M. Granato, B. Chiozzi, M.R. Filardi, L.V. Lotti, L. Di Renzo, A. Faggioni, M. Cirone, Tyrosine kinase inhibitor tyrphostin AG490 triggers both apoptosis and autophagy by reducing HSF1 and mcl-1 in PEL cells. Cancer Lett 366, 191–197 (2015)CrossRefPubMed
28.
Zurück zum Zitat L.A. Gillies, T. Kuwana, Apoptosis regulation at the mitochondrial outer membrane. J Cell Biochem 115, 632–640 (2014)CrossRefPubMed L.A. Gillies, T. Kuwana, Apoptosis regulation at the mitochondrial outer membrane. J Cell Biochem 115, 632–640 (2014)CrossRefPubMed
29.
Zurück zum Zitat J. Han, L.A. Goldstein, B.R. Gastman, H. Rabinowich, Interrelated roles for mcl-1 and BIM in regulation of TRAIL-mediated mitochondrial apoptosis. J Biol Chem 281, 10153–10163 (2006)CrossRefPubMed J. Han, L.A. Goldstein, B.R. Gastman, H. Rabinowich, Interrelated roles for mcl-1 and BIM in regulation of TRAIL-mediated mitochondrial apoptosis. J Biol Chem 281, 10153–10163 (2006)CrossRefPubMed
30.
Zurück zum Zitat G.J. Griffiths, L. Dubrez, C.P. Morgan, N.A. Jones, J. Whitehouse, B.M. Corfe, C. Dive, J.A. Hickman, Cell damage-induced conformational changes of the pro-apoptotic protein Bak in vivo precede the onset of apoptosis. J Cell Biol 144, 903–914 (1999)CrossRefPubMedPubMedCentral G.J. Griffiths, L. Dubrez, C.P. Morgan, N.A. Jones, J. Whitehouse, B.M. Corfe, C. Dive, J.A. Hickman, Cell damage-induced conformational changes of the pro-apoptotic protein Bak in vivo precede the onset of apoptosis. J Cell Biol 144, 903–914 (1999)CrossRefPubMedPubMedCentral
31.
Zurück zum Zitat L. Lalier, P.F. Cartron, P. Juin, S. Nedelkina, S. Manon, B. Bechinger, F.M. Vallette, Bax activation and mitochondrial insertion during apoptosis. Apoptosis 12, 887–896 (2007)CrossRefPubMed L. Lalier, P.F. Cartron, P. Juin, S. Nedelkina, S. Manon, B. Bechinger, F.M. Vallette, Bax activation and mitochondrial insertion during apoptosis. Apoptosis 12, 887–896 (2007)CrossRefPubMed
32.
Zurück zum Zitat I. Ischenko, O. Petrenko, M.J. Hayman, A MEK/PI3K/HDAC inhibitor combination therapy for KRAS mutant pancreatic cancer cells. Oncotarget 6, 15814–15827 (2015)CrossRefPubMedPubMedCentral I. Ischenko, O. Petrenko, M.J. Hayman, A MEK/PI3K/HDAC inhibitor combination therapy for KRAS mutant pancreatic cancer cells. Oncotarget 6, 15814–15827 (2015)CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat R. Sears, G. Leone, J. DeGregori, J.R. Nevins, Ras enhances Myc protein stability. Mol Cell 3, 169–179 (1999)CrossRefPubMed R. Sears, G. Leone, J. DeGregori, J.R. Nevins, Ras enhances Myc protein stability. Mol Cell 3, 169–179 (1999)CrossRefPubMed
34.
Zurück zum Zitat E. Hessmann, G. Schneider, V. Ellenrieder, J.T. Siveke, MYC in pancreatic cancer: novel mechanistic insights and their translation into therapeutic strategies. Oncogene 35, 1609–1618 (2016)CrossRefPubMed E. Hessmann, G. Schneider, V. Ellenrieder, J.T. Siveke, MYC in pancreatic cancer: novel mechanistic insights and their translation into therapeutic strategies. Oncogene 35, 1609–1618 (2016)CrossRefPubMed
35.
Zurück zum Zitat N. Angela, V. Carafa, M. Conte, F.P. Tambaro, C. Abbondanza, J.H. Martens, M. Nees, R. Benedetti, I. Pallavicini, S. Minucci, G. Garcia-Manero, F. Iovino, G. Lania, C. Ingenito, V. Belsito Petrizzi, H.G. Stunnenberg and L. Altucci, c-Myc modulation & acetylation is a key HDAC inhibitor target in cancer. Clin Cancer Res (2016) N. Angela, V. Carafa, M. Conte, F.P. Tambaro, C. Abbondanza, J.H. Martens, M. Nees, R. Benedetti, I. Pallavicini, S. Minucci, G. Garcia-Manero, F. Iovino, G. Lania, C. Ingenito, V. Belsito Petrizzi, H.G. Stunnenberg and L. Altucci, c-Myc modulation & acetylation is a key HDAC inhibitor target in cancer. Clin Cancer Res (2016)
36.
Zurück zum Zitat P.S. Moore, B. Sipos, S. Orlandini, C. Sorio, F.X. Real, N.R. Lemoine, T. Gress, C. Bassi, G. Kloppel, H. Kalthoff, H. Ungefroren, M. Lohr, A. Scarpa, Genetic profile of 22 pancreatic carcinoma cell lines. Analysis of K-ras, p53, p16 and DPC4/Smad4. Virchows Arch 439, 798–802 (2001)CrossRefPubMed P.S. Moore, B. Sipos, S. Orlandini, C. Sorio, F.X. Real, N.R. Lemoine, T. Gress, C. Bassi, G. Kloppel, H. Kalthoff, H. Ungefroren, M. Lohr, A. Scarpa, Genetic profile of 22 pancreatic carcinoma cell lines. Analysis of K-ras, p53, p16 and DPC4/Smad4. Virchows Arch 439, 798–802 (2001)CrossRefPubMed
37.
Zurück zum Zitat R. Brosh, V. Rotter, When mutants gain new powers: news from the mutant p53 field. Nat Rev Cancer 9, 701–713 (2009)PubMed R. Brosh, V. Rotter, When mutants gain new powers: news from the mutant p53 field. Nat Rev Cancer 9, 701–713 (2009)PubMed
38.
39.
Zurück zum Zitat C.J. Brown, C.F. Cheok, C.S. Verma, D.P. Lane, Reactivation of p53: from peptides to small molecules. Trends Pharmacol Sci 32, 53–62 (2011)CrossRefPubMed C.J. Brown, C.F. Cheok, C.S. Verma, D.P. Lane, Reactivation of p53: from peptides to small molecules. Trends Pharmacol Sci 32, 53–62 (2011)CrossRefPubMed
40.
Zurück zum Zitat C. Fiorini, M. Cordani, C. Padroni, G. Blandino, S. Di Agostino, M. Donadelli, Mutant p53 stimulates chemoresistance of pancreatic adenocarcinoma cells to gemcitabine. Biochim Biophys Acta 1853, 89–100 (2015)CrossRefPubMed C. Fiorini, M. Cordani, C. Padroni, G. Blandino, S. Di Agostino, M. Donadelli, Mutant p53 stimulates chemoresistance of pancreatic adenocarcinoma cells to gemcitabine. Biochim Biophys Acta 1853, 89–100 (2015)CrossRefPubMed
41.
Zurück zum Zitat C.M. Aliouat-Denis, N. Dendouga, I. Van den Wyngaert, H. Goehlmann, U. Steller, I. van de Weyer, N. Van Slycken, L. Andries, S. Kass, W. Luyten, M. Janicot, J.E. Vialard, p53-independent regulation of p21Waf1/Cip1 expression and senescence by Chk2. Mol Cancer Res 3, 627–634 (2005)CrossRefPubMed C.M. Aliouat-Denis, N. Dendouga, I. Van den Wyngaert, H. Goehlmann, U. Steller, I. van de Weyer, N. Van Slycken, L. Andries, S. Kass, W. Luyten, M. Janicot, J.E. Vialard, p53-independent regulation of p21Waf1/Cip1 expression and senescence by Chk2. Mol Cancer Res 3, 627–634 (2005)CrossRefPubMed
42.
Zurück zum Zitat J.R. Jeffers, E. Parganas, Y. Lee, C. Yang, J. Wang, J. Brennan, K.H. MacLean, J. Han, T. Chittenden, J.N. Ihle, P.J. McKinnon, J.L. Cleveland, G.P. Zambetti, Puma is an essential mediator of p53-dependent and -independent apoptotic pathways. Cancer Cell 4, 321–328 (2003)CrossRefPubMed J.R. Jeffers, E. Parganas, Y. Lee, C. Yang, J. Wang, J. Brennan, K.H. MacLean, J. Han, T. Chittenden, J.N. Ihle, P.J. McKinnon, J.L. Cleveland, G.P. Zambetti, Puma is an essential mediator of p53-dependent and -independent apoptotic pathways. Cancer Cell 4, 321–328 (2003)CrossRefPubMed
43.
Zurück zum Zitat R. Scherz-Shouval, Z. Elazar, ROS, mitochondria and the regulation of autophagy. Trends Cell Biol 17, 422–427 (2007)CrossRefPubMed R. Scherz-Shouval, Z. Elazar, ROS, mitochondria and the regulation of autophagy. Trends Cell Biol 17, 422–427 (2007)CrossRefPubMed
44.
Zurück zum Zitat J.S. Ungerstedt, Y. Sowa, W.S. Xu, Y. Shao, M. Dokmanovic, G. Perez, L. Ngo, A. Holmgren, X. Jiang, P.A. Marks, Role of thioredoxin in the response of normal and transformed cells to histone deacetylase inhibitors. Proc Natl Acad Sci U S A 102, 673–678 (2005)CrossRefPubMedPubMedCentral J.S. Ungerstedt, Y. Sowa, W.S. Xu, Y. Shao, M. Dokmanovic, G. Perez, L. Ngo, A. Holmgren, X. Jiang, P.A. Marks, Role of thioredoxin in the response of normal and transformed cells to histone deacetylase inhibitors. Proc Natl Acad Sci U S A 102, 673–678 (2005)CrossRefPubMedPubMedCentral
45.
Zurück zum Zitat C. Fiorini, M. Cordani, G. Gotte, D. Picone, M. Donadelli, Onconase induces autophagy sensitizing pancreatic cancer cells to gemcitabine and activates Akt/mTOR pathway in a ROS-dependent manner. Biochim Biophys Acta 1853, 549–560 (2015)CrossRefPubMed C. Fiorini, M. Cordani, G. Gotte, D. Picone, M. Donadelli, Onconase induces autophagy sensitizing pancreatic cancer cells to gemcitabine and activates Akt/mTOR pathway in a ROS-dependent manner. Biochim Biophys Acta 1853, 549–560 (2015)CrossRefPubMed
46.
Zurück zum Zitat R. Scherz-Shouval, Z. Elazar, Regulation of autophagy by ROS: physiology and pathology. Trends Biochem Sci 36, 30–38 (2011)CrossRefPubMed R. Scherz-Shouval, Z. Elazar, Regulation of autophagy by ROS: physiology and pathology. Trends Biochem Sci 36, 30–38 (2011)CrossRefPubMed
47.
Zurück zum Zitat D.J. Klionsky, K. Abdelmohsen, A. Abe, et al., Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy 12, 1–222 (2016) D.J. Klionsky, K. Abdelmohsen, A. Abe, et al., Guidelines for the use and interpretation of assays for monitoring autophagy (3rd edition). Autophagy 12, 1–222 (2016)
49.
Zurück zum Zitat M. Cirone, L. Di Renzo, L.V. Lotti, V. Conte, P. Trivedi, R. Santarelli, R. Gonnella, L. Frati, A. Faggioni, Primary effusion lymphoma cell death induced by bortezomib and AG 490 activates dendritic cells through CD91. PLoS One 7, e31732 (2012)CrossRefPubMedPubMedCentral M. Cirone, L. Di Renzo, L.V. Lotti, V. Conte, P. Trivedi, R. Santarelli, R. Gonnella, L. Frati, A. Faggioni, Primary effusion lymphoma cell death induced by bortezomib and AG 490 activates dendritic cells through CD91. PLoS One 7, e31732 (2012)CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat D. Cecconi, M. Donadelli, A. Scarpa, A. Milli, M. Palmieri, M. Hamdan, L.B. Areces, J. Rappsilber, P.G. Righetti, Proteomic analysis of pancreatic ductal carcinoma cells after combined treatment with gemcitabine and trichostatin a. J Proteome Res 4, 1909–1916 (2005)CrossRefPubMed D. Cecconi, M. Donadelli, A. Scarpa, A. Milli, M. Palmieri, M. Hamdan, L.B. Areces, J. Rappsilber, P.G. Righetti, Proteomic analysis of pancreatic ductal carcinoma cells after combined treatment with gemcitabine and trichostatin a. J Proteome Res 4, 1909–1916 (2005)CrossRefPubMed
51.
Zurück zum Zitat A.R. Goloudina, O.N. Demidov, C. Garrido, Inhibition of HSP70: a challenging anti-cancer strategy. Cancer Lett 325, 117–124 (2012)CrossRefPubMed A.R. Goloudina, O.N. Demidov, C. Garrido, Inhibition of HSP70: a challenging anti-cancer strategy. Cancer Lett 325, 117–124 (2012)CrossRefPubMed
52.
Zurück zum Zitat M. Granato, V. Lacconi, M. Peddis, L.V. Lotti, L. Di Renzo, R. Gonnella, R. Santarelli, P. Trivedi, L. Frati, G. D'Orazi, A. Faggioni, M. Cirone, HSP70 inhibition by 2-phenylethynesulfonamide induces lysosomal cathepsin D release and immunogenic cell death in primary effusion lymphoma. Cell Death Dis 4, e730 (2013)CrossRefPubMedPubMedCentral M. Granato, V. Lacconi, M. Peddis, L.V. Lotti, L. Di Renzo, R. Gonnella, R. Santarelli, P. Trivedi, L. Frati, G. D'Orazi, A. Faggioni, M. Cirone, HSP70 inhibition by 2-phenylethynesulfonamide induces lysosomal cathepsin D release and immunogenic cell death in primary effusion lymphoma. Cell Death Dis 4, e730 (2013)CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat K.J. Falkenberg, R.W. Johnstone, Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders. Nat Rev Drug Discov 13, 673–691 (2014)CrossRefPubMed K.J. Falkenberg, R.W. Johnstone, Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders. Nat Rev Drug Discov 13, 673–691 (2014)CrossRefPubMed
55.
Zurück zum Zitat J. Roche, P. Bertrand, Inside HDACs with more selective HDAC inhibitors. Eur J Med Chem 121, 451–483 (2016)CrossRefPubMed J. Roche, P. Bertrand, Inside HDACs with more selective HDAC inhibitors. Eur J Med Chem 121, 451–483 (2016)CrossRefPubMed
58.
Zurück zum Zitat L. Weisz, M. Oren, V. Rotter, Transcription regulation by mutant p53. Oncogene 26, 2202–2211 (2007)CrossRefPubMed L. Weisz, M. Oren, V. Rotter, Transcription regulation by mutant p53. Oncogene 26, 2202–2211 (2007)CrossRefPubMed
59.
Zurück zum Zitat G. Bougeard, R. Sesboue, S. Baert-Desurmont, S. Vasseur, C. Martin, J. Tinat, L. Brugieres, A. Chompret, B.B. de Paillerets, D. Stoppa-Lyonnet, C. Bonaiti-Pellie, T. Frebourg, L.F.S.w.g. French, Molecular basis of the Li-Fraumeni syndrome: an update from the French LFS families. J Med Genet 45, 535–538 (2008) G. Bougeard, R. Sesboue, S. Baert-Desurmont, S. Vasseur, C. Martin, J. Tinat, L. Brugieres, A. Chompret, B.B. de Paillerets, D. Stoppa-Lyonnet, C. Bonaiti-Pellie, T. Frebourg, L.F.S.w.g. French, Molecular basis of the Li-Fraumeni syndrome: an update from the French LFS families. J Med Genet 45, 535–538 (2008)
60.
Zurück zum Zitat Y. Zerdoumi, J. Aury-Landas, C. Bonaiti-Pellie, C. Derambure, R. Sesboue, M. Renaux-Petel, T. Frebourg, G. Bougeard, J.M. Flaman, Drastic effect of germline TP53 missense mutations in Li-Fraumeni patients. Hum Mutat 34, 453–461 (2013)CrossRefPubMed Y. Zerdoumi, J. Aury-Landas, C. Bonaiti-Pellie, C. Derambure, R. Sesboue, M. Renaux-Petel, T. Frebourg, G. Bougeard, J.M. Flaman, Drastic effect of germline TP53 missense mutations in Li-Fraumeni patients. Hum Mutat 34, 453–461 (2013)CrossRefPubMed
61.
Zurück zum Zitat A. Garufi, D. Pucci, V. D'Orazi, M. Cirone, G. Bossi, M.L. Avantaggiati, G. D'Orazi, Degradation of mutant p53H175 protein by Zn(II) through autophagy. Cell Death Dis 5, e1271 (2014)CrossRefPubMedPubMedCentral A. Garufi, D. Pucci, V. D'Orazi, M. Cirone, G. Bossi, M.L. Avantaggiati, G. D'Orazi, Degradation of mutant p53H175 protein by Zn(II) through autophagy. Cell Death Dis 5, e1271 (2014)CrossRefPubMedPubMedCentral
62.
Zurück zum Zitat A. Garufi, V. D'Orazi, A. Crispini, G. D'Orazi, Zn(II)-curc targets p53 in thyroid cancer cells. Int J Oncol 47, 1241–1248 (2015)PubMedPubMedCentral A. Garufi, V. D'Orazi, A. Crispini, G. D'Orazi, Zn(II)-curc targets p53 in thyroid cancer cells. Int J Oncol 47, 1241–1248 (2015)PubMedPubMedCentral
63.
Zurück zum Zitat A. Willis, E.J. Jung, T. Wakefield, X. Chen, Mutant p53 exerts a dominant negative effect by preventing wild-type p53 from binding to the promoter of its target genes. Oncogene 23, 2330–2338 (2004)CrossRefPubMed A. Willis, E.J. Jung, T. Wakefield, X. Chen, Mutant p53 exerts a dominant negative effect by preventing wild-type p53 from binding to the promoter of its target genes. Oncogene 23, 2330–2338 (2004)CrossRefPubMed
64.
Zurück zum Zitat G. Filomeni, E. Desideri, S. Cardaci, G. Rotilio, M.R. Ciriolo, Under the ROS...thiol network is the principal suspect for autophagy commitment. Autophagy 6, 999–1005 (2010)CrossRefPubMed G. Filomeni, E. Desideri, S. Cardaci, G. Rotilio, M.R. Ciriolo, Under the ROS...thiol network is the principal suspect for autophagy commitment. Autophagy 6, 999–1005 (2010)CrossRefPubMed
65.
Zurück zum Zitat S. Fulda, D. Kogel, Cell death by autophagy: emerging molecular mechanisms and implications for cancer therapy. Oncogene 34, 5105–5113 (2015)CrossRefPubMed S. Fulda, D. Kogel, Cell death by autophagy: emerging molecular mechanisms and implications for cancer therapy. Oncogene 34, 5105–5113 (2015)CrossRefPubMed
66.
Zurück zum Zitat G. He, G. Chen, W. Chen, W. Zhang, J. Cao, Q. Ye, Lack of association of XRCC1 rs1799782 genetic polymorphism with risk of pancreatic cancer: a meta-analysis. Tumour Biol 35, 4545–4550 (2014)CrossRefPubMed G. He, G. Chen, W. Chen, W. Zhang, J. Cao, Q. Ye, Lack of association of XRCC1 rs1799782 genetic polymorphism with risk of pancreatic cancer: a meta-analysis. Tumour Biol 35, 4545–4550 (2014)CrossRefPubMed
68.
Zurück zum Zitat N. Gammoh, D. Lam, C. Puente, I. Ganley, P.A. Marks, X. Jiang, Role of autophagy in histone deacetylase inhibitor-induced apoptotic and nonapoptotic cell death. Proc Natl Acad Sci U S A 109, 6561–6565 (2012)CrossRefPubMedPubMedCentral N. Gammoh, D. Lam, C. Puente, I. Ganley, P.A. Marks, X. Jiang, Role of autophagy in histone deacetylase inhibitor-induced apoptotic and nonapoptotic cell death. Proc Natl Acad Sci U S A 109, 6561–6565 (2012)CrossRefPubMedPubMedCentral
69.
Zurück zum Zitat J. Zhang, S. Ng, J. Wang, J. Zhou, S.H. Tan, N. Yang, Q. Lin, D. Xia, H.M. Shen, Histone deacetylase inhibitors induce autophagy through FOXO1-dependent pathways. Autophagy 11, 629–642 (2015)CrossRefPubMedPubMedCentral J. Zhang, S. Ng, J. Wang, J. Zhou, S.H. Tan, N. Yang, Q. Lin, D. Xia, H.M. Shen, Histone deacetylase inhibitors induce autophagy through FOXO1-dependent pathways. Autophagy 11, 629–642 (2015)CrossRefPubMedPubMedCentral
70.
Zurück zum Zitat G.M. Matthews, A. Newbold, R.W. Johnstone, Intrinsic and extrinsic apoptotic pathway signaling as determinants of histone deacetylase inhibitor antitumor activity. Adv Cancer Res 116, 165–197 (2012)CrossRefPubMed G.M. Matthews, A. Newbold, R.W. Johnstone, Intrinsic and extrinsic apoptotic pathway signaling as determinants of histone deacetylase inhibitor antitumor activity. Adv Cancer Res 116, 165–197 (2012)CrossRefPubMed
71.
Zurück zum Zitat S. Mukhopadhyay, P.K. Panda, N. Sinha, D.N. Das, S.K. Bhutia, Autophagy and apoptosis: where do they meet? Apoptosis 19, 555–566 (2014)CrossRefPubMed S. Mukhopadhyay, P.K. Panda, N. Sinha, D.N. Das, S.K. Bhutia, Autophagy and apoptosis: where do they meet? Apoptosis 19, 555–566 (2014)CrossRefPubMed
72.
Zurück zum Zitat R. Santarelli, R. Gonnella, G. Di Giovenale, L. Cuomo, A. Capobianchi, M. Granato, G. Gentile, A. Faggioni, M. Cirone, STAT3 activation by KSHV correlates with IL-10, IL-6 and IL-23 release and an autophagic block in dendritic cells. Sci Rep 4, 4241 (2014)CrossRefPubMedPubMedCentral R. Santarelli, R. Gonnella, G. Di Giovenale, L. Cuomo, A. Capobianchi, M. Granato, G. Gentile, A. Faggioni, M. Cirone, STAT3 activation by KSHV correlates with IL-10, IL-6 and IL-23 release and an autophagic block in dendritic cells. Sci Rep 4, 4241 (2014)CrossRefPubMedPubMedCentral
73.
Zurück zum Zitat M. Granato, R. Santarelli, L.V. Lotti, L. Di Renzo, R. Gonnella, A. Garufi, P. Trivedi, L. Frati, G. D'Orazi, A. Faggioni, M. Cirone, JNK and macroautophagy activation by bortezomib has a pro-survival effect in primary effusion lymphoma cells. PLoS One 8, e75965 (2013)CrossRefPubMedPubMedCentral M. Granato, R. Santarelli, L.V. Lotti, L. Di Renzo, R. Gonnella, A. Garufi, P. Trivedi, L. Frati, G. D'Orazi, A. Faggioni, M. Cirone, JNK and macroautophagy activation by bortezomib has a pro-survival effect in primary effusion lymphoma cells. PLoS One 8, e75965 (2013)CrossRefPubMedPubMedCentral
74.
Zurück zum Zitat M. Granato, C. Rizzello, M.A. Romeo, S. Yadav, R. Santarelli, G. D'Orazi, A. Faggioni, M. Cirone, Concomitant reduction of c-Myc expression and PI3K/AKT/mTOR signaling by quercetin induces a strong cytotoxic effect against Burkitt's lymphoma. Int J Biochem Cell Biol 79, 393–400 (2016)CrossRefPubMed M. Granato, C. Rizzello, M.A. Romeo, S. Yadav, R. Santarelli, G. D'Orazi, A. Faggioni, M. Cirone, Concomitant reduction of c-Myc expression and PI3K/AKT/mTOR signaling by quercetin induces a strong cytotoxic effect against Burkitt's lymphoma. Int J Biochem Cell Biol 79, 393–400 (2016)CrossRefPubMed
75.
Zurück zum Zitat A. Garufi, G. Pistritto, M. Cirone, G. D'Orazi, Reactivation of mutant p53 by capsaicin, the major constituent of peppers. J Exp Clin Cancer Res 35, 136 (2016)CrossRefPubMedPubMedCentral A. Garufi, G. Pistritto, M. Cirone, G. D'Orazi, Reactivation of mutant p53 by capsaicin, the major constituent of peppers. J Exp Clin Cancer Res 35, 136 (2016)CrossRefPubMedPubMedCentral
76.
Zurück zum Zitat S.T. Nawrocki, J.S. Carew, M.S. Pino, R.A. Highshaw, R.H. Andtbacka, K. Dunner Jr., A. Pal, W.G. Bornmann, P.J. Chiao, P. Huang, H. Xiong, J.L. Abbruzzese, D.J. McConkey, Aggresome disruption: a novel strategy to enhance bortezomib-induced apoptosis in pancreatic cancer cells. Cancer Res 66, 3773–3781 (2006) S.T. Nawrocki, J.S. Carew, M.S. Pino, R.A. Highshaw, R.H. Andtbacka, K. Dunner Jr., A. Pal, W.G. Bornmann, P.J. Chiao, P. Huang, H. Xiong, J.L. Abbruzzese, D.J. McConkey, Aggresome disruption: a novel strategy to enhance bortezomib-induced apoptosis in pancreatic cancer cells. Cancer Res 66, 3773–3781 (2006)
Metadaten
Titel
Histone deacetylase inhibitors VPA and TSA induce apoptosis and autophagy in pancreatic cancer cells
verfasst von
Maria Saveria Gilardini Montani
Marisa Granato
Claudio Santoni
Paola Del Porto
Nicolò Merendino
Gabriella D’Orazi
Alberto Faggioni
Mara Cirone
Publikationsdatum
03.02.2017
Verlag
Springer Netherlands
Erschienen in
Cellular Oncology / Ausgabe 2/2017
Print ISSN: 2211-3428
Elektronische ISSN: 2211-3436
DOI
https://doi.org/10.1007/s13402-017-0314-z

Weitere Artikel der Ausgabe 2/2017

Cellular Oncology 2/2017 Zur Ausgabe

Neu im Fachgebiet Pathologie