Cellular mechanisms triggered by the cotreatment of resveratrol and doxorubicin in breast cancer: A translational in vitro–In silico model

Doxorubicin (Doxo) is the most effective chemotherapeutic agent for the treatment of breast cancer. However, resistance to Doxo is common. Adjuvant compounds capable of modulating mechanisms involved in Doxo resistance may potentiate the effectiveness of the drug. Resveratrol (Rsv) has been tested a...

Full description

Autores:
Vargas, José Eduardo
Puga, Renato
Lenz, Guido
Trindade, Cristiano
Filippi-Chiela, Eduardo
Tipo de recurso:
Fecha de publicación:
2020
Institución:
Universidad Simón Bolívar
Repositorio:
Repositorio Digital USB
Idioma:
eng
OAI Identifier:
oai:bonga.unisimon.edu.co:20.500.12442/6761
Acceso en línea:
https://hdl.handle.net/20.500.12442/6761
https://doi.org/10.1155/2020/5432651
https://www.hindawi.com/journals/omcl/2020/5432651/
Palabra clave:
Rights
openAccess
License
Attribution-NonCommercial-NoDerivatives 4.0 Internacional
id USIMONBOL2_54844e2ff501caef2970e576fe46d1c6
oai_identifier_str oai:bonga.unisimon.edu.co:20.500.12442/6761
network_acronym_str USIMONBOL2
network_name_str Repositorio Digital USB
repository_id_str
dc.title.eng.fl_str_mv Cellular mechanisms triggered by the cotreatment of resveratrol and doxorubicin in breast cancer: A translational in vitro–In silico model
title Cellular mechanisms triggered by the cotreatment of resveratrol and doxorubicin in breast cancer: A translational in vitro–In silico model
spellingShingle Cellular mechanisms triggered by the cotreatment of resveratrol and doxorubicin in breast cancer: A translational in vitro–In silico model
title_short Cellular mechanisms triggered by the cotreatment of resveratrol and doxorubicin in breast cancer: A translational in vitro–In silico model
title_full Cellular mechanisms triggered by the cotreatment of resveratrol and doxorubicin in breast cancer: A translational in vitro–In silico model
title_fullStr Cellular mechanisms triggered by the cotreatment of resveratrol and doxorubicin in breast cancer: A translational in vitro–In silico model
title_full_unstemmed Cellular mechanisms triggered by the cotreatment of resveratrol and doxorubicin in breast cancer: A translational in vitro–In silico model
title_sort Cellular mechanisms triggered by the cotreatment of resveratrol and doxorubicin in breast cancer: A translational in vitro–In silico model
dc.creator.fl_str_mv Vargas, José Eduardo
Puga, Renato
Lenz, Guido
Trindade, Cristiano
Filippi-Chiela, Eduardo
dc.contributor.author.none.fl_str_mv Vargas, José Eduardo
Puga, Renato
Lenz, Guido
Trindade, Cristiano
Filippi-Chiela, Eduardo
description Doxorubicin (Doxo) is the most effective chemotherapeutic agent for the treatment of breast cancer. However, resistance to Doxo is common. Adjuvant compounds capable of modulating mechanisms involved in Doxo resistance may potentiate the effectiveness of the drug. Resveratrol (Rsv) has been tested as an adjuvant in mammary malignancies. However, the cellular and molecular mechanisms underlying the effects of cotreatment with Doxo and Rsv in breast cancer are poorly understood. Here, we combined in vitro and in silico analysis to characterize these mechanisms. In vitro, we employed a clinically relevant experimental design consisting of acute (24 h) treatment followed by 15 days of analysis. Acute Rsv potentiated the long-lasting effect of Doxo through the induction of apoptosis and senescence. Cells that survived to the cotreatment triggered high levels of autophagy. Autophagy inhibition during its peak of activation but not concomitant with Doxo+Rsv increased the long-term toxicity of the cotreatment. To uncover key proteins potentially associated with in vitro effects, an in silico multistep strategy was implemented. Chemical-protein networks were predicted based on constitutive gene expression of MCF7 cells and interatomic data from breast cancer. Topological analysis, KM survival analysis, and a quantitative model based on the connectivity between apoptosis, senescence, and autophagy were performed. We found seven putative genes predicted to be modulated by Rsv in the context of Doxo treatment: CCND1, CDH1, ESR1, HSP90AA1, MAPK3, PTPN11, and RPS6KB1. Six out of these seven genes have been experimentally proven to be modulated by Rsv in cancer cells, with 4 of the 6 genes in MCF7 cells. In conclusion, acute Rsv potentiated the long-term toxicity of Doxo in breast cancer potentially through the modulation of genes and mechanisms involved in Doxo resistance. Rational autophagy inhibition potentiated the effects of Rsv+Doxo, a strategy that should be further tested in animal models.
publishDate 2020
dc.date.accessioned.none.fl_str_mv 2020-11-05T17:55:44Z
dc.date.available.none.fl_str_mv 2020-11-05T17:55:44Z
dc.date.issued.none.fl_str_mv 2020
dc.type.coarversion.fl_str_mv http://purl.org/coar/version/c_970fb48d4fbd8a85
dc.type.coar.fl_str_mv http://purl.org/coar/resource_type/c_2df8fbb1
dc.type.driver.eng.fl_str_mv info:eu-repo/semantics/article
dc.type.spa.eng.fl_str_mv Artículo científico
dc.identifier.issn.none.fl_str_mv 19420994
dc.identifier.uri.none.fl_str_mv https://hdl.handle.net/20.500.12442/6761
dc.identifier.doi.none.fl_str_mv https://doi.org/10.1155/2020/5432651
dc.identifier.url.none.fl_str_mv https://www.hindawi.com/journals/omcl/2020/5432651/
identifier_str_mv 19420994
url https://hdl.handle.net/20.500.12442/6761
https://doi.org/10.1155/2020/5432651
https://www.hindawi.com/journals/omcl/2020/5432651/
dc.language.iso.eng.fl_str_mv eng
language eng
dc.rights.*.fl_str_mv Attribution-NonCommercial-NoDerivatives 4.0 Internacional
dc.rights.coar.fl_str_mv http://purl.org/coar/access_right/c_abf2
dc.rights.uri.*.fl_str_mv http://creativecommons.org/licenses/by-nc-nd/4.0/
dc.rights.accessrights.spa.fl_str_mv info:eu-repo/semantics/openAccess
rights_invalid_str_mv Attribution-NonCommercial-NoDerivatives 4.0 Internacional
http://creativecommons.org/licenses/by-nc-nd/4.0/
http://purl.org/coar/access_right/c_abf2
eu_rights_str_mv openAccess
dc.format.mimetype.eng.fl_str_mv pdf
dc.publisher.eng.fl_str_mv Hindawi
dc.source.eng.fl_str_mv Oxidative Medicine and Cellular Longevity
institution Universidad Simón Bolívar
bitstream.url.fl_str_mv https://bonga.unisimon.edu.co/bitstreams/4874ba94-23b7-448f-af72-7fe0ef622a92/download
https://bonga.unisimon.edu.co/bitstreams/1a40dd1f-5e6a-4bf4-8b9e-cb0ed07560e1/download
https://bonga.unisimon.edu.co/bitstreams/d91817e0-9269-400c-89eb-c54179d5772f/download
https://bonga.unisimon.edu.co/bitstreams/79f517a4-c246-4fa6-90b6-f0c6181f58f0/download
https://bonga.unisimon.edu.co/bitstreams/e0b98c75-b522-44cc-b8c7-59b12cb18ffe/download
https://bonga.unisimon.edu.co/bitstreams/f6c75568-38ce-4931-9274-be7622d2d0bd/download
https://bonga.unisimon.edu.co/bitstreams/4d420d49-8adf-47ed-9824-4154346fce60/download
bitstream.checksum.fl_str_mv 5b445794b89cef77bcc38d8ea9874957
733bec43a0bf5ade4d97db708e29b185
4460e5956bc1d1639be9ae6146a50347
f6739d98c69ed53d48ad946faafd8b9a
0e8ba9b88304756eff623bd98cae8c06
46dd0a23a51209cebd423ee444cac591
a6a8ad12522728a363ac732bbbd0265f
bitstream.checksumAlgorithm.fl_str_mv MD5
MD5
MD5
MD5
MD5
MD5
MD5
repository.name.fl_str_mv Repositorio Digital Universidad Simón Bolívar
repository.mail.fl_str_mv repositorio.digital@unisimon.edu.co
_version_ 1812100486100156416
spelling Vargas, José Eduardo416f0def-6df6-4f2d-ad69-3c969af5664dPuga, Renato27951978-47c5-484c-a747-a0d418c2eb19Lenz, Guido46ea5e9c-875b-4eac-ba9f-1df73d830e75Trindade, Cristiano30b50065-9290-4b8a-837b-01634d3219d7Filippi-Chiela, Eduardo869c9e6c-3e06-445d-8891-f7ad72cafff12020-11-05T17:55:44Z2020-11-05T17:55:44Z202019420994https://hdl.handle.net/20.500.12442/6761https://doi.org/10.1155/2020/5432651https://www.hindawi.com/journals/omcl/2020/5432651/Doxorubicin (Doxo) is the most effective chemotherapeutic agent for the treatment of breast cancer. However, resistance to Doxo is common. Adjuvant compounds capable of modulating mechanisms involved in Doxo resistance may potentiate the effectiveness of the drug. Resveratrol (Rsv) has been tested as an adjuvant in mammary malignancies. However, the cellular and molecular mechanisms underlying the effects of cotreatment with Doxo and Rsv in breast cancer are poorly understood. Here, we combined in vitro and in silico analysis to characterize these mechanisms. In vitro, we employed a clinically relevant experimental design consisting of acute (24 h) treatment followed by 15 days of analysis. Acute Rsv potentiated the long-lasting effect of Doxo through the induction of apoptosis and senescence. Cells that survived to the cotreatment triggered high levels of autophagy. Autophagy inhibition during its peak of activation but not concomitant with Doxo+Rsv increased the long-term toxicity of the cotreatment. To uncover key proteins potentially associated with in vitro effects, an in silico multistep strategy was implemented. Chemical-protein networks were predicted based on constitutive gene expression of MCF7 cells and interatomic data from breast cancer. Topological analysis, KM survival analysis, and a quantitative model based on the connectivity between apoptosis, senescence, and autophagy were performed. We found seven putative genes predicted to be modulated by Rsv in the context of Doxo treatment: CCND1, CDH1, ESR1, HSP90AA1, MAPK3, PTPN11, and RPS6KB1. Six out of these seven genes have been experimentally proven to be modulated by Rsv in cancer cells, with 4 of the 6 genes in MCF7 cells. In conclusion, acute Rsv potentiated the long-term toxicity of Doxo in breast cancer potentially through the modulation of genes and mechanisms involved in Doxo resistance. Rational autophagy inhibition potentiated the effects of Rsv+Doxo, a strategy that should be further tested in animal models.pdfengHindawiAttribution-NonCommercial-NoDerivatives 4.0 Internacionalhttp://creativecommons.org/licenses/by-nc-nd/4.0/info:eu-repo/semantics/openAccesshttp://purl.org/coar/access_right/c_abf2Oxidative Medicine and Cellular LongevityCellular mechanisms triggered by the cotreatment of resveratrol and doxorubicin in breast cancer: A translational in vitro–In silico modelinfo:eu-repo/semantics/articleArtículo científicohttp://purl.org/coar/version/c_970fb48d4fbd8a85http://purl.org/coar/resource_type/c_2df8fbb1L. A. Torre, F. Islami, R. L. Siegel, E. M. Ward, and A. Jemal, “Global cancer in women: burden and trends,” Cancer Epidemiology, Biomarkers & Prevention, vol. 26, no. 4, pp. 444– 457, 2017.O. Abe, R. Abe, K. Enomoto, K. Kikuchi, H. Koyama et al., “Effects of chemotherapy and hormonal therapy for early breast cancer on recurrence and 15-year survival: an overview of the randomised trials,” Lancet, vol. 365, no. 9472, pp. 1687–1717, 2005.N. Howlader, S. F. Altekruse, C. I. Li et al., “US Incidence of Breast Cancer Subtypes Defined by Joint Hormone Receptor and HER2 Status,” JNCI: Journal of the National Cancer Institute, vol. 106, no. 5, 2014.N. Howlader, K. A. Cronin, A. W. Kurian, and R. Andridge, “Differences in breast cancer survival by molecular subtypes in the United States,” Cancer Epidemiology, Biomarkers & Prevention, vol. 27, no. 6, pp. 619–626, 2018.M. Videira, R. L. Reis, and M. A. Brito, “Deconstructing breast cancer cell biology and the mechanisms of multidrug resistance,” Biochim. Biophys. Acta - Rev. Cancer., vol. 1846, no. 2, pp. 312–325, 2014.The Cancer Genome Atlas Network, “Comprehensive molecular portraits of human breast tumours,” Nature, vol. 490, no. 7418, pp. 61–70, 2012.K. Kalecky, R. Modisette, S. Pena, Y. R. Cho, and J. Taube, “Integrative analysis of breast cancer profiles in TCGA by TNBC subgrouping reveals novel microRNA-specific clusters, including miR-17-92a, distinguishing basal-like 1 and basal-like 2 TNBC subtypes,” BMC Cancer, vol. 20, no. 1, 2020.J. A. Mestres, A. B. iMolins, L. C. Martínez et al., “Defining the optimal sequence for the systemic treatment of metastatic breast cancer,” Clinical and Translational Oncology, vol. 19, no. 2, pp. 149–161, 2017.G. Minotti, P. Menna, E. Salvatorelli, G. Cairo, and L. Gianni, “Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity,” Pharmacological Reviews, vol. 56, no. 2, pp. 185–229, 2004.L. Smith, M. B. Watson, S. L. O'Kane, P. J. Drew, M. J. Lind, and L. Cawkwell, “The analysis of doxorubicin resistance in human breast cancer cells using antibody microarrays,” Molecular Cancer Therapeutics, vol. 5, no. 8, pp. 2115–2120, 2006.X. Jin, Y. Wei, Y. Liu et al., “Resveratrol promotes sensitization to doxorubicin by inhibiting epithelial-mesenchymal transition and modulating SIRT1/β-catenin signaling pathway in breast cancer,” Cancer Medicine, vol. 8, no. 3, pp. 1246–1257, 2019.T. H. Kim, Y. J. Shin, A. J. Won et al., “Resveratrol enhances chemosensitivity of doxorubicin in multidrug-resistant human breast cancer cells via increased cellular influx of doxorubicin,” Biochim. Biophys. Acta - Gen. Subj., vol. 1840, no. 1, pp. 615–625, 2014.Y. Zhao, M. L. Huan, M. Liu et al., “Doxorubicin and resveratrol co-delivery nanoparticle to overcome doxorubicin resistance,” Scientific Reports, vol. 6, no. 1, pp. 1–15, 2016.D. Sinha, N. Sarkar, J. Biswas, and A. Bishayee, “Resveratrol for breast cancer prevention and therapy: Preclinical evidence and molecular mechanisms,” Seminars in Cancer Biology, vol. 40-41, pp. 209–232, 2016.P. A. Volkart, R. B. Gassen, B. M. Nogueira, B. N. Porto, J. E. Vargas, and A. A. Souto, “Antitumor activity of resveratrol is independent of Cu(II) complex formation in MCF-7 cell line,” Bioorganic & Medicinal Chemistry Letters, vol. 27, no. 15, pp. 3238–3242, 2017.A. Chimento, R. Sirianni, C. Saturnino, A. Caruso, M. Stefania Sinicropi, and V. Pezzi, “Resveratrol and Its Analogs As Antitumoral Agents For Breast Cancer Treatment,” Mini-Reviews in Medicinal Chemistry, vol. 16, no. 9, pp. 699–709, 2016.B. S. Vinod, H. H. Nair, V. Vijayakurup et al., “Resveratrol chemosensitizes HER-2-overexpressing breast cancer cells to docetaxel chemoresistance by inhibiting docetaxelmediated activation of HER-2–Akt axis,” Cell Death Discov., vol. 1, no. 1, pp. 1–9, 2015.M. Fukui, N. Yamabe, and B. T. Zhu, “Resveratrol attenuates the anticancer efficacy of paclitaxel in human breast cancer cells in vitro and in vivo,” European Journal of Cancer, vol. 46, no. 10, pp. 1882–1891, 2010.S. E. Al-Harthi, O. M. Alarabi, W. S. Ramadan et al., “Amelioration of doxorubicin-induced cardiotoxicity by resveratrol,” Molecular Medicine Reports, vol. 10, no. 3, pp. 1455–1460, 2014.E. Tatlidede, O. Sehirli, A. Velioğlu-Oğünc et al., “Resveratrol treatment protects against doxorubicin-induced cardiotoxicity by alleviating oxidative damage,” Free Radical Research, vol. 43, no. 3, pp. 195–205, 2009.W. Zhu, W. Qin, K. Zhang et al., “Trans-resveratrol alters mammary promoter hypermethylation in women at increased risk for breast cancer,” Nutrition and Cancer, vol. 64, no. 3, pp. 393–400, 2012.W. Strober, “Trypan Blue Exclusion Test of Cell Viability,” Current Protocols in Immunology, vol. 111, no. 1, pp. - A3.B.1–A3.B.3, 2015.A. O. Silva, E. Dalsin, G. R. Onzi, E. C. Filippi-Chiela, and G. Lenz, “The regrowth kinetic of the surviving population is independent of acute and chronic responses to temozolomide in glioblastoma cell lines,” Experimental Cell Research, vol. 348, no. 2, pp. 177–183, 2016.M. P. Thomé, E. C. Filippi-Chiela, E. S. Villodre et al., “Ratiometric analysis of acridine orange staining in the study of acidic organelles and autophagy,” Journal of Cell Science, vol. 129, no. 24, pp. 4622–4632, 2016.L. L. Zamin, E. C. Filippi-Chiela, P. Dillenburg-Pilla, F. Horn, C. Salbego, and G. Lenz, “Resveratrol and quercetin cooperate to induce senescence-like growth arrest in C6 rat glioma cells,” Cancer Science, vol. 100, no. 9, pp. 1655–1662, 2009.E. C. Filippi-Chiela, M. M. Oliveira, B. Jurkovski, S. M. Callegari-Jacques, V. D. d. Silva, and G. Lenz, “Nuclear Morphometric Analysis (NMA): Screening of Senescence, Apoptosis and Nuclear Irregularities,” PLoS ONE, vol. 7, no. 8, p. e42522, 2012.G. K. Smyth, “Limma: linear models for microarray dat,” in Bioinformatics and computational biology solutions using R and Bioconductor, pp. 397–420, Springer, New York, NY, 2005, http://link.springer.com/chapter/10.1007/0-387- 29362-0_23%5Cnpapers3://publication/uuid/7617F4B3- 24B5-4568-9C76-1C22000E7796.N. T. Doncheva, J. H. Morris, J. Gorodkin, and L. J. Jensen, “Cytoscape StringApp: Network Analysis and Visualization of Proteomics Data,” Journal of Proteome Research, vol. 18, no. 2, pp. 623–632, 2018.P. Shannon, A. Markiel, O. Ozier et al., “Cytoscape: a software environment for integrated models,” Genome Research, vol. 13, p. 426, 1971.S. Pletscher-Frankild, A. Pallejà, K. Tsafou, J. X. Binder, and L. J. Jensen, “DISEASES: text mining and data integration of disease-gene associations,” Methods, vol. 74, pp. 83–89, 2015.D. Szklarczyk, A. Santos, C. Von Mering, L. J. Jensen, P. Bork, and M. Kuhn, “STITCH 5: augmenting protein-chemical interaction networks with tissue and affinity data,” Nucleic Acids Research, vol. 44, no. D1, pp. D380–D384, 2016.Y. Assenov, F. Ramírez, S.-E. Schelhorn, T. Lengauer, and M. Albrecht, “Computing topological parameters of biological networks,” Bioinformatics, vol. 24, no. 2, pp. 282–284, 2008.M. Kanehisa, M. Furumichi, M. Tanabe, Y. Sato, and K. Morishima, “KEGG: new perspectives on genomes, pathways, diseases and drugs,” Nucleic Acids Research, vol. 45, no. D1, pp. D353–D361, 2017.G. Scardoni, M. Petterlini, and C. Laudanna, “Analyzing biological network parameters with CentiScaPe,” Bioinformatics, vol. 25, no. 21, pp. 2857–2859, 2009.M. E. J. Newman, “A measure of betweenness centrality based on random walks,” Social Networks, vol. 27, no. 1, pp. 39–54, 2005.J. E. Vargas, R. Puga, J. d. F. Poloni et al., “A Network Flow Approach to Predict Protein Targets and Flavonoid Backbones to Treat Respiratory Syncytial Virus Infection,” BioMed Research International, vol. 2015, Article ID 301635, 9 pages, 2015.G. Bindea, B. Mlecnik, H. Hackl et al., “ClueGO: a Cytoscape plug-in to decipher functionally grouped gene ontology and pathway annotation networks,” Bioinformatics, vol. 25, no. 8, pp. 1091–1093, 2009.D. S. Chandrashekar, B. Bashel, S. A. H. Balasubramanya et al., “UALCAN: a portal for facilitating tumor subgroup gene expression and survival analyses,” Neoplasia, vol. 19, no. 8, pp. 649–658, 2017.B. Györffy, A. Lanczky, A. C. Eklund et al., “An online survival analysis tool to rapidly assess the effect of 22,277 genes on breast cancer prognosis using microarray data of 1,809 patients,” Breast Cancer Research and Treatment, vol. 123, no. 3, pp. 725–731, 2010.Y. Han, S. Fan, T. Qin et al., “Role of autophagy in breast cancer and breast cancer stem cells (review),” International Journal of Oncology, vol. 52, pp. 1057–1070, 2018.J. I. G. Solari, E. Filippi-Chiela, E. S. Pilar et al., “Damageassociated molecular patterns (DAMPs) related to immunogenic cell death are differentially triggered by clinically relevant chemotherapeutics in lung adenocarcinoma cells,” BMC Cancer, vol. 20, no. 1, 2020.M. Szostakowska, A. Trębińska-Stryjewska, E. A. Grzybowska, and A. Fabisiewicz, “Resistance to endocrine therapy in breast cancer: molecular mechanisms and future goals,” Breast Cancer Research and Treatment, vol. 173, no. 3, pp. 489–497, 2019.C. Chen, L. Lu, S. Yan et al., “Autophagy and doxorubicin resistance in cancer,” Anti-Cancer Drugs, vol. 29, no. 1, pp. 1–9, 2018.N. Matsumura, B. N. Zordoky, I. M. Robertson, and S. M. Hamza, “Co-administration of resveratrol with doxorubicin in young mice attenuates detrimental late-occurring cardiovascular changes,” Cardiovascular Research, vol. 114, no. 10, 2017.Y. Fu, H. Chang, X. Peng et al., “Resveratrol inhibits breast cancer stem-like cells and induces autophagy via suppressing Wnt/β-catenin signaling pathway,” PLoS One, vol. 9, no. 7, pp. e102535–e102538, 2014.Y. N. Zhao, Y. N. Cao, J. Sun et al., “Anti-breast cancer activity of resveratrol encapsulated in liposomes,” Journal of Materials Chemistry B, vol. 8, no. 1, pp. 27–37, 2020.Y.-A. Kim, B. Choi, Y. Lee et al., “Resveratrol inhibits cell proliferation and induces apoptosis of human breast carcinoma MCF-7 cells,” Oncology Reports, vol. 11, pp. 441–446, 2004.E. Pozo-Guisado, J. M. Merino, S. Mulero-Navarro et al., “Resveratrol-induced apoptosis in MCF-7 human breast cancer cells involves a caspase-independent mechanism with downregulation of Bcl-2 and NF-?B,” International Journal of Cancer, vol. 115, no. 1, pp. 74–84, 2005.R. Venkatadri, T. Muni, A. K. V. Iyer, J. S. Yakisich, and N. Azad, “Role of apoptosis-related miRNAs in resveratrolinduced breast cancer cell death,” Cell Death & Disease, vol. 7, no. 2, p. e2104, 2016.E. Pozo-Guisado, A. Alvarez-Barrientos, S. Mulero-Navarro, B. Santiago-Josefat, and P. M. Fernandez-Salguero, “The antiproliferative activity of resveratrol results in apoptosis in MCF-7 but not in MDA-MB-231 human breast cancer cells: cell-specific alteration of the cell cycle,” Biochemical Pharmacology, vol. 64, no. 9, pp. 1375–1386, 2002.J. Suh, D. H. Kim, and Y. J. Surh, “Resveratrol suppresses migration, invasion and stemness of human breast cancer cells by interfering with tumor-stromal cross-talk,” Archives of Biochemistry and Biophysics, vol. 643, pp. 62–71, 2018.D. Park, H. Jeong, M. N. Lee et al., “Resveratrol induces autophagy by directly inhibiting mTOR through ATP competition,” Scientific Reports, vol. 6, no. 1, pp. 1–11, 2016.J. H. Ko, G. Sethi, J. Y. Um et al., “The role of resveratrol in cancer therapy,” International Journal of Molecular Sciences, vol. 18, no. 12, p. 2589, 2017.Y. Umekita, Y. Ohi, Y. Sagara, and H. Yoshida, “Overexpression of cyclinD1 predicts for poor prognosis in estrogen receptor-negative breast cancer patients,” International Journal of Cancer, vol. 98, no. 3, pp. 415–418, 2002.N. Sausgruber, M. M. Coissieux, A. Britschgi et al., “Tyrosine phosphatase SHP2 increases cell motility in triple-negative breast cancer through the activation of SRC-family kinases,” Oncogene, vol. 34, no. 17, pp. 2272–2278, 2015J. A. van der Hage, L. J. C. M. van den Broek, C. Legrand et al., “Overexpression of P70 S6 kinase protein is associated with increased risk of locoregional recurrence in node-negative premenopausal early breast cancer patients,” British Journal of Cancer, vol. 90, no. 8, pp. 1543–1550, 2004.L. Yang, X.‑. W. Wang, L.‑. P. Zhu et al., “Significance and prognosis of epithelial-cadherin expression in invasive breast carcinoma,” Oncology Letters, vol. 16, pp. 1659–1665, 2018.S. Yu, M. Zhang, L. Huang et al., “ERK1 indicates good prognosis and inhibits breast cancer progression by suppressing YAP1 signaling,” Aging (Albany NY), vol. 11, no. 24, pp. 12295–12314, 2019.E. Mechetner, A. Kyshtoobayeva, S. Zonis et al., “Levels of multidrug resistance (MDR1) P-glycoprotein expression by human breast cancer correlate with in vitro resistance to taxol and doxorubicin,” Clinical Cancer Research, vol. 4, no. 2, pp. 389–398, 1998.X. J. Fang, H. Jiang, Y. Q. Zhu, L. Y. Zhang, Q. H. Fan, and Y. Tian, “Doxorubicin induces drug resistance and expression of the novel CD44st via NF-κB in human breast cancerMCF-7 cells,” Oncology Reports, vol. 31, no. 6, pp. 2735–2742, 2014.P. C. Marinello, C. Panis, T. N. X. Silva et al., “Metformin prevention of doxorubicin resistance in MCF-7 and MDA-MB-231 involves oxidative stress generation and modulation of cell adaptation genes,” Scientific Reports, vol. 9, no. 1, 2019.J. Huang, H. Li, and G. Ren, “Epithelial-mesenchymal transition and drug resistance in breast cancer (review),” International Journal of Oncology, vol. 47, no. 3, pp. 840–848, 2015.L. Ponnusamy, P. K. S. Mahalingaiah, and K. P. Singh, “Treatment schedule and estrogen receptor-status influence acquisition of doxorubicin resistance in breast cancer cells,” European Journal of Pharmaceutical Sciences, vol. 104, pp. 424–433, 2017.S. Chen, H. Wang, Z. Li et al., “Interaction of WBP2 with ERα increases doxorubicin resistance of breast cancer cells by modulating MDR1 transcription,” British Journal of Cancer, vol. 119, no. 2, pp. 182–192, 2018.S. AbuHammad and M. Zihlif, “Gene expression alterations in doxorubicin resistant MCF7 breast cancer cell line,” Genomics, vol. 101, no. 4, pp. 213–220, 2013.F. Ashour, M. H. Awwad, H. E. L. Sharawy, and M. Kamal, “Estrogen receptor positive breast tumors resist chemotherapy by the overexpression of P53 in cancer stem cells,” Journal of the Egyptian National Cancer Institute, vol. 30, no. 2, pp. 45–48, 2018.B. D. Gehm, J. M. McAndrews, P. Y. Chien, and J. L. Jameson, “Resveratrol, a polyphenolic compound found in grapes and wine, is an agonist for the estrogen receptor,” Proceedings of the National Academy of Sciences of the United States of America, vol. 94, no. 25, pp. 14138–14143, 1997.J. Saluzzo, K. M. Hallman, K. Aleck et al., “The regulation of tumor suppressor protein, p53, and estrogen receptor (ERα) by resveratrol in breast cancer cells,” Genes and Cancer., vol. 7, no. 11-12, pp. 414–425, 2016.E. M. Ciruelos Gil, “Targeting the PI3K/AKT/mTOR pathway in estrogen receptor-positive breast cancer,” Cancer Treatment Reviews, vol. 40, no. 7, pp. 862–871, 2014.K. A. West, S. S. Castillo, and P. A. Dennis, “Activation of the PI3K/Akt pathway and chemotherapeutic resistance,” Drug Resistance Updates, vol. 5, no. 6, pp. 234–248, 2002.Y. Li, J. Liu, X. Liu et al., “Resveratrol-induced cell inhibition of growth and apoptosis in MCF7 human breast cancer cells are associated with modulation of phosphorylated Akt and caspase-9,” Applied Biochemistry and Biotechnology, vol. 135, no. 3, pp. 181–192, 2006.A. M. Gao, Z. P. Ke, J. N. Wang, J. Y. Yang, S. Y. Chen, and H. Chen, “Apigenin sensitizes doxorubicin-resistant hepatocellular carcinoma BEL-7402/ADM cells to doxorubicin via inhibiting PI3K/Akt/Nrf2 pathway,” Carcinogenesis, vol. 34, no. 8, pp. 1806–1814, 2013.R. L. Yamnik, A. Digilova, D. C. Davis, Z. N. Brodt, C. J. Murphy, and M. K. Holz, “S6 kinase 1 regulates estrogen receptor α in control of breast cancer cell proliferation,” The Journal of Biological Chemistry, vol. 284, no. 10, pp. 6361– 6369, 2009.Y. B. Khotskaya, A. Goverdhan, J. Shen et al., “S6k1 promotes invasiveness of breast cancer cells in a model of metastasis of triple-negative breast cancer,” American Journal of Translational Research, vol. 6, no. 4, pp. 361–376, 2014.D. M. Maruani, T. N. Spiegel, E. N. Harris et al., “Estrogenic regulation of S6K1 expression creates a positive regulatory loop in control of breast cancer cell proliferation,” Oncogene, vol. 31, no. 49, pp. 5073–5080, 2012.A. Alayev, P. F. Doubleday, S. M. Berger, B. A. Ballif, and M. K. Holz, “Phosphoproteomics reveals resveratroldependent inhibition of Akt/mTORC1/S6K1 signaling,” Journal of Proteome Research, vol. 13, no. 12, pp. 5734– 5742, 2014.J. S. Shin, S. H. Woo, and H. C. Lee, “Low doses of ionizing radiation suppress doxorubicin-induced senescence-like phenotypes by activation of ERK1/2 and suppression of p38 kinase in MCF7 human breast cancer cells,” International Journal of Oncology, vol. 36, no. 6, pp. 1445–1452, 2010.C. Ahlin, C. Lundgren, E. Embretsén-Varro, K. Jirström, C. Blomqvist, and M. -L. Fjällskog, “High expression of cyclin D1 is associated to high proliferation rate and increased risk of mortality in women with ER-positive but not in ERnegative breast cancers,” Breast Cancer Research and Treatment, vol. 164, no. 3, pp. 667–678, 2017.M. Stendahl, À. Kronblad, L. Rydén, S. Emdin, N. O. Bengtsson, and G. Landberg, “Cyclin D1 overexpression is a negative predictive factor for tamoxifen response in postmenopausal breast cancer patients,” British Journal of Cancer, vol. 90, no. 10, pp. 1942–1948, 2004.N. V. Rusetskaya, N. Y. Lukyanova, and V. F. Chekhun, “Molecular profile and cell cycle in MCF-7 and MCF-7/Dox cells exposed to conventional and liposomal forms of doxorubicin,” Experimental Oncology, vol. 31, no. 3, pp. 140–143, 2009.A. Mondal and L. L. Bennett, “Resveratrol enhances the efficacy of sorafenib mediated apoptosis in human breast cancer MCF7 cells through ROS, cell cycle inhibition, caspase 3 and PARP cleavage,” Biomedicine & Pharmacotherapy, vol. 84, pp. 1906–1914, 2016.R. Medina-Aguilar, L. A. Marchat, E. A. Ocampo et al., “Resveratrol inhibits cell cycle progression by targeting aurora kinase A and polo-like kinase 1 in breast cancer cells,” Oncology Reports, vol. 35, no. 6, pp. 3696–3704, 2016.D. Luo, D. J. Liao, and Y. Sun, “CyclinD1 protein plays different roles in modulating chemoresponses in MCF7 and MDAMB231 cells,” Journal of Carcinogenesis, vol. 11, no. 1, p. 12, 2012.Z.-P. Ji, L. Qiang, and J.-L. Zhang, “Transcription activated p73-modulated cyclin D1 expression leads to doxorubicin resistance in gastric cancer,” Experimental and Therapeutic Medicine, vol. 15, pp. 1831–1833, 2017.F. Xu, F. Wang, T. Yang, Y. Sheng, T. Zhong, and Y. Chen, “Differential drug resistance acquisition to doxorubicin and paclitaxel in breast cancer cells,” Cancer Cell International, vol. 14, no. 1, 2014.G. Solanas, M. Porta-de-la-Riva, C. Agusti et al., “E-cadherin controls -catenin and NF- B transcriptional activity in mesenchymal gene expression,” Journal of Cell Science, vol. 121, no. 13, pp. 2224–2234, 2008.Z. Hu, H. Fang, X. Wang, D. Chen, Z. Chen, and S. Wang, “Overexpression of SHP2 tyrosine phosphatase promotes the tumorigenesis of breast carcinoma,” Oncology Reports, vol. 32, no. 1, pp. 205–212, 2014.L. Lan, J. D. Holland, J. Qi et al., “Shp2 signaling suppresses senescence in PyMT‐induced mammary gland cancer in mice,” The EMBO Journal, vol. 34, no. 18, pp. 2383–2383, 2015.D. A. Jerjees, M. Alabdullah, M. Alkaabi et al., “ERK1/2 is related to oestrogen receptor and predicts outcome in hormone-treated breast cancer,” Breast Cancer Research and Treatment, vol. 147, no. 1, pp. 25–37, 2014.J. Xu, D. Liu, H. Niu et al., “Resveratrol reverses doxorubicin resistance by inhibiting epithelial-mesenchymal transition (EMT) through modulating PTEN/Akt signaling pathway in gastric cancer,” Journal of Experimental & Clinical Cancer Research, vol. 36, no. 1, 2017.G. Rai, S. Suman, S. Mishra, and Y. Shukla, “Evaluation of growth inhibitory response of resveratrol and salinomycin combinations against triple negative breast cancer cells,” Biomedicine & Pharmacotherapy, vol. 89, pp. 1142–1151, 2017.I. C. Iser, S. M. Ceschini, G. R. Onzi, A. P. S. Bertoni, G. Lenz, and M. R. Wink, “Conditioned medium from adiposederived stem cells (ADSCs) promotes epithelial-to-mesenchymal- like transition (EMT-like) in glioma cells in vitro,” Molecular Neurobiology, vol. 53, no. 10, pp. 7184–7199, 2016.X. D. Zhou and Y. M. Agazie, “Inhibition of SHP2 leads to mesenchymal to epithelial transition in breast cancer cells,” Cell Death and Differentiation, vol. 15, no. 6, pp. 988–996, 2008.Y. Sun, Q. M. Zhou, Y. Y. Lu et al., “Resveratrol inhibits the migration and metastasis ofMDA-MB-231 human breast cancer by reversing TGF-β1-induced epithelial-mesenchymal transition,” Molecules, vol. 24, no. 6, p. 1131, 2019.V. Padmanaban, I. Krol, Y. Suhail et al., “E-cadherin is required for metastasis in multiple models of breast cancer,” Nature, vol. 573, no. 7774, pp. 439–444, 2019.M. Lu, S. Marsters, X. Ye, E. Luis, L. Gonzalez, and A. Ashkenazi, “E-cadherin couples death receptors to the cytoskeleton to regulate apoptosis,” Molecular Cell, vol. 54, no. 6, pp. 987–998, 2014.L. Wang, Z. Li, C. Wang et al., “E-cadherin decreased human breast cancer cells sensitivity to staurosporine by upregulating Bcl-2 expression,” Archives of Biochemistry and Biophysics, vol. 481, no. 1, pp. 116–122, 2009.J. Liu, X. Sun, S. Qin et al., “CDH1 promoter methylation correlates with decreased gene expression and poor prognosis in patients with breast cancer,” Oncology Letters, vol. 11, no. 4, pp. 2635–2643, 2016.T. Han, S. Jiang, H. Zheng et al., “Interplay between c-Src and the APC/C co-activator Cdh1 regulates mammary tumorigenesis,” Nature Communications, vol. 10, no. 1, p. 3716, 2019.N. Matsuyoshi, S. Hirano, M. Takeichi, H. Tahara, T. Kobayashi, and T. Mori, “Expression of E-cadherin cell adhesion molecules in human breast cancer tissues and its relationship to metastasis,” Cancer Research, vol. 53, pp. 1696–1701, 1993.Y. Zhou, Y. Hu, M. Yang et al., “The miR-106b∼25 cluster promotes bypass of doxorubicin-induced senescence and increase in motility and invasion by targeting the Ecadherin transcriptional activator EP300,” Cell Death and Differentiation, vol. 21, no. 3, pp. 462–474, 2014.D. Vergara, C. M. Valente, A. Tinelli et al., “Resveratrol inhibits the epidermal growth factor-induced epithelial mesenchymal transition in MCF-7 cells,” Cancer Letters, vol. 310, no. 1, pp. 1–8, 2011.E. Tili, J. J. Michaille, H. Alder et al., “Resveratrol modulates the levels of microRNAs targeting genes encoding tumorsuppressors and effectors of TGFβ signaling pathway in SW480 cells,” Biochemical Pharmacology, vol. 80, no. 12, pp. 2057–2065, 2010.X. Zhou, J. Coad, B. Ducatman, and Y. M. Agazie, “SHP2 is up-regulated in breast cancer cells and in infiltrating ductal carcinoma of the breast, implying its involvement in breast oncogenesis,” Histopathology, vol. 53, no. 4, pp. 389–402, 2008.N. Aceto, N. Sausgruber, H. Brinkhaus et al., “Tyrosine phosphatase SHP2 promotes breast cancer progression and maintains tumor-initiating cells via activation of key transcription factors and a positive feedback signaling loop,” Nature Medicine, vol. 18, no. 4, pp. 529–537, 2012.X. Zhou and Y. M. Agazie, “Molecular mechanism for SHP2 in promoting HER2-induced signaling and transformation,” The Journal of Biological Chemistry, vol. 284, no. 18, pp. 12226–12234, 2009.L. Zhang, X. Wen, M. Li, S. Li, and H. Zhao, “Targeting cancer stem cells and signaling pathways by resveratrol and pterostilbene,” BioFactors, vol. 44, no. 1, pp. 61–68, 2018.G. Bousquet, M. E. Bouchtaoui, T. Sophie et al., “Targeting autophagic cancer stem-cells to reverse chemoresistance in human triple negative breast cancer,” Oncotarget, vol. 8, no. 21, pp. 35205–35221, 2017.N. E. Bhola, V. M. Jansen, J. P. Koch et al., “Treatment of triple-negative breast cancer with TORC1/2 inhibitors sustains a drug-resistant and notch-dependent cancer stem cell population,” Cancer Research, vol. 76, no. 2, pp. 440–452, 2016.A. Rodina, T. Wang, P. Yan et al., “The epichaperome is an integrated chaperome network that facilitates tumour survival,” Nature, vol. 538, no. 7625, pp. 397–401, 2016.X. Lu, L. Xiao, L. Wang, and D. M. Ruden, “Hsp90 inhibitors and drug resistance in cancer: the potential benefits of combination therapies ofHsp90 inhibitors and other anti-cancer drugs,” Biochemical Pharmacology, vol. 83, no. 8, pp. 995–1004, 2012.M. Klimczak, P. Biecek, A. Zylicz, and M. Zylicz, “Heat shock proteins create a signature to predict the clinical outcome in breast cancer,” Scientific Reports, vol. 9, no. 1, pp. 7507–7515, 2019.C. H. Lai, K. S. Park, D. H. Lee et al., “HSP-90 inhibitor ganetespib is synergistic with doxorubicin in small cell lung cancer,” Oncogene, vol. 33, no. 40, pp. 4867–4876, 2014.J. Díaz-Chávez, M. A. Fonseca-Sánchez, E. Arechaga- Ocampo et al., “Proteomic profiling reveals that resveratrol inhibits HSP27 expression and sensitizes breast cancer cells to doxorubicin therapy,” PLoS One, vol. 8, no. 5, p. e64378, 2013.L. Liu, C. Fu, M. Yan et al., “Resveratrol modulates intestinal morphology and HSP70/90, NF-κB and EGF expression in the jejunal mucosa of black-boned chickens on exposure to circular heat stress,” Food & Function, vol. 7, no. 3, pp. 1329–1338, 2016.K. Sahin, C. Orhan, F. Akdemir, M. Tuzcu, C. Iben, and N. Sahin, “Resveratrol protects quail hepatocytes against heat stress: modulation of the Nrf2 transcription factor and heat shock proteins,” Journal of Animal Physiology and Animal Nutrition, vol. 96, no. 1, pp. 66–74, 2012.ORIGINALPDF.pdfPDF.pdfPDFapplication/pdf9705757https://bonga.unisimon.edu.co/bitstreams/4874ba94-23b7-448f-af72-7fe0ef622a92/download5b445794b89cef77bcc38d8ea9874957MD51LICENSElicense.txtlicense.txttext/plain; charset=utf-8381https://bonga.unisimon.edu.co/bitstreams/1a40dd1f-5e6a-4bf4-8b9e-cb0ed07560e1/download733bec43a0bf5ade4d97db708e29b185MD53CC-LICENSElicense_rdflicense_rdfapplication/rdf+xml; charset=utf-8805https://bonga.unisimon.edu.co/bitstreams/d91817e0-9269-400c-89eb-c54179d5772f/download4460e5956bc1d1639be9ae6146a50347MD52TEXTCellular Mechanisms Triggered by the Cotreatment of Resveratrol and Doxorubicin in Breast Cancer A translational in vitro-in silico model.pdf.txtCellular Mechanisms Triggered by the Cotreatment of Resveratrol and Doxorubicin in Breast Cancer A translational in vitro-in silico model.pdf.txtExtracted texttext/plain98891https://bonga.unisimon.edu.co/bitstreams/79f517a4-c246-4fa6-90b6-f0c6181f58f0/downloadf6739d98c69ed53d48ad946faafd8b9aMD54PDF.pdf.txtPDF.pdf.txtExtracted texttext/plain101144https://bonga.unisimon.edu.co/bitstreams/e0b98c75-b522-44cc-b8c7-59b12cb18ffe/download0e8ba9b88304756eff623bd98cae8c06MD56THUMBNAILCellular Mechanisms Triggered by the Cotreatment of Resveratrol and Doxorubicin in Breast Cancer A translational in vitro-in silico model.pdf.jpgCellular Mechanisms Triggered by the Cotreatment of Resveratrol and Doxorubicin in Breast Cancer A translational in vitro-in silico model.pdf.jpgGenerated Thumbnailimage/jpeg1617https://bonga.unisimon.edu.co/bitstreams/f6c75568-38ce-4931-9274-be7622d2d0bd/download46dd0a23a51209cebd423ee444cac591MD55PDF.pdf.jpgPDF.pdf.jpgGenerated Thumbnailimage/jpeg5662https://bonga.unisimon.edu.co/bitstreams/4d420d49-8adf-47ed-9824-4154346fce60/downloada6a8ad12522728a363ac732bbbd0265fMD5720.500.12442/6761oai:bonga.unisimon.edu.co:20.500.12442/67612024-08-14 21:52:56.311http://creativecommons.org/licenses/by-nc-nd/4.0/Attribution-NonCommercial-NoDerivatives 4.0 Internacionalopen.accesshttps://bonga.unisimon.edu.coRepositorio Digital Universidad Simón Bolívarrepositorio.digital@unisimon.edu.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