Skip to main content
Top
Published in: Investigational New Drugs 6/2021

Open Access 01-12-2021 | PRECLINICAL STUDIES

Network pharmacology of triptolide in cancer cells: implications for transcription factor binding

Authors: Ean-Jeong Seo, Mona Dawood, Annika K. Hult, Martin L. Olsson, Thomas Efferth

Published in: Investigational New Drugs | Issue 6/2021

Login to get access

Summary

Background Triptolide is an active natural product, which inhibits cell proliferation, induces cell apoptosis, suppresses tumor metastasis and improves the effect of other therapeutic treatments in several cancer cell lines by affecting multiple molecules and signaling pathways, such as caspases, heat-shock proteins, DNA damage and NF-ĸB. Purpose We investigated the effect of triptolide towards NF-ĸB and GATA1. Methods We used cell viability assay, compare and cluster analyses of microarray-based mRNA transcriptome-wide expression data, gene promoter binding motif analysis, molecular docking, Ingenuity pathway analysis, NF-ĸB reporter cell assay, and electrophoretic mobility shift assay (EMSA) of GATA1. Results Triptolide inhibited the growth of drug-sensitive (CCRF-CEM, U87.MG) and drug-resistant cell lines (CEM/ADR5000, U87.MGΔEGFR). Hierarchical cluster analysis showed six major clusters in dendrogram. The sensitive and resistant cell lines were statistically significant (p = 0.65 × 10–2) distributed. The binding motifs of NF-κB (Rel) and of GATA1 proteins were significantly enriched in regions of 25 kb upstream promoter of all genes. IPA showed the networks, biological functions, and canonical pathways influencing the activity of triptolide towards tumor cells. Interestingly, upstream analysis for the 40 genes identified by compare analysis revealed ZFPM1 (friend of GATA protein 1) as top transcription regulator. However, we did not observe any effect of triptolide to the binding of GATA1 in vitro. We confirmed that triptolide inhibited NF-κB activity, and it strongly bound to the pharmacophores of IκB kinase β and NF-κB in silico. Conclusion Triptolide showed promising inhibitory effect toward NF-κB, making it a potential candidate for targeting NF-κB.
Literature
1.
go back to reference Kupchan SM et al (1972) Triptolide and tripdiolide, novel antileukemic diterpenoid triepoxides from Tripterygium wilfordii. J Am Chem Soc 94(20):7194–7195PubMedCrossRef Kupchan SM et al (1972) Triptolide and tripdiolide, novel antileukemic diterpenoid triepoxides from Tripterygium wilfordii. J Am Chem Soc 94(20):7194–7195PubMedCrossRef
2.
go back to reference Chugh R et al (2012) A Preclinical Evaluation of Minnelide as a Therapeutic Agent Against Pancreatic Cancer. Science Translational Medicine 4(156) Chugh R et al (2012) A Preclinical Evaluation of Minnelide as a Therapeutic Agent Against Pancreatic Cancer. Science Translational Medicine 4(156)
3.
go back to reference Guo X et al (2016) Protective effects of triptolide on TLR4 mediated autoimmune and inflammatory response induced myocardial fibrosis in diabetic cardiomyopathy. J Ethnopharmacol 193:333–344PubMedCrossRef Guo X et al (2016) Protective effects of triptolide on TLR4 mediated autoimmune and inflammatory response induced myocardial fibrosis in diabetic cardiomyopathy. J Ethnopharmacol 193:333–344PubMedCrossRef
4.
go back to reference Long C et al (2016) Triptolide decreases expression of latency-associated nuclear antigen 1 and reduces viral titers in Kaposi’s sarcoma-associated and herpesvirus-related primary effusion lymphoma cells. Int J Oncol 48(4):1519–1530PubMedCrossRef Long C et al (2016) Triptolide decreases expression of latency-associated nuclear antigen 1 and reduces viral titers in Kaposi’s sarcoma-associated and herpesvirus-related primary effusion lymphoma cells. Int J Oncol 48(4):1519–1530PubMedCrossRef
5.
go back to reference Liu Z et al (2015) Computational prediction and experimental validation of a novel synthesized pan-PIM inhibitor PI003 and its apoptosis-inducing mechanisms in cervical cancer. Oncotarget 6(10):8019–8035PubMedPubMedCentralCrossRef Liu Z et al (2015) Computational prediction and experimental validation of a novel synthesized pan-PIM inhibitor PI003 and its apoptosis-inducing mechanisms in cervical cancer. Oncotarget 6(10):8019–8035PubMedPubMedCentralCrossRef
6.
go back to reference Zhao Q et al (2015) Natural products triptolide, celastrol, and withaferin A inhibit the chaperone activity of peroxiredoxin I. Chem Sci 6(7):4124–4130PubMedPubMedCentralCrossRef Zhao Q et al (2015) Natural products triptolide, celastrol, and withaferin A inhibit the chaperone activity of peroxiredoxin I. Chem Sci 6(7):4124–4130PubMedPubMedCentralCrossRef
8.
9.
go back to reference Yi JM et al (2016) Triptolide Induces Cell Killing in Multidrug-Resistant Tumor Cells via CDK7/RPB1 Rather than XPB or p44. Mol Cancer Ther 15(7):1495–1503PubMedCrossRef Yi JM et al (2016) Triptolide Induces Cell Killing in Multidrug-Resistant Tumor Cells via CDK7/RPB1 Rather than XPB or p44. Mol Cancer Ther 15(7):1495–1503PubMedCrossRef
10.
go back to reference Park SW, Kim YI (2013) Triptolide induces apoptosis of PMA-treated THP-1 cells through activation of caspases, inhibition of NF-kappa B and activation of MAPKs. Int J Oncol 43(4):1169–1175PubMedCrossRef Park SW, Kim YI (2013) Triptolide induces apoptosis of PMA-treated THP-1 cells through activation of caspases, inhibition of NF-kappa B and activation of MAPKs. Int J Oncol 43(4):1169–1175PubMedCrossRef
11.
go back to reference Yang SM et al (2003) Triptolide inhibits the growth and metastasis of solid tumors. Mol Cancer Ther 2(1):65–72PubMed Yang SM et al (2003) Triptolide inhibits the growth and metastasis of solid tumors. Mol Cancer Ther 2(1):65–72PubMed
12.
go back to reference Meng CC et al (2014) Targets and molecular mechanisms of triptolide in cancer therapy. Chin J Cancer Res 26(5):622–626PubMedPubMedCentral Meng CC et al (2014) Targets and molecular mechanisms of triptolide in cancer therapy. Chin J Cancer Res 26(5):622–626PubMedPubMedCentral
13.
go back to reference Darnell JE (2002) Transcription factors as targets for cancer therapy. Nat Rev Cancer 2(10):740–749PubMedCrossRef Darnell JE (2002) Transcription factors as targets for cancer therapy. Nat Rev Cancer 2(10):740–749PubMedCrossRef
15.
16.
go back to reference Baud V, Karin M (2009) OPINION Is NF-kappa B a good target for cancer therapy? Hopes and pitfalls. Nat Rev Drug Discovery 8(1):33–40PubMedCrossRef Baud V, Karin M (2009) OPINION Is NF-kappa B a good target for cancer therapy? Hopes and pitfalls. Nat Rev Drug Discovery 8(1):33–40PubMedCrossRef
17.
go back to reference Ohneda K et al (2002) A minigene containing four discrete cis elements recapitulates GATA-1 gene expression in vivo. Genes Cells 7(12):1243–1254PubMedCrossRef Ohneda K et al (2002) A minigene containing four discrete cis elements recapitulates GATA-1 gene expression in vivo. Genes Cells 7(12):1243–1254PubMedCrossRef
18.
go back to reference Bresnick EH et al (2005) Developmental control via GATA factor interplay at chromatin domains. J Cell Physiol 205(1):1–9PubMedCrossRef Bresnick EH et al (2005) Developmental control via GATA factor interplay at chromatin domains. J Cell Physiol 205(1):1–9PubMedCrossRef
19.
20.
go back to reference Tsang SY et al (2015) Glioma Association and Balancing Selection of ZFPM2. Plos One. 10(7). Tsang SY et al (2015) Glioma Association and Balancing Selection of ZFPM2. Plos One. 10(7).
21.
22.
go back to reference Efferth T et al (2003) Molecular modes of action of artesunate in tumor cell lines. Mol Pharmacol 64(2):382–394PubMedCrossRef Efferth T et al (2003) Molecular modes of action of artesunate in tumor cell lines. Mol Pharmacol 64(2):382–394PubMedCrossRef
23.
go back to reference Saeed M et al (2015) Activity of the dietary flavonoid, apigenin, against multidrug-resistant tumor cells as determined by pharmacogenomics and molecular docking. J Nutr Biochem 26(1):44–56PubMedCrossRef Saeed M et al (2015) Activity of the dietary flavonoid, apigenin, against multidrug-resistant tumor cells as determined by pharmacogenomics and molecular docking. J Nutr Biochem 26(1):44–56PubMedCrossRef
24.
go back to reference Alley MC et al (1988) Feasibility of Drug Screening with Panels of Human-Tumor Cell-Lines Using a Microculture Tetrazolium Assay. Can Res 48(3):589–601 Alley MC et al (1988) Feasibility of Drug Screening with Panels of Human-Tumor Cell-Lines Using a Microculture Tetrazolium Assay. Can Res 48(3):589–601
25.
go back to reference O’Brien J et al (2000) Investigation of the Alamar Blue (resazurin) fluorescent dye for the assessment of mammalian cell cytotoxicity. Eur J Biochem 267(17):5421–5426PubMedCrossRef O’Brien J et al (2000) Investigation of the Alamar Blue (resazurin) fluorescent dye for the assessment of mammalian cell cytotoxicity. Eur J Biochem 267(17):5421–5426PubMedCrossRef
26.
go back to reference Kuete V et al (2016) Cytotoxicity of methanol extracts of Annona muricata, Passiflora edulis and nine other Cameroonian medicinal plants towards multi-factorial drug-resistant cancer cell lines. Springerplus 5 Kuete V et al (2016) Cytotoxicity of methanol extracts of Annona muricata, Passiflora edulis and nine other Cameroonian medicinal plants towards multi-factorial drug-resistant cancer cell lines. Springerplus 5
27.
go back to reference Kuete V et al (2016) Cytotoxicity of methanol extracts of 10 Cameroonian medicinal plants towards multi-factorial drug-resistant cancer cell lines. Bmc Complementary and Alternative Medicine 16 Kuete V et al (2016) Cytotoxicity of methanol extracts of 10 Cameroonian medicinal plants towards multi-factorial drug-resistant cancer cell lines. Bmc Complementary and Alternative Medicine 16
28.
go back to reference Scherf U et al (2000) A gene expression database for the molecular pharmacology of cancer. Nat Genet 24(3):236–244PubMedCrossRef Scherf U et al (2000) A gene expression database for the molecular pharmacology of cancer. Nat Genet 24(3):236–244PubMedCrossRef
29.
go back to reference Amundson SA et al (2008) Integrating global gene expression and radiation survival parameters across the 60 cell lines of the National Cancer Institute Anticancer Drug Screen. Can Res 68(2):415–424CrossRef Amundson SA et al (2008) Integrating global gene expression and radiation survival parameters across the 60 cell lines of the National Cancer Institute Anticancer Drug Screen. Can Res 68(2):415–424CrossRef
30.
go back to reference Wosikowski K et al (1997) Identification of epidermal growth factor receptor and c-erbB2 pathway inhibitors by correlation with gene expression patterns. Jnci-Journal of the National Cancer Institute 89(20):1505–1515CrossRef Wosikowski K et al (1997) Identification of epidermal growth factor receptor and c-erbB2 pathway inhibitors by correlation with gene expression patterns. Jnci-Journal of the National Cancer Institute 89(20):1505–1515CrossRef
31.
go back to reference Evans A et al (2008) Glut-1 as a therapeutic target: increased chemoresistance and HIF-1-independent link with cell turnover is revealed through COMPARE analysis and metabolomic studies. Cancer Chemother Pharmacol 61(3):377–393PubMedCrossRef Evans A et al (2008) Glut-1 as a therapeutic target: increased chemoresistance and HIF-1-independent link with cell turnover is revealed through COMPARE analysis and metabolomic studies. Cancer Chemother Pharmacol 61(3):377–393PubMedCrossRef
32.
go back to reference Fagan V et al (2010) One-pot double intramolecular homolytic aromatic substitution routes to dialicyclic ring fused imidazobenzimidazolequinones and preliminary analysis of anticancer activity. Org Biomol Chem 8(14):3149–3156PubMedCrossRef Fagan V et al (2010) One-pot double intramolecular homolytic aromatic substitution routes to dialicyclic ring fused imidazobenzimidazolequinones and preliminary analysis of anticancer activity. Org Biomol Chem 8(14):3149–3156PubMedCrossRef
33.
go back to reference Efferth T et al (2011) Cytotoxic activity of secondary metabolites derived from Artemisia annua L. towards cancer cells in comparison to its designated active constituent artemisinin. Phytomedicine 18(11):959–969 Efferth T et al (2011) Cytotoxic activity of secondary metabolites derived from Artemisia annua L. towards cancer cells in comparison to its designated active constituent artemisinin. Phytomedicine 18(11):959–969
34.
go back to reference Weinstein JN et al (1997) An information-intensive approach to the molecular pharmacology of cancer. Science 275(5298):343–349PubMedCrossRef Weinstein JN et al (1997) An information-intensive approach to the molecular pharmacology of cancer. Science 275(5298):343–349PubMedCrossRef
35.
go back to reference Liu T et al (2011) Cistrome: an integrative platform for transcriptional regulation studies. Genome Biology 12(8) Liu T et al (2011) Cistrome: an integrative platform for transcriptional regulation studies. Genome Biology 12(8)
36.
go back to reference Zeino M et al (2014) The ability of molecular docking to unravel the controversy and challenges related to P-glycoprotein-a well-known, yet poorly understood drug transporter. Invest New Drugs 32(4):618–625PubMedCrossRef Zeino M et al (2014) The ability of molecular docking to unravel the controversy and challenges related to P-glycoprotein-a well-known, yet poorly understood drug transporter. Invest New Drugs 32(4):618–625PubMedCrossRef
37.
go back to reference Dawood M, Hamdoun S, Efferth T (2018) Multifactorial Modes of Action of Arsenic Trioxide in Cancer Cells as Analyzed by Classical and Network Pharmacology. Front Pharmacol 9:143PubMedPubMedCentralCrossRef Dawood M, Hamdoun S, Efferth T (2018) Multifactorial Modes of Action of Arsenic Trioxide in Cancer Cells as Analyzed by Classical and Network Pharmacology. Front Pharmacol 9:143PubMedPubMedCentralCrossRef
38.
go back to reference Ye J et al (2019) Transcriptome analysis identifies key regulators and networks in Acute myeloid leukemia. Hematology 24(1):487–491PubMedCrossRef Ye J et al (2019) Transcriptome analysis identifies key regulators and networks in Acute myeloid leukemia. Hematology 24(1):487–491PubMedCrossRef
39.
go back to reference Kadioglu O, Efferth T (2015) Pharmacogenomic Characterization of Cytotoxic Compounds from Salvia officinalis in Cancer Cells. J Nat Prod 78(4):762–775PubMedCrossRef Kadioglu O, Efferth T (2015) Pharmacogenomic Characterization of Cytotoxic Compounds from Salvia officinalis in Cancer Cells. J Nat Prod 78(4):762–775PubMedCrossRef
40.
go back to reference Kadioglu O et al (2016) Evaluating ancient Egyptian prescriptions today: Anti-inflammatory activity of Ziziphus spina-christi. Phytomedicine 23(3):293–306PubMedCrossRef Kadioglu O et al (2016) Evaluating ancient Egyptian prescriptions today: Anti-inflammatory activity of Ziziphus spina-christi. Phytomedicine 23(3):293–306PubMedCrossRef
41.
go back to reference Dawood M, Ooko E, Efferth T (2019) Collateral Sensitivity of Parthenolide via NF-kappaB and HIF-alpha Inhibition and Epigenetic Changes in Drug-Resistant Cancer Cell Lines. Front Pharmacol 10:542PubMedPubMedCentralCrossRef Dawood M, Ooko E, Efferth T (2019) Collateral Sensitivity of Parthenolide via NF-kappaB and HIF-alpha Inhibition and Epigenetic Changes in Drug-Resistant Cancer Cell Lines. Front Pharmacol 10:542PubMedPubMedCentralCrossRef
42.
go back to reference Moller M et al (2018) Disruption of a GATA1-binding motif upstream of XG/PBDX abolishes Xg(a) expression and resolves the Xg blood group system. Blood 132(3):334–338PubMedCrossRef Moller M et al (2018) Disruption of a GATA1-binding motif upstream of XG/PBDX abolishes Xg(a) expression and resolves the Xg blood group system. Blood 132(3):334–338PubMedCrossRef
43.
go back to reference Andrews NC, Faller DV (1991) A rapid micropreparation technique for extraction of DNA-binding proteins from limiting numbers of mammalian cells. Nucleic Acids Res 19(9):2499PubMedPubMedCentralCrossRef Andrews NC, Faller DV (1991) A rapid micropreparation technique for extraction of DNA-binding proteins from limiting numbers of mammalian cells. Nucleic Acids Res 19(9):2499PubMedPubMedCentralCrossRef
44.
go back to reference Kiviharju TM et al (2002) Antiproliferative and proapoptotic activities of triptolide (PG490), a natural product entering clinical trials, on primary cultures of human prostatic epithelial cells. Clin Cancer Res 8(8):2666–2674PubMed Kiviharju TM et al (2002) Antiproliferative and proapoptotic activities of triptolide (PG490), a natural product entering clinical trials, on primary cultures of human prostatic epithelial cells. Clin Cancer Res 8(8):2666–2674PubMed
45.
go back to reference Joshi P, Vishwakarma RA, Bharate SB (2017) Natural alkaloids as P-gp inhibitors for multidrug resistance reversal in cancer. Eur J Med Chem 138:273–292PubMedCrossRef Joshi P, Vishwakarma RA, Bharate SB (2017) Natural alkaloids as P-gp inhibitors for multidrug resistance reversal in cancer. Eur J Med Chem 138:273–292PubMedCrossRef
46.
go back to reference Chen M et al (2015) Triptolide inhibits TGF-beta1-induced cell proliferation in rat airway smooth muscle cells by suppressing Smad signaling. Exp Cell Res 331(2):362–368PubMedCrossRef Chen M et al (2015) Triptolide inhibits TGF-beta1-induced cell proliferation in rat airway smooth muscle cells by suppressing Smad signaling. Exp Cell Res 331(2):362–368PubMedCrossRef
47.
go back to reference Hutchison DJ (1963) Cross Resistance and Collateral Sensitivity Studies in Cancer Chemotherapy. Adv Cancer Res 7:235–350PubMedCrossRef Hutchison DJ (1963) Cross Resistance and Collateral Sensitivity Studies in Cancer Chemotherapy. Adv Cancer Res 7:235–350PubMedCrossRef
48.
go back to reference Gottesman MM, Pastan I (1993) Biochemistry of Multidrug-Resistance Mediated by the Multidrug Transporter. Annu Rev Biochem 62:385–427PubMedCrossRef Gottesman MM, Pastan I (1993) Biochemistry of Multidrug-Resistance Mediated by the Multidrug Transporter. Annu Rev Biochem 62:385–427PubMedCrossRef
49.
51.
go back to reference Shinojima N et al (2003) Prognostic value of epidermal growth factor receptor in patients with glioblastoma multiforme. Can Res 63(20):6962–6970 Shinojima N et al (2003) Prognostic value of epidermal growth factor receptor in patients with glioblastoma multiforme. Can Res 63(20):6962–6970
52.
go back to reference Sertel S et al (2012) Factors Determining Sensitivity and Resistance of Tumor Cells to Arsenic Trioxide. Plos One 7(5) Sertel S et al (2012) Factors Determining Sensitivity and Resistance of Tumor Cells to Arsenic Trioxide. Plos One 7(5)
53.
54.
go back to reference Li M et al (2014) NF-kappaB signaling inhibition and anticancer activities of LLDT-246 on human colorectal cancer HCT-116 cells in vitro. Biomed Pharmacother 68(5):527–535PubMedCrossRef Li M et al (2014) NF-kappaB signaling inhibition and anticancer activities of LLDT-246 on human colorectal cancer HCT-116 cells in vitro. Biomed Pharmacother 68(5):527–535PubMedCrossRef
55.
go back to reference Ou CC et al (2012) Triptolide Transcriptionally Represses HER2 in Ovarian Cancer Cells by Targeting NF-kappaB. Evid Based Complement Alternat Med p. 350239 Ou CC et al (2012) Triptolide Transcriptionally Represses HER2 in Ovarian Cancer Cells by Targeting NF-kappaB. Evid Based Complement Alternat Med p. 350239
56.
go back to reference Li CZ et al (2015) The role of breast cancer resistance protein (Bcrp/Abcg2) in triptolide-induced testis toxicity. Toxicology Research 4(5):1260–1268CrossRef Li CZ et al (2015) The role of breast cancer resistance protein (Bcrp/Abcg2) in triptolide-induced testis toxicity. Toxicology Research 4(5):1260–1268CrossRef
Metadata
Title
Network pharmacology of triptolide in cancer cells: implications for transcription factor binding
Authors
Ean-Jeong Seo
Mona Dawood
Annika K. Hult
Martin L. Olsson
Thomas Efferth
Publication date
01-12-2021
Publisher
Springer US
Published in
Investigational New Drugs / Issue 6/2021
Print ISSN: 0167-6997
Electronic ISSN: 1573-0646
DOI
https://doi.org/10.1007/s10637-021-01137-y

Other articles of this Issue 6/2021

Investigational New Drugs 6/2021 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