1
|
Gu T, Raval R, Bashkin Z, Zhou C, Ko S, Kong N, Hong S, Bhaskara A, Shah S, Joshi A, Thellakal S, Rim K, Marimuthu A, Venkatesan S, Wang E, Li S, Jayabalan A, Tao A, Fang Y, Xia L, Chui A, Shu E, Zhang T, Chen Z, Njoo E. Synthesis, antiproliferative activity, and biological profiling of C-19 trityl and silyl ether andrographolide analogs in colon cancer and breast cancer cells. Bioorg Med Chem Lett 2025; 121:130163. [PMID: 40043819 DOI: 10.1016/j.bmcl.2025.130163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 02/08/2025] [Accepted: 02/28/2025] [Indexed: 03/10/2025]
Abstract
Andrographolide, a labdane diterpenoid isolated from Andrographis paniculata, putatively functions through covalent inhibition of NF-κB, a transcription factor that modulates tumor survival and metastasis. Previous studies have found that functionalization of the C-19 hydroxyl alters the primary mode of action from inhibition of NF-κB to the modulation of the Wnt1/β-catenin signaling pathway. Here, we synthesized a series of twelve C-19 trityl and silyl ether analogs, including three novel substituted trityl analogs and four novel substituted silyl analogs of andrographolide. MTT assays revealed cell line selectivity between colorectal and breast cancer cells, which is consistent with known mechanisms of β-catenin-driven cell proliferation in colorectal cancer cell lines. Most compounds exhibited cell line specific antiproliferative activity in HCT-116 and HT-29 colorectal cancer cell lines. Specifically, within 24 h, C-19 analogs of andrographolide exhibit far more limited antiproliferative activity in MCF-7 breast cancer cells compared to HCT-116, HT-29, and MDA-MB-231 cells. Through in vitro TNF-α-dependent NF-κB reporter and Wnt1-dependent luciferase reporter assays, we observed that several analogs generally exhibit greater inhibitory activity compared to andrographolide. Fluorescence imaging demonstrated that cells treated with andrographolide and its C-19 analogs retained similar distributions of active β-catenin, but notable differences in antiproliferative potency upon co-delivery with GSK-3β inhibitor CHIR99021 indicate that several lead compounds exhibit attenuated biological activity selectively in HT-29 cells. Collectively, this work indicates that modest structural modifications at C-19 of andrographolide can have profound implications for its biological activity in mechanisms connected to its anticancer activity.
Collapse
Affiliation(s)
- Tiffany Gu
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Rushika Raval
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Zachary Bashkin
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Carina Zhou
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Sanghyuk Ko
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Natalie Kong
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Seoyeon Hong
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Aditya Bhaskara
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Samarth Shah
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Aditi Joshi
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Samahith Thellakal
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Kaitlyn Rim
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Anushree Marimuthu
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Srishti Venkatesan
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Emma Wang
- Department of Computer Science & Engineering, Aspiring Scholars Directed Research Program, USA
| | - Sophia Li
- Department of Computer Science & Engineering, Aspiring Scholars Directed Research Program, USA
| | - Aditi Jayabalan
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Alice Tao
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Yilin Fang
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Lorelei Xia
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Aidan Chui
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Emily Shu
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA
| | - Tracy Zhang
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Zhan Chen
- Department of Biological and Life Sciences, Aspiring Scholars Directed Research Program, USA
| | - Edward Njoo
- Department of Chemistry, Aspiring Scholars Directed Research Program, USA.
| |
Collapse
|
2
|
Hu J, Li Y, Xie X, Song Y, Yan W, Luo Y, Jiang Y. The therapeutic potential of andrographolide in cancer treatment. Biomed Pharmacother 2024; 180:117438. [PMID: 39298908 DOI: 10.1016/j.biopha.2024.117438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024] Open
Abstract
Cancer poses a substantial global health challenge, necessitating the widespread use of chemotherapy and radiotherapy. Despite these efforts, issues like resistance development and severe side effects remain. As such, the search for more effective alternatives is critical. Andrographolide, a naturally occurring compound, has recently gained attention for its extensive biological activities. This review explores the role of andrographolide in cancer therapy, especially focusing on the molecular mechanisms that drive its anti-tumor properties. It also examines innovative methods to enhance andrographolide's bioavailability, thus boosting its effectiveness against cancer. Notably, andrographolide has potential for use in combination with various clinical drugs, and both preclinical and clinical studies provide strong evidence supporting its broader anticancer applications. Additionally, this paper proposes future research directions for andrographolide's anti-cancer effects and discusses the challenges in its clinical usage along with current research efforts to address these issues. In summary, this review underscores andrographolide's potential roles and contributes to the development of improved cancer treatment strategies.
Collapse
Affiliation(s)
- Jiaxuan Hu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Jiangxi Provincal Key Laboratory of Tissue Engineering, Scientific Research Center, Gannan Medical University, Ganzhou 341000, China
| | - Yi Li
- Department of Anesthesiology, Ganzhou Key Laboratory of Anesthesiology, Ganzhou Key Laboratory of Osteoporosis Research, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Xin Xie
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Jiangxi Provincal Key Laboratory of Tissue Engineering, School of Pharmacy, Gannan Medical University, Ganzhou 341000, China
| | - Yunlei Song
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Jiangxi Provincal Key Laboratory of Tissue Engineering, Scientific Research Center, Gannan Medical University, Ganzhou 341000, China
| | - Wenjing Yan
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Jiangxi Provincal Key Laboratory of Tissue Engineering, Scientific Research Center, Gannan Medical University, Ganzhou 341000, China
| | - Yan Luo
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Jiangxi Provincal Key Laboratory of Tissue Engineering, Scientific Research Center, Gannan Medical University, Ganzhou 341000, China
| | - Yumao Jiang
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Jiangxi Provincal Key Laboratory of Tissue Engineering, Scientific Research Center, Gannan Medical University, Ganzhou 341000, China.
| |
Collapse
|
3
|
Panayides JL, Riley DL, Hasenmaile F, van Otterlo WAL. The role of silicon in drug discovery: a review. RSC Med Chem 2024; 15:3286-3344. [PMID: 39430101 PMCID: PMC11484438 DOI: 10.1039/d4md00169a] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/07/2024] [Indexed: 10/22/2024] Open
Abstract
This review aims to highlight the role of silicon in drug discovery. Silicon and carbon are often regarded as being similar with silicon located directly beneath carbon in the same group in the periodic table. That being noted, in many instances a clear dichotomy also exists between silicon and carbon, and these differences often lead to vastly different physiochemical and biological properties. As a result, the utility of silicon in drug discovery has attracted significant attention and has grown rapidly over the past decade. This review showcases some recent advances in synthetic organosilicon chemistry and examples of the ways in which silicon has been employed in the drug-discovery field.
Collapse
Affiliation(s)
- Jenny-Lee Panayides
- Pharmaceutical Technologies, Future Production: Chemicals, Council for Scientific and Industrial Research (CSIR) Meiring Naude Road, Brummeria Pretoria South Africa
| | - Darren Lyall Riley
- Department of Chemistry, Faculty of Natural and Agricultural Sciences, University of Pretoria Lynnwood Road Pretoria South Africa
| | - Felix Hasenmaile
- Department of Chemistry and Polymer Science, Stellenbosch University Matieland Stellenbosch 7600 South Africa
| | - Willem A L van Otterlo
- Department of Chemistry and Polymer Science, Stellenbosch University Matieland Stellenbosch 7600 South Africa
| |
Collapse
|
4
|
Messire G, Rollin P, Gillaizeau I, Berteina-Raboin S. Synthetic Modifications of Andrographolide Targeting New Potential Anticancer Drug Candidates: A Comprehensive Overview. Molecules 2024; 29:2884. [PMID: 38930949 PMCID: PMC11206892 DOI: 10.3390/molecules29122884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/10/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
This review collects the synthetic modifications performed on andrographolide, a natural molecule derived from Andrographis paniculata, for oncology applications. Various pharmacomodulations were carried out, and the products were tested on different cancer cell lines. The impact of these modifications was analyzed with the aim of mapping the positions essential for activity to facilitate future research in this field. However, this study makes it clear that, in addition to structural modifications of the molecule, which can result in varying degrees of effectiveness in targeting interactions, the lipophilic capacity of the structures obtained through hemisynthesis is of significant importance.
Collapse
Affiliation(s)
| | | | | | - Sabine Berteina-Raboin
- Institut de Chimie Organique et Analytique (ICOA), Université d’Orléans, UMR-CNRS 7311, BP 6759, rue de Chartres, 45067 Orléans, Cedex 2, France; (G.M.); (P.R.); (I.G.)
| |
Collapse
|
5
|
Sharma NK, Bahot A, Sekar G, Bansode M, Khunteta K, Sonar PV, Hebale A, Salokhe V, Sinha BK. Understanding Cancer's Defense against Topoisomerase-Active Drugs: A Comprehensive Review. Cancers (Basel) 2024; 16:680. [PMID: 38398072 PMCID: PMC10886629 DOI: 10.3390/cancers16040680] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
In recent years, the emergence of cancer drug resistance has been one of the crucial tumor hallmarks that are supported by the level of genetic heterogeneity and complexities at cellular levels. Oxidative stress, immune evasion, metabolic reprogramming, overexpression of ABC transporters, and stemness are among the several key contributing molecular and cellular response mechanisms. Topo-active drugs, e.g., doxorubicin and topotecan, are clinically active and are utilized extensively against a wide variety of human tumors and often result in the development of resistance and failure to therapy. Thus, there is an urgent need for an incremental and comprehensive understanding of mechanisms of cancer drug resistance specifically in the context of topo-active drugs. This review delves into the intricate mechanistic aspects of these intracellular and extracellular topo-active drug resistance mechanisms and explores the use of potential combinatorial approaches by utilizing various topo-active drugs and inhibitors of pathways involved in drug resistance. We believe that this review will help guide basic scientists, pre-clinicians, clinicians, and policymakers toward holistic and interdisciplinary strategies that transcend resistance, renewing optimism in the ongoing battle against cancer.
Collapse
Affiliation(s)
- Nilesh Kumar Sharma
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Anjali Bahot
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Gopinath Sekar
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Mahima Bansode
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Kratika Khunteta
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Priyanka Vijay Sonar
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Ameya Hebale
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Vaishnavi Salokhe
- Cancer and Translational Research Centre Dr. D.Y. Patil Biotechnology & Bioinformatics Institute, Dr. D.Y. Patil Vidyapeeth, Pune 411033, Maharashtra, India; (N.K.S.); (A.B.); (G.S.); (M.B.); (K.K.); (P.V.S.); (A.H.); (V.S.)
| | - Birandra Kumar Sinha
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA
| |
Collapse
|
6
|
Rosin Derivative IDOAMP Inhibits Prostate Cancer Growth via Activating RIPK1/RIPK3/MLKL Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9325973. [PMID: 35965682 PMCID: PMC9371855 DOI: 10.1155/2022/9325973] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 07/25/2022] [Indexed: 12/05/2022]
Abstract
Rosin derivatives such as dehydroabietic acid and dehydroabietic amine belonging to diterpenoids have similar structure with androgen that inhibited the occurrence and development of prostate cancer. In this study, the effects and possible mechanism of the rosin derivative IDOAMP on prostate cancer were investigated. Our results showed that IDOAMP effectively inhibited cell viabilities of LNCaP, PC3, and DU145 prostate cells. After the treatment with IDOAMP, the levels of cleaved-PARP, LC3BII/I, and HMGB1 were increased, whereas the expression of GPX4 was decreased. Interestingly, cell viability was reversed by the supplements of necrostatin-1 and necrosulfonamide. Meanwhile, the IDOAMP downregulated the expression of human Aurora kinase A that was overexpressed in prostate cancer. In addition, co-IP results showed that IDOAMP inhibited the binding of Aurora kinase A to the receptor-interacting serine/threonine kinase 1 (RIPK1) and RIPK3. However, the binding of RIPK1 to FADD, RIPK3, or MLKL was significantly promoted. Further studies showed that the phosphorylation levels of RIPK1, RIPK, and MLKL were increased in a concentration-dependent manner. In in vivo model, IDOAMP reduced the tumor volumes and weights. In conclusion, IDOAMP directly inhibited Aurora kinase A and promoted the RIPK1/RIPK3/MLKL necrosome activation to inhibit the prostate cancer.
Collapse
|
7
|
Mitra Ghosh T, Kansom T, Mazumder S, Davis J, Alnaim AS, Jasper SL, Zhang C, Bird A, Opanasopit P, Mitra AK, Arnold RD. The Andrographolide Analogue 3A.1 Synergizes with Taxane Derivatives in Aggressive Metastatic Prostate Cancers by Upregulation of Heat Shock Proteins and Downregulation of MAT2A-Mediated Cell Migration and Invasion. J Pharmacol Exp Ther 2022; 380:180-201. [PMID: 34949650 DOI: 10.1124/jpet.121.000898] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 12/06/2021] [Indexed: 11/22/2022] Open
Abstract
Conventional treatment with taxanes (docetaxel-DTX or cabazitaxel-CBZ) increases the survival rates of patients with aggressive metastatic castration-resistant prostate cancer (mCRPC); however, most patients acquire resistance to taxanes. The andrographolide analog, 19-tert-butyldiphenylsilyl-8,7-epoxy andrographolide (3A.1), has shown anticancer activity against various cancers. In this study, we investigated the effect of 3A.1 alone and in combination with DTX/CBZ against mCRPC and their mechanism of action. Exposure to 3A.1 alone exhibited a dose- and time-dependent antitumor activity in mCRPC. Chou-Talalay's combination index (CI) values of all 3A.1 + TX combinations were less than 0.5, indicating synergism. Co-treatment of 3A.1 with TX reduced the required dose of DTX and CBZ (P < 0.05). Caspase assay (apoptosis) results concurred with in vitro cytotoxicity data. RNA sequencing (RNAseq), followed by ingenuity pathway analysis (IPA), identified that upregulation of heat-shock proteins (Hsp70, Hsp40, Hsp27, and Hsp90) and downregulation of MAT2A as the key player for 3A.1 response. Furthermore, the top treatment-induced differentially expressed genes (DEGs) belong to DNA damage, cell migration, hypoxia, autophagy (MMP1, MMP9, HIF-1α, Bag-3, H2AX, HMOX1, PSRC1), and cancer progression pathways. Most importantly, top downregulated DEG MAT2A has earlier been shown to be involved in cell migration and invasion. Furthermore, using in silico analysis on the Cancer Genome Atlas (TCGA) database, this study found that MAT2A and highly co-expressed (r > 0.7) genes, TRA2B and SF1, were associated with worse Gleason score and nodal metastasis status in prostate adenocarcinoma patients (PRAD-TCGA). Immunoblotting, comet, and migration assays corroborated these findings. These results suggest that 3A.1 may be useful in increasing the anticancer efficacy of taxanes to treat aggressive PCa. SIGNIFICANCE STATEMENT: The andrographolide analogue, 19-tert-butyldiphenylsilyl-8,7-epoxy andrographolide (3A.1), showed anticancer activity against metastatic castration-resistant and neuroendocrine variant prostate cancers (mCRPC/NEPC). Additionally, 3A.1 exhibited synergistic anticancer effect in combination with standard chemotherapy drugs docetaxel and cabazitaxel in mCRPC/NEPC. Post-treatment gene expression studies revealed that heat shock proteins (Hsp70, Hsp40, Hsp27, and Hsp90) and MAT2A are important in the mechanism of 3A.1 action and drug response. Furthermore, DNA damage, cell migration, hypoxia, and autophagy were crucial pathways for the anticancer activity of 3A.1.
Collapse
Affiliation(s)
- Taraswi Mitra Ghosh
- Department of Drug Discovery and Development, Harrison School of Pharmacy (T.M.G., T.K., S.M., J.D., A.S.A., S.L.J., C.Z., A.B., A.K.M., R.D.A.) and Center for Pharmacogenomics and Single-Cell Omics Initiative (S.M., A.K.M.), Auburn University, Auburn, Alabama; Department of Pharmaceutical Technology, Faculty of Pharmacy, Silpakorn University, Bangkok, Thailand (T.K., P.O.); and University of Alabama at Birmingham O'Neal Comprehensive Cancer Center, Birmingham, Alabama (A.K.M., R.D.A.)
| | - Teeratas Kansom
- Department of Drug Discovery and Development, Harrison School of Pharmacy (T.M.G., T.K., S.M., J.D., A.S.A., S.L.J., C.Z., A.B., A.K.M., R.D.A.) and Center for Pharmacogenomics and Single-Cell Omics Initiative (S.M., A.K.M.), Auburn University, Auburn, Alabama; Department of Pharmaceutical Technology, Faculty of Pharmacy, Silpakorn University, Bangkok, Thailand (T.K., P.O.); and University of Alabama at Birmingham O'Neal Comprehensive Cancer Center, Birmingham, Alabama (A.K.M., R.D.A.)
| | - Suman Mazumder
- Department of Drug Discovery and Development, Harrison School of Pharmacy (T.M.G., T.K., S.M., J.D., A.S.A., S.L.J., C.Z., A.B., A.K.M., R.D.A.) and Center for Pharmacogenomics and Single-Cell Omics Initiative (S.M., A.K.M.), Auburn University, Auburn, Alabama; Department of Pharmaceutical Technology, Faculty of Pharmacy, Silpakorn University, Bangkok, Thailand (T.K., P.O.); and University of Alabama at Birmingham O'Neal Comprehensive Cancer Center, Birmingham, Alabama (A.K.M., R.D.A.)
| | - Joshua Davis
- Department of Drug Discovery and Development, Harrison School of Pharmacy (T.M.G., T.K., S.M., J.D., A.S.A., S.L.J., C.Z., A.B., A.K.M., R.D.A.) and Center for Pharmacogenomics and Single-Cell Omics Initiative (S.M., A.K.M.), Auburn University, Auburn, Alabama; Department of Pharmaceutical Technology, Faculty of Pharmacy, Silpakorn University, Bangkok, Thailand (T.K., P.O.); and University of Alabama at Birmingham O'Neal Comprehensive Cancer Center, Birmingham, Alabama (A.K.M., R.D.A.)
| | - Ahmed S Alnaim
- Department of Drug Discovery and Development, Harrison School of Pharmacy (T.M.G., T.K., S.M., J.D., A.S.A., S.L.J., C.Z., A.B., A.K.M., R.D.A.) and Center for Pharmacogenomics and Single-Cell Omics Initiative (S.M., A.K.M.), Auburn University, Auburn, Alabama; Department of Pharmaceutical Technology, Faculty of Pharmacy, Silpakorn University, Bangkok, Thailand (T.K., P.O.); and University of Alabama at Birmingham O'Neal Comprehensive Cancer Center, Birmingham, Alabama (A.K.M., R.D.A.)
| | - Shanese L Jasper
- Department of Drug Discovery and Development, Harrison School of Pharmacy (T.M.G., T.K., S.M., J.D., A.S.A., S.L.J., C.Z., A.B., A.K.M., R.D.A.) and Center for Pharmacogenomics and Single-Cell Omics Initiative (S.M., A.K.M.), Auburn University, Auburn, Alabama; Department of Pharmaceutical Technology, Faculty of Pharmacy, Silpakorn University, Bangkok, Thailand (T.K., P.O.); and University of Alabama at Birmingham O'Neal Comprehensive Cancer Center, Birmingham, Alabama (A.K.M., R.D.A.)
| | - Chu Zhang
- Department of Drug Discovery and Development, Harrison School of Pharmacy (T.M.G., T.K., S.M., J.D., A.S.A., S.L.J., C.Z., A.B., A.K.M., R.D.A.) and Center for Pharmacogenomics and Single-Cell Omics Initiative (S.M., A.K.M.), Auburn University, Auburn, Alabama; Department of Pharmaceutical Technology, Faculty of Pharmacy, Silpakorn University, Bangkok, Thailand (T.K., P.O.); and University of Alabama at Birmingham O'Neal Comprehensive Cancer Center, Birmingham, Alabama (A.K.M., R.D.A.)
| | - Aedan Bird
- Department of Drug Discovery and Development, Harrison School of Pharmacy (T.M.G., T.K., S.M., J.D., A.S.A., S.L.J., C.Z., A.B., A.K.M., R.D.A.) and Center for Pharmacogenomics and Single-Cell Omics Initiative (S.M., A.K.M.), Auburn University, Auburn, Alabama; Department of Pharmaceutical Technology, Faculty of Pharmacy, Silpakorn University, Bangkok, Thailand (T.K., P.O.); and University of Alabama at Birmingham O'Neal Comprehensive Cancer Center, Birmingham, Alabama (A.K.M., R.D.A.)
| | - Praneet Opanasopit
- Department of Drug Discovery and Development, Harrison School of Pharmacy (T.M.G., T.K., S.M., J.D., A.S.A., S.L.J., C.Z., A.B., A.K.M., R.D.A.) and Center for Pharmacogenomics and Single-Cell Omics Initiative (S.M., A.K.M.), Auburn University, Auburn, Alabama; Department of Pharmaceutical Technology, Faculty of Pharmacy, Silpakorn University, Bangkok, Thailand (T.K., P.O.); and University of Alabama at Birmingham O'Neal Comprehensive Cancer Center, Birmingham, Alabama (A.K.M., R.D.A.)
| | - Amit K Mitra
- Department of Drug Discovery and Development, Harrison School of Pharmacy (T.M.G., T.K., S.M., J.D., A.S.A., S.L.J., C.Z., A.B., A.K.M., R.D.A.) and Center for Pharmacogenomics and Single-Cell Omics Initiative (S.M., A.K.M.), Auburn University, Auburn, Alabama; Department of Pharmaceutical Technology, Faculty of Pharmacy, Silpakorn University, Bangkok, Thailand (T.K., P.O.); and University of Alabama at Birmingham O'Neal Comprehensive Cancer Center, Birmingham, Alabama (A.K.M., R.D.A.)
| | - Robert D Arnold
- Department of Drug Discovery and Development, Harrison School of Pharmacy (T.M.G., T.K., S.M., J.D., A.S.A., S.L.J., C.Z., A.B., A.K.M., R.D.A.) and Center for Pharmacogenomics and Single-Cell Omics Initiative (S.M., A.K.M.), Auburn University, Auburn, Alabama; Department of Pharmaceutical Technology, Faculty of Pharmacy, Silpakorn University, Bangkok, Thailand (T.K., P.O.); and University of Alabama at Birmingham O'Neal Comprehensive Cancer Center, Birmingham, Alabama (A.K.M., R.D.A.)
| |
Collapse
|
8
|
Malat P, Ekalaksananan T, Heawchaiyaphum C, Suebsasana S, Roytrakul S, Yingchutrakul Y, Pientong C. Andrographolide Inhibits Lytic Reactivation of Epstein-Barr Virus by Modulating Transcription Factors in Gastric Cancer. Microorganisms 2021; 9:microorganisms9122561. [PMID: 34946164 PMCID: PMC8708910 DOI: 10.3390/microorganisms9122561] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/24/2022] Open
Abstract
Andrographolide is the principal bioactive chemical constituent of Andrographis paniculata and exhibits activity against several viruses, including Epstein–Barr virus (EBV). However, the particular mechanism by which andrographolide exerts an anti-EBV effect in EBV-associated gastric cancer (EBVaGC) cells remains unclear. We investigated the molecular mechanism by which andrographolide inhibits lytic reactivation of EBV in EBVaGC cells (AGS-EBV cell line) using proteomics and bioinformatics approaches. An andrographolide treatment altered EBV protein-expression patterns in AGS-EBV cells by suppressing the expression of EBV lytic protein. Interestingly cellular transcription factors (TFs), activators for EBV lytic reactivation, such as MEF2D and SP1, were significantly abolished in AGS-EBV cells treated with andrographolide and sodium butyrate (NaB) compared with NaB-treated cells. In contrast, the suppressors of EBV lytic reactivation, such as EZH2 and HDAC6, were significantly up-regulated in cells treated with both andrographolide and NaB compared with NaB treatment alone. In addition, bioinformatics predicted that HDAC6 could interact directly with MEF2D and SP1. Furthermore, andrographolide significantly induced cell cytotoxicity and apoptosis of AGS-EBV cells by induction of apoptosis-related protein expression. Our results suggest that andrographolide inhibits EBV lytic reactivation by inhibition of host TFs, partially through the interaction of HDAC6 with TFs, and induces apoptosis of EBVaGC cells.
Collapse
Affiliation(s)
- Praphatson Malat
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (P.M.); (T.E.); (C.H.)
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Tipaya Ekalaksananan
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (P.M.); (T.E.); (C.H.)
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Chukkris Heawchaiyaphum
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (P.M.); (T.E.); (C.H.)
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Supawadee Suebsasana
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Thammasat University, Bangkok 10200, Thailand;
| | - Sittiruk Roytrakul
- Genome Technology Research Unit, Proteomics Research Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), Pathum Thani 12120, Thailand; (S.R.); (Y.Y.)
| | - Yodying Yingchutrakul
- Genome Technology Research Unit, Proteomics Research Laboratory, National Center for Genetic Engineering and Biotechnology (BIOTEC), Pathum Thani 12120, Thailand; (S.R.); (Y.Y.)
| | - Chamsai Pientong
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (P.M.); (T.E.); (C.H.)
- HPV & EBV and Carcinogenesis Research Group, Khon Kaen University, Khon Kaen 40002, Thailand
- Correspondence:
| |
Collapse
|
9
|
Paul S, Roy D, Pati S, Sa G. The Adroitness of Andrographolide as a Natural Weapon Against Colorectal Cancer. Front Pharmacol 2021; 12:731492. [PMID: 34795581 PMCID: PMC8592893 DOI: 10.3389/fphar.2021.731492] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 09/28/2021] [Indexed: 12/15/2022] Open
Abstract
The conventional carcinoma treatment generally encompasses the employment of radiotherapy, chemotherapy, surgery or use of cytotoxic drugs. However, recent advances in pharmacological research have divulged the importance of traditional treatments in cancer. The aim of the present review is to provide an overview of the importance of one such medicinal herb of Chinese and Indian origin: Andrographis paniculate on colorectal cancer with special emphasis on its principal bioactive component andrographolide (AGP) and its underlying mechanisms of action. AGP has long been known to possess medicinal properties. Studies led by numerous groups of researchers shed light on its molecular mechanism of action. AGP has been shown to act in a multi-faceted manner in context of colorectal cancer by targeting matrix metalloproteinase-9, Toll-like receptor or NFκB signaling pathways. In this review, we highlighted the recent studies that show that AGP can act as an effective immunomodulator by harnessing effective anti-tumor immune response. Recent studies strongly recommend further research on this compound and its analogues, especially under in-vivo condition to assess its actual potential as a prospective and efficient candidate against colorectal cancer. The current review deals with the roles of this phytomedicine in context of colorectal cancer and briefly describes its perspectives to emerge as an essential anti-cancer drug candidate. Finally, we also point out the drawbacks and difficulties in administration of AGP and indicate the use of nano-formulations of this phytomedicine for better therapeutic efficacy.
Collapse
Affiliation(s)
- Silpita Paul
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Dia Roy
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Subhadip Pati
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| | - Gaurisankar Sa
- Division of Molecular Medicine, Bose Institute, Kolkata, India
| |
Collapse
|
10
|
Udomwan P, Pientong C, Tongchai P, Burassakarn A, Sunthamala N, Roytrakul S, Suebsasana S, Ekalaksananan T. Proteomics Analysis of Andrographolide-Induced Apoptosis via the Regulation of Tumor Suppressor p53 Proteolysis in Cervical Cancer-Derived Human Papillomavirus 16-Positive Cell Lines. Int J Mol Sci 2021; 22:ijms22136806. [PMID: 34202736 PMCID: PMC8268713 DOI: 10.3390/ijms22136806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 06/22/2021] [Accepted: 06/22/2021] [Indexed: 12/22/2022] Open
Abstract
Regardless of the prophylactic vaccine accessibility, persistent infections of high-risk human papillomaviruses (hr-HPVs), recognized as an etiology of cervical cancers, continues to represent a major health problem for the world population. An overexpression of viral early protein 6 (E6) is linked to carcinogenesis. E6 induces anti-apoptosis by degrading tumor suppressor proteins p53 (p53) via E6-E6-associated protein (E6AP)-mediated polyubiquitination. Thus, the restoration of apoptosis by interfering with the E6 function has been proposed as a selective medicinal strategy. This study aimed to determine the activities of andrographolide (Androg) on the disturbance of E6-mediated p53 degradation in cervical cancer cell lines using a proteomic approach. These results demonstrated that Androg could restore the intracellular p53 level, leading to apoptosis-induced cell death in HPV16-positive cervical cancer cell lines, SiHa and CaSki. Mechanistically, the anti-tumor activity of Androg essentially relied on the reduction in host cell proteins, which are associated with ubiquitin-mediated proteolysis pathways, particularly HERC4 and SMURF2. They are gradually suppressed in Androg-treated HPV16-positive cervical cancer cells. Collectively, the restoration of p53 in HPV16-positive cervical cancer cells might be achieved by disruption of E3 ubiquitin ligase activity by Androg, which could be an alternative treatment for HPV-associated epithelial lesions.
Collapse
Affiliation(s)
- Pariyakorn Udomwan
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (P.U.); (C.P.); (P.T.); (A.B.)
- HPV & EBV and Carcinogenesis Research (HEC) Group, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Chamsai Pientong
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (P.U.); (C.P.); (P.T.); (A.B.)
- HPV & EBV and Carcinogenesis Research (HEC) Group, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Panwad Tongchai
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (P.U.); (C.P.); (P.T.); (A.B.)
- HPV & EBV and Carcinogenesis Research (HEC) Group, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Ati Burassakarn
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (P.U.); (C.P.); (P.T.); (A.B.)
- HPV & EBV and Carcinogenesis Research (HEC) Group, Khon Kaen University, Khon Kaen 40002, Thailand;
| | - Nuchsupha Sunthamala
- HPV & EBV and Carcinogenesis Research (HEC) Group, Khon Kaen University, Khon Kaen 40002, Thailand;
- Department of Biology, Faculty of Science, Mahasarakham University, Mahasarakham 44150, Thailand
| | - Sittiruk Roytrakul
- Functional Ingredients and Food Innovation Research Group, National Center for Genetic Engineering and Biotechnology, National Science and Technology Development Agency, Pathum Thani 12120, Thailand;
| | - Supawadee Suebsasana
- Faculty of Pharmacy, Thammasat University (Rangsit campus), Pathum Thani 12120, Thailand;
| | - Tipaya Ekalaksananan
- Department of Microbiology, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; (P.U.); (C.P.); (P.T.); (A.B.)
- HPV & EBV and Carcinogenesis Research (HEC) Group, Khon Kaen University, Khon Kaen 40002, Thailand;
- Correspondence: ; Tel./Fax: +66-4334-8385
| |
Collapse
|
11
|
Suriyo T, Chotirat S, Rangkadilok N, Pholphana N, Satayavivad J. Interactive effects of Andrographis paniculata extracts and cancer chemotherapeutic 5-Fluorouracil on cytochrome P450s expression in human hepatocellular carcinoma HepG2 cells. J Herb Med 2021. [DOI: 10.1016/j.hermed.2021.100421] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
12
|
Shakya B, Yadav PN. Thiosemicarbazones as Potent Anticancer Agents and their Modes of Action. Mini Rev Med Chem 2020; 20:638-661. [DOI: 10.2174/1389557519666191029130310] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Revised: 09/17/2019] [Accepted: 10/03/2019] [Indexed: 11/22/2022]
Abstract
:Thiosemicarbazones (TSCs) are a class of Schiff bases usually obtained by the condensation of thiosemicarbazide with a suitable aldehyde or ketone. TSCs have been the focus of chemists and biologists due to their wide range of pharmacological effects. One of the promising areas in which these excellent metal chelators are being developed is their use against cancer. TSCs have a wide clinical antitumor spectrum with efficacy in various tumor types such as leukemia, pancreatic cancer, breast cancer, non-small cell lung cancer, cervical cancer, prostate cancer and bladder cancer. To obtain better activity, different series of TSCs have been developed by modifying the heteroaromatic system in their molecules. These compounds possessed significant antineoplastic activity when the carbonyl attachment of the side chain was located at a position α to the ring nitrogen atom, whereas attachment of the side chain β or γ to the heterocyclic N atom resulted in inactive antitumor agents. In addition, replacement of the heterocyclic ring N with C also resulted in a biologically inactive compound suggesting that a conjugated N,N,S-tridentate donor set is essential for the biological activities of thiosemicarbazones. Several possible mechanisms have been implemented for the anticancer activity of thiosemicarbazones.
Collapse
Affiliation(s)
- Bhushan Shakya
- Amrit Campus, Tribhuvan University, Thamel, Kathmandu, Nepal
| | - Paras Nath Yadav
- Central Department of Chemistry, Tribhuvan University, Kirtipur, Kathmandu, Nepal
| |
Collapse
|
13
|
Kitdumrongthum S, Reabroi S, Suksen K, Tuchinda P, Munyoo B, Mahalapbutr P, Rungrotmongkol T, Ounjai P, Chairoungdua A. Inhibition of topoisomerase IIα and induction of DNA damage in cholangiocarcinoma cells by altholactone and its halogenated benzoate derivatives. Biomed Pharmacother 2020; 127:110149. [PMID: 32344256 DOI: 10.1016/j.biopha.2020.110149] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 04/01/2020] [Accepted: 04/04/2020] [Indexed: 01/02/2023] Open
Abstract
Topoisomerase IIα enzyme (Topo IIα) plays a critical function in DNA replication process and is considered to be a promising target of anti-cancer drugs. In the present study, we reported that the altholactone derivatives modified by adding a halogenated benzoate group showed greater inhibitory activity on Topo IIα enzyme in cell-free system concomitant with cytotoxicity against the CCA cell lines (KKU-M055 and KKU-M213) than those of the parent altholactone. However, the cytotoxic activities of four halogenated benzoate altholactone derivatives including iodo-, fluoro-, chloro-, and bromobenzoate derivatives (compound 1, 2, 3, and 4, respectively) were of equal potency. The fluorobenzoate derivative (compound 2) was chosen for investigating the underlying mechanism in CCA cells. Compound 2 arrested CCA cell cycle at sub G1 phase and induced apoptotic cell death. It markedly inhibited Topo IIα protein expression in both KKU-M055 and KKU-M213 cells, which was accompanied by DNA double-strand breaks demonstrated by an increase in phosphorylated H2A.X protein. Interestingly, KKU-M055 cells, which express higher Topo IIα mRNA compared to KKU-M213 cells, showed greater sensitivity to the compound, indicating the selectivity of the compound to Topo IIα enzyme. By computational docking analysis, the binding affinity of altholactone (-52.5 kcal/mol) and compound 2 (-56.7 kcal/mol) were similar to that of the Topo II poison salvicine (-53.7 kcal/mol). The aromatic moiety of both altholactones embedded in a hydrophobic pocket of Topo II ATPase domain. In addition, compound 2 induced the formation of linear DNA in Topo II-mediated cleavage assay. Collectively, our results demonstrate that the addition of fluorobenzoyl group to altholactone enhances potency and selectivity to inhibit type IIα topoisomerases. Atholactone and fluorobenzoate derivative act as Topo II cleavage complexes stabilizing compounds or Topo II poisons preferentially through binding at ATPase domain of Topo IIα, leading to DNA double-strand breaks and apoptosis induction. Such activity of 3-fluorobenzoate derivative of altholactone should be further explored for the development of an anti-cancer drug for CCA.
Collapse
Affiliation(s)
- Sarunya Kitdumrongthum
- Toxicology Graduate Program, Faculty of Science, Mahidol University, Bangkok, Thailand; Excellent Center for Drug Discovery (ECDD), Mahidol University, Bangkok, Thailand
| | - Somrudee Reabroi
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Kanoknetr Suksen
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Patoomratana Tuchinda
- Excellent Center for Drug Discovery (ECDD), Mahidol University, Bangkok, Thailand; Department of Chemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Bamroong Munyoo
- Excellent Center for Drug Discovery (ECDD), Mahidol University, Bangkok, Thailand; Department of Chemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Panupong Mahalapbutr
- Structural and Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Thanyada Rungrotmongkol
- Structural and Computational Biology Research Unit, Department of Biochemistry, Faculty of Science, Chulalongkorn University, Bangkok, Thailand; Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Puey Ounjai
- Department of Biology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Arthit Chairoungdua
- Toxicology Graduate Program, Faculty of Science, Mahidol University, Bangkok, Thailand; Excellent Center for Drug Discovery (ECDD), Mahidol University, Bangkok, Thailand; Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
14
|
Graidist P, Tedasen A, Khoka A, Madla S, Sriwiriyajan S. Anticancer effects of piperine-free Piper nigrum extract on cholangiocarcinoma cell lines. Pharmacogn Mag 2020. [DOI: 10.4103/pm.pm_288_19] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
15
|
Fabrication and characterization of andrographolide analogue (3A.1) nanosuspensions stabilized by amphiphilic chitosan derivatives for colorectal cancer therapy. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101287] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
16
|
Li H, Long J, Xie F, Kang K, Shi Y, Xu W, Wu X, Lin J, Xu H, Du S, Xu Y, Zhao H, Zheng Y, Gu J. Transcriptomic analysis and identification of prognostic biomarkers in cholangiocarcinoma. Oncol Rep 2019; 42:1833-1842. [PMID: 31545466 PMCID: PMC6787946 DOI: 10.3892/or.2019.7318] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 08/20/2019] [Indexed: 12/27/2022] Open
Abstract
Cholangiocarcinoma (CCA) is acknowledged as the second most commonly diagnosed primary liver tumor and is associated with a poor patient prognosis. The present study aimed to explore the biological functions, signaling pathways and potential prognostic biomarkers involved in CCA through transcriptomic analysis. Based on the transcriptomic dataset of CCA from The Cancer Genome Atlas (TCGA), differentially expressed protein-coding genes (DEGs) were identified. Biological function enrichment analysis, including Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, was applied. Through protein-protein interaction (PPI) network analysis, hub genes were identified and further verified using open-access datasets and qRT-PCR. Finally, a survival analysis was conducted. A total of 1,463 DEGs were distinguished, including 267 upregulated genes and 1,196 downregulated genes. For the GO analysis, the upregulated DEGs were enriched in ‘cadherin binding in cell-cell adhesion’, ‘extracellular matrix (ECM) organization’ and ‘cell-cell adherens junctions’. Correspondingly, the downregulated DEGs were enriched in the ‘oxidation-reduction process’, ‘extracellular exosomes’ and ‘blood microparticles’. In regards to the KEGG pathway analysis, the upregulated DEGs were enriched in ‘ECM-receptor interactions’, ‘focal adhesions’ and ‘small cell lung cancer’. The downregulated DEGs were enriched in ‘metabolic pathways’, ‘complement and coagulation cascades’ and ‘biosynthesis of antibiotics’. The PPI network suggested that CDK1 and another 20 genes were hub genes. Furthermore, survival analysis suggested that CDK1, MKI67, TOP2A and PRC1 were significantly associated with patient prognosis. These results enhance the current understanding of CCA development and provide new insight into distinguishing candidate biomarkers for predicting the prognosis of CCA.
Collapse
Affiliation(s)
- Hanyu Li
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Junyu Long
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Fucun Xie
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Kai Kang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Yue Shi
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Weiyu Xu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Xiaoqian Wu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Jianzhen Lin
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Haifeng Xu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Shunda Du
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Yiyao Xu
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Haitao Zhao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Yongchang Zheng
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Jin Gu
- MOE Key Laboratory of Bioinformatics, BNIRST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, P.R. China
| |
Collapse
|
17
|
Kandanur SGS, Tamang N, Golakoti NR, Nanduri S. Andrographolide: A natural product template for the generation of structurally and biologically diverse diterpenes. Eur J Med Chem 2019; 176:513-533. [DOI: 10.1016/j.ejmech.2019.05.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 04/22/2019] [Accepted: 05/06/2019] [Indexed: 01/11/2023]
|
18
|
Pearngam P, Kumkate S, Okada S, Janvilisri T. Andrographolide Inhibits Cholangiocarcinoma Cell Migration by Down-Regulation of Claudin-1 via the p-38 Signaling Pathway. Front Pharmacol 2019; 10:827. [PMID: 31404237 PMCID: PMC6669233 DOI: 10.3389/fphar.2019.00827] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 06/27/2019] [Indexed: 12/13/2022] Open
Abstract
Andrographolide, a bioactive phytochemical from Andrographis paniculata, is emerging as a promising anticancer agent against various cancers. This study aims to investigate anticancer activities of andrographolide against cholangiocarcinoma (CCA) and to understand the underlying mechanism. The anti-proliferative activity of andrographolide was evaluated in a range of cholangiocarcinoma (CCA) cell lines including HuCCA-1, KKU-100, KKU-M213, and RMCCA-1. The anti-migration activity and the corresponding mechanism were studied in highly metastatic KKU-M213 cells. The results indicated that andrographolide significantly inhibited the proliferation of CCA cells with the 50% inhibitory growth concentration (IC50) of ∼120 µM. Andrographolide also inhibited CCA cell migration and invasion. Our further explorations demonstrated that andrographolide decreased the expression of claudin-1, a major tight junction protein, while it up-regulated the expression of Snail, a transcriptional repressor of claudin-1. Moreover, andrographolide induced the phosphorylation of Jun N-terminus kinase (JNK) and p-38 Mitogen-activated protein kinase (MAPK). Treatment with the p-38-specific inhibitor recovered the claudin-1 expression and migration ability of CCA cells. This work demonstrated the potential anticancer effects of andrographolide, indicating that andrographolide could inhibit CCA cell migration via suppression of claudin-1 through the activation of p-38 MAPK signaling pathway. This compound would be useful for development of alternative therapeutic agent for CCA.
Collapse
Affiliation(s)
- Phorutai Pearngam
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Supeecha Kumkate
- Department of Biology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection & Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tavan Janvilisri
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand
| |
Collapse
|
19
|
Wang Y, Wang Y, Wang S, Tong Y, Jin L, Zong H, Zheng R, Yang J, Zhang Z, Ouyang E, Zhou M, Zhang X. GIDB: a knowledge database for the automated curation and multidimensional analysis of molecular signatures in gastrointestinal cancer. DATABASE-THE JOURNAL OF BIOLOGICAL DATABASES AND CURATION 2019; 2019:5487627. [PMID: 31089686 PMCID: PMC6517830 DOI: 10.1093/database/baz051] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 01/08/2019] [Accepted: 03/29/2019] [Indexed: 01/12/2023]
Abstract
Gastrointestinal (GI) cancer is common, characterized by high mortality, and includes oesophagus, gastric, liver, bile duct, pancreas, rectal and colon cancers. The insufficient specificity and sensitivity of biomarkers is still a key clinical hindrance for GI cancer diagnosis and successful treatment. The emergence of `precision medicine', `basket trial' and `field cancerization' concepts calls for an urgent need and importance for the understanding of how organ system cancers occur at the molecular levels. Knowledge from both the literature and data available in public databases is informative in elucidating the molecular alterations underlying GI cancer. Currently, most available cancer databases have not offered a comprehensive discovery of gene-disease associations, molecular alterations and clinical information by integrated text mining and data mining in GI cancer. We develop GIDB, a panoptic knowledge database that attempts to automate the curation of molecular signatures using natural language processing approaches and multidimensional analyses. GIDB covers information on 8730 genes with both literature and data supporting evidence, 248 miRNAs, 58 lncRNAs, 320 copy number variations, 49 fusion genes and 2381 semantic networks. It presents a comprehensive database, not only in parallelizing supporting evidence and data integration for signatures associated with GI cancer but also in providing the timeline feature of major molecular discoveries. It highlights the most comprehensive overview, research hotspots and the development of historical knowledge of genes in GI cancer. Furthermore, GIDB characterizes genomic abnormalities in multilevel analysis, including simple somatic mutations, gene expression, DNA methylation and prognosis. GIDB offers a user-friendly interface and two customizable online tools (Heatmap and Network) for experimental researchers and clinicians to explore data and help them shorten the learning curve and broaden the scope of knowledge. More importantly, GIDB is an ongoing research project that will continue to be updated and improve the automated method for reducing manual work.
Collapse
Affiliation(s)
- Ying Wang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Department of Laboratory Medicine, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Yueqian Wang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Shuangkuai Wang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yuantao Tong
- Research Center for Translational Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Ling Jin
- Research Center for Translational Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Hui Zong
- Research Center for Translational Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Rongbin Zheng
- Research Center for Translational Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jinxuan Yang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zeyu Zhang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - En Ouyang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Mengyan Zhou
- Research Center for Translational Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiaoyan Zhang
- Research Center for Translational Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
20
|
Kansom T, Sajomsang W, Saeeng R, Charoensuksai P, Opanasopit P, Tonglairoum P. Apoptosis Induction and Antimigratory Activity of Andrographolide Analog (3A.1)-Incorporated Self-Assembled Nanoparticles in Cancer Cells. AAPS PharmSciTech 2018; 19:3123-3133. [PMID: 30117042 DOI: 10.1208/s12249-018-1139-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 07/31/2018] [Indexed: 11/30/2022] Open
Abstract
Andrographolide analog, namely 19-tert-butyldiphenylsilyl-8,17-epoxy andrographolide (or 3A.1) has been reported to be a potential anticancer agent for several types of cancer. Due to its poor aqueous solubility, 3A.1 was incorporated within self-assembly polymeric nanoparticles made of naphthyl-grafted succinyl chitosan (NSC), octyl-grafted succinyl chitosan (OSC), and benzyl-grafted succinyl chitosan (BSC). These 3A.1-loaded nanoparticles were nanosized (< 200 nm) and spherical in shape with a negative surface charge. 3A.1-loaded nanoparticles were produced using a dropping method, which 40% initial drug adding exhibited the highest entrapment efficiency. The release of 3A.1 from the 3A.1-loaded nanoparticles displayed a delayed release pattern. Under acidic conditions (pH 1.2), there was no free drug release. After the pH was adjusted to 6.8, a high cumulative 3A.1 release was obtained which was dependent on the hydrophobic moieties. These 3A.1-loaded pH-sensitive nanoparticles proved to be beneficial for specifically delivering anticancer drugs to the targeted colon cancer sites. In vitro anticancer activity against HT-29 found that the 3A.1-loaded nanoparticles had significantly lower IC50 than that of the free drug and promoted apoptosis. Additionally, in vitro wound-healing migration on HN-22 revealed that free 3A.1 and the 3A.1-loaded nanoparticles inhibited cell motility compared with untreated cells. These pH-sensitive amphiphilic chitosan nanoparticles may be promising nanocarriers for oral anticancer drug delivery to colorectal cancer cells. Graphical abstract ᅟ.
Collapse
|
21
|
Reabroi S, Chairoungdua A, Saeeng R, Kasemsuk T, Saengsawang W, Zhu W, Piyachaturawat P. A silyl andrographolide analogue suppresses Wnt/β-catenin signaling pathway in colon cancer. Biomed Pharmacother 2018; 101:414-421. [DOI: 10.1016/j.biopha.2018.02.119] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 02/16/2018] [Accepted: 02/23/2018] [Indexed: 11/16/2022] Open
|
22
|
Islam MT, Ali ES, Uddin SJ, Islam MA, Shaw S, Khan IN, Saravi SSS, Ahmad S, Rehman S, Gupta VK, Găman MA, Găman AM, Yele S, Das AK, de Castro E Sousa JM, de Moura Dantas SMM, Rolim HML, de Carvalho Melo-Cavalcante AA, Mubarak MS, Yarla NS, Shilpi JA, Mishra SK, Atanasov AG, Kamal MA. Andrographolide, a diterpene lactone from Andrographis paniculata and its therapeutic promises in cancer. Cancer Lett 2018; 420:129-145. [PMID: 29408515 DOI: 10.1016/j.canlet.2018.01.074] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2017] [Revised: 01/26/2018] [Accepted: 01/29/2018] [Indexed: 12/17/2022]
Abstract
The diterpene lactone andrographolide, isolated from Andrographis paniculata, has been proven to possess several important protective biological activities, including antioxidant, anti-inflammatory, immunomodulatory, antiseptic, antimicrobial, cytotoxic, hypolipidemic, cardioprotective, hepatoprotective, and neuroprotective effects. In addition, it has been reported to play a therapeutic role in the treatment of major human diseases, such as Parkinson's disease, rheumatoid arthritis, and colitis. This systematic review aims to highlight andrographolide as a promising agent in cancer treatment. To this purpose, a number of databases were used to search for the cytotoxic/anticancer effects of andrographolide in pre-clinical and clinical studies. Among 1703 identified literature articles, 139 were included in this review; 109 were investigated as non-clinical, whereas 24, 3, and 3 were pre-clinical, clinical, and non-pre-clinical trials, respectively. Among the model systems, cultured cell lines appeared as the most frequently (79.14%) used, followed by in vivo models using rodents, among others. Furthermore, andrographolide was found to exert cytotoxic/anticancer effects on almost all types of cell lines with the underlying mechanisms involving oxidative stress, cell cycle arrest, anti-inflammatory and immune system mediated effects, apoptosis, necrosis, autophagy, inhibition of cell adhesion, proliferation, migration, invasion, anti-angiogenic activity, and other miscellaneous actions. After careful consideration of the relevant evidence, we suggest that andrographolide can be one of the potential agents in the treatment of cancer in the near future.
Collapse
Affiliation(s)
- Muhammad Torequl Islam
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, 700000, Vietnam; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, 700000, Vietnam; Department of Pharmacy, Ranada Prasad Shaha University, Narayanganj, 1400, Bangladesh
| | - Eunüs S Ali
- Gaco Pharmaceuticals and Research Laboratory, Dhaka, 1000, Bangladesh; College of Medicine and Public Health, Flinders University, Bedford Park, Adelaide, 5042, Australia
| | - Shaikh Jamal Uddin
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, 9208, Bangladesh
| | - Md Amirul Islam
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, 9208, Bangladesh
| | - Subrata Shaw
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA, 02142, USA
| | - Ishaq N Khan
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, 25100, Pakistan
| | - Seyed Soheil Saeedi Saravi
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, USA; Department of Toxicology-Pharmacology, Faculty of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
| | - Saheem Ahmad
- Department of Bio-Sciences, Integral University, Lucknow, U.P., 226026, India
| | - Shahnawaz Rehman
- Department of Bio-Sciences, Integral University, Lucknow, U.P., 226026, India
| | - Vijai Kumar Gupta
- Department of Chemistry and Biotechnology, Tallinn University of Technology, 12618, Tallinn, Estonia
| | - Mihnea-Alexandru Găman
- "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania; Facoltà di Medicina e Chirurgia, Università degli Studi di Bari "Aldo Moro", Bari, Italy
| | - Amelia Maria Găman
- Department of Pathophysiology, Research Center of Experimental and Clinical Medicine, University of Medicine and Pharmacy of Craiova, Romania; Department of Haematology, Filantropia City Hospital of Craiova, Craiova, Romania
| | - Santosh Yele
- School of Pharmacy and Technology Management, SVKM's NMIMS, Shirpur, India
| | - Asish Kumar Das
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, 9208, Bangladesh
| | | | | | - Hercília Maria Lins Rolim
- Laboratory of Pharmaceutical Nanosystems (NANOSFAR), Postgraduate Pharmaceutical Sciences, Federal University of Piauí, Teresina, Piauí, Brazil
| | | | | | - Nagendra Sastry Yarla
- Department of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad, 500003, T.N., India
| | - Jamil A Shilpi
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, 9208, Bangladesh
| | - Siddhartha Kumar Mishra
- Cancer Biology Laboratory, School of Biological Sciences (Zoology), Dr. Harisingh Gour Central University, Sagar, 470003, M.P., India
| | - Atanas G Atanasov
- Institute of Genetics and Animal Breeding of the Polish Academy of Sciences, Jastrzebiec, Poland; Department of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Althanstrasse 14, A-1090, Vienna, Austria.
| | - Mohammad Amjad Kamal
- King Fahd Medical Research Center, King Abdulaziz University, Saudi Arabia; Enzymoics, 7 Peterlee Place, Hebersham, NSW, 2770, Australia; Novel Global Community Educational Foundation, Australia.
| |
Collapse
|
23
|
de Almeida SMV, Ribeiro AG, de Lima Silva GC, Ferreira Alves JE, Beltrão EIC, de Oliveira JF, de Carvalho LB, Alves de Lima MDC. DNA binding and Topoisomerase inhibition: How can these mechanisms be explored to design more specific anticancer agents? Biomed Pharmacother 2017; 96:1538-1556. [DOI: 10.1016/j.biopha.2017.11.054] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 11/07/2017] [Accepted: 11/07/2017] [Indexed: 12/11/2022] Open
|
24
|
Monger A, Boonmuen N, Suksen K, Saeeng R, Kasemsuk T, Piyachaturawat P, Saengsawang W, Chairoungdua A. Inhibition of Topoisomerase IIα and Induction of Apoptosis in Gastric Cancer Cells by 19-Triisopropyl Andrographolide. Asian Pac J Cancer Prev 2017; 18:2845-2851. [PMID: 29072435 PMCID: PMC5747413 DOI: 10.22034/apjcp.2017.18.10.2845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Gastric cancer is the most common cancer in Eastern Asia. Increasing chemoresistance and general systemic
toxicities have complicated the current chemotherapy leading to an urgent need of more effective agents. The present
study reported a potent DNA topoisomerase IIα inhibitory activity of an andrographolide analogue (19-triisopropyl
andrographolide, analogue-6) in gastric cancer cells; MKN-45, and AGS cells. The analogue was potently cytotoxic to
both gastric cancer cell lines with the half maximal inhibitory concentration (IC50 values) of 6.3±0.7 μM, and 1.7±0.05
μM at 48 h for MKN-45, and AGS cells, respectively. It was more potent than the parent andrographolide and the
clinically used, etoposide with the IC50 values of >50 μM in MKN-45 and 11.3±2.9 μM in AGS cells for andrographolide
and 28.5±4.4 μM in MKN-45 and 4.08±0.5 μM in AGS cells for etoposide. Analogue-6 at 2 μM significantly inhibited
DNA topoisomerase IIα enzyme in AGS cells, induced DNA damage, activated cleaved PARP-1, and Caspase3 leading
to late cellular apoptosis. Interestingly, the expression of tumor suppressor p53 was not activated. These results show
the importance of 19-triisopropyl-andrographolide in its emerging selectivity to primary target on topoisomerase IIα
enzyme, inducing DNA damage and apoptosis by p53- independent mechanism. Thereby, the results provide insights of
the potential of 19-triisopropyl andrographolide as an anticancer agent for gastric cancer. The chemical transformation
of andrographolide is a promising strategy in drug discovery of a novel class of anticancer drugs from bioactive natural
products.
Collapse
Affiliation(s)
- Adeep Monger
- Toxicology Graduate Program, Faculty of Science, Mahidol University, Bangkok 10400, Thailand.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Kasemsuk T, Piyachaturawat P, Bunthawong R, Sirion U, Suksen K, Suksamrarn A, Saeeng R. One-pot three steps cascade synthesis of novel isoandrographolide analogues and their cytotoxic activity. Eur J Med Chem 2017; 138:952-963. [DOI: 10.1016/j.ejmech.2017.07.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 07/18/2017] [Accepted: 07/20/2017] [Indexed: 01/20/2023]
|
26
|
de Oliveira JF, Lima TS, Vendramini-Costa DB, de Lacerda Pedrosa SCB, Lafayette EA, da Silva RMF, de Almeida SMV, de Moura RO, Ruiz ALTG, de Carvalho JE, de Lima MDCA. Thiosemicarbazones and 4-thiazolidinones indole-based derivatives: Synthesis, evaluation of antiproliferative activity, cell death mechanisms and topoisomerase inhibition assay. Eur J Med Chem 2017; 136:305-314. [DOI: 10.1016/j.ejmech.2017.05.023] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 05/04/2017] [Accepted: 05/06/2017] [Indexed: 12/13/2022]
|
27
|
In vivo inhibitory activity of andrographolide derivative ADN-9 against liver cancer and its mechanisms involved in inhibition of tumor angiogenesis. Toxicol Appl Pharmacol 2017; 327:1-12. [DOI: 10.1016/j.taap.2017.04.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/15/2017] [Accepted: 04/20/2017] [Indexed: 02/07/2023]
|
28
|
Lim SC, Jeon HJ, Kee KH, Lee MJ, Hong R, Han SI. Andrographolide induces apoptotic and non-apoptotic death and enhances tumor necrosis factor-related apoptosis-inducing ligand-mediated apoptosis in gastric cancer cells. Oncol Lett 2017; 13:3837-3844. [PMID: 28529596 PMCID: PMC5431559 DOI: 10.3892/ol.2017.5923] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 02/27/2017] [Indexed: 02/07/2023] Open
Abstract
Andrographolide, a natural compound isolated from Andrographis paniculata, has been reported to possess antitumor activity. In the present study, the effect of andrographolide in human gastric cancer (GC) cells was investigated. Andrographolide induced cell death with apoptotic and non-apoptotic features. At a low concentration, andrographolide potentiated apoptosis and reduction of clonogenicity triggered by recombinant human tumor necrosis factor-related apoptosis-inducing ligand (rhTRAIL). Exposure of GC cells to andrographolide altered the expression level of several growth-inhibiting and apoptosis-regulating proteins, including death receptors. It was demonstrated that activity of the TRAIL-R2 (DR5) pathway was critical in the development of andrographolide-mediated rhTRAIL sensitization, since its inhibition significantly reduced the extent of apoptosis induced by the combination of rhTRAIL and andrographolide. In addition, andrographolide increased reactive oxygen species (ROS) generation in a dose-dependent manner. N-acetyl cysteine prevented andrographolide-mediated DR5 induction and the apoptotic effect induced by the combination of rhTRAIL and andrographolide. Collectively, the present study demonstrated that andrographolide enhances TRAIL-induced apoptosis through induction of DR5 expression. This effect appears to involve ROS generation in GCs.
Collapse
Affiliation(s)
- Sung-Chul Lim
- Department of Pathology, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea
| | - Ho Jong Jeon
- Department of Pathology, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea
| | - Keun Hong Kee
- Department of Pathology, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea
| | - Mi Ja Lee
- Department of Pathology, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea
| | - Ran Hong
- Department of Pathology, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea
| | - Song Iy Han
- Division of Premedical Science, College of Medicine, Chosun University, Gwangju 61452, Republic of Korea
| |
Collapse
|
29
|
Aromdee C. Andrographolide: progression in its modifications and applications - a patent review (2012 - 2014). Expert Opin Ther Pat 2015; 24:1129-38. [PMID: 25231887 DOI: 10.1517/13543776.2014.956084] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Extraction, isolation and modifications of andrographolide (Androg) is extensively investigated and patented. The prominent activities were vastly modified for anticancer and antivirals. Many products related to Androg are commercially available, thus the section 'Interaction of Androg and Andrographis paniculata dried extract with drugs' is included. AREAS COVERED The data in this review are searched and selected from SciFinder and Espacenet for the patents, with the keywords: Andrographolide and Andrographolide analogs, and the results were refined by the years. EXPERT OPINION Modifications of Androg have been done to nearly all of the possible sites, and now screening tests for any new activities had been settled down. Categorizing the analogs that have been developed is not clear cut since some diseases can develop into others, for example, inflammation and some viral infections can develop into cancer. Currently, investigation of the mode of action and the mechanisms at the molecular level are intensively ongoing. Producing new chemotherapeutic agents from Androg looks promising. The main problem of using Androg in therapeutic applications is its insolubility in aqueous media. Those modified analogs' esters, ethers or salts, have to be considered for the stability of pharmaceutical preparations, and transformation in biological fluids after administration. Further stages of drug development are required for those promising analogs.
Collapse
Affiliation(s)
- Chantana Aromdee
- Khon Kaen University, Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences , 123 Mitraparp Rd, Muang, Khon Kaen, 40002 , Thailand +66 043 362095 ; +66 043 202379 ;
| |
Collapse
|
30
|
CAO BO, CHEN HONG, GAO YING, NIU CONG, ZHANG YUAN, LI LING. CIP-36, a novel topoisomerase II-targeting agent, induces the apoptosis of multidrug-resistant cancer cells in vitro. Int J Mol Med 2014; 35:771-6. [DOI: 10.3892/ijmm.2015.2068] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Accepted: 01/08/2015] [Indexed: 11/05/2022] Open
|