1
|
Stutz C, Fontão APGA, Silva GWDSE, Seito LN, Perdomo RT, Sampaio ALF. Betulinic Acid Acts in Synergism with Imatinib Mesylate, Triggering Apoptosis in MDR Leukemia Cells. PLANTA MEDICA 2025; 91:19-28. [PMID: 39395407 DOI: 10.1055/a-2440-4847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2024]
Abstract
Chronic myeloid leukemia (CML) is a myeloproliferative disease, characterized by the presence of the oncogene BCR-ABL. Imatinib mesylate (IMA) is the first-line treatment for CML, and some treatment resistance has been reported. Natural products are rich sources of bioactive compounds with biological effects, opening a possibility to alter cell susceptibility to drugs such as imatinib. Herein, we evaluated the interference of betulinic acid and ursolic acid in glycoprotein P (P-gp) activity and the possible synergistic effect when associated with IMA by the Chou-Talalay method. Ursolic acid presented an IC50 of 14.0 µM and 19.6 µM for K562 and Lucena 1, respectively, whilst betulinic acid presented an IC50 of 8.6 µM and 12.5 µM for these cell lines. Evaluation of the combination of terpenoids and imatinib mesylate revealed that ursolic acid or betulinic acid acts in synergism with IMA, as indicated by the combination indexes (CI<1). Analysis of annexin V labeling demonstrated that a combination of IMA with betulinic acid enhances the inhibition on cell proliferation via the apoptosis pathway, with caspases 3/7 activation after 24 hours of treatment and inhibition of the STAT5/survivin pathway, decreasing cell viability. The combination of natural products and IMA on a multidrug-resistant leukemia cell line is a promising strategy for CML treatment.
Collapse
Affiliation(s)
- Claudia Stutz
- Fundação Oswaldo Cruz, Eusébio, CE, Brasil
- Fundação Oswaldo Cruz, Campo Grande, MS, Brasil
| | | | | | - Leonardo Noboru Seito
- Laboratório de Farmacologia Aplicada, Instituto de Tecnologia em Fármacos; Fiocruz, Rio de Janeiro, RJ, Brasil
| | - Renata Trentin Perdomo
- Laboratório de Biologia Molecular e Culturas Celulares, Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição; UFMS, Campo Grande, MS, Brasil
| | - André Luiz Franco Sampaio
- Laboratório de Farmacologia Molecular, Instituto de Tecnologia em Fármacos; Fiocruz, Rio de Janeiro, RJ, Brasil
| |
Collapse
|
2
|
Iweala EJ, Oluwapelumi AE, Dania OE, Ugbogu EA. Bioactive Phytoconstituents and Their Therapeutic Potentials in the Treatment of Haematological Cancers: A Review. Life (Basel) 2023; 13:1422. [PMID: 37511797 PMCID: PMC10381774 DOI: 10.3390/life13071422] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/12/2023] [Accepted: 02/15/2023] [Indexed: 07/30/2023] Open
Abstract
Haematological (blood) cancers are the cancers of the blood and lymphoid forming tissues which represents approximately 10% of all cancers. It has been reported that approximately 60% of all blood cancers are incurable. Despite substantial improvement in access to detection/diagnosis, chemotherapy and bone marrow transplantation, there is still high recurrence and unpredictable but clearly defined relapses indicating that effective therapies are still lacking. Over the past two decades, medicinal plants and their biologically active compounds are being used as potential remedies and alternative therapies for the treatment of cancer. This is due to their anti-oxidant, anti-inflammatory, anti-mutagenic, anti-angiogenic, anti-cancer activities and negligible side effects. These bioactive compounds have the capacity to reduce proliferation of haematological cancers via various mechanisms such as promoting apoptosis, transcription regulation, inhibition of signalling pathways, downregulating receptors and blocking cell cycle. This review study highlights the mechanistic and beneficial effects of nine bioactive compounds (quercetin, ursolic acid, fisetin, resveratrol, epigallocatechin gallate, curcumin, gambogic acid, butein and celastrol) as potential remedies for chemoprevention of haematological cancers. The study provides useful insights on the effectiveness of the use of bioactive compounds from plants for chemoprevention of haematological cancers.
Collapse
Affiliation(s)
- Emeka J Iweala
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota PMB 1023, Ogun State, Nigeria
- Covenant Applied Informatics and Communication African Centre of Excellence (CApIC-ACE), Covenant University, Ota PMB 1023, Ogun State, Nigeria
| | - Adurosakin E Oluwapelumi
- Department of Microbiology, Ladoke Akintola University of Technology, Ogbomoso PMB 4000, Oyo State, Nigeria
| | - Omoremime E Dania
- Department of Biochemistry, College of Science and Technology, Covenant University, Ota PMB 1023, Ogun State, Nigeria
| | | |
Collapse
|
3
|
Egbuna C, Patrick-Iwuanyanwu KC, Onyeike EN, Khan J, Alshehri B. FMS-like tyrosine kinase-3 (FLT3) inhibitors with better binding affinity and ADMET properties than sorafenib and gilteritinib against acute myeloid leukemia: in silico studies. J Biomol Struct Dyn 2022; 40:12248-12259. [PMID: 34486940 DOI: 10.1080/07391102.2021.1969286] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Over 30-35% of patients down with AML are caused by mutations of FLT3-ITD and FLT3-TKD which keeps the protein activated while it activates other signaling proteins downstream that are involved in cell proliferation, differentiation, and survival. As drug targets, many inhibitors are already in clinical practice. Unfortunately, the average overall survival rate for patients on medication suffering from AML is 5 years despite the huge efforts in this field. To perform docking simulation and ADMET studies on selected phytochemicals against FLT3 protein receptor for drug discovery against FLT3 induced AML, molecular docking simulation was performed using human FLT3 protein target (PDB ID: 6JQR) and 313 phytochemicals with standard anticancer drugs (Sorafenib and Gilteritinib in addition to other anticancer drugs). The crystal structure of the protein was downloaded from the protein data bank and prepared using Biovia Discovery Studio. The chemical structures of the phytochemicals were downloaded from the NCBI PubChem database and prepared using Open Babel and VConf softwares. Molecular docking was performed using PyRx on Autodock Vina. The ADMET properties of the best performing compounds were calculated using SwissADME and pkCMS web servers. The results obtained showed that glabridin, ellipticine and derivatives (elliptinium and 9-methoxyellipticine), mezerein, ursolic acid, formononetin, cycloartocarpesin, hypericin, silymarin, and indirubin are the best performing compounds better than sorafenib and gilteritinib based on their binding affinities. The top-performing compounds which had better binding and ADMET properties than sorafenib and gilteritinib could serve as scaffolds or leads for new drug discovery against FLT3 induced AML.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Chukwuebuka Egbuna
- Africa Centre of Excellence in Public Health and Toxicological Research (ACE-PUTOR), University of Port-Harcourt, Port Harcourt, Rivers State, Nigeria.,Department of Biochemistry, Faculty of Science, University of Port Harcourt, Port Harcourt, Rivers State, Nigeria.,Department of Biochemistry, Faculty of Natural Sciences, Chukwuemeka Odumegwu Ojukwu University, Uli, Anambra State, Nigeria
| | - Kingsley C Patrick-Iwuanyanwu
- Africa Centre of Excellence in Public Health and Toxicological Research (ACE-PUTOR), University of Port-Harcourt, Port Harcourt, Rivers State, Nigeria.,Department of Biochemistry, Faculty of Science, University of Port Harcourt, Port Harcourt, Rivers State, Nigeria
| | - Eugene N Onyeike
- Africa Centre of Excellence in Public Health and Toxicological Research (ACE-PUTOR), University of Port-Harcourt, Port Harcourt, Rivers State, Nigeria.,Department of Biochemistry, Faculty of Science, University of Port Harcourt, Port Harcourt, Rivers State, Nigeria
| | - Johra Khan
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, Saudia Arabia.,Health and Basic Sciences Research Center, Majmaah University, Majmaah, Saudi Arabia
| | - Bader Alshehri
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Majmaah University, Majmaah, Saudia Arabia.,Health and Basic Sciences Research Center, Majmaah University, Majmaah, Saudi Arabia
| |
Collapse
|
4
|
Haque A, Brazeau D, Amin AR. Perspectives on natural compounds in chemoprevention and treatment of cancer: an update with new promising compounds. Eur J Cancer 2021; 149:165-183. [PMID: 33865202 PMCID: PMC8113151 DOI: 10.1016/j.ejca.2021.03.009] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/04/2021] [Accepted: 03/13/2021] [Indexed: 12/21/2022]
Abstract
Cancer is the second deadliest disease worldwide. Although recent advances applying precision treatments with targeted (molecular and immune) agents are promising, the histological and molecular heterogeneity of cancer cells and huge mutational burdens (intrinsic or acquired after therapy) leading to drug resistance and treatment failure are posing continuous challenges. These recent advances do not negate the need for alternative approaches such as chemoprevention, the pharmacological approach to reverse, suppress or prevent the initial phases of carcinogenesis or the progression of premalignant cells to invasive disease by using non-toxic agents. Although data are limited, the success of several clinical trials in preventing cancer in high-risk populations suggests that chemoprevention is a rational, appealing and viable strategy to prevent carcinogenesis. Particularly among higher-risk groups, the use of safe, non-toxic agents is the utmost consideration because these individuals have not yet developed invasive disease. Natural dietary compounds present in fruits, vegetables and spices are especially attractive for chemoprevention and treatment because of their easy availability, high margin of safety, relatively low cost and widespread human consumption. Hundreds of such compounds have been widely investigated for chemoprevention and treatment in the last few decades. Previously, we reviewed the most widely studied natural compounds and their molecular mechanisms, which were highly exploited by the cancer research community. In the time since our initial review, many promising new compounds have been identified. In this review, we critically review these promising new natural compounds, their molecular targets and mechanisms of anticancer activity that may create novel opportunities for further design and conduct of preclinical and clinical studies.
Collapse
Affiliation(s)
- Abedul Haque
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Daniel Brazeau
- Department of Pharmacy Practice, Administration and Research, School of Pharmacy, Marshall University, Huntington, WV, 25701, USA
| | - Arm R Amin
- Department of Pharmaceutical Sciences and Research, School of Pharmacy, Marshall University, Huntington, WV, 25701, USA.
| |
Collapse
|
5
|
Barreto Vianna DR, Gotardi J, Baggio Gnoatto SC, Pilger DA. Natural and Semisynthetic Pentacyclic Triterpenes for Chronic Myeloid Leukemia Therapy: Reality, Challenges and Perspectives. ChemMedChem 2021; 16:1835-1860. [PMID: 33682360 DOI: 10.1002/cmdc.202100038] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/05/2021] [Indexed: 01/11/2023]
Abstract
Chronic myeloid leukemia (CML) is a neoplasm characterized by BCR-ABL1, an oncoprotein with vital role in leukemogenesis. Its inhibition by tyrosine kinase inhibitors represents the main choice of treatment. However, therapeutic failure is worrying given the lack of pharmacological options. Pentacyclic triterpenes are phytochemicals with outstanding antitumoral properties and have also been explored as a basis for the design of potential leads. In this review, we have gathered and discuss data regarding both natural and semisynthetic pentacyclic triterpenes applied to CML cell treatment. We found consistent evidence that the class of pentacyclic triterpenes in general exerts promising pro-apoptotic and antiproliferative activities in sensitive and resistant CML cells, and thus represents a rich source for drug development. We also analyze the predicted drug-like properties of the molecules, discuss the structural changes with biological implications and show the great opportunities this class represents, as well as the perspectives they provide on drug discovery for CML treatment.
Collapse
Affiliation(s)
- Débora Renz Barreto Vianna
- Laboratory of Biochemical and Cytological Analysis, Postgraduate Program in Pharmaceutical Sciences, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul, Avenida Ipiranga, 2752 CEP, 90610-000, Porto Alegre, Brazil
| | - Jessica Gotardi
- Laboratory of Phytochemistry and Organic Synthesis, Postgraduate Program in Pharmaceutical Sciences, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul (Brazil), Avenida Ipiranga 2752, 90610-000, Porto Alegre, Brazil
| | - Simone Cristina Baggio Gnoatto
- Laboratory of Phytochemistry and Organic Synthesis, Postgraduate Program in Pharmaceutical Sciences, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul (Brazil), Avenida Ipiranga 2752, 90610-000, Porto Alegre, Brazil
| | - Diogo André Pilger
- Laboratory of Biochemical and Cytological Analysis, Postgraduate Program in Pharmaceutical Sciences, Faculty of Pharmacy, Universidade Federal do Rio Grande do Sul, Avenida Ipiranga, 2752 CEP, 90610-000, Porto Alegre, Brazil
| |
Collapse
|
6
|
Lee NR, Meng RY, Rah SY, Jin H, Ray N, Kim SH, Park BH, Kim SM. Reactive Oxygen Species-Mediated Autophagy by Ursolic Acid Inhibits Growth and Metastasis of Esophageal Cancer Cells. Int J Mol Sci 2020; 21:E9409. [PMID: 33321911 PMCID: PMC7764507 DOI: 10.3390/ijms21249409] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/30/2020] [Accepted: 12/07/2020] [Indexed: 12/13/2022] Open
Abstract
Ursolic acid (UA) possesses various pharmacological activities, such as antitumorigenic and anti-inflammatory effects. In the present study, we investigated the mechanisms underlying the effects of UA against esophageal squamous cell carcinoma (ESCC) (TE-8 cells and TE-12 cells). The cell viability assay showed that UA decreased the viability of ESCC in a dose-dependent manner. In the soft agar colony formation assay, the colony numbers and size were reduced in a dose-dependent manner after UA treatment. UA caused the accumulation of vacuoles and LC3 puncta, a marker of autophagosome, in a dose-dependent manner. Autophagy induction was confirmed by measuring the expression levels of LC3 and p62 protein in ESCC cells. UA increased LC3-II protein levels and decreased p62 levels in ESCC cells. When autophagy was hampered using 3-methyladenine (3-MA), the effect of UA on cell viability was reversed. UA also significantly inhibited protein kinase B (Akt) activation and increased p-Akt expression in a dose-dependent manner in ESCC cells. Accumulated LC3 puncta by UA was reversed after wortmannin treatment. LC3-II protein levels were also decreased after treatment with Akt inhibitor and wortmannin. Moreover, UA treatment increased cellular reactive oxygen species (ROS) levels in ESCC in a time- and dose-dependent manner. Diphenyleneiodonium (an ROS production inhibitor) blocked the ROS and UA induced accumulation of LC3-II levels in ESCC cells, suggesting that UA-induced cell death and autophagy are mediated by ROS. Therefore, our data indicate that UA inhibits the growth of ESCC cells by inducing ROS-dependent autophagy.
Collapse
Affiliation(s)
- Na-Ri Lee
- Division of Hematology and Oncology, Jeonbuk National University Medical School, Jeonju 54907, Korea;
- Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju 54907, Korea;
- Research Institute of Clinical Medicine, Biomedical Research Institute of Jeonbuk National University Medical School, Jeonju 54907, Korea
| | - Ruo Yu Meng
- Department of Physiology and Institute of Medical Science, Jeonbuk National University Medical School, Jeonju 54907, Korea; (R.Y.M.); (N.R.)
| | - So-Young Rah
- Department of Biochemistry, Jeonbuk National University Medical School, Jeonju 54907, Korea; (S.-Y.R.); (B.H.P.)
| | - Hua Jin
- School of Pharmaceutical Sciences, Tsinghua University, Beijing 100084, China;
| | - Navin Ray
- Department of Physiology and Institute of Medical Science, Jeonbuk National University Medical School, Jeonju 54907, Korea; (R.Y.M.); (N.R.)
| | - Seong-Hun Kim
- Department of Internal Medicine, Jeonbuk National University Medical School, Jeonju 54907, Korea;
| | - Byung Hyun Park
- Department of Biochemistry, Jeonbuk National University Medical School, Jeonju 54907, Korea; (S.-Y.R.); (B.H.P.)
| | - Soo Mi Kim
- Department of Physiology and Institute of Medical Science, Jeonbuk National University Medical School, Jeonju 54907, Korea; (R.Y.M.); (N.R.)
| |
Collapse
|
7
|
Guo W, Xu B, Wang X, Zheng B, Du J, Liu S. The Analysis of the Anti-Tumor Mechanism of Ursolic Acid Using Connectively Map Approach in Breast Cancer Cells Line MCF-7. Cancer Manag Res 2020; 12:3469-3476. [PMID: 32523377 PMCID: PMC7237111 DOI: 10.2147/cmar.s241957] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 04/23/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Ursolic acid (UA), a primary bioactive triterpenoid, was reported as an anti-cancer agent. However, the current knowledge of UA and its potential anti-cancer mechanisms and targets in breast cancer cells are limited. In this study, we aimed to illustrate the potential mechanisms and targets of UA in breast cancer cells MCF-7. METHODS The effect of UA on cell growth was determined in MCF-7 cells by MTT assay. The anti-tumor mechanism of UA was evaluated by microarray, CAMP, and Western blot. Moreover, the molecular docking between UA and potential receptors were predicted by iGEMDOCK software. RESULTS The result of MTT assay demonstrated that UA could inhibit MCF-7 cell growth with IC50 values of 20 μM. Microarray and CMAP analysis, validated by Western blot, indicated that UA significantly modulated IKK/NF-κB, RAF/ERK pathways, and down-regulated the phosphorylation level of PLK1 in MCF-7 cells. CONCLUSION Our data indicated that the anti-tumor effects of UA are due to the inhibited RAF/ERK pathway and IKK/NF-κB pathway. It could also be explained by the reduced phosphorylation of PLK1 in MCF-7 cells. This study provides a new insight for deep understanding of the new anti-cancer mechanisms of UA in MCF-7 breast cancer cells.
Collapse
Affiliation(s)
- Weiqiang Guo
- School of Chemistry, Biology and Material Engineering, Suzhou University of Science and Technology, Suzhou215009, People’s Republic of China
| | - Bin Xu
- School of Chemistry, Biology and Material Engineering, Suzhou University of Science and Technology, Suzhou215009, People’s Republic of China
| | - Xiaoxiao Wang
- Suzhou Key Laboratory for Medical Biotechnology, Suzhou Vocational Health College, Suzhou215009, People’s Republic of China
| | - Bo Zheng
- School of Chemistry, Biology and Material Engineering, Suzhou University of Science and Technology, Suzhou215009, People’s Republic of China
| | - Jiahui Du
- Suzhou Key Laboratory for Medical Biotechnology, Suzhou Vocational Health College, Suzhou215009, People’s Republic of China
| | - Songbai Liu
- Suzhou Key Laboratory for Medical Biotechnology, Suzhou Vocational Health College, Suzhou215009, People’s Republic of China
| |
Collapse
|
8
|
Lin CW, Chin HK, Lee SL, Chiu CF, Chung JG, Lin ZY, Wu CY, Liu YC, Hsiao YT, Feng CH, Bai LY, Weng JR. Ursolic acid induces apoptosis and autophagy in oral cancer cells. ENVIRONMENTAL TOXICOLOGY 2019; 34:983-991. [PMID: 31062913 DOI: 10.1002/tox.22769] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 04/19/2019] [Accepted: 04/24/2019] [Indexed: 06/09/2023]
Abstract
Oral squamous cell carcinoma (OSCC) is the fifth common cause of cancer mortality in Taiwan with high incidence and recurrence and needs new therapeutic strategies. In this study, ursolic acid (UA), a triterpenoid, was examined the antitumor potency in OSCC cells. Our results showed that UA inhibited the proliferation of OSCC cells in a dose- and time-dependent manner in both Ca922 and SCC2095 oral cancer cells. UA induced caspase-dependent apoptosis accompanied with the modulation of various biological biomarkers including downregulating Akt/mTOR/NF-κB signaling, ERK, and p38. In addition, UA inhibited angiogenesis as evidenced by abrogation of migration/invasion and blocking MMP-2 secretion in Ca922 cells. Interestingly, UA induced autophagy in OSCC cells, as manifested by LC3B-II conversion and increased p62 expression and accumulation of autophagosomes. Inhibition by autophagy inhibitor enhanced UA-mediated apoptosis in Ca922 cells. The experiment provides a rationale for using triterpenoid in the treatment of OSCC.
Collapse
Affiliation(s)
- Cheng-Wen Lin
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Wufeng, Taichung, Taiwan
| | - Hsien-Kuo Chin
- Division of Cardiovascular Surgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Shou-Lun Lee
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chang-Fang Chiu
- College of Medicine, China Medical University, Taichung, Taiwan
- Cancer Center, China Medical University Hospital, Taichung, Taiwan
| | - Jing-Gung Chung
- Department of Biotechnology, Asia University, Wufeng, Taichung, Taiwan
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Zi-Yin Lin
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chia-Yung Wu
- Cancer Center, China Medical University Hospital, Taichung, Taiwan
| | - Ying-Chen Liu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Yung-Ting Hsiao
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chia-Hsien Feng
- Department of Fragrance and Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Li-Yuan Bai
- College of Medicine, China Medical University, Taichung, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Jing-Ru Weng
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
9
|
Zhang T, He B, Yuan H, Feng G, Chen F, Wu A, Zhang L, Lin H, Zhuo Z, Wang T. Synthesis and Antitumor Evaluation in Vitro of NO-Donating Ursolic Acid-Benzylidene Derivatives. Chem Biodivers 2019; 16:e1900111. [PMID: 30977577 DOI: 10.1002/cbdv.201900111] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 04/11/2019] [Indexed: 12/20/2022]
Abstract
Antitumor activity of triterpenoid and its derivatives has attracted great attention recently. Our previous efforts led to the discovery of a series of NO-donor betulin derivatives with potent antitumor activity. Herein, we prepared eight compounds derived from ursolic acid (UA). All the compounds were evaluated for their in vitro cytotoxicity against four human cancer cell lines (HepG-2, MCF-7, HT-29 and A549). Among the compounds tested, compound 4a was found to be most active against HT-29 (IC50 =4.28 μm). Further biological assays demonstrated that compound 4a could induce cell cycle arrest at G1 phase and apoptosis in a dose-dependent manner. In addition, compound 4a was found to upregulate pro-apoptotic Bax, p53 and downregulate anti-apoptotic Bcl-2. All these results suggested that compound 4a is a potential candidate drug for the therapy of colon cancer.
Collapse
Affiliation(s)
- Te Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China
| | - Baoen He
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China
| | - Huan Yuan
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China
| | - Gaili Feng
- Research and Development Office, Yangling Chairisma Bio-Pharmaceutical Co., Ltd., Xianyang, 712100, P. R. China
| | - Fenglian Chen
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China
| | - Aizhi Wu
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China
| | - Lili Zhang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China
| | - Huiran Lin
- Laboratory Animal Management Office, Public Technology Service Platform, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Zhenjian Zhuo
- School of Chinese Medicine, The Chinese University of Hong Kong, Hong Kong, 999077, P. R. China
| | - Tao Wang
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, P. R. China
| |
Collapse
|
10
|
Kim EY, Sudini K, Singh AK, Haque M, Leaman D, Khuder S, Ahmed S. Ursolic acid facilitates apoptosis in rheumatoid arthritis synovial fibroblasts by inducing SP1-mediated Noxa expression and proteasomal degradation of Mcl-1. FASEB J 2018; 32:fj201800425R. [PMID: 29799788 PMCID: PMC6181629 DOI: 10.1096/fj.201800425r] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
Rheumatoid arthritis (RA) is characterized by hyperplastic pannus formation mediated by activated synovial fibroblasts (RASFs) that cause joint destruction. We have shown earlier that RASFs exhibit resistance to apoptosis, primarily as a result of enhanced expression of myeloid cell leukemia-1 (Mcl-1). In this study, we discovered that ursolic acid (UA), a plant-derived pentacyclic triterpenoid, selectively induces B-cell lymphoma 2 homology 3-only protein Noxa in human RASFs. We observed that UA-induced Noxa expression was followed by a consequent decrease in Mcl-1 expression in a dose-dependent manner. Subsequent evaluation of the signaling pathways showed that UA-induced Noxa is primarily mediated by the JNK pathway in human RASFs. Chromatin immunoprecipitation (IP) studies into the promoter region of Noxa indicated the role of transcription factor specificity protein 1 in JNK-mediated Noxa expression. Furthermore, the results from IP studies and proximity ligation assays indicated that UA-induced Noxa colocalizes and associates with Mcl-1 to prime it for proteasomal degradation through K48-linked ubiquitination by the selective recruitment of Mcl-1 ubiquitin ligase E3, a homologous to E6-associated protein C terminus domain-containing E3 ubiquitin ligase. These findings unveil a novel mechanism of inducing apoptosis in RASFs and a potential adjunct therapeutic strategy of regulating synovial hyperplasia in RA.-Kim, E. Y., Sudini, K., Singh, A. K., Haque, M., Leaman, D., Khuder, S., Ahmed, S. Ursolic acid facilitates apoptosis in rheumatoid arthritis synovial fibroblasts by inducing SP1-mediated Noxa expression and proteasomal degradation of Mcl-1.
Collapse
Affiliation(s)
- Eugene Y. Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington, USA
| | - Kuladeep Sudini
- Department of Pharmacology, University of Toledo, Toledo, Ohio, USA
| | - Anil K. Singh
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington, USA
| | - Mahamudul Haque
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington, USA
| | - Douglas Leaman
- Department of Pharmacology, University of Toledo, Toledo, Ohio, USA
| | - Sadik Khuder
- Department of Medicine, University of Toledo, Toledo, Ohio, USA
- Department of Public Health, University of Toledo, Toledo, Ohio, USA
| | - Salahuddin Ahmed
- Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, Spokane, Washington, USA
- Division of Rheumatology, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
11
|
Lewinska A, Adamczyk-Grochala J, Kwasniewicz E, Deregowska A, Wnuk M. Ursolic acid-mediated changes in glycolytic pathway promote cytotoxic autophagy and apoptosis in phenotypically different breast cancer cells. Apoptosis 2017; 22:800-815. [PMID: 28213701 PMCID: PMC5401707 DOI: 10.1007/s10495-017-1353-7] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Plant-derived pentacyclic triterpenotids with multiple biological activities are considered as promising candidates for cancer therapy and prevention. However, their mechanisms of action are not fully understood. In the present study, we have analyzed the effects of low dose treatment (5-20 µM) of ursolic acid (UA) and betulinic acid (BA) on breast cancer cells of different receptor status, namely MCF-7 (ER+, PR+/-, HER2-), MDA-MB-231 (ER-, PR-, HER2-) and SK-BR-3 (ER-, PR-, HER2+). UA-mediated response was more potent than BA-mediated response. Triterpenotids (5-10 µM) caused G0/G1 cell cycle arrest, an increase in p21 levels and SA-beta-galactosidase staining that was accompanied by oxidative stress and DNA damage. UA (20 µM) also diminished AKT signaling that affected glycolysis as judged by decreased levels of HK2, PKM2, ATP and lactate. UA-induced energy stress activated AMPK that resulted in cytotoxic autophagy and apoptosis. UA-mediated elevation in nitric oxide levels and ATM activation may also account for AMPK activation-mediated cytotoxic response. Moreover, UA-promoted apoptosis was associated with decreased pERK1/2 signals and the depolarization of mitochondrial membrane potential. Taken together, we have shown for the first time that UA at low micromolar range may promote its anticancer action by targeting glycolysis in phenotypically distinct breast cancer cells.
Collapse
Affiliation(s)
- Anna Lewinska
- Department of Genetics, University of Rzeszow, Werynia 502, 36-100, Kolbuszowa, Poland.
| | | | - Ewa Kwasniewicz
- Department of Genetics, University of Rzeszow, Werynia 502, 36-100, Kolbuszowa, Poland
| | - Anna Deregowska
- Department of Genetics, University of Rzeszow, Werynia 502, 36-100, Kolbuszowa, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Maciej Wnuk
- Department of Genetics, University of Rzeszow, Werynia 502, 36-100, Kolbuszowa, Poland
| |
Collapse
|