1
|
Haciosmanoglu Aldogan E, Önsü KA, Saylan CC, Günçer B, Baday S, Bektaş M. Depolymerization of actin filaments by Cucurbitacin I through binding G-actin. Food Sci Nutr 2024; 12:881-889. [PMID: 38370084 PMCID: PMC10867458 DOI: 10.1002/fsn3.3804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/12/2023] [Accepted: 10/18/2023] [Indexed: 02/20/2024] Open
Abstract
Cucurbitacins have high economic value as they are a major source of food and have pharmacological properties. Cucurbitacin I (CuI) is a plant-derived natural tetracyclic triterpenoid compound that shows an anticancer effect via inhibiting the JAK2-STAT3 signaling pathway. The actin cytoskeleton is the most abundant protein in cells and regulates critical events through reorganization in cells. In this study, it is aimed at determining the direct effect of CuI on actin dynamics. The fluorescence profile of G-actin in the presence of CuI (1-200 nM) shifted to a higher temperature, suggesting that G-actin binds CuI and that G-actin-CuI is more thermally stable than the ligand-free form. CuI dose-dependently inhibited the polymerization of F-actin in vitro and disrupted actin filaments in endothelial cells. Docking and MD simulations suggested that CuI binds to the binding site formed by residues I136, I175, D154, and A138 that are at the interface of monomers in F-actin. The migration ability of cells treated with CuI for 24 h was significantly lower than the control group (p < .001). This study reveals the molecular mechanisms of CuI in the regulation of actin dynamics by binding G-actin. More importantly, this study indicates a novel role of CuI as an actin-targeting drug by binding directly to G-actin and may contribute to the mode of action of CuI on anticancer activities.
Collapse
Affiliation(s)
| | - Kemal Alper Önsü
- Department of Biophysics, Istanbul Faculty of MedicineIstanbul UniversityIstanbulTurkey
| | - Cemil Can Saylan
- Chair of Experimental Bioinformatics, TUM School of Life SciencesTechnical University of MunichFreisingGermany
| | - Başak Günçer
- Department of Biophysics, Istanbul Faculty of MedicineIstanbul UniversityIstanbulTurkey
| | - Sefer Baday
- Applied Informatics Department, Informatics InstituteIstanbul Technical UniversityIstanbulTurkey
| | - Muhammet Bektaş
- Department of Biophysics, Istanbul Faculty of MedicineIstanbul UniversityIstanbulTurkey
| |
Collapse
|
2
|
Grădinaru TC, Vlad A, Gilca M. Bitter Phytochemicals as Novel Candidates for Skin Disease Treatment. Curr Issues Mol Biol 2023; 46:299-326. [PMID: 38248322 PMCID: PMC10814078 DOI: 10.3390/cimb46010020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 01/23/2024] Open
Abstract
Skin diseases represent a global healthcare challenge due to their rising incidence and substantial socio-economic burden. While biological, immunological, and targeted therapies have brought a revolution in improving quality of life and survival rates for certain dermatological conditions, there remains a stringent demand for new remedies. Nature has long served as an inspiration for drug development. Recent studies have identified bitter taste receptors (TAS2Rs) in both skin cell lines and human skin. Additionally, bitter natural compounds have shown promising benefits in addressing skin aging, wound healing, inflammatory skin conditions, and even skin cancer. Thus, TAS2Rs may represent a promising target in all these processes. In this review, we summarize evidence supporting the presence of TAS2Rs in the skin and emphasize their potential as drug targets for addressing skin aging, wound healing, inflammatory skin conditions, and skin carcinogenesis. To our knowledge, this is a pioneering work in connecting information on TAS2Rs expression in skin and skin cells with the impact of bitter phytochemicals on various beneficial effects related to skin disorders.
Collapse
Affiliation(s)
- Teodora-Cristiana Grădinaru
- Department of Functional Sciences I/Biochemistry, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (T.-C.G.); (M.G.)
| | - Adelina Vlad
- Department of Functional Sciences I/Physiology, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Marilena Gilca
- Department of Functional Sciences I/Biochemistry, Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (T.-C.G.); (M.G.)
| |
Collapse
|
3
|
Chen M, Zhang M, Lu X, Li Y, Lu C. Diselenium-linked dimeric prodrug nanomedicine breaking the intracellular redox balance for triple-negative breast cancer targeted therapy. Eur J Pharm Biopharm 2023; 193:16-27. [PMID: 37865134 DOI: 10.1016/j.ejpb.2023.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 10/04/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
Triple-negative breast cancer (TNBC) has been regarded as the strongest malignancy in cases of breast cancer with a poor prognosis. The development of effective treatment strategies for TNBC has always been an urgent and unmet need. The intracellular redox balance is essential for maintaining TNBC cell malignancy. Disrupting intracellular redox balance by enlarging reactive oxygen species (ROS) generation and facilitating glutathione (GSH) depletion to amplify intracellular oxidative stress may be an alternative strategy to eliminate TNBC cells. However, inducing ROS generation and GSH depletion concurrently may be challenging. Herein, a diselenium linked-dimeric prodrug nanomedicine FA-SeSe-NPs was developed to break the intracellular redox homeostasis for TNBC targeted therapy. The dimeric prodrug was synthesized by conjugating two cucurbitacin B (CuB) molecules via one diselenium bond, which was subsequently assembled with FA-PEG-DSPE to form the final nanomedicine FA-SeSe-NPs. Using the active targeting potential of folic acid (FA), FA-SeSe-NPs could accumulate in tumor tissue with elevated levels and then be specifically internalized by cancer cells. In the high ROS and GSH conditions of TNBC cells, the diselenium bond can specifically respond to ROS to produce selenium free radicals to increase ROS and react with GSH to generate S-Se bond to deplete GSH. The released CuB further induced ROS production in TNBC cells. The diselenium bond and CuB functioned synergistically to amplify oxidative stress to kill the TNBC cells. Here, we provide a promising strategy to disrupt the intracellular redox balance of cancer cells for effective TNBC therapy.
Collapse
Affiliation(s)
- Mie Chen
- Department of Mastopathy, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Min Zhang
- Department of Mastopathy, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China
| | - Xun Lu
- School of Public Health Yale University, New Haven, CT 06510-3201, USA; Graduate School of Arts and Science, Columbia University, New York, NY 10027, USA
| | - Yongfei Li
- Department of Mastopathy, The Affiliated Hospital of Nanjing University of Chinese Medicine (Jiangsu Province Hospital of TCM), Nanjing 210029, China
| | - Cheng Lu
- Department of Mastopathy, Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing 210004, China.
| |
Collapse
|
4
|
Grudzińska M, Stachnik B, Galanty A, Sołtys A, Podolak I. Progress in Antimelanoma Research of Natural Triterpenoids and Their Derivatives: Mechanisms of Action, Bioavailability Enhancement and Structure Modifications. Molecules 2023; 28:7763. [PMID: 38067491 PMCID: PMC10707933 DOI: 10.3390/molecules28237763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/17/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Melanoma is one of the most dangerous forms of skin cancer, characterized by early metastasis and rapid development. In search for effective treatment options, much attention is given to triterpenoids of plant origin, which are considered promising drug candidates due to their well described anticancer properties and relatively low toxicity. This paper comprehensively summarizes the antimelanoma potential of natural triterpenoids, that are also used as scaffolds for the development of more effective derivatives. These include betulin, betulinic acid, ursolic acid, maslinic acid, oleanolic acid, celastrol and lupeol. Some lesser-known triterpenoids that deserve attention in this context are 22β-hydroxytingenone, cucurbitacins, geoditin A and ganoderic acids. Recently described mechanisms of action are presented, together with the results of preclinical in vitro and in vivo studies, as well as the use of drug delivery systems and pharmaceutical technologies to improve the bioavailability of triterpenoids. This paper also reviews the most promising structural modifications, based on structure-activity observations. In conclusion, triterpenoids of plant origin and some of their semi-synthetic derivatives exert significant cytotoxic, antiproliferative and chemopreventive effects that can be beneficial for melanoma treatment. Recent data indicate that their poor solubility in water, and thus low bioavailability, can be overcome by complexing with cyclodextrins, or the use of nanoparticles and ethosomes, thus making these compounds promising antimelanoma drug candidates for further development.
Collapse
Affiliation(s)
- Marta Grudzińska
- Department of Pharmacognosy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (M.G.); (B.S.); (A.S.); (I.P.)
- Department of Food Chemistry and Nutrition, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
- Doctoral School of Medical and Health Sciences, Jagiellonian University Medical College, Łazarza 16, 31-530 Kraków, Poland
| | - Bogna Stachnik
- Department of Pharmacognosy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (M.G.); (B.S.); (A.S.); (I.P.)
| | - Agnieszka Galanty
- Department of Pharmacognosy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (M.G.); (B.S.); (A.S.); (I.P.)
| | - Agnieszka Sołtys
- Department of Pharmacognosy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (M.G.); (B.S.); (A.S.); (I.P.)
| | - Irma Podolak
- Department of Pharmacognosy, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland; (M.G.); (B.S.); (A.S.); (I.P.)
| |
Collapse
|
5
|
Tsai TH, Lee KT, Hsu YC. JSI-124 Induces Cell Cycle Arrest and Regulates the Apoptosis in Glioblastoma Cells. Biomedicines 2023; 11:2999. [PMID: 38001999 PMCID: PMC10669163 DOI: 10.3390/biomedicines11112999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 10/30/2023] [Accepted: 11/04/2023] [Indexed: 11/26/2023] Open
Abstract
Cucurbitacin I (JSI-124), derived from Cucurbitaceae, has shown the potential to induce apoptosis and cell cycle arrest in some cancer cells. However, the effect of JSI-124 on glioblastoma multiforme (GBM) cell cycle and apoptosis is still unclear. Our investigation revealed that JSI-124 effectively reduced cell viability in GBM cells, leading to apoptosis and increased caspase-3 activity. Intriguingly, JSI-124 caused the accumulation of G2/M phase to regulate cell cycle, confirmed by MPM-2 staining and increased protein synthesis during mitosis by mitotic index analysis. Western blot analysis found that JSI-124 affected the progression of G2/M arrest by downregulating the CDK1 and upregulating the cyclinB1, suggesting that JSI-124 disrupted the formation and function of the cyclin B1/CDK1 complex in GBM8401 and U87MG cells. However, we found the JSI-124-regulated cell cycle G2/M and apoptosis-relative gene in GBM8401 and U87MG cells by NGS data analysis. Notably, we found that the GBM8401 and U87MG cells observed regulation of apoptosis and cell-cycle-related signaling pathways. Taken together, JSI-124 exhibited the ability to induce G2/M arrest, effectively arresting the cell cycle at critical stages. This arrest is accompanied by the initiation of apoptosis, highlighting the dual mechanism of action of JSI-124. Collectively, our findings emphasize that JSI-124 holds potential as a therapeutic agent for GBM by impeding cell cycle progression, inhibiting cell proliferation, and promoting apoptosis. As demonstrated by our in vitro experiments, these effects are mediated through modulation of key molecular targets.
Collapse
Affiliation(s)
- Tai-Hsin Tsai
- Department of Surgery, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan;
- Division of Neurosurgery, Department of Surgery, Kaohsiung Medical University Hospital, Kaohsiung 807378, Taiwan
- Graduate Institutes of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan;
- Division of Neurosurgery, Department of Surgery, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Kuan-Ting Lee
- Graduate Institutes of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807378, Taiwan;
- Division of Neurosurgery, Department of Surgery, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 80145, Taiwan
| | - Yi-Chiang Hsu
- School of Medicine, I-Shou University, Kaohsiung 82445, Taiwan
| |
Collapse
|
6
|
Kumar A, Sharma B, Sharma U, Parashar G, Parashar NC, Rani I, Ramniwas S, Kaur S, Haque S, Tuli HS. Apoptotic and antimetastatic effect of cucurbitacins in cancer: recent trends and advancement. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:1867-1878. [PMID: 37010571 DOI: 10.1007/s00210-023-02471-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 03/21/2023] [Indexed: 04/04/2023]
Abstract
The Cucurbitaceae family produces a class of secondary metabolites known as cucurbitacins. The eight cucurbitacin subunits are cucurbitacin B, D, E, I, IIa, L glucoside, Q, and R with the most significant anticancer activity. They are reported to inhibit cell proliferation, invasion, and migration; induce apoptosis; and encourage cell cycle arrest, as some of their modes of action. The JAK-STAT3, Wnt, PI3K/Akt, and MAPK signaling pathways, which are essential for the survival and apoptosis of cancer cells, have also been shown to be suppressed by cucurbitacins. The goal of the current study is to summarize potential molecular targets that cucurbitacins could inhibit in order to suppress various malignant processes. The review is noteworthy since it presents all putative molecular targets for cucurbitacins in cancer on a single podium.
Collapse
Affiliation(s)
- Ajay Kumar
- Punjab Biotechnology Incubator (PBTI), Phase VIII, Mohali, 160071, India
| | - Bunty Sharma
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India
| | - Ujjawal Sharma
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Gaurav Parashar
- Division of Biomedical and Life Sciences, School of Science, Navrachana University Vadodara, Gujrat, 391410, India
| | - Nidarshana Chaturvedi Parashar
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India
| | - Isha Rani
- Department of Biochemistry, maharishi markendashwar college of medical sciences and Research (MMCMSR), Sadopur, Ambala, 134007, India
| | - Seema Ramniwas
- University Centre for Research and Development, University Institute of Pharmaceutical Sciences, Chandigarh University, Gharuan, Mohali, 140413, India
| | - Satwinderjeet Kaur
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, 45142, Saudi Arabia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India.
| |
Collapse
|
7
|
Aimvijarn P, Payuhakrit W, Charoenchon N, Okada S, Suwannalert P. Riceberry Rice Germination and UVB Radiation Enhance Protocatechuic Acid and Vanillic Acid to Reduce Cellular Oxidative Stress and Suppress B16F10 Melanogenesis Relating to F-Actin Rearrangement. PLANTS (BASEL, SWITZERLAND) 2023; 12:484. [PMID: 36771569 PMCID: PMC9920603 DOI: 10.3390/plants12030484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/12/2023] [Accepted: 01/17/2023] [Indexed: 06/18/2023]
Abstract
Ultraviolet type B (UVB) radiation plays an important role in hyperpigmentation disorder, which induces cellular oxidative stress and causes abnormal melanin production and secretion. The stress condition plays an essential role in actin polymerization relating to F-actin rearrangement and forms dendrite to send melanin pigment to the uppermost layer of the skin. Phenolic compounds are secondary metabolites that mainly synthesize under stress conditions to protect plants from harmful environments and have been reported as effective agents in anti-oxidant and anti-melanogenesis. However, the influence of phenolic compounds on F-actin rearrangement-associated dendrite formation has not been studied so far. Hence, this study aimed to investigate the enhancing phytophenolic targets in riceberry rice (Oryza sativa L.) germination and UVB radiation (RR-GR) to suppress melanogenesis relating to F-rearrangement. As a result, the RR-GR had the potential to enhance phenolic acids such as protocatechuic and vanillic acid, which have been proven to possess anti-oxidant activity and anti-tyrosinase properties. Riceberry rice's modification showed the potential to reduce cellular oxidative stress and suppress B16F10 melanogenesis relating to F-actin rearrangement that is associated with dendrite formation.
Collapse
Affiliation(s)
- Parichaya Aimvijarn
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Witchuda Payuhakrit
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Pathobiology Information and Learning Center, Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Nisamanee Charoenchon
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Graduate School of Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Prasit Suwannalert
- Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Pathobiology Information and Learning Center, Department of Pathobiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
8
|
Dai S, Wang C, Zhao X, Ma C, Fu K, Liu Y, Peng C, Li Y. Cucurbitacin B: A review of its pharmacology, toxicity, and pharmacokinetics. Pharmacol Res 2023; 187:106587. [PMID: 36460279 DOI: 10.1016/j.phrs.2022.106587] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/18/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022]
Abstract
Cucurbitacin B (CuB, C32H46O8), the most abundant and active member of cucurbitacins, which are highly oxidized tetracyclic triterpenoids. Cucurbitacins are widely distributed in a variety of plants and mainly isolated from plants in the Cucurbitaceae family. CuB is mostly obtained from the pedicel of Cucumis melo L. Modern pharmacological studies have confirmed that CuB has a broad range of pharmacological activities, with significant therapeutic effects on a variety of diseases including inflammatory diseases, neurodegenerative diseases, diabetes mellitus, and cancers. In this study the PubMed, Web of Science, Science Direct, and China National Knowledge Infrastructure (CNKI) databases were searched from 1986 to 2022. After inclusion and exclusion criteria were applied, 98 out of 2484 articles were selected for a systematic review to comprehensively summarize the pharmacological activity, toxicity, and pharmacokinetic properties of CuB. The results showed that CuB exhibits potent anti-inflammatory, antioxidant, antiviral, hypoglycemic, hepatoprotective, neuroprotective, and anti-cancer activities mainly via regulating various signaling pathways, such as the Janus kinase/signal transducer and activator of transcription-3 (JAK/STAT3), nuclear factor erythroid 2-related factor-2/antioxidant responsive element (Nrf2/ARE), nuclear factor (NF)-κB, AMP-activated protein kinase (AMPK), mitogen-activated protein kinase (MAPK), phosphoinositide 3-kinase (PI3K)/Akt, cancerous inhibitor of protein phosphatase-2A/protein phosphatase-2A (CIP2A/PP2A), Wnt, focal adhesion kinase (FAK), Notch, and Hippo-Yes-associated protein (YAP) pathways. Studies of its toxicity and pharmacokinetic properties showed that CuB has non-specific toxicity and low bioavailability. In addition, derivatives and clinical applications of CuB are discussed in this paper.
Collapse
Affiliation(s)
- Shu Dai
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - XingTao Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ke Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yanfang Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
9
|
A comprehensive review on the botany, traditional uses, phytochemistry, pharmacology and toxicity of Anagallis arvensis (L).: A wild edible medicinal food plant. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.102328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
10
|
Li Y, He XL, Zhou LP, Huang XZ, Li S, Guan S, Li J, Zhang L. Asiatic acid alleviates liver fibrosis via multiple signaling pathways based on integrated network pharmacology and lipidomics. Eur J Pharmacol 2022; 931:175193. [PMID: 35963324 DOI: 10.1016/j.ejphar.2022.175193] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 07/30/2022] [Accepted: 08/05/2022] [Indexed: 11/30/2022]
Abstract
Liver fibrosis is characterized by the abnormal deposition of the extracellular matrix with a severe inflammatory response and/or metabolic disorder. Asiatic acid (AA), a natural compound derived from Centella asiatica, exhibited potent anti-fibrosis effects. This investigation first confirmed the anti-fibrosis effects of AA in TGF-β-LX-2 cells and CCl4-induced liver fibrosis mice, and then sought to elucidate a novel mechanism of action by integrating network pharmacology and lipidomics. Network pharmacology was used to find potential targets of AA, while lipidomics was used to identify differential metabolites between fibrosis and recovered cohorts. AA could suppress hepatic stellate cell activation in vitro and improve liver fibrosis in vivo. Network pharmacology unveiled the genes involved in pathways in cancer, peroxisome proliferators-activated receptors signaling pathway, and arachidonic acid metabolism pathway. Furthermore, five key genes were found in the both human and mouse databases, indicating that arachidonic acid metabolism was important. Changes in lyso-phosphocholine (22:5), prostaglandin F2α, and other related lipid metabolites also suggested the involvement of arachidonic acid metabolism the anti-fibrotic effect. In summary, our integrated strategies demonstrated that AA targeted multiple targets and impeded the progression of liver fibrosis by ameliorating arachidonic acid metabolism.
Collapse
Affiliation(s)
- Yong Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, PR China; Department of Pharmacology & Toxicology, Guangdong Sunshine Lake Pharma Co. Ltd, Dongguan, 523000, PR China
| | - Xu-Lin He
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, PR China
| | - Li-Ping Zhou
- Evaluation and Monitoring Center of Occupational Health, Guangzhou Twelfth People's Hospital, Guangzhou, 510006, PR China.
| | - Xiao-Zhong Huang
- Department of Pharmacology & Toxicology, Guangdong Sunshine Lake Pharma Co. Ltd, Dongguan, 523000, PR China
| | - Shan Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, PR China
| | - Su Guan
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, PR China
| | - Jing Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, PR China
| | - Lei Zhang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, PR China.
| |
Collapse
|
11
|
Cucurbitacin B Down-Regulates TNF Receptor 1 Expression and Inhibits the TNF-α-Dependent Nuclear Factor κB Signaling Pathway in Human Lung Adenocarcinoma A549 Cells. Int J Mol Sci 2022; 23:ijms23137130. [PMID: 35806134 PMCID: PMC9267118 DOI: 10.3390/ijms23137130] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/20/2022] [Accepted: 06/24/2022] [Indexed: 12/30/2022] Open
Abstract
Pro-inflammatory cytokines, such as tumor necrosis factor-α (TNF-α), induce the expression of intracellular adhesion molecule-1 (ICAM-1) by activating the nuclear factor κB (NF-κB) signaling pathway. In the present study, we found that cucurbitacin B decreased the expression of ICAM-1 in human lung adenocarcinoma A549 cells stimulated with TNF-α or interleukin-1α. We further investigated the mechanisms by which cucurbitacin B down-regulates TNF-α-induced ICAM-1 expression. Cucurbitacin B inhibited the nuclear translocation of the NF-κB subunit RelA and the phosphorylation of IκBα in A549 cells stimulated with TNF-α. Cucurbitacin B selectively down-regulated the expression of TNF receptor 1 (TNF-R1) without affecting three adaptor proteins (i.e., TRADD, RIPK1, and TRAF2). The TNF-α-converting enzyme inhibitor suppressed the down-regulation of TNF-R1 expression by cucurbitacin B. Glutathione, N-acetyl-L-cysteine, and, to a lesser extent, L-cysteine attenuated the inhibitory effects of cucurbitacin B on the TNF-α-induced expression of ICAM-1, suggesting that an α,β-unsaturated carbonyl moiety is essential for anti-inflammatory activity. The present results revealed that cucurbitacin B down-regulated the expression of TNF-R1 at the initial step in the TNF-α-dependent NF-κB signaling pathway.
Collapse
|
12
|
Ueno M, Kariya R, Sittithumcharee G, Okada S. Cucurbitacin B induces apoptosis of primary effusion lymphoma via disruption of cytoskeletal organization. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2021; 85:153545. [PMID: 33799222 DOI: 10.1016/j.phymed.2021.153545] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 02/09/2021] [Accepted: 03/05/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND Primary effusion lymphoma (PEL) is an aggressive B cell non-Hodgkin lymphoma that develops especially in AIDS patients and immunocompromised patients infected with human herpes virus-8 (HHV-8)/Kaposi's sarcoma-associated herpesvirus (KSHV). PEL has a poor prognosis in patients despite conventional chemotherapeutic treatment, and a safe and efficient therapy is required. PURPOSE To examine the effects on PEL of cucurbitacin B (CuB), a triterpene found in plants of the Cucurbitaceae family that has several anti-cancer activities. STUDY DESIGN We evaluated the anti-cancer activities of CuB in vitro and in vivo. METHODS Cell proliferation of PEL cell lines was measured by MTT assay. Cleaved caspases and signaling transduction associated proteins were analyzed by western blotting. Wright and Giemsa staining and immunofluorescence staining were carried out to observe cell morphology. Cell cycles were analyzed by flow cytometry. RT-PCR was performed to detect viral gene expressions. A xenograft mouse model was employed to evaluate the anti-cancer activity of CuB in vivo. RESULTS CuB inhibited cell proliferation of PEL cell lines (BCBL-1, BC-1, GTO and TY-1) in a dose-dependent manner (0-50 nM) and induced apoptosis of BCBL-1 cells via caspase activation in a dose- and time-dependent manner. In addition, CuB caused cell-shape disruption by inducing actin aggregation and suppressing the p-cofilin level, resulting in BCBL-1 cell arrest at the G2/M phase. In contrast, CuB showed almost no suppression of p-STAT3 and p-Akt activation, which were constitutively activated by KSHV-derived proteins. Furthermore, CuB (0.5 mg/kg) via intraperitoneal injection significantly (p < 0.05) suppressed solid tumor growth in the xenograft mouse model. CONCLUSION This study suggests that CuB is a promising agent for PEL treatment.
Collapse
Affiliation(s)
- Mikinori Ueno
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Ryusho Kariya
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Gunya Sittithumcharee
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan; Division of Hematopoiesis, Graduate School of Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan
| | - Seiji Okada
- Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan; Division of Hematopoiesis, Graduate School of Medical Sciences, Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto 860-0811, Japan.
| |
Collapse
|
13
|
Mesas C, Fuel M, Martínez R, Prados J, Melguizo C, Porres JM. In vitro evidence of the antitumor capacity of Solanaceae and Cucurbitaceae in colon cancer: A systematic review. Crit Rev Food Sci Nutr 2021; 62:6293-6314. [PMID: 33739207 DOI: 10.1080/10408398.2021.1900058] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Colon cancer is the fourth leading cause of cancer deaths around the world. Despite advances in understanding its etiology and in diagnosis and treatment, new therapeutic strategies are still required. In this sense, the Solanaceae and Cucurbitaceae families have been widely used to treat various pathologies, including cancer, for their bioactive components. The objective of this systematic review was to analyze the antitumor activity of the bioactive components present in extracts from Solanaceae and Cucurbitaceae families using different in in vitro models of colon cancer. 241 publications have been identified (published from January 2008 to January 2020) from different electronic data base. 44 articles were included, 26 of which examined the Solanaceae family. The antitumor activity exhibited by this family was due to the withanolide-type steroid compounds they harbor. 18 articles were related to the Cucurbitaceae family. This family is characterized by their production of cucurbitacin-type triterpenoid compounds and their derivatives, which confer antitumor activity. In conclusion, the different genera belonging to both families are an important source of bioactive compounds with relevant activity against colon cancer. More experimental and in vivo studies will be required to corroborate their antitumor activity and to leverage them in future clinical practice.
Collapse
Affiliation(s)
- Cristina Mesas
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain.,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Marco Fuel
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Rosario Martínez
- Department of Physiology, Institute of Nutrition and Food Technology (INyTA), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain.,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain.,Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), Granada, Spain
| | - Jesús M Porres
- Department of Physiology, Institute of Nutrition and Food Technology (INyTA), Center of Biomedical Research (CIBM), University of Granada, Granada, Spain
| |
Collapse
|
14
|
Ramezani M, Hasani M, Ramezani F, Karimi Abdolmaleki M. Cucurbitacins: A Focus on Cucurbitacin E As A Natural Product and Their Biological Activities. PHARMACEUTICAL SCIENCES 2020. [DOI: 10.34172/ps.2020.66] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
For the last years, different types of cucurbitacins have been extracted from various species of Cucurbitaceae family. For this review, all related papers were accumulated by searching electronic databases in the English language, including PubMed, Scopus, and Google Scholar. The keywords of cucurbitacin, cucumber anticancer therapy, cytotoxic effects, chemotherapy, and inhibitor effect were searched until February 2020. According to the result of this review, cucurbitacin E as a tetracyclic triterpenes compound, has been exhibited cell cycle arrest, anti-inflammatory and anticancer activities. It showed tumor proliferation prevention, induction of apoptosis or synergistically acts with other established antitumor compounds and cytokines throughout many molecular mechanisms. In a function-structure association manner, cucurbitacin E can inhibit Janus kinas2 (JAK2) phosphorylation, the signal transducer activator of transcription 3 (STAT3) and subsequently block these pathways, which seems to be the main mechanism of its activity. Future studies could target its detection in uninvestigated sources, subsequently its derivatives to improve their anticancer activity.
Collapse
Affiliation(s)
| | | | - Fatemeh Ramezani
- Physiology Research Center, Iran University of Medical Science, Tehran, Iran
| | | |
Collapse
|
15
|
Zhong Y, Xu H, Zhong Y, Zhang X, Zeng T, Li L, Xu G, Li M, Liu J, Yang T. Identification and characterization of the Cucurbitacins, a novel class of small-molecule inhibitors of Tropomyosin receptor kinase a. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 19:295. [PMID: 31694615 PMCID: PMC6836411 DOI: 10.1186/s12906-019-2709-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 10/03/2019] [Indexed: 02/07/2023]
Abstract
Background NGF-TrkA is well known to play a key role in propagating and sustaining pruritogenic signals, which form the pathology of chronic pruritus. Inhibition of NGF-TrkA is a known strategy for the treatment of pruritus. In the present paper, we describe the identification, in vitro characterization, structure–activity analysis, and inhibitory evaluation of a novel TrkA inhibitory scaffold exemplified by Cucurbitacins (Cus). Methods Cus were identified as TrkA inhibitors in a large-scale kinase library screen. To obtain structural models of Cus as TrkA inhibitors, AutoDock was used to explore their binding to TrkA. Furthermore, PC12 cell culture systems have been used to study the effects of Cus and traditional Chinese medicinal plants (Tian Gua Di and bitter gourd leaf) extracts on the kinase activity of TrkA. Results Cus block the phosphorylation of TrkA on several tyrosine sites, including Tyr490, Tyr674/675, and Tyr785, and inhibit downstream Akt and MAPK phosphorylation in response to NGF in PC12 cell model systems. Furthermore, traditional Chinese medicinal plants (Tian Gua Di and bitter gourd leaf) containing Cu extracts were shown to inhibit the phosphorylation of TrkA and Akt. These data reveal mechanisms, at least partly, of the anti-pruritus bioactivity of Cus. Conclusion Taken together, with the recent discovery of the important role of TrkA as a therapeutic target, Cus could be the basis for the design of improved TrkA kinase inhibitors, which could someday help treat pruritus.
Collapse
|
16
|
Ge W, Chen X, Han F, Liu Z, Wang T, Wang M, Chen Y, Ding Y, Zhang Q. Synthesis of Cucurbitacin B Derivatives as Potential Anti-Hepatocellular Carcinoma Agents. Molecules 2018; 23:E3345. [PMID: 30567327 PMCID: PMC6321601 DOI: 10.3390/molecules23123345] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/07/2018] [Accepted: 12/14/2018] [Indexed: 01/11/2023] Open
Abstract
Cucurbitacin B shows potent activity against tumor cells, but its high toxicity limits its application in the clinic. A series of cucurbitacin B derivatives was synthesized and evaluated for their anti-hepatocellular carcinoma (HCC) activities against the HepG-2 cell line. These compounds were also tested for their toxicity against the L-O2 normal cell line. The compound with the most potential, 10b, exhibited potent activity against the HepG-2 cell line with an IC50 value of 0.63 μM. Moreover, compound 10b showed the highest TI value (4.71), which is a 14.7-fold improvement compared to its parent compound cucurbitacin B. A preliminary molecular mechanism study of 10b indicated that 10b could inhibit P-STAT3 to induce the activation of mitochondrial apoptotic pathways. An in vivo acute toxicity study indicated that the compound 10b has preferable safety and tolerability compared with cucurbitacin B. These findings indicate that compound 10b might be considered as a lead compound for exploring effective anti-HCC drugs.
Collapse
Affiliation(s)
- Weizhi Ge
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China.
| | - Xinyi Chen
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China.
| | - Fangzhi Han
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China.
| | - Zhongquan Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China.
| | - Tianpeng Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China.
| | | | - Yue Chen
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China.
| | - Yahui Ding
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China.
| | - Quan Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300353, China.
| |
Collapse
|
17
|
Qin Y, Chen K, Gu W, Dong X, Lei R, Chang Y, Bai X, Xia S, Zeng L, Zhang J, Ma S, Li J, Li S, Xing G. Small size fullerenol nanoparticles suppress lung metastasis of breast cancer cell by disrupting actin dynamics. J Nanobiotechnology 2018; 16:54. [PMID: 29935539 PMCID: PMC6015447 DOI: 10.1186/s12951-018-0380-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 06/14/2018] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Tumor metastasis is the primary cause of mortality in cancer patients. Migratory breast cancer cells in lymphatic and blood vessels seek new sites and form metastatic colonies in the lung and bone, and then these cancer cells often wreak considerable havoc. With advances in nanotechnology, nanomaterials and nanotechnologies are widely applied in tumor therapy. In this paper, small size fullerenol nanoparticles, which are separated by isoelectric focusing electrophoresis (IFE) for discrepancy of isoelectric point (pI), are used in the study of tumor metastasis. RESULTS In this study, the commendable inhibition of tumor metastasis was uncovered by intravenous injection of purified fullerenol fraction with special surface charge and functional groups, which was separated by IFE for discrepancy of pI. By investigating the actin dynamics in several cancer cell lines, we found these small size fullerenol nanoparticles disturbed actin dynamics. Young's modulus detection and cell migration assays revealed that fullerenol lowered stiffness and restrained migration of breast cancer cells. Filopodia, the main supporting structures of actin bundles, are important for cell motility and adhesion. Scanning electron microscopy showed that fullerenol reduced the number and length of filopodia. Simultaneously, the inhibition of integrin to form clusters on filopodias, which was likely induced by reorganizing of actin cytoskeleton, impacted cancer cell adhesion and motility. CONCLUSIONS With intravenous injection of these fullerenol nanoparticles, tumor metastasis is well inhibited in vivo. The underlying mechanism most likely to be attributed to the effect of fullerenol nanoparticles on disturbing actin dynamics. With the disordered actin fiber, cell function is varied, including decreased cell stiffness, reduced filopodia formation, and inactivated integrin.
Collapse
Affiliation(s)
- Yanxia Qin
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006 China
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049 China
| | - Kui Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049 China
- University of Chinese Academy of Sciences, Beijing, 100049 People’s Republic of China
| | - Weihong Gu
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049 China
- University of Chinese Academy of Sciences, Beijing, 100049 People’s Republic of China
| | - Xinghua Dong
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049 China
- University of Chinese Academy of Sciences, Beijing, 100049 People’s Republic of China
| | - Ruihong Lei
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049 China
| | - Yanan Chang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049 China
| | - Xue Bai
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049 China
- University of Chinese Academy of Sciences, Beijing, 100049 People’s Republic of China
| | - Shibo Xia
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049 China
- University of Chinese Academy of Sciences, Beijing, 100049 People’s Republic of China
| | - Li Zeng
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049 China
| | - Jiaxin Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049 China
- University of Chinese Academy of Sciences, Beijing, 100049 People’s Republic of China
| | - Sihan Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049 China
| | - Juan Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049 China
| | - Shan Li
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006 China
| | - Gengmei Xing
- CAS Key Laboratory for Biomedical Effects of Nanomaterial & Nanosafety, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing, 100049 China
| |
Collapse
|
18
|
Sui HH, Zhou YJ, Wang H, Li L, Cao M, Huang JJ. Effects of sulindac sulfide on proliferation and apoptosis of human breast cancer cell. Oncol Lett 2018; 15:7981-7986. [PMID: 29849803 DOI: 10.3892/ol.2018.8331] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 06/02/2017] [Indexed: 11/06/2022] Open
Abstract
The present study aimed to observe the effects of sulindac sulfide on the proliferation and apoptosis of human breast cancer cells MCF-7, and to explore the potential underlying molecular mechanism. The inhibitory ratio was detected using a cell counting kit-8 assay. The changes in cell cycle distribution were assessed using flow cytometry (FCM). Furthermore, the changes in cell apoptosis rates were detected by Hoechst 33258 staining and FCM coupled with Annexin V-FITC/propidium iodide (PI) staining. In addition, the protein expression was detected using western blotting. Sulindac sulfide was able to inhibit the proliferation of breast cancer in a dose- and time-dependent manner. In addition, sulindac sulfide altered the cell cycle of breast cancer cells. The results of Hoechst 33258 staining and FCM coupled with Annexin V-FITC/PI staining demonstrated that sulindac sulfide could significantly induce the apoptosis of MCF-7 cells in a dose-dependent, and time-dependent manner. The western blot analysis demonstrated the protein expression of Bcl-2 was downregulated, and Bax and cleaved caspase-3 were upregulated. The results of the present study suggest that sulindac sulfide can inhibit the proliferation and induce the apoptosis of MCF-7 cells.
Collapse
Affiliation(s)
- He-Huan Sui
- Department of Pharmacology, Chongqing Medical University, Chongqing 400016, P.R. China.,Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yun-Jiang Zhou
- Department of Pharmacology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Hu Wang
- Department of Pharmacology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Li Li
- Department of Pharmacology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Min Cao
- Department of Pharmacology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jia-Jun Huang
- Department of Pharmacology, Chongqing Medical University, Chongqing 400016, P.R. China.,Chongqing Key Laboratory of Biochemistry and Molecular Pharmacology, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
19
|
Cucurbitacin I inhibits STAT3, but enhances STAT1 signaling in human cancer cells in vitro through disrupting actin filaments. Acta Pharmacol Sin 2018; 39:425-437. [PMID: 29119966 DOI: 10.1038/aps.2017.99] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2017] [Accepted: 07/09/2017] [Indexed: 12/11/2022] Open
Abstract
STAT1 and STAT3 are two important members of the STAT (signal transducers and activators of transcription) protein family and play opposing roles in regulating cancer cell growth. Suppressing STAT3 and/or enhancing STAT1 signaling are considered to be attractive anticancer strategies. Cucurbitacin I (CuI) isolated from the cucurbitacin family was reported to be an inhibitor of STAT3 signaling and a disruptor of actin cytoskeleton. In this study we investigated the function and mechanisms of CuI in regulating STAT signaling in human cancer cells in vitro. CuI (0.1-10 mmol/L) dose-dependently inhibited the phosphorylation of STAT3, and enhanced the phosphorylation of STAT1 in lung adenocarcinoma A549 cells possibly through disrupting actin filaments. We further demonstrated that actin filaments physically associated with JAK2 and STAT3 in A549 cells and regulated their phosphorylation through two signaling complexes, the IL-6 receptor complex and the focal adhesion complex. Actin filaments also interacted with STAT1 in A549 cells and regulated its dephosphorylation. Taken together, this study reveals the molecular mechanisms of CuI in the regulation of STAT signaling and in a possible inhibition of human cancer cell growth. More importantly, this study uncovers a novel role of actin and actin-associated signaling complexes in regulating STAT signaling.
Collapse
|
20
|
Lee PJ, Tsai TY, Chen S. Analysis of NO-suppressing activity of Strawberry Wine supplemented with ball-milled achenes. Journal of Food Science and Technology 2018; 55:1285-1294. [PMID: 29606742 DOI: 10.1007/s13197-018-3039-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 05/17/2017] [Accepted: 01/12/2018] [Indexed: 10/18/2022]
Abstract
Inflammation is generally thought to be involved in the development of several chronical diseases, therefore, phytochemicals to modulate immune responses has attracted great interests. The objective of the present study was to evaluate the potential anti-inflammatory effects of wine supplemented using ball-milled achene on modulating NO production and inducible nitric oxide synthase (iNOS) expression. Ball-milled achenes were added in strawberry must prior to fermentation, and the wine samples were then concentrated and extracted with water and/or ethanol prior to analysis. Bioactivities of wine extracts were evaluated using the cell viability assay, cell cycle measurements, NO production and iNOS expression in lipopolysaccharide (LPS)-stimulated RAW264.7 cells. Treatments of achenes supplemented strawberry wine extract up to 100 μg/mL inhibited the proliferation of LPS-stimulated RAW264.7 cell via affecting the progression of cell cycle. Moreover, no detectable cytotoxicity in RAW264.7 cells was observed. The supplemented wine extract suppressed the action of LPS and led to a decreased NO production in stimulated cells. The inhibitory effect of the wine extract on NO production was determined to be a 25-40% decrease in the level of 25-100 μg/mL, in contrast to a 10% decrease for conventional wine samples. Additionally, an alcoholic wine extract (100 μg/mL) led to a 40.31% decrease in iNOS expression in LPS-stimulated cells, which was more effective than the same dose of tocopherol. The results show that strawberry wine supplemented with ball-milled achenes causes a substantial inhibition of NO production, and this biofunction is exerted via the down-regulation of iNOS expression.
Collapse
Affiliation(s)
- Pao-Ju Lee
- 1Ph.D. Program in Nutrition and Food Sciences, Fu Jen Catholic University, New Taipei City, 24205 Taiwan
| | - Tsung-Yu Tsai
- 1Ph.D. Program in Nutrition and Food Sciences, Fu Jen Catholic University, New Taipei City, 24205 Taiwan.,2EP 305, Department of Food Science, Fu Jen Catholic University, # 510 Chungcheng Road, Hsinchuan District, New Taipei City, 24205 Taiwan
| | - Shaun Chen
- 1Ph.D. Program in Nutrition and Food Sciences, Fu Jen Catholic University, New Taipei City, 24205 Taiwan.,2EP 305, Department of Food Science, Fu Jen Catholic University, # 510 Chungcheng Road, Hsinchuan District, New Taipei City, 24205 Taiwan
| |
Collapse
|
21
|
LUO WW, ZHAO WW, LU JJ, WANG YT, CHEN XP. Cucurbitacin B suppresses metastasis mediated by reactive oxygen species (ROS) via focal adhesion kinase (FAK) in breast cancer MDA-MB-231 cells. Chin J Nat Med 2018; 16:10-19. [DOI: 10.1016/s1875-5364(18)30025-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Indexed: 12/15/2022]
|
22
|
Garg S, Kaul SC, Wadhwa R. Cucurbitacin B and cancer intervention: Chemistry, biology and mechanisms (Review). Int J Oncol 2017; 52:19-37. [PMID: 29138804 DOI: 10.3892/ijo.2017.4203] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/23/2017] [Indexed: 11/06/2022] Open
Abstract
Cancer is one of the most important healthcare matters, with the worst prognosis but the best possibilities for scientific development. It is likely to increase in the future and cause global havoc designating it as an epidemic. Cancer development requires urgent intervention. Past few decades have witnessed extensive research to challenge carcinogenesis. Treatment involving synthetic discipline is often associated with severe adverse effects, or even worsened prognosis. Accordingly, newer economic and patient friendly molecules are warranted. Many natural substances have proved their potential so far. Cucurbitacin B against cancer and other diseases has achieved towering popularity among the researchers around the world, as detailed in the below sections with summarized tables. In line with the fascinating role of cucurbitacin B against various types of cancers, through various molecular signaling pathways, it is justifiable to propose cucurbitacin B as a mainline chemotherapy before the onset and after the diagnosis of cancer.
Collapse
Affiliation(s)
- Sukant Garg
- Drug Discovery and Assets Innovation Lab, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Japan
| | - Sunil C Kaul
- Drug Discovery and Assets Innovation Lab, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Japan
| | - Renu Wadhwa
- Drug Discovery and Assets Innovation Lab, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Japan
| |
Collapse
|
23
|
Cucurbitacin B inhibits breast cancer metastasis and angiogenesis through VEGF-mediated suppression of FAK/MMP-9 signaling axis. Int J Biochem Cell Biol 2016; 77:41-56. [DOI: 10.1016/j.biocel.2016.05.014] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 04/28/2016] [Accepted: 05/17/2016] [Indexed: 12/19/2022]
|
24
|
Zhang Y, Lapidus RG, Liu P, Choi EY, Adediran S, Hussain A, Wang X, Liu X, Dan HC. Targeting IκB Kinase β/NF-κB Signaling in Human Prostate Cancer by a Novel IκB Kinase β Inhibitor CmpdA. Mol Cancer Ther 2016; 15:1504-14. [PMID: 27196761 DOI: 10.1158/1535-7163.mct-15-0999] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Accepted: 03/28/2016] [Indexed: 12/13/2022]
Abstract
NF-κB plays an important role in many types of cancer, including prostate cancer, but the role of the upstream kinase of NF-κB, IKKβ, in prostate cancer has neither been fully documented nor are there any effective IKKβ inhibitors used in clinical settings. Here, we have shown that IKKβ activity is mediated by multiple kinases including IKKα in human prostate cancer cell lines that express activated IKKβ. IHC analysis (IHC) of human prostate cancer tissue microarrays (TMA) demonstrates that phosphorylation of IKKα/β within its activation loop gradually increases in low to higher stage tumors as compared with normal tissue. The expression of cell proliferation and survival markers (Ki-67, Survivin) and epithelial-to-mesenchymal transition (EMT) markers (Slug, Snail), as well as cancer stem cell (CSC)-related transcription factors (Nanog, Sox2, Oct-4), also increase in parallel among the respective TMA samples analyzed. IKKβ, but not NF-κB, is found to regulate Nanog, which, in turn, modulates the levels of Oct4, Sox2, Snail, and Slug, indicating an essential role of IKKβ in regulating CSCs and EMT. The novel IKKβ inhibitor CmpdA inhibits constitutively activated IKKβ/NF-κB signaling, leading to induction of apoptosis and inhibition of proliferation, migration, and stemness in these cells. CmpdA also significantly inhibits tumor growth in xenografts without causing apparent in vivo toxicity. Furthermore, CmpdA and docetaxel act synergistically to inhibit proliferation of prostate cancer cells. These results indicate that IKKβ plays a pivotal role in prostate cancer, and targeting IKKβ, including in combination with docetaxel, may be a potentially useful strategy for treating advanced prostate cancer. Mol Cancer Ther; 15(7); 1504-14. ©2016 AACR.
Collapse
Affiliation(s)
- Yanting Zhang
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Rena G Lapidus
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Peiyan Liu
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Eun Yong Choi
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Samusi Adediran
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Arif Hussain
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland. Baltimore VA Medical Center, Baltimore, Maryland
| | - Xinghuan Wang
- Department of Urology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, P.R. China
| | - Xuefeng Liu
- Department of Pathology, Georgetown University Medical Center, Washington, DC
| | - Han C Dan
- Marlene and Stewart Greenebaum Cancer Center, University of Maryland School of Medicine, Baltimore, Maryland. Department of Pathology, University of Maryland School of Medicine, Baltimore, Maryland.
| |
Collapse
|
25
|
Wang L, Chen L, Yu M, Xu LH, Cheng B, Lin YS, Gu Q, He XH, Xu J. Discovering new mTOR inhibitors for cancer treatment through virtual screening methods and in vitro assays. Sci Rep 2016; 6:18987. [PMID: 26732172 PMCID: PMC4702177 DOI: 10.1038/srep18987] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2014] [Accepted: 11/27/2015] [Indexed: 12/31/2022] Open
Abstract
Mammalian target of rapamycin (mTOR) is an attractive target for new anticancer drug development. We recently developed in silico models to distinguish mTOR inhibitors and non-inhibitors. In this study, we developed an integrated strategy for identifying new mTOR inhibitors using cascaded in silico screening models. With this strategy, fifteen new mTOR kinase inhibitors including four compounds with IC50 values below 10 μM were discovered. In particular, compound 17 exhibited potent anticancer activities against four tumor cell lines, including MCF-7, HeLa, MGC-803, and C6, with IC50 values of 1.90, 2.74, 3.50 and 11.05 μM. Furthermore, cellular studies and western blot analyses revealed that 17 induces cell death via apoptosis by targeting both mTORC1 and mTORC2 within cells and arrests the cell cycle of HeLa at the G1/G0-phase. Finally, multi-nanosecond explicit solvent simulations and MM/GBSA analyses were carried out to study the inhibitory mechanisms of 13, 17, and 40 for mTOR. The potent compounds presented here are worthy of further investigation.
Collapse
Affiliation(s)
- Ling Wang
- Research Center for Drug Discovery &Institute of Human Virology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China.,Pre-Incubator for Innovative Drugs &Medicine, School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Lei Chen
- Research Center for Drug Discovery &Institute of Human Virology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Miao Yu
- Research Center for Drug Discovery &Institute of Human Virology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Li-Hui Xu
- Department of Cell Biology, Jinan University, Guangzhou, 510632, China
| | - Bao Cheng
- Research Center for Drug Discovery &Institute of Human Virology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Yong-Sheng Lin
- Research Center for Drug Discovery &Institute of Human Virology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Qiong Gu
- Research Center for Drug Discovery &Institute of Human Virology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| | - Xian-Hui He
- Department of Immunobiology, Jinan University, Guangzhou, 510632, China
| | - Jun Xu
- Research Center for Drug Discovery &Institute of Human Virology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510006, China
| |
Collapse
|
26
|
Improvement of the Antitumor Efficacy of Intratumoral Administration of Cucurbitacin Poly(Lactic-co-Glycolic Acid) Microspheres Incorporated in In Situ-Forming Sucrose Acetate Isobutyrate Depots. J Pharm Sci 2016; 105:205-11. [DOI: 10.1002/jps.24695] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 08/27/2015] [Accepted: 09/22/2015] [Indexed: 12/31/2022]
|
27
|
Sari-Hassoun M, Clement MJ, Hamdi I, Bollot G, Bauvais C, Joshi V, Toma F, Burgo A, Cailleret M, Rosales-Hernández MC, Macias Pérez ME, Chabane-Sari D, Curmi PA. Cucurbitacin I elicits the formation of actin/phospho-myosin II co-aggregates by stimulation of the RhoA/ROCK pathway and inhibition of LIM-kinase. Biochem Pharmacol 2015; 102:45-63. [PMID: 26707799 DOI: 10.1016/j.bcp.2015.12.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 12/15/2015] [Indexed: 12/11/2022]
Abstract
Cucurbitacins are cytotoxic triterpenoid sterols isolated from plants. One of their earliest cellular effect is the aggregation of actin associated with blockage of cell migration and division that eventually lead to apoptosis. We unravel here that cucurbitacin I actually induces the co-aggregation of actin with phospho-myosin II. This co-aggregation most probably results from the stimulation of the Rho/ROCK pathway and the direct inhibition of the LIMKinase. We further provide data that suggest that the formation of these co-aggregates is independent of a putative pro-oxidant status of cucurbitacin I. The results help to understand the impact of cucurbitacins on signal transduction and actin dynamics and open novel perspectives to use it as drug candidates for cancer research.
Collapse
Affiliation(s)
- Meryem Sari-Hassoun
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France; Laboratoire des Produits Naturels, LAPRONA, Université Abou Bekr Belkaid, Tlemcen 13000, Algeria
| | - Marie-Jeanne Clement
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France
| | - Imane Hamdi
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France
| | | | | | - Vandana Joshi
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France
| | - Flavio Toma
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France
| | - Andrea Burgo
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France
| | - Michel Cailleret
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France
| | - Martha Cecilia Rosales-Hernández
- Laboratorio de Biofísica y Biocatalisis, Sección de Estudios de Posgrado e Investigación de la Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, México D.F. 11340, México
| | - Martha Edith Macias Pérez
- Laboratorio de Biofísica y Biocatalisis, Sección de Estudios de Posgrado e Investigación de la Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, México D.F. 11340, México
| | - Daoudi Chabane-Sari
- Laboratoire des Produits Naturels, LAPRONA, Université Abou Bekr Belkaid, Tlemcen 13000, Algeria
| | - Patrick A Curmi
- Institut National de la Santé et de la Recherche Médicale, UMR1204, Laboratoire Structure-Activité des Biomolécules Normales et Pathologiques, Université d'Evry-Val d'Essonne, Evry 91025, France.
| |
Collapse
|
28
|
Cai Y, Fang X, He C, Li P, Xiao F, Wang Y, Chen M. Cucurbitacins: A Systematic Review of the Phytochemistry and Anticancer Activity. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2015; 43:1331-50. [PMID: 26503558 DOI: 10.1142/s0192415x15500755] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Cucurbitacins are highly oxidized tetracyclic triterpenoids that are widely present in traditional Chinese medicines (Cucurbitaceae family), possess strong anticancer activity, and are divided into 12 classes from A to T with over 200 derivatives. The eight most active cucurbitacin components against cancer are cucurbitacin B, D, E, I, IIa, L glucoside, Q, and R. Their mechanisms of action include antiproliferation, inhibition of migration and invasion, proapoptosis, and cell cycle arrest promotion. Cucurbitacins are also found to be the inhibitors of JAK-STAT3, Wnt, PI3K/Akt, and MAPK signaling pathways, which play important roles in the apoptosis and survival of cancer cells. Recently, new studies have discovered synergistic anticancer effects by using cucurbitacins together with clinically approved chemotherapeutic drugs, such as docetaxel and methotrexate. This paper provides a summary of recent research progress on the anticancer property of cucurbitacins and the various intracellular signaling pathways involved in the regulation of cancer cell proliferation, death, invasion, and migration. Therefore, cucurbitacins are a class of promising anticancer drugs to be used alone or be intergraded in current chemotherapies and radiotherapies to treat many types of cancers.
Collapse
Affiliation(s)
- Yuee Cai
- * State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P.R. China
| | - Xiefan Fang
- † Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Chengwei He
- * State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P.R. China
| | - Peng Li
- * State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P.R. China
| | - Fei Xiao
- * State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P.R. China.,‡ Department of Pharmacology, School of Medicine, Jinan University, Guangzhou 510632, P.R. China
| | - Yitao Wang
- * State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P.R. China
| | - Meiwan Chen
- * State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P.R. China
| |
Collapse
|
29
|
Yar Saglam AS, Alp E, Elmazoglu Z, Menevse S. Treatment with cucurbitacin B alone and in combination with gefitinib induces cell cycle inhibition and apoptosis via EGFR and JAK/STAT pathway in human colorectal cancer cell lines. Hum Exp Toxicol 2015; 35:526-43. [PMID: 26183715 DOI: 10.1177/0960327115595686] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The epidermal growth factor receptor (EGFR) associated with signaling pathways, such as Janus kinase (JAK)/signal transducer and activator of transcription (STAT), plays an important role in colorectal cancers (CRCs). Gefitinib (Gef) is an orally active inhibitor targeting the adenosine tri phosphate-binding domain of EGFR, and cucurbitacin B (CuB) is a selective inhibitor of JAK/STAT signaling with potent antitumor activity via suppression of STAT3 phosphorylation, but the underlying mechanism is not clear. We aimed to investigate the apoptotic and antiproliferative effects of CuB as a single agent and in combination with Gef on both HT-29 and HCT-116 cell lines. Cell proliferation, cell cycle distribution, and apoptosis were evaluated using viability assay, fluorescent microscopy, cytotoxicity assay, proliferation, DNA fragmentation, and cleaved caspase 3 levels. Real-time polymerase chain reaction and Western blot analyses were performed to determine the expression of relevant genes and proteins including antiapoptotic, proapoptotic, and cell cycle regulation. EGFR, phosphorylated EGFR (pEGFR), STAT3, and pSTAT3 proteins were evalutaed with Western blot analysis. Our results showed that, compared to CuB alone, CuB plus Gef treatment caused a significant growth and cell cycle inhibition and induced apoptosis in both cell lines. Also CuB plus Gef treatment decreased DNA synthesis rate more effectively than CuB alone. Treatment with CuB alone and in combination with Gef decreased the expression levels of B-Cell CLL/Lymphoma 2 (Bcl-2), BCL2-like 1 (BCL2L1), cyclin D1, pSTAT3, and pEGFR and increased the expression levels of Bcl-2-like protein 4, Bcl-2 homologous antagonist/killer, Bcl-2-associated death promoter, Bcl-2-like protein 11, and p27kip1 levels. Our results suggest that treatment with CuB alone and more likely in combination with Gef may be a considerable alternative therapeutic approach for CRC, at least in vitro.
Collapse
Affiliation(s)
- A S Yar Saglam
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Besevler, Ankara, Turkey
| | - E Alp
- Department of Medical Biology, Faculty of Medicine, Giresun University, Giresun, Turkey
| | - Z Elmazoglu
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Besevler, Ankara, Turkey
| | - S Menevse
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Besevler, Ankara, Turkey
| |
Collapse
|
30
|
Wang L, Li C, Lin Q, Zhang X, Pan H, Xu L, Shi Z, Ouyang D, He X. Cucurbitacin E suppresses cytokine expression in human Jurkat T cells through down-regulating the NF-κB signaling. Acta Biochim Biophys Sin (Shanghai) 2015; 47:459-65. [PMID: 25921411 DOI: 10.1093/abbs/gmv030] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2014] [Accepted: 03/08/2015] [Indexed: 12/15/2022] Open
Abstract
Cucurbitacin E (CucE), a triterpenoid isolated from Cucurbitaceae plants, has been shown to possess an anti-inflammatory or immunosuppressive activity in vitro and in vivo, yet the underlying mechanism has been incompletely understood. The aim of the present study was to explore its effect on cytokine expression and the underlying mechanism in human Jurkat T cells as a cellular model. The results showed that CucE significantly inhibited the production of interleukin-2, tumor necrosis factor-α, and interferon-γ in culture medium of cells treated with phorbol 12,13-dibutyrate (PDB) plus ionomycin (Ion). Furthermore, the mRNA levels of these cytokines in activated Jurkat T cells were also decreased upon CucE treatment, suggesting a potential modulatory effect on the critical signaling pathways for cytokine expression, including nuclear factor-κB (NF-κB) or mitogen-activated protein kinases (MAPKs). In support of its effect on the NF-κB signaling pathway, CucE decreased the phosphorylation levels of inhibitor of κB (IκB) and NF-κB/p65 in PDB + Ion-stimulated cells. Further supporting this, the nuclear translocation of NF-κB/p65 was significantly suppressed in response to PDB plus Ion stimulation in the presence of CucE. The phosphorylation of p38MAPK, c-Jun N-terminal kinase (JNK), and Erk1/2, however, was not decreased or slightly increased at some time points by CucE treatment. Collectively, these data suggest that CucE may exhibit immunosuppressive effect by attenuating critical cytokine expression through down-regulating the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Lixian Wang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China Key Laboratory of Physical and Training Adapted Control System, Guangdong Provincial Institute of Sports Science, Guangzhou 510663, China
| | - Chenguang Li
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Qiuru Lin
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xiaoyu Zhang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Hao Pan
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Lihui Xu
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Zijian Shi
- Department of Fetal Medicine, the First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Dongyun Ouyang
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Xianhui He
- Department of Immunobiology, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| |
Collapse
|
31
|
Ren G, Sha T, Guo J, Li W, Lu J, Chen X. Cucurbitacin B induces DNA damage and autophagy mediated by reactive oxygen species (ROS) in MCF-7 breast cancer cells. J Nat Med 2015; 69:522-30. [PMID: 26018422 DOI: 10.1007/s11418-015-0918-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 05/06/2015] [Indexed: 01/11/2023]
Abstract
Cucurbitacin B (Cuc B), a natural compound extracted from cucurbitaceous plants, demonstrated potent anticancer activities, while the underlying mechanisms remain unclear. We investigated the anticancer effect of Cuc B on MCF-7 breast cancer cells. Cuc B drastically decreased cell viability in a concentration-dependent manner. Cuc B treatment caused DNA damage, as shown by long tails in the comet assay and increased γH2AX protein expression. Immunofluorescence staining showed that Cuc B treatment induced nuclear γH2AX foci. Cuc B activated DNA damage pathways by phosphorylation of ATM/ATR [two large phosphatidylinositol-3-kinase-like kinase family (PIKKs) members]. Furthermore, it also induced autophagy, as evidenced by monodansylcadaverine (MDC) staining and autophagic protein expression. In addition, Cuc B treatment led to increased reactive oxygen species (ROS) formation, which was inhibited by N-acetyl-L-cysteine (NAC) pretreatment. NAC pretreatment inhibited Cuc-B-induced DNA damage and autophagy. Taken together, these results suggest that ROS-mediated Cuc-B-induced DNA damage and autophagy in MCF-7 cells, which provides new insights into the anticancer molecular mechanism of Cuc B.
Collapse
Affiliation(s)
- Guowen Ren
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | | | | | | | | | | |
Collapse
|
32
|
Zha QB, Zhang XY, Lin QR, Xu LH, Zhao GX, Pan H, Zhou D, Ouyang DY, Liu ZH, He XH. Cucurbitacin E Induces Autophagy via Downregulating mTORC1 Signaling and Upregulating AMPK Activity. PLoS One 2015; 10:e0124355. [PMID: 25970614 PMCID: PMC4430304 DOI: 10.1371/journal.pone.0124355] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 03/01/2015] [Indexed: 12/25/2022] Open
Abstract
Cucurbitacins, the natural triterpenoids possessing many biological activities, have been reported to suppress the mTORC1/p70S6K pathway and to induce autophagy. However, the correlation between such activities is largely unknown. In this study, we addressed this issue in human cancer cells in response to cucurbitacin E (CuE) treatment. Our results showed that CuE induced autophagy as evidenced by the formation of LC3-II and colocalization of punctate LC3 with the lysosomal marker LAMP2 in HeLa and MCF7 cells. However, CuE induced much lower levels of autophagy in ATG5-knocked down cells and failed to induce autophagy in DU145 cells lacking functional ATG5 expression, suggesting the dependence of CuE-induced autophagy on ATG5. Consistent with autophagy induction, mTORC1 activity (as reflected by p70S6K and ULK1S758 phosphorylation) was inhibited by CuE treatment. The suppression of mTORC1 activity was further confirmed by reduced recruitment of mTOR to the lysosome, which is the activation site of mTORC1. In contrast, CuE rapidly activated AMPK leading to increased phosphorylation of its substrates. AMPK activation contributed to CuE-induced suppression of mTORC1/p70S6K signaling and autophagy induction, since AMPK knockdown diminished these effects. Collectively, our data suggested that CuE induced autophagy in human cancer cells at least partly via downregulation of mTORC1 signaling and upregulation of AMPK activity.
Collapse
Affiliation(s)
- Qing-Bing Zha
- College of Life Science and Technology, Jinan University, Guangzhou, China
- Department of Fetal Medicine, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiao-Yu Zhang
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Qiu-Ru Lin
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Li-Hui Xu
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Gao-Xiang Zhao
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Hao Pan
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Dan Zhou
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Dong-Yun Ouyang
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Ze-Huan Liu
- College of Life Science and Technology, Jinan University, Guangzhou, China
- * E-mail: (ZL); (XH)
| | - Xian-Hui He
- College of Life Science and Technology, Jinan University, Guangzhou, China
- * E-mail: (ZL); (XH)
| |
Collapse
|
33
|
Fang S, Chen L, Yu M, Cheng B, Lin Y, Morris-Natschke SL, Lee KH, Gu Q, Xu J. Synthesis, antitumor activity, and mechanism of action of 6-acrylic phenethyl ester-2-pyranone derivatives. Org Biomol Chem 2015; 13:4714-26. [PMID: 25800703 PMCID: PMC4390547 DOI: 10.1039/c5ob00007f] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Based on the scaffolds of caffeic acid phenethyl ester (CAPE) as well as bioactive lactone-containing compounds, 6-acrylic phenethyl ester-2-pyranone derivatives were synthesized and evaluated against five tumor cell lines (HeLa, C6, MCF-7, A549, and HSC-2). Most of the new derivatives exhibited moderate to potent cytotoxic activity. Moreover, HeLa cell lines showed higher sensitivity to these compounds. In particular, compound showed potent cytotoxic activity (IC50 = 0.50-3.45 μM) against the five cell lines. Further investigation on the mechanism of action showed that induced apoptosis, arrested the cell cycle at G2/M phases in HeLa cells, and inhibited migration through disruption of the actin cytoskeleton. In addition, ADMET properties were also calculated in silico, and compound showed good ADMET properties with good absorption, low hepatotoxicity, and good solubility, and thus, could easily be bound to carrier proteins, without inhibition of CYP2D6. A structure-activity relationship (SAR) analysis indicated that compounds with ortho-substitution on the benzene ring exhibited obviously increased cytotoxic potency. This study indicated that compound is a promising compound as an antitumor agent.
Collapse
Affiliation(s)
- Sai Fang
- Research Center for Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, People's Republic of China.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Shukla S, Khan S, Kumar S, Sinha S, Farhan M, Bora HK, Maurya R, Meeran SM. Cucurbitacin B Alters the Expression of Tumor-Related Genes by Epigenetic Modifications in NSCLC and Inhibits NNK-Induced Lung Tumorigenesis. Cancer Prev Res (Phila) 2015; 8:552-62. [PMID: 25813524 DOI: 10.1158/1940-6207.capr-14-0286] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 03/22/2015] [Indexed: 11/16/2022]
Abstract
Non-small cell lung cancer (NSCLC) represents almost 85% of total diagnosed lung cancer. Studies have shown that combination of DNA methyltransferase (DNMT) and histone deacetylase (HDAC) inhibitors is effective against various cancers, including lung cancer. However, optimizing the synergistic dose regime is very difficult and involves adverse side effects. Therefore, in this study, we have shown that cucurbitacin B (CuB), a single bioactive triterpenoid compound, inhibits both DNMTs and HDACs starting at a very low dose of 60 nmol/L in NSCLC H1299 cells. The CuB-mediated inhibition of DNMTs and HDACs in H1299 cells leads to the reactivation of key tumor suppressor genes (TSG) such as CDKN1A and CDKN2A, as well as downregulation of oncogenes c-MYC and K-RAS and key tumor promoter gene (TPG), human telomerase reverse transcriptase (hTERT). The upregulation of TSGs and downregulation of TPG were consistently correlated with the alterations in their promoter methylation and histone modifications. This altered expression of TPG and TSGs is, at least in part, responsible for the inhibition of cellular proliferation and induction of cellular apoptosis in NSCLC. Furthermore, CuB treatment significantly inhibited the tumor incidence and multiplicity in 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-induced lung tumorigenesis in A/J mice, which was associated with the induction of apoptosis and inhibition of hyperproliferation in the lung tissues. Together, our study provides new insight into the CuB-mediated epigenetic alterations and its chemotherapeutic effects on lung cancer.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Blotting, Southwestern
- Blotting, Western
- Carcinogens/toxicity
- Carcinoma, Non-Small-Cell Lung/chemically induced
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/prevention & control
- Cell Cycle/drug effects
- Cell Proliferation/drug effects
- Cell Transformation, Neoplastic/drug effects
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/pathology
- Chromatin Immunoprecipitation
- DNA Methylation/drug effects
- Epigenesis, Genetic
- Female
- Gene Expression Regulation, Neoplastic/drug effects
- Genes, Neoplasm
- Histone Deacetylases/chemistry
- Humans
- Immunoenzyme Techniques
- Lung Neoplasms/chemically induced
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Lung Neoplasms/prevention & control
- Mice
- Mice, Inbred A
- Nitrosamines/toxicity
- Promoter Regions, Genetic
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- Triterpenes/pharmacology
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Samriddhi Shukla
- Laboratory of Cancer Epigenetics, Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sajid Khan
- Laboratory of Cancer Epigenetics, Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sudhir Kumar
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, India
| | - Sonam Sinha
- Laboratory of Cancer Epigenetics, Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Mohd Farhan
- Laboratory of Cancer Epigenetics, Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Himangsu K Bora
- Division of Laboratory Animals, CSIR-Central Drug Research Institute, Lucknow, India
| | - Rakesh Maurya
- Division of Medicinal and Process Chemistry, CSIR-Central Drug Research Institute, Lucknow, India. Academy of Scientific and Innovative Research (AcSIR), New Delhi, India
| | - Syed Musthapa Meeran
- Laboratory of Cancer Epigenetics, Division of Endocrinology, CSIR-Central Drug Research Institute, Lucknow, India. Academy of Scientific and Innovative Research (AcSIR), New Delhi, India.
| |
Collapse
|
35
|
Gupta P, Srivastava SK. Inhibition of Integrin-HER2 signaling by Cucurbitacin B leads to in vitro and in vivo breast tumor growth suppression. Oncotarget 2015; 5:1812-28. [PMID: 24729020 PMCID: PMC4039119 DOI: 10.18632/oncotarget.1743] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
HER2, an oncogenic receptor is overexpressed in about 25-30% of breast cancer patients. HER2 has been shown to play role in tumor promotion by having cross-talk with multiple oncogenic pathways in cancer cells. Our results show that Cucurbitacin B (CuB), a triterpenoid steroidal compound inhibited the growth of various breast cancer cells with an IC50 ranging from 18-50nM after 48 and 72 h of treatment. Our study also revealed the significant inhibitory effects of CuB on HER2 and integrin signaling in breast cancer. Notably, CuB inhibited ITGA6 and ITGB4 (integrin α6 & integrin β4), which are overexpressed in breast cancer. Furthermore, CuB also induced the expression of major ITGB1and ITGB3, which are known to cause integrin-mediated cell death. In addition, we observed that TGFβ treatment resulted in the increased association of HER2 with ITGA6 and this association was inhibited by CuB treatment. Efficacy of CuB was tested in vivo using two different orthotopic models of breast cancer. MDA-MB-231 and 4T-1 cells were injected orthotopically in the mammary fat pad of female athymic nude mice or BALB/c mice respectively. Our results showed that CuB administration inhibited MDA-MB-231 orthotopic tumors by 55%, and 4T-1 tumors by 40%. The 4T-1 cells represent stage IV breast cancer and form very aggressive tumors. CuB mediated breast tumor growth suppression was associated with the inhibition of HER2/integrin signaling. Our results suggest novel targets of CuB in breast cancer in vitro and in vivo.
Collapse
Affiliation(s)
- Parul Gupta
- Department of Biomedical Sciences and Cancer Biology Center, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | | |
Collapse
|
36
|
ZHENG QIAN, LIU YUNYI, LIU WEIWEI, MA FENGYUN, ZHOU YI, CHEN MINGJIE, CHANG JUNLI, WANG YUESHENG, YANG GUANGXIAO, HE GUANGYUAN. Cucurbitacin B inhibits growth and induces apoptosis through the JAK2/STAT3 and MAPK pathways in SH-SY5Y human neuroblastoma cells. Mol Med Rep 2014; 10:89-94. [DOI: 10.3892/mmr.2014.2175] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 03/21/2014] [Indexed: 11/05/2022] Open
|
37
|
Zhang YT, Xu LH, Lu Q, Liu KP, Liu PY, Ji F, Liu XM, Ouyang DY, He XH. VASP activation via the Gα13/RhoA/PKA pathway mediates cucurbitacin-B-induced actin aggregation and cofilin-actin rod formation. PLoS One 2014; 9:e93547. [PMID: 24691407 PMCID: PMC3972149 DOI: 10.1371/journal.pone.0093547] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2013] [Accepted: 03/06/2014] [Indexed: 11/25/2022] Open
Abstract
Cucurbitacin B (CuB), a potent antineoplastic agent of cucurbitacin triterpenoids, induces rapid disruption of actin cytoskeleton and aberrant cell cycle inhibiting carcinogenesis. However, the underlying molecular mechanism of such anticancer effects remains incompletely understood. In this study, we showed that CuB treatment rapidly induced vasodilator-stimulated phosphoprotein (VASP) phosphorylation (i.e. activation) at the Ser157 residue and generated VASP clumps which were co-localized with amorphous actin aggregates prior to the formation of highly-ordered cofilin-actin rods in melanoma cells. Knockdown of VASP or inhibition of VASP activation using PKA-specific inhibitor H89 suppressed CuB-induced VASP activation, actin aggregation and cofilin-actin rod formation. The VASP activation was mediated by cAMP-independent PKA activation as CuB decreased the levels of cAMP while MDL12330A, an inhibitor of adenylyl cyclase, had weak effect on VASP activation. Knockdown of either Gα13 or RhoA not only suppressed VASP activation, but also ameliorated CuB-induced actin aggregation and abrogated cofilin-actin rod formation. Collectively, our studies highlighted that the CuB-induced actin aggregation and cofilin-actin rod formation was mediated via the Gα13/RhoA/PKA/VASP pathway.
Collapse
Affiliation(s)
- Yan-Ting Zhang
- Department of Immunobiology, Jinan University, Guangzhou, China
| | - Li-Hui Xu
- Department of Cell Biology, Jinan University, Guangzhou, China
| | - Qun Lu
- Department of Anatomy and Cell Biology, East Carolina University Brody School of Medicine, Greenville, North Carolina, United States of America
| | - Kun-Peng Liu
- Department of Immunobiology, Jinan University, Guangzhou, China
| | - Pei-Yan Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Fang Ji
- Guangdong Entomological Institute, Guangzhou, China
| | - Xiao-Ming Liu
- Southern China Primate Research Center, Guangzhou, China
| | - Dong-Yun Ouyang
- Department of Immunobiology, Jinan University, Guangzhou, China
- * E-mail: (DO); (XH)
| | - Xian-Hui He
- Department of Immunobiology, Jinan University, Guangzhou, China
- * E-mail: (DO); (XH)
| |
Collapse
|
38
|
Cucurbitacin B induced ATM-mediated DNA damage causes G2/M cell cycle arrest in a ROS-dependent manner. PLoS One 2014; 9:e88140. [PMID: 24505404 PMCID: PMC3913755 DOI: 10.1371/journal.pone.0088140] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Accepted: 01/05/2014] [Indexed: 01/11/2023] Open
Abstract
Cucurbitacins are a class of triterpenoids widely distributed in plant kingdom with potent anti-cancer activities both in vitro and in vivo by inducing cycle arrest, autophagy, and apoptosis. Cucurbitacin B (Cuc B), could induce S or G2/M cell cycle arrest in cancer cells while the detailed mechanisms remain to be clear. This study was designed to precisely dissect the signaling pathway(s) responsible for Cuc B induced cell cycle arrest in human lung adenocarcinoma epithelial A549 cells. We demonstrated that low concentrations of Cuc B dramatically induced G2/M phase arrest in A549 cells. Cuc B treatment caused DNA double-strand breaks (DSBs) without affecting the signal transducer and activator of transcription 3 (STAT3), the potential molecular target for Cuc B. Cuc B triggers ATM-activated Chk1-Cdc25C-Cdk1, which could be reversed by both ATM siRNA and Chk1 siRNA. Cuc B also triggers ATM-activated p53-14-3-3-σ pathways, which could be reversed by ATM siRNA. Cuc B treatment also led to increased intracellular reactive oxygen species (ROS) formation, which was inhibited by N-acetyl-l-cysteine (NAC) pretreatment. Furthermore, NAC pretreatment inhibited Cuc B induced DNA damage and G2/M phase arrest. Taken together, these results suggested that Cuc B induces DNA damage in A549 cells mediated by increasing intracellular ROS formation, which lead to G2/M cell phase arrest through ATM-activated Chk1-Cdc25C-Cdk1 and p53-14-3-3-σ parallel branches. These observations provide novel mechanisms and potential targets for better understanding of the anti-cancer mechanisms of cucurbitacins.
Collapse
|
39
|
Mo H, Ouyang D, Xu L, Gao Q, He X. Human endogenous retroviral syncytin exerts inhibitory effect on invasive phenotype of B16F10 melanoma cells. Chin J Cancer Res 2013; 25:556-64. [PMID: 24255580 DOI: 10.3978/j.issn.1000-9604.2013.10.06] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2012] [Accepted: 10/31/2012] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE Fusogenic endogenous retroviral syncytin plays an important role in the formation of syncytiotrophoblasts in human placenta. Apart from its expression in placenta, brain and testis, syncytin has also been found in many cancers. Although syncytin has been proposed to serve as a positive prognostic marker in some cancers, the underlying mechanism is unclear. The aim of this study is to evaluate the effects of syncytin expression on the invasive phenotype of melanoma cells. METHODS The eukaryotic expression plasmid for syncytin-EGFP was constructed and transfected into B16F10 melanoma cells. The effect of syncytin on the invasion potential of tumor cells was evaluated in B16F10 subline cells that stably expressed syncytin-EGFP fusion protein or EGFP alone. RESULTS The B16F10 sublines that stably expressed syncytin-EGFP or EGFP alone were established respectively and confirmed by immunofluorescent and immunoblotting assay. Syncytin expression in B16F10 cells was associated with decreased cell proliferation, migration and invasion. Multinucleated giant cells that contained as many as five nuclei were induced in syncytin-expressing cells. In addition, syncytin expression did not alter the sensitivity of B16F10 cells to trichosanthin, a toxin that damages syncytiotrophoblasts more efficiently than other tissues. CONCLUSIONS These results suggest that syncytin expression in some cancers may confine their invasion potential and thus serve as a positive prognostic factor.
Collapse
Affiliation(s)
- Hongbo Mo
- Institute of Tissue Transplantation and Immunology, Jinan University, Guangzhou 510632, China
| | | | | | | | | |
Collapse
|
40
|
Zhang YT, Ouyang DY, Xu LH, Zha QB, He XH. Formation of cofilin-actin rods following cucurbitacin-B-induced actin aggregation depends on slingshot homolog 1-mediated cofilin hyperactivation. J Cell Biochem 2013; 114:2415-29. [DOI: 10.1002/jcb.24587] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 05/02/2013] [Indexed: 01/15/2023]
Affiliation(s)
- Yan-Ting Zhang
- Department of Immunobiology; Jinan University; Guangzhou; 510632; China
| | - Dong-Yun Ouyang
- Department of Immunobiology; Jinan University; Guangzhou; 510632; China
| | | | - Qing-Bing Zha
- Department of Immunobiology; Jinan University; Guangzhou; 510632; China
| | - Xian-Hui He
- Department of Immunobiology; Jinan University; Guangzhou; 510632; China
| |
Collapse
|
41
|
Jianbo G, Xue L, Hongdan Y, Zhaohui T, Xing T, Chenchen C, Jinghua X, Hui X. The Anti-Melanoma Efficiency of the Intratumoral Injection of Cucurbitacin-Loaded Sustained-Release Carriers: A PLGA Particle System. J Pharm Sci 2013; 102:2550-63. [DOI: 10.1002/jps.23604] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2013] [Revised: 04/06/2013] [Accepted: 04/23/2013] [Indexed: 11/11/2022]
|
42
|
Kausar H, Munagala R, Bansal SS, Aqil F, Vadhanam MV, Gupta RC. Cucurbitacin B potently suppresses non-small-cell lung cancer growth: Identification of intracellular thiols as critical targets. Cancer Lett 2013; 332:35-45. [DOI: 10.1016/j.canlet.2013.01.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 12/24/2012] [Accepted: 01/04/2013] [Indexed: 01/06/2023]
|
43
|
Cucurbitacin IIa induces caspase-3-dependent apoptosis and enhances autophagy in lipopolysaccharide-stimulated RAW 264.7 macrophages. Int Immunopharmacol 2013; 16:27-34. [PMID: 23541744 DOI: 10.1016/j.intimp.2013.03.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2012] [Revised: 01/19/2013] [Accepted: 03/13/2013] [Indexed: 01/22/2023]
Abstract
Cucurbitacin IIa (CuIIa), a member of cucurbitacin family, is isolated from the root of Hemsleya amabilis which has been used as an ancient remedy for bacillary dysentery and gastroenteritis. The anti-inflammatory properties of CuIIa have long been recognized but the underlying mechanism is largely unknown. In this study, we investigated the anti-inflammatory effect of CuIIa on lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophage cells. The results showed that CuIIa inhibited the proliferation and migration of RAW 264.7 cells in a dose-dependent manner. Whereas CuIIa did not cause apoptosis in unstimulated RAW 264.7 cells, it did induce a significant apoptosis in LPS-stimulated cells, which was caspase-3-dependent and associated with downregulation of survivin. Furthermore, LPS induced autophagy in RAW 264.7 cells and this effect was further enhanced by CuIIa as evidenced by increased levels of LC3-II conjugates and formation of LC3 puncta. In addition, CuIIa disrupted actin cytoskeleton via inducing actin aggregation. However, neither the synthesis of tumor necrosis factor-α, nor the activation of the mitogen-activated protein kinases and NF-κB pathways in LPS-stimulated cells was suppressed by CuIIa treatment. Collectively, these results suggested that induction of apoptosis and enhancement of autophagy contributed to the anti-inflammatory activity of CuIIa against inflammation-related diseases.
Collapse
|
44
|
Lopez-Haber C, Kazanietz MG. Cucurbitacin I inhibits Rac1 activation in breast cancer cells by a reactive oxygen species-mediated mechanism and independently of Janus tyrosine kinase 2 and P-Rex1. Mol Pharmacol 2013; 83:1141-54. [PMID: 23478800 DOI: 10.1124/mol.112.084293] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The small GTPase Rac1 has been widely implicated in mammary tumorigenesis and metastasis. Previous studies established that stimulation of ErbB receptors in breast cancer cells activates Rac1 and enhances motility via the Rac-guanine nucleotide exchange factor P-Rex1. As the Janus tyrosine kinase 2 (Jak2)/signal transducer and activator of transcription 3 (Stat3) pathway has been shown to be functionally associated with ErbB receptors, we asked if this pathway could mediate P-Rex1/Rac1 activation in response to ErbB ligands. Here we found that the anticancer agent cucurbitacin I, a Jak2 inhibitor, reduced the activation of Rac1 and motility in response to the ErbB3 ligand heregulin in breast cancer cells. However, Rac1 activation was not affected by Jak2 or Stat3 RNA interference, suggesting that the effect of cucurbitacin I occurs through a Jak2-independent mechanism. Cucurbitacin I also failed to affect the activation of P-Rex1 by heregulin. Subsequent analysis revealed that cucurbitacin I strongly activates RhoA and the Rho effector Rho kinase (ROCK) in breast cancer cells and induces the formation of stress fibers. Interestingly, disruption of the RhoA-ROCK pathway prevented the inhibitory effect of cucurbitacin I on Rac1 activation by heregulin. Lastly, we found that RhoA activation by cucurbitacin I is mediated by reactive oxygen species (ROS). The ROS scavenger N-acetyl L-cysteine and the mitochondrial antioxidant Mito-TEMPO rescued the inhibitory effect of cucurbitacin I on Rac1 activation. In conclusion, these results indicate that ErbB-driven Rac1 activation in breast cancer cells proceeds independently of the Jak2 pathway. Moreover, they established that the inhibitory effect of cucurbitacin I on Rac1 activity involves the alteration of the balance between Rho and Rac.
Collapse
Affiliation(s)
- Cynthia Lopez-Haber
- Department of Pharmacology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6160, USA
| | | |
Collapse
|
45
|
Cucurbitacin E exhibits anti-inflammatory effect in RAW 264.7 cells via suppression of NF-κB nuclear translocation. Inflamm Res 2013; 62:461-9. [DOI: 10.1007/s00011-013-0598-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 12/31/2012] [Accepted: 01/16/2013] [Indexed: 02/01/2023] Open
|
46
|
Kapoor S. Cucurbitacin B and its rapidly emerging role in the management of systemic malignancies besides lung carcinomas. Cancer Biother Radiopharm 2013; 28:359. [PMID: 23350897 DOI: 10.1089/cbr.2012.1373] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
47
|
Biological activities and potential molecular targets of cucurbitacins: a focus on cancer. Anticancer Drugs 2012; 23:777-87. [PMID: 22561419 DOI: 10.1097/cad.0b013e3283541384] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Cucurbitacin and its derivatives (cucurbitacins) are a class of highly oxidized tetracyclic triterpenoids. They are widely distributed in the plant kingdom, where they act as heterologous chemical pheromones that protect plants from external biological insults. Their bioactivities first attracted attention in the 1960s. Documented data demonstrate that cucurbitacins possess strong pharmacological properties, such as antitumor, anti-inflammatory, and hepatoprotective effects, etc. Several molecular targets for cucurbitacins have been discovered, such as fibrous-actin, signal transducer and activator of transcription 3, cyclooxygenase-2, etc. The present study summarizes the achievements of the 50 years of research on cucurbitacins. The aim was to systematically analyze their bioactivities with an emphasis on their anticancer effects. Research and development has shed new insight into the beneficial properties of these compounds.
Collapse
|
48
|
Duangmano S, Sae-lim P, Suksamrarn A, Domann FE, Patmasiriwat P. Cucurbitacin B inhibits human breast cancer cell proliferation through disruption of microtubule polymerization and nucleophosmin/B23 translocation. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 12:185. [PMID: 23062075 PMCID: PMC3527297 DOI: 10.1186/1472-6882-12-185] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 10/09/2012] [Indexed: 12/21/2022]
Abstract
Background Cucurbitacin B, an oxygenated tetracyclic triterpenoid compound extracted from the Thai medicinal plant Trichosanthes cucumerina L., has been reported to have several biological activities including anti-inflammatory, antimicrobial and anticancer. Cucurbitacin B is great of interest because of its biological activity. This agent inhibits growth of various types of human cancer cells lines. Methods In this study, we explored the novel molecular response of cucurbitacin B in human breast cancer cells, MCF-7 and MDA-MB-231. The growth inhibitory effect of cucurbitacin B on breast cancer cells was assessed by MTT assay. The effects of cucurbitacin B on microtubules morphological structure and tubulin polymerization were analyzed using immunofluorescence technique and tubulin polymerization assay kit, respectively. Proteomic analysis was used to identify the target-specific proteins that involved in cucurbitacin B treatment. Some of the differentially expressed genes and protein products were validated by real-time RT-PCR and western blot analysis. Cell cycle distributions and apoptosis were investigated using flow cytometry. Results Cucurbitacin B exhibited strong antiproliferative effects against breast cancer cells in a dose-dependent manner. We show that cucurbitacin B prominently alters the cytoskeletal network of breast cancer cells, inducing rapid morphologic changes and improper polymerization of the microtubule network. Moreover, the results of 2D-PAGE, real-time RT-PCR, and western blot analysis revealed that the expression of nucleophosmin/B23 and c-Myc decreased markedly after cucurbitacin B treatment. Immunofluorescence microscopy showed that cucurbitacin B induced translocation of nucleophosmin/B23 from the nucleolus to nucleoplasm. Treatment with cucurbitacin B resulted in cell cycle arrest at G2/M phase and the enhancement of apoptosis. Conclusions Our findings suggest that cucurbitacin B may inhibit the proliferation of human breast cancer cells through disruption of the microtubule network and down-regulation of c-Myc and nucleophosmin/B23 as well as the perturbation in nucleophosmin/B23 trafficking from the nucleolus to nucleoplasm, resulting in G2/M arrest.
Collapse
|
49
|
Zhu JS, Ouyang DY, Shi ZJ, Xu LH, Zhang YT, He XH. Cucurbitacin B induces cell cycle arrest, apoptosis and autophagy associated with G actin reduction and persistent activation of cofilin in Jurkat cells. Pharmacology 2012; 89:348-6. [PMID: 22699368 DOI: 10.1159/000338757] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Accepted: 03/21/2012] [Indexed: 01/11/2023]
Abstract
AIM The present study aimed to explore the antitumor effect and action mechanism of cucurbitacin B (CuB) on human T-cell leukemia Jurkat cells. METHODS Cell proliferation was measured by the MTS assay. Cell cycle distribution, mitochondrial membrane potential and annexin V staining were analyzed using flow cytometry. Western blotting was used to determine the levels of apoptosis- and autophagy-related proteins. RESULTS CuB inhibited the proliferation of Jurkat cells in a dose-dependent manner and induced G 2 /M phase arrest as well as formation of tetraploid cells. Accompanied with these effects, the actin dynamics was disrupted, and cofilin, a key regulator of actin dynamics, was persistently activated (dephosphorylated). Although CuB induced around 10% cells undergoing apoptosis, most of the cells were alive after CuB treatment for 24 h. Induction of autophagy was also evident by accumulation of LC3-II. CuB-induced autophagy seemed to be a prosurvival response, since suppression of CuB-induced autophagy significantly increased the activation of caspase-3. CONCLUSION Our results demonstrated that CuB exhibited antitumor activity in Jurkat cells through induction of cell cycle arrest and apoptosis which was at least partly due to the disruption of actin dynamics.
Collapse
Affiliation(s)
- Jun-Shan Zhu
- Department of Immunobiology, Jinan University, Guangzhou, PR China
| | | | | | | | | | | |
Collapse
|
50
|
Khabbazi S, Jacques R, Moyano Cardaba C, Mueller A. Janus kinase 2 and signal transducer and activator of transcription 3 activation is not essential for CCL3-, CCL5- or CCL8-induced chemotaxis. Cell Biochem Funct 2012; 31:312-8. [DOI: 10.1002/cbf.2901] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 08/28/2012] [Accepted: 08/30/2012] [Indexed: 01/08/2023]
Affiliation(s)
- S. Khabbazi
- School of Pharmacy; University of East Anglia; Norwich; UK
| | - R.O. Jacques
- School of Pharmacy; University of East Anglia; Norwich; UK
| | | | - A. Mueller
- School of Pharmacy; University of East Anglia; Norwich; UK
| |
Collapse
|