1
|
Luo J, Chen K, Nong X. Potential regulation of artesunate on bone metabolism through suppressing inflammatory infiltration in type 2 diabetes mellitus. Immunopharmacol Immunotoxicol 2025; 47:147-158. [PMID: 39762719 DOI: 10.1080/08923973.2024.2444953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 12/15/2024] [Indexed: 03/29/2025]
Abstract
OBJECTIVE Osteoimmunology is an emerging field that explores the interplay between bone and the immune system. The immune system plays a critical role in the pathogenesis of diabetes and significantly affects bone homeostasis. Artesunate, a first-line treatment for malaria, is known for its low toxicity and multifunctional properties. Increasing evidence suggests that artesunate possesses anti-inflammatory, immunoregulatory, and osteogenic effects. This review aims to explore the relationship between immune regulation and bone metabolism in type 2 diabetes (T2DM) and to investigate the potential therapeutic application of artesunate. METHODS This review systematically examines literature from PubMed/Medline, Elsevier, Web of Science, Embase, the International Diabetes Federation, and other relevant databases. RESULTS This review synthesizes evidence from multiple sources to delineate the relationship between T lymphocytes and T2DM, the regulation of T lymphocyte subsets in bone metabolism, and the effects of artesunate on both T lymphocytes and bone metabolism. Recent studies suggest a bidirectional regulatory relationship between T2DM and T lymphocytes (CD4+ T and CD8+ T) during the onset and progression of the disease, with inflammatory and anti-inflammatory cytokines serving as key mediators. T lymphocyte subsets and their cytokines play a pivotal role in regulating osteogenesis and osteoclastogenesis in pathological conditions. Furthermore, artesunate has shown promise in modulating inflammatory infiltration and bone metabolism. CONCLUSION The accumulated evidence indicates that artesunate exerts regulatory effects on bone metabolism in T2DM by influencing T lymphocyte differentiation.
Collapse
Affiliation(s)
- Jinghong Luo
- Department of Oral & Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Kun Chen
- Department of Oral & Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiaolin Nong
- Department of Oral & Maxillofacial Surgery, College of Stomatology, Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Key Laboratory of Oral and Maxillofacial Rehabilitation and Reconstruction, Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
2
|
Ghosh S, Das SK, Sinha K, Ghosh B, Sen K, Ghosh N, Sil PC. The Emerging Role of Natural Products in Cancer Treatment. Arch Toxicol 2024; 98:2353-2391. [PMID: 38795134 DOI: 10.1007/s00204-024-03786-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/08/2024] [Indexed: 05/27/2024]
Abstract
The exploration of natural products as potential agents for cancer treatment has garnered significant attention in recent years. In this comprehensive review, we delve into the diverse array of natural compounds, including alkaloids, carbohydrates, flavonoids, lignans, polyketides, saponins, tannins, and terpenoids, highlighting their emerging roles in cancer therapy. These compounds, derived from various botanical sources, exhibit a wide range of mechanisms of action, targeting critical pathways involved in cancer progression such as cell proliferation, apoptosis, angiogenesis, and metastasis. Through a meticulous examination of preclinical and clinical studies, we provide insights into the therapeutic potential of these natural products across different cancer types. Furthermore, we discuss the advantages and challenges associated with their use in cancer treatment, emphasizing the need for further research to optimize their efficacy, pharmacokinetics, and delivery methods. Overall, this review underscores the importance of natural products in advancing cancer therapeutics and paves the way for future investigations into their clinical applications.
Collapse
Affiliation(s)
- Sumit Ghosh
- Department of Zoology, Ramakrishna Mission Vidyamandira, Belur Math, Howrah, 711202, India
- Division of Molecular Medicine, Bose Institute, Kolkata, 700054, India
| | - Sanjib Kumar Das
- Department of Zoology, Jhargram Raj College, Jhargram, 721507, India
| | - Krishnendu Sinha
- Department of Zoology, Jhargram Raj College, Jhargram, 721507, India.
| | - Biswatosh Ghosh
- Department of Zoology, Bidhannagar College, Kolkata, 700064, India
| | - Koushik Sen
- Department of Zoology, Jhargram Raj College, Jhargram, 721507, India
| | - Nabanita Ghosh
- Department of Zoology, Maulana Azad College, Kolkata, 700013, India
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, Kolkata, 700054, India.
| |
Collapse
|
3
|
Ashoub MH, Razavi R, Heydaryan K, Salavati-Niasari M, Amiri M. Targeting ferroptosis for leukemia therapy: exploring novel strategies from its mechanisms and role in leukemia based on nanotechnology. Eur J Med Res 2024; 29:224. [PMID: 38594732 PMCID: PMC11003188 DOI: 10.1186/s40001-024-01822-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 03/30/2024] [Indexed: 04/11/2024] Open
Abstract
The latest findings in iron metabolism and the newly uncovered process of ferroptosis have paved the way for new potential strategies in anti-leukemia treatments. In the current project, we reviewed and summarized the current role of nanomedicine in the treatment and diagnosis of leukemia through a comparison made between traditional approaches applied in the treatment and diagnosis of leukemia via the existing investigations about the ferroptosis molecular mechanisms involved in various anti-tumor treatments. The application of nanotechnology and other novel technologies may provide a new direction in ferroptosis-driven leukemia therapies. The article explores the potential of targeting ferroptosis, a new form of regulated cell death, as a new therapeutic strategy for leukemia. It discusses the mechanisms of ferroptosis and its role in leukemia and how nanotechnology can enhance the delivery and efficacy of ferroptosis-inducing agents. The article not only highlights the promise of ferroptosis-targeted therapies and nanotechnology in revolutionizing leukemia treatment, but also calls for further research to overcome challenges and fully realize the clinical potential of this innovative approach. Finally, it discusses the challenges and opportunities in clinical applications of ferroptosis.
Collapse
Affiliation(s)
- Muhammad Hossein Ashoub
- Department of Hematology and Medical Laboratory Sciences, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Stem Cells and Regenerative Medicine Innovation Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Razieh Razavi
- Department of Chemistry, Faculty of Science, University of Jiroft, Jiroft, Iran
| | - Kamran Heydaryan
- Department of Medical Biochemical Analysis, Cihan University-Erbil, Kurdistan Region, Iraq
| | - Masoud Salavati-Niasari
- Institute of Nano Science and Nano Technology, University of Kashan, P.O. Box 87317-51167, Kashan, Iran
| | - Mahnaz Amiri
- Student Research Committee, Faculty of Allied Medicine, Kerman University of Medical Sciences, Kerman, Iran.
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Science, Kerman, Iran.
| |
Collapse
|
4
|
Terragno M, Vetrova A, Semenov O, Sayan AE, Kriajevska M, Tulchinsky E. Mesenchymal-epithelial transition and AXL inhibitor TP-0903 sensitise triple-negative breast cancer cells to the antimalarial compound, artesunate. Sci Rep 2024; 14:425. [PMID: 38172210 PMCID: PMC10764797 DOI: 10.1038/s41598-023-50710-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 12/23/2023] [Indexed: 01/05/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a difficult-to-treat, aggressive cancer type. TNBC is often associated with the cellular program of epithelial-mesenchymal transition (EMT) that confers drug resistance and metastasis. EMT and reverse mesenchymal-epithelial transition (MET) programs are regulated by several signaling pathways which converge on a group of transcription factors, EMT- TFs. Therapy approaches could rely on the EMT reversal to sensitise mesenchymal tumours to compounds effective against epithelial cancers. Here, we show that the antimalarial ROS-generating compound artesunate (ART) exhibits higher cytotoxicity in epithelial than mesenchymal breast cancer cell lines. Ectopic expression of EMT-TF ZEB1 in epithelial or ZEB1 depletion in mesenchymal cells, respectively, reduced or increased ART-generated ROS levels, DNA damage and apoptotic cell death. In epithelial cells, ZEB1 enhanced expression of superoxide dismutase 2 (SOD2) and glutathione peroxidase 8 (GPX8) implicated in ROS scavenging. Although SOD2 or GPX8 levels were unaffected in mesenchymal cells in response to ZEB1 depletion, stable ZEB1 knockdown enhanced total ROS. Receptor tyrosine kinase AXL maintains a mesenchymal phenotype and is overexpressed in TNBC. The clinically-relevant AXL inhibitor TP-0903 induced MET and synergised with ART to generate ROS, DNA damage and apoptosis in TNBC cells. TP-0903 reduced the expression of GPX8 and SOD2. Thus, TP-0903 and ZEB1 knockdown sensitised TNBC cells to ART, likely via different pathways. Synergistic interactions between TP-0903 and ART indicate that combination approaches involving these compounds can have therapeutic prospects for TNBC treatment.
Collapse
Affiliation(s)
- Mirko Terragno
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana, Kazakhstan.
| | - Anastassiya Vetrova
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana, Kazakhstan
| | - Oleg Semenov
- Regulation of Gene Expression Laboratory, Institute of Cytology RAS, Saint Petersburg, Russia
| | - A Emre Sayan
- Cancer Sciences Division, University of Southampton, Southampton, UK
| | - Marina Kriajevska
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana, Kazakhstan
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK
| | - Eugene Tulchinsky
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Astana, Kazakhstan.
- Department of Genetics and Genome Biology, University of Leicester, Leicester, UK.
| |
Collapse
|
5
|
Taheriazam A, Abad GGY, Hajimazdarany S, Imani MH, Ziaolhagh S, Zandieh MA, Bayanzadeh SD, Mirzaei S, Hamblin MR, Entezari M, Aref AR, Zarrabi A, Ertas YN, Ren J, Rajabi R, Paskeh MDA, Hashemi M, Hushmandi K. Graphene oxide nanoarchitectures in cancer biology: Nano-modulators of autophagy and apoptosis. J Control Release 2023; 354:503-522. [PMID: 36641122 DOI: 10.1016/j.jconrel.2023.01.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/08/2023] [Accepted: 01/09/2023] [Indexed: 01/16/2023]
Abstract
Nanotechnology is a growing field, with many potential biomedical applications of nanomedicine for the treatment of different diseases, particularly cancer, on the horizon. Graphene oxide (GO) nanoparticles can act as carbon-based nanocarriers with advantages such as a large surface area, good mechanical strength, and the capacity for surface modification. These nanostructures have been extensively used in cancer therapy for drug and gene delivery, photothermal therapy, overcoming chemotherapy resistance, and for imaging procedures. In the current review, we focus on the biological functions of GO nanoparticles as regulators of apoptosis and autophagy, the two major forms of programmed cell death. GO nanoparticles can either induce or inhibit autophagy in cancer cells, depending on the conditions. By stimulating autophagy, GO nanocarriers can promote the sensitivity of cancer cells to chemotherapy. However, by impairing autophagy flux, GO nanoparticles can reduce cell survival and enhance inflammation. Similarly, GO nanomaterials can increase ROS production and induce DNA damage, thereby sensitizing cancer cells to apoptosis. In vitro and in vivo experiments have investigated whether GO nanomaterials show any toxicity in major body organs, such as the brain, liver, spleen, and heart. Molecular pathways, such as ATG, MAPK, JNK, and Akt, can be regulated by GO nanomaterials, leading to effects on autophagy and apoptosis. These topics are discussed in this review to shed some lights towards the biomedical potential of GO nanoparticles and their biocompatibility, paving the way for their future application in clinical trials.
Collapse
Affiliation(s)
- Afshin Taheriazam
- Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran
| | - Ghazaleh Gholamiyan Yousef Abad
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Shima Hajimazdarany
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Hassan Imani
- Department of Clinical Science, Faculty of Veterinary Medicine, Islamic Azad University, Shahr-e kord Branch, Chaharmahal and Bakhtiari, Iran
| | - Setayesh Ziaolhagh
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Mohammad Arad Zandieh
- Department of Food Hygiene and Quality Control, Division of epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein 2028, South Africa; Radiation Biology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Maliheh Entezari
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Amir Reza Aref
- Belfer Center for Applied Cancer Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Vice President at Translational Sciences, Xsphera Biosciences Inc., 6 Tide Street, Boston, MA, 02210, USA
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkey
| | - Yavuz Nuri Ertas
- Department of Biomedical Engineering, Erciyes University, Kayseri, Turkey; ERNAM-Nanotechnology Research and Application Center, Erciyes University, Kayseri, Turkey
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Romina Rajabi
- Faculty of Veterinary Medicine, Islamic Azad University, Science and Research Branch, Tehran, Iran.
| | - Mahshid Deldar Abad Paskeh
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
6
|
Sandhu SS, Rouz SK, Kumar S, Swamy N, Deshmukh L, Hussain A, Haque S, Tuli HS. Ursolic acid: a pentacyclic triterpenoid that exhibits anticancer therapeutic potential by modulating multiple oncogenic targets. Biotechnol Genet Eng Rev 2023:1-31. [PMID: 36600517 DOI: 10.1080/02648725.2022.2162257] [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: 08/29/2022] [Accepted: 12/20/2022] [Indexed: 01/06/2023]
Abstract
The world is currently facing a global challenge against neoplastic diseases. Chemotherapy, hormonal therapy, surgery, and radiation therapy are some approaches used to treat cancer. However, these treatments are frequently causing side effects in patients, such as multidrug resistance, fever, weakness, and allergy, among others side effects. As a result, current research has focused on phytochemical compounds isolated from plants to treat deadly cancers. Plants are excellent resources of bioactive molecules, and many natural molecules have exceptional anticancer properties. They produce diverse anticancer derivatives such as alkaloids, terpenoids, flavonoids, pigments, and tannins, which have powerful anticancer activities against various cancer cell lines and animal models. Because of their safety, eco-friendly, and cost-effective nature, research communities have recently focused on various phytochemical bioactive molecules. Ursolic acid (UA) and its derivative compounds have anti-inflammatory, anticancer, apoptosis induction, anti-carcinogenic, and anti-breast cancer proliferation properties. Ursolic acid (UA) can improve the clinical management of human cancer because it inhibits cancer cell viability and proliferation, preventing tumour angiogenesis and metastatic activity. Therefore, the present article focuses on numerous bioactivities of Ursolic acid (UA), which can inhibit cancer cell production, mechanism of action, and modulation of anticancer properties via regulating various cellular processes.
Collapse
Affiliation(s)
| | - Sharareh Khorami Rouz
- School of Life Sciences, Manipal Academy of Higher Education, Dubai, United Arab Emirates
| | - Suneel Kumar
- Bio-Design Innovation Centre, Rani Durgavati University, Jabalpur, India
| | - Nitin Swamy
- Fungal Biotechnology and Invertebrate Pathology Laboratory Department of Biological Sciences, Rani Durgavati University, Jabalpur, India
| | - Loknath Deshmukh
- School of Life and Allied Science, ITM University, Raipur, India
| | - Arif Hussain
- School of Life Sciences, Manipal Academy of Higher Education, Dubai, United Arab Emirates
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
- Arabia and Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, India
| |
Collapse
|
7
|
Naliyadhara N, Kumar A, Kumar Gangwar S, Nair Devanarayanan T, Hegde M, Alqahtani MS, Abbas M, Sethi G, Kunnumakara A. Interplay of dietary antioxidants and gut microbiome in human health: What has been learnt thus far? J Funct Foods 2023. [DOI: 10.1016/j.jff.2022.105365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
|
8
|
Jung YY, Um JY, Sethi G, Ahn KS. Fangchinoline abrogates growth and survival of hepatocellular carcinoma by negative regulation of c-met/HGF and its associated downstream signaling pathways. Phytother Res 2022; 36:4542-4557. [PMID: 35867025 DOI: 10.1002/ptr.7573] [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] [Received: 01/04/2022] [Revised: 06/21/2022] [Accepted: 07/09/2022] [Indexed: 12/13/2022]
Abstract
Among all cancers, hepatocellular carcinoma (HCC) remains a lethal disease with limited treatment options. In this study, we have analyzed the possible inhibitory effects of Fangchinoline (FCN) on c-Met, a protein known to regulate the rapid phosphorylation of downstream signals, as well as mediate aberrant growth, metastasis, survival, and motility in cancer. FCN inhibited the activation of c-Met and its downstream signals PI3K, AKT, mTOR, MEK, and ERK under in vitro settings. Moreover, c-Met gene silencing lead to suppression of PI3K/AKT/mTOR and MEK/ERK signaling pathways, and induced apoptotic cell death upon exposure to FCN. In addition, FCN markedly inhibited the expression of the various oncogenic proteins such as Bcl-2/xl, survivin, IAP-1/2, cyclin D1, and COX-2. In vivo studies in HepG2 cells xenograft mouse model showed that FCN could significantly attenuate the tumor volume and weight, without affecting significant loss in the body weight. Similar to in vitro studies, expression level of c-Met and PI3K/AKT/mTOR, MEK/ERK signals was also suppressed by FCN in the tissues obtained from mice. Therefore, the novel findings of this study suggest that FCN can potentially function as a potent anticancer agent against HCC.
Collapse
Affiliation(s)
- Young Yun Jung
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Jae-Young Um
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, South Korea
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
9
|
Luo L, Xu G. Fascaplysin Induces Apoptosis and Ferroptosis, and Enhances Anti-PD-1 Immunotherapy in Non-Small Cell Lung Cancer (NSCLC) by Promoting PD-L1 Expression. Int J Mol Sci 2022; 23:13774. [PMID: 36430250 PMCID: PMC9699238 DOI: 10.3390/ijms232213774] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/25/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022] Open
Abstract
Fascaplysin is a natural product isolated from sponges with a wide range of anticancer activities. However, the mechanism of fascaplysin against NSCLC has not been clearly studied. In this study, fascaplysin was found to inhibit migration by regulating the wnt/β-catenin signaling pathway and reversing the epithelial-mesenchymal transition phenotype. Further research showed that the anti-NSCLC effect of fascaplysin was mainly through the induction of ferroptosis and apoptosis. Fascaplysin-induced ferroptosis in lung cancer cells, evidenced by increased levels of ROS and Fe2+ and downregulation of ferroptosis-associated protein and endoplasmic reticulum stress, was involved in fascaplysin-induced ferroptosis. In addition, ROS was found to mediate fascaplysin-induced apoptosis. Fascaplysin significantly upregulated the expression of PD-L1 in lung cancer cells, and enhanced anti-PD-1 antitumor efficacy in a syngeneic mouse model. Therefore, these results suggest that fascaplysin exerts anticancer effects by inducing apoptosis and ferroptosis in vitro, and improving the sensitivity of anti-PD-1 immunotherapy in vivo. Fascaplysin is a promising compound for the treatment of NSCLC.
Collapse
Affiliation(s)
- Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang 524023, China
| | - Guangxiang Xu
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang 524023, China
| |
Collapse
|
10
|
Sajeev A, Hegde M, Daimary UD, Kumar A, Girisa S, Sethi G, Kunnumakkara AB. Modulation of diverse oncogenic signaling pathways by oroxylin A: An important strategy for both cancer prevention and treatment. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 105:154369. [PMID: 35985182 DOI: 10.1016/j.phymed.2022.154369] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 07/14/2022] [Accepted: 07/31/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Regardless of major advances in diagnosis, prevention and treatment strategies, cancer is still a foreboding cause due to factors like chemoresistance, radioresistance, adverse side effects and cancer recurrence. Therefore, continuous development of unconventional approaches is a prerequisite to overcome foregoing glitches. Natural products have found their way into treatment of serious health conditions, including cancer since ancient times. The compound oroxylin A (OA) is one among those with enormous potential against different malignancies. It is a flavonoid obtained from the several plants such as Oroxylum indicum, Scutellaria baicalensis and S. lateriflora, Anchietea pyrifolia, and Aster himalaicus. PURPOSE The main purpose of this study is to comprehensively elucidate the anticancerous effects of OA against various malignancies and unravel their chemosensitization and radiosensitization potential. Pharmacokinetic and pharmacodynamic studies of OA have also been investigated. METHOD The literature on antineoplastic effects of OA was searched in PubMed and Scopus, including in vitro and in vivo studies and is summarized based on a systematic review protocol prepared according to Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. The term "oroxylin A" was used in combination with "cancer" and all the title, abstracts and keywords appeared were considered. RESULTS In Scopus, a total of 157 articles appeared out of which 103 articles that did not meet the eligibility criteria were eliminated and 54 were critically evaluated. In PubMed, from the 85 results obtained, 26 articles were eliminated and 59 were included in the preparation of this review. Mounting number of studies have illustrated the anticancer effects of OA, and its mechanism of action. CONCLUSION OA is a promising natural flavonoid possessing wide range of pleiotropic properties and is a potential anticancer agent. It has a great potential in the treatment of multiple cancers including brain, breast, cervical, colon, esophageal, gall bladder, gastric, hematological, liver, lung, oral, ovarian, pancreatic and skin. However, lack of pharmacokinetic studies, toxicity assessments, and dose standardization studies and adverse effects limit the optimization of this compound as a therapeutic agent.
Collapse
Affiliation(s)
- Anjana Sajeev
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology, Guwahati, 781039, Assam, India
| | - Mangala Hegde
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology, Guwahati, 781039, Assam, India
| | - Uzini Devi Daimary
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology, Guwahati, 781039, Assam, India
| | - Aviral Kumar
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology, Guwahati, 781039, Assam, India
| | - Sosmitha Girisa
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology, Guwahati, 781039, Assam, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology, Guwahati, 781039, Assam, India.
| |
Collapse
|
11
|
Leelamine Modulates STAT5 Pathway Causing Both Autophagy and Apoptosis in Chronic Myelogenous Leukemia Cells. BIOLOGY 2022; 11:biology11030366. [PMID: 35336740 PMCID: PMC8945775 DOI: 10.3390/biology11030366] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/23/2022] [Accepted: 02/23/2022] [Indexed: 01/07/2023]
Abstract
Simple Summary Autophagy is a cellular mechanism that is essential for removing misfolded proteins and damaged organelles. Moreover, the aberrant activation of signal transducer and activator of transcription 5 (STAT5), which can regulate cellular survival and homeostasis, has been often observed in different malignancies. In this study, we demonstrate that leelamine inhibits the STAT5 phosphorylation while inducing autophagy as well as apoptosis in chronic myeloid leukemia cells. Leelamine induces autophagy by stimulating the expression of Atg7, beclin-1, and the production of autophagosomes, which leads to substantial inhibition of STAT5 activation. Abstract Leelamine (LEE) has recently attracted significant attention for its growth inhibitory effects against melanoma, breast cancer, and prostate cancer cells; however, its impact on hematological malignancies remains unclear. Here, we first investigate the cytotoxic effects of LEE on several human chronic myeloid leukemia (CML) cells. We noted that LEE stimulated both apoptosis and autophagy in CML cells. In addition, the constitutive activation of signal transducer and activator of transcription 5 (STAT5) was suppressed substantially upon LEE treatment. Moreover, STAT5 knockdown with small interfering RNA (siRNA) increased LEE-induced apoptosis as well as autophagy and affected the levels of various oncogenic proteins. Thus, the targeted mitigation of STAT5 activation by LEE can contribute to its diverse anticancer effects by enhancing two distinct cell death pathways.
Collapse
|
12
|
Kamran S, Sinniah A, Abdulghani MAM, Alshawsh MA. Therapeutic Potential of Certain Terpenoids as Anticancer Agents: A Scoping Review. Cancers (Basel) 2022; 14:1100. [PMID: 35267408 PMCID: PMC8909202 DOI: 10.3390/cancers14051100] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/19/2022] [Accepted: 02/05/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer is a life-threatening disease and is considered to be among the leading causes of death worldwide. Chemoresistance, severe toxicity, relapse and metastasis are the major obstacles in cancer therapy. Therefore, introducing new therapeutic agents for cancer remains a priority to increase the range of effective treatments. Terpenoids, a large group of secondary metabolites, are derived from plant sources and are composed of several isoprene units. The high diversity of terpenoids has drawn attention to their potential anticancer and pharmacological activities. Some terpenoids exhibit an anticancer effect by triggering various stages of cancer progression, for example, suppressing the early stage of tumorigenesis via induction of cell cycle arrest, inhibiting cancer cell differentiation and activating apoptosis. At the late stage of cancer development, certain terpenoids are able to inhibit angiogenesis and metastasis via modulation of different intracellular signaling pathways. Significant progress in the identification of the mechanism of action and signaling pathways through which terpenoids exert their anticancer effects has been highlighted. Hence, in this review, the anticancer activities of twenty-five terpenoids are discussed in detail. In addition, this review provides insights on the current clinical trials and future directions towards the development of certain terpenoids as potential anticancer agents.
Collapse
Affiliation(s)
- Sareh Kamran
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia; (S.K.); (A.S.)
| | - Ajantha Sinniah
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia; (S.K.); (A.S.)
| | - Mahfoudh A. M. Abdulghani
- Department of Pharmacology and Toxicology, Unaizah College of Pharmacy, Qassim University, Al Qassim 51911, Saudi Arabia;
| | - Mohammed Abdullah Alshawsh
- Department of Pharmacology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia; (S.K.); (A.S.)
| |
Collapse
|
13
|
Ma Z, Woon CYN, Liu CG, Cheng JT, You M, Sethi G, Wong ALA, Ho PCL, Zhang D, Ong P, Wang L, Goh BC. Repurposing Artemisinin and its Derivatives as Anticancer Drugs: A Chance or Challenge? Front Pharmacol 2022; 12:828856. [PMID: 35035355 PMCID: PMC8758560 DOI: 10.3389/fphar.2021.828856] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 12/13/2021] [Indexed: 11/30/2022] Open
Abstract
Cancer has become a global health problem, accounting for one out of six deaths. Despite the recent advances in cancer therapy, there is still an ever-growing need for readily accessible new therapies. The process of drug discovery and development is arduous and takes many years, and while it is ongoing, the time for the current lead compounds to reach clinical trial phase is very long. Drug repurposing has recently gained significant attention as it expedites the process of discovering new entities for anticancer therapy. One such potential candidate is the antimalarial drug, artemisinin that has shown anticancer activities in vitro and in vivo. In this review, major molecular and cellular mechanisms underlying the anticancer effect of artemisinin and its derivatives are summarised. Furthermore, major mechanisms of action and some key signaling pathways of this group of compounds have been reviewed to explore potential targets that contribute to the proliferation and metastasis of tumor cells. Despite its established profile in malaria treatment, pharmacokinetic properties, anticancer potency, and current formulations that hinder the clinical translation of artemisinin as an anticancer agent, have been discussed. Finally, potential solutions or new strategies are identified to overcome the bottlenecks in repurposing artemisinin-type compounds as anticancer drugs.
Collapse
Affiliation(s)
- Zhaowu Ma
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Clariis Yi-Ning Woon
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Chen-Guang Liu
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Jun-Ting Cheng
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Mingliang You
- Hangzhou Cancer Institute, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou, China.,Affiliated Hangzhou Cancer Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Andrea Li-Ann Wong
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Paul Chi-Lui Ho
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Daping Zhang
- School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, China
| | - Peishi Ong
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore
| | - Boon-Cher Goh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Cancer Science Institute of Singapore, National University of Singapore, Singapore, Singapore.,Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| |
Collapse
|
14
|
Zhao Y, Huang Z, Peng H. Molecular Mechanisms of Ferroptosis and Its Roles in Hematologic Malignancies. Front Oncol 2021; 11:743006. [PMID: 34778060 PMCID: PMC8582018 DOI: 10.3389/fonc.2021.743006] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 10/05/2021] [Indexed: 01/08/2023] Open
Abstract
Cell death is essential for the normal metabolism of human organisms. Ferroptosis is a unique regulated cell death (RCD) mode characterized by excess accumulation of iron-dependent lipid peroxide and reactive oxygen species (ROS) compared with other well-known programmed cell death modes. It has been currently recognized that ferroptosis plays a rather important role in the occurrence, development, and treatment of traumatic brain injury, stroke, acute kidney injury, liver damage, ischemia-reperfusion injury, tumor, etc. Of note, ferroptosis may be explained by the expression of various molecules and signaling components, among which iron, lipid, and amino acid metabolism are the key regulatory mechanisms of ferroptosis. Meanwhile, tumor cells of hematological malignancies, such as leukemia, lymphoma, and multiple myeloma (MM), are identified to be sensitive to ferroptosis. Targeting potential regulatory factors in the ferroptosis pathway may promote or inhibit the disease progression of these malignancies. In this review, a systematic summary was conducted on the key molecular mechanisms of ferroptosis and the current potential relationships of ferroptosis with leukemia, lymphoma, and MM. It is expected to provide novel potential therapeutic approaches and targets for hematological malignancies.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Hematology, The Second Xiangya Hospital, Hunan Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
- Institute of Hematology, Central South University, Changsha, China
| | - Zineng Huang
- Department of Hematology, The Second Xiangya Hospital, Hunan Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
- Institute of Hematology, Central South University, Changsha, China
| | - Hongling Peng
- Department of Hematology, The Second Xiangya Hospital, Hunan Key Laboratory of Basic and Applied Hematology, Central South University, Changsha, China
- Institute of Hematology, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, Changsha, China
| |
Collapse
|
15
|
Yang X, Zheng Y, Liu L, Huang J, Wang F, Zhang J. Progress on the study of the anticancer effects of artesunate. Oncol Lett 2021; 22:750. [PMID: 34539854 PMCID: PMC8436334 DOI: 10.3892/ol.2021.13011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 07/30/2021] [Indexed: 12/16/2022] Open
Abstract
Artesunate (ART) is a derivative of artemisinin that is extracted from the wormwood plant Artemisia annua. ART is an antimalarial drug that has been shown to be safe and effective for clinical use. In addition to its antimalarial properties, ART has been attracting attention over recent years due to its reported inhibitory effects on cancer cell proliferation, invasion and migration. Therefore, ART has a wider range of potential clinical applications than first hypothesized. The aim of the present review was to summarize the latest research progress on the possible anticancer effects of ART, in order to lay a theoretical foundation for the further development of ART as a therapeutic option for cancer.
Collapse
Affiliation(s)
- Xiulan Yang
- Department of Pharmacology, The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Yudong Zheng
- Department of Pharmacology, The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Lian Liu
- Department of Pharmacology, The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Jiangrong Huang
- Department of Pharmacology, The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Fei Wang
- Center of Experiment and Training, Hubei College of Chinese Medicine, Jingzhou, Hubei 434020, P.R. China
| | - Jie Zhang
- Department of Pharmacology, The School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| |
Collapse
|
16
|
Jung YY, Kim C, Ha IJ, Lee SG, Lee J, Um JY, Ahn KS. Pyrimethamine Modulates Interplay between Apoptosis and Autophagy in Chronic Myelogenous Leukemia Cells. Int J Mol Sci 2021; 22:8147. [PMID: 34360911 PMCID: PMC8347135 DOI: 10.3390/ijms22158147] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/23/2021] [Accepted: 07/23/2021] [Indexed: 12/15/2022] Open
Abstract
Pyrimethamine (Pyri) is being used in combination with other medications to treat serious parasitic infections of the body, brain, or eye and to also reduce toxoplasmosis infection in the patients with HIV infection. Additionally, Pyri can display significant anti-cancer potential in different tumor models, but the possible mode of its actions remains unclear. Hence, in this study, the possible anti-tumoral impact of Pyri on human chronic myeloid leukemia (CML) was deciphered. Pyri inhibited cell growth in various types of tumor cells and exhibited a marked inhibitory action on CML cells. In addition to apoptosis, Pyri also triggered sustained autophagy. Targeted inhibition of autophagy sensitized the tumor cells to Pyri-induced apoptotic cell death. Moreover, the activation of signal transducer and activator of transcription 5 (STAT5) and its downstream target gene Bcl-2 was attenuated by Pyri. Accordingly, small interfering RNA (siRNA)-mediated STAT5 knockdown augmented Pyri-induced autophagy and apoptosis and promoted the suppressive action of Pyri on cell viability. Moreover, ectopic overexpression of Bcl-2 protected the cells from Pyri-mediated autophagy and apoptosis. Overall, the data indicated that the attenuation of STAT5-Bcl-2 cascade by Pyri can regulate its growth inhibitory properties by simultaneously targeting both apoptosis and autophagy cell death mechanism(s).
Collapse
Affiliation(s)
- Young Yun Jung
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (Y.Y.J.); (C.K.); (S.-G.L.); (J.-Y.U.)
| | - Chulwon Kim
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (Y.Y.J.); (C.K.); (S.-G.L.); (J.-Y.U.)
| | - In Jin Ha
- Korean Medicine Clinical Trial Center (K-CTC), Korean Medicine Hospital, Kyung Hee University, Seoul 02447, Korea; (I.J.H.); (J.L.)
| | - Seok-Geun Lee
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (Y.Y.J.); (C.K.); (S.-G.L.); (J.-Y.U.)
- Korean Medicine Clinical Trial Center (K-CTC), Korean Medicine Hospital, Kyung Hee University, Seoul 02447, Korea; (I.J.H.); (J.L.)
| | - Junhee Lee
- Korean Medicine Clinical Trial Center (K-CTC), Korean Medicine Hospital, Kyung Hee University, Seoul 02447, Korea; (I.J.H.); (J.L.)
| | - Jae-Young Um
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (Y.Y.J.); (C.K.); (S.-G.L.); (J.-Y.U.)
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea; (Y.Y.J.); (C.K.); (S.-G.L.); (J.-Y.U.)
| |
Collapse
|
17
|
Mancuso RI, Foglio MA, Olalla Saad ST. Artemisinin-type drugs for the treatment of hematological malignancies. Cancer Chemother Pharmacol 2020; 87:1-22. [PMID: 33141328 DOI: 10.1007/s00280-020-04170-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/06/2020] [Indexed: 12/19/2022]
Abstract
Qinghaosu, known as artemisinin (ARS), has been for over two millennia, one of the most common herbs prescribed in traditional Chinese medicine (TCM). ARS was developed as an antimalarial drug and currently belongs to the established standard treatments of malaria as a combination therapy worldwide. In addition to the antimalarial bioactivity of ARS, anticancer activities have been shown both in vitro and in vivo. Like other natural products, ARS acts in a multi-specific manner also against hematological malignancies. The chemical structure of ARS is a sesquiterpene lactone, which contains an endoperoxide bridge essential for activity. The main mechanism of action of ARS and its derivatives (artesunate, dihydroartemisinin, artemether) toward leukemia, multiple myeloma, and lymphoma cells comprises oxidative stress response, inhibition of proliferation, induction of various types of cell death as apoptosis, autophagy, ferroptosis, inhibition of angiogenesis, and signal transducers, as NF-κB, MYC, amongst others. Therefore, new pharmaceutically active compounds, dimers, trimers, and hybrid molecules, could enhance the existing therapeutic alternatives in combating hematologic malignancies. Owing to the high potency and good tolerance without side effects of ARS-type drugs, combination therapies with standard chemotherapies could be applied in the future after further clinical trials in hematological malignancies.
Collapse
Affiliation(s)
- R I Mancuso
- Hematology and Hemotherapy Center, University of Campinas, HEMOCENTRO UNICAMP, Campinas, São Paulo, Brazil
| | - M A Foglio
- Faculty of Pharmaceutical Science, University of Campinas-UNICAMP, Campinas, São Paulo, Brazil
| | - S T Olalla Saad
- Hematology and Hemotherapy Center, University of Campinas, HEMOCENTRO UNICAMP, Campinas, São Paulo, Brazil.
| |
Collapse
|
18
|
STAT3 transcription factor as target for anti-cancer therapy. Pharmacol Rep 2020; 72:1101-1124. [PMID: 32880101 DOI: 10.1007/s43440-020-00156-5] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 08/23/2020] [Accepted: 08/25/2020] [Indexed: 12/17/2022]
Abstract
STATs constitute a large family of transcription activators and transducers of signals that have an important role in many cell functions as regulation of proliferation and differentiation of the cell also regulation of apoptosis and angiogenesis. STAT3 as a member of that family, recently was discovered to have a vital role in progression of different types of cancers. The activation of STAT3 was observed to regulate multiple gene functions during cancer-like cell proliferation, differentiation, apoptosis, metastasis, inflammation, immunity, cell survival, and angiogenesis. The inhibition of STAT3 activation has been an important target for cancer therapy. Inhibitors of STAT3 have been used for a long time for treatment of many types of cancers like leukemia, melanoma, colon, and renal cancer. In this review article, we summarize and discuss different drugs inhibiting the action of STAT3 and used in treatment of different types of cancer.
Collapse
|
19
|
Involvement of STAT5 in Oncogenesis. Biomedicines 2020; 8:biomedicines8090316. [PMID: 32872372 PMCID: PMC7555335 DOI: 10.3390/biomedicines8090316] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/19/2020] [Accepted: 08/26/2020] [Indexed: 12/24/2022] Open
Abstract
Signal transducer and activator of transcription (STAT) proteins, and in particular STAT3, have been established as heavily implicated in cancer. Recently, the involvement of STAT5 signalling in the pathology of cancer has been shown to be of increasing importance. STAT5 plays a crucial role in the development of the mammary gland and the homeostasis of the immune system. However, in various cancers, aberrant STAT5 signalling promotes the expression of target genes, such as cyclin D, Bcl-2 and MMP-2, that result in increased cell proliferation, survival and metastasis. To target constitutive STAT5 signalling in cancers, there are several STAT5 inhibitors that can prevent STAT5 phosphorylation, dimerisation, or its transcriptional activity. Tyrosine kinase inhibitors (TKIs) that target molecules upstream of STAT5 could also be utilised. Consequently, since STAT5 contributes to tumour aggressiveness and cancer progression, inhibiting STAT5 constitutive activation in cancers that rely on its signalling makes for a promising targeted treatment option.
Collapse
|
20
|
Parama D, Boruah M, Yachna K, Rana V, Banik K, Harsha C, Thakur KK, Dutta U, Arya A, Mao X, Ahn KS, Kunnumakkara AB. Diosgenin, a steroidal saponin, and its analogs: Effective therapies against different chronic diseases. Life Sci 2020; 260:118182. [PMID: 32781063 DOI: 10.1016/j.lfs.2020.118182] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Chronic diseases are a major cause of mortality worldwide, and despite the recent development in treatment modalities, synthetic drugs have continued to show toxic side effects and development of chemoresistance, thereby limiting their application. The use of phytochemicals has gained attention as they show minimal side effects. Diosgenin is one such phytochemical which has gained importance for its efficacy against the life-threatening diseases, such as cardiovascular diseases, cancer, nervous system disorders, asthma, arthritis, diabetes, and many more. AIM To evaluate the literature available on the potential of diosgenin and its analogs in modulating different molecular targets leading to the prevention and treatment of chronic diseases. METHOD A detailed literature search has been carried out on PubMed for gathering information related to the sources, biosynthesis, physicochemical properties, biological activities, pharmacokinetics, bioavailability and toxicity of diosgenin and its analogs. KEY FINDINGS The literature search resulted in many in vitro, in vivo and clinical trials that reported the efficacy of diosgenin and its analogs in modulating important molecular targets and signaling pathways such as PI3K/AKT/mTOR, JAK/STAT, NF-κB, MAPK, etc., which play a crucial role in the development of most of the diseases. Reports have also revealed the safety of the compound and the adaptation of nanotechnological approaches for enhancing its bioavailability and pharmacokinetic properties. SIGNIFICANCE Thus, the review summarizes the efficacy of diosgenin and its analogs for developing as a potent drug against several chronic diseases.
Collapse
Affiliation(s)
- Dey Parama
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Monikongkona Boruah
- Cell and Molecular Biology Lab, Department of Zoology, Cotton University, Guwahati, Assam 781001, India
| | - Kumari Yachna
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Varsha Rana
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Kishore Banik
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Choudhary Harsha
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Krishan Kumar Thakur
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India
| | - Uma Dutta
- Cell and Molecular Biology Lab, Department of Zoology, Cotton University, Guwahati, Assam 781001, India
| | - Aditya Arya
- Department of Pharmacology and Therapeutics, School of Medicine, Faculty of Health and Medical Sciences, Taylor's University, Subang Jaya, Malaysia
| | - Xinliang Mao
- Institute of Clinical Pharmacology, Guangzhou University of Chinese Medicine, 12 Jichang Road, Baiyun District, Guangzhou 510405, China; Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, 199 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea.
| | - Ajaikumar B Kunnumakkara
- Cancer Biology Laboratory and DBT-AIST International Center for Translational and Environmental Research (DAICENTER), Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Assam 781039, India.
| |
Collapse
|
21
|
Sin ZW, Bhardwaj V, Pandey AK, Garg M. A brief overview of antitumoral actions of bruceine D. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2020; 1:200-217. [PMID: 36046775 PMCID: PMC9400783 DOI: 10.37349/etat.2020.00013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Accepted: 06/30/2020] [Indexed: 12/25/2022] Open
Abstract
Cancer remains the second leading cause of mortality globally. In combating cancer, conventional chemotherapy and/or radiotherapy are administered as first-line therapy. However, these are usually accompanied with adverse side effects that decrease the quality of patient’s lives. As such, natural bioactive compounds have gained an attraction in the scientific and medical community as evidence of their anticancer properties and attenuation of side effects mounted. In particular, quassinoids have been found to exhibit a plethora of inhibitory activities such as anti-proliferative effects on tumor development and metastasis. Recently, bruceine D, a quassinoid isolated from the shrub Brucea javanica (L.) Merr. (Simaroubaceae), has come under immense investigation on its antineoplastic properties in various human cancers including pancreas, breast, lung, blood, bone, and liver. In this review, we have highlighted the antineoplastic effects of bruceine D and its mode of actions in different tumor models.
Collapse
Affiliation(s)
- Zi Wayne Sin
- Department of Biological Sciences, National University of Singapore, Singapore 117600, Singapore
| | - Vipul Bhardwaj
- Amity Institute of Molecular Medicine and Stem cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida 201313, India
| | - Amit Kumar Pandey
- Amity Institute of Biotechnology, Amity University Haryana, Manesar, Haryana 122413, India
| | - Manoj Garg
- Amity Institute of Molecular Medicine and Stem cell Research (AIMMSCR), Amity University Uttar Pradesh, Noida 201313, India
| |
Collapse
|
22
|
Abstract
Artemisinin (ART) and its derivatives are one of the most important classes of antimalarial agents, originally derived from a Chinese medicinal plant called Artemisia annua L. Beyond their outstanding antimalarial and antischistosomal activities, ART and its derivatives also possess both in-vitro and in-vivo activities against various types of cancer. Their anticancer effects range from initiation of apoptotic cell death to inhibition of cancer proliferation, metastasis and angiogenesis, and even modulation of the cell signal transduction pathway. This review provides a comprehensive update on ART and its derivatives, their mechanisms of action, and their synergistic effects with other chemicals in targeting leukemia cells. Combined with limited evidence of drug resistance and low toxicity profile, we conclude that ART and its derivatives, including dimers, trimers, and hybrids, might be a potential therapeutic alternative to current chemotherapies in combating leukemia, although more studies are necessary before they can be applied clinically.
Collapse
|
23
|
Aggarwal V, Tuli HS, Kaur J, Aggarwal D, Parashar G, Chaturvedi Parashar N, Kulkarni S, Kaur G, Sak K, Kumar M, Ahn KS. Garcinol Exhibits Anti-Neoplastic Effects by Targeting Diverse Oncogenic Factors in Tumor Cells. Biomedicines 2020; 8:103. [PMID: 32365899 PMCID: PMC7277375 DOI: 10.3390/biomedicines8050103] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/27/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022] Open
Abstract
Garcinol, a polyisoprenylated benzophenone, is the medicinal component obtained from fruits and leaves of Garcinia indica (G. indica) and has traditionally been extensively used for its antioxidant and anti-inflammatory properties. In addition, it has been also been experimentally illustrated to elicit anti-cancer properties. Several in vitro and in vivo studies have illustrated the potential therapeutic efficiency of garcinol in management of different malignancies. It mainly acts as an inhibitor of cellular processes via regulation of transcription factors NF-κB and JAK/STAT3 in tumor cells and have been demonstrated to effectively inhibit growth of malignant cell population. Numerous studies have highlighted the anti-neoplastic potential of garcinol in different oncological transformations including colon cancer, breast cancer, prostate cancer, head and neck cancer, hepatocellular carcinoma, etc. However, use of garcinol is still in its pre-clinical stage and this is mainly attributed to the limitations of conclusive evaluation of pharmacological parameters. This necessitates evaluation of garcinol pharmacokinetics to precisely identify an appropriate dose and route of administration, tolerability, and potency under physiological conditions along with characterization of a therapeutic index. Hence, the research is presently ongoing in the dimension of exploring the precise metabolic mechanism of garcinol. Despite various lacunae, garcinol has presented with promising anti-cancer effects. Hence, this review is motivated by the constantly emerging and promising positive anti-cancerous effects of garcinol. This review is the first effort to summarize the mechanism of action of garcinol in modulation of anti-cancer effect via regulation of different cellular processes.
Collapse
Affiliation(s)
- Vaishali Aggarwal
- Department of Histopathology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, Punjab 160012, India;
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India; (D.A.); or (G.P.); (N.C.P.)
| | - Jagjit Kaur
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics (CNBP), Faculty of Engineering, The University of New South Wales, Sydney 2052, Australia;
| | - Diwakar Aggarwal
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India; (D.A.); or (G.P.); (N.C.P.)
| | - Gaurav Parashar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India; (D.A.); or (G.P.); (N.C.P.)
| | - Nidarshana Chaturvedi Parashar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India; (D.A.); or (G.P.); (N.C.P.)
| | - Samruddhi Kulkarni
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’S NMIMS, Vileparle-West, Mumbai 400056, India; (S.K.); (G.K.)
| | - Ginpreet Kaur
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’S NMIMS, Vileparle-West, Mumbai 400056, India; (S.K.); (G.K.)
| | | | - Manoj Kumar
- Department of Chemistry, Maharishi Markandeshwar University, Sadopur 133001, India;
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| |
Collapse
|
24
|
Li Y, Shan NN, Sui XH. Research Progress on Artemisinin and Its Derivatives against Hematological Malignancies. Chin J Integr Med 2020; 26:947-955. [PMID: 32048169 DOI: 10.1007/s11655-019-3207-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2019] [Indexed: 12/19/2022]
Abstract
Although current therapeutic methods against hematological malignancies are effective in the early stage, they usually lose their effectiveness because of the development of drug resistances. Seeking new drugs with significant therapeutic effects is one of the current research hotspots. Artemisinin, an extract from the plant Artemisia annua Linne, and its derivatives have excellent antimalarial effects in clinical applications as well as excellent safety. Recent studies have documented that artemisinin and its derivatives (ARTs) also have significant effects against multiple types of tumours, including hematological malignancies. This review focuses on the latest research achievements of ARTs in the treatment of hematological malignancies as well as its mechanisms and future applications. The mechanisms of ARTs against different types of hematological malignancies mainly include cell cycle arrest, induction autophagy and apoptosis, inhibition of angiogenesis, production of reactive oxygen species, and induction of differentiation. Additionally, the review also summarizes the anticancer effects of ARTs in many drug-resistant hematological malignancies and its synergistic effects with other drugs.
Collapse
Affiliation(s)
- Ying Li
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China.
| | - Ning-Ning Shan
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China
| | - Xiao-Hui Sui
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China
| |
Collapse
|
25
|
Ma L, Fei H. Antimalarial drug artesunate is effective against chemoresistant anaplastic thyroid carcinoma via targeting mitochondrial metabolism. J Bioenerg Biomembr 2020; 52:123-130. [PMID: 32036542 DOI: 10.1007/s10863-020-09824-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 02/02/2020] [Indexed: 12/29/2022]
Abstract
Anaplastic thyroid carcinoma (ATC) is the most aggressive type of thyroid malignancies and resistant to chemotherapy. Novel therapeutic strategy is required for better management of ATC. In this work, we show that artesunate, an antimalarial drug, is active against chemoresistant ATC cells. Artesunate dose-dependently inhibits growth and induces apoptosis in chemo-sensitive (8505C and KAT-4) and -resistant (8505C-r and KAT-4-r) ATC cells, and acts synergistically with doxorubicin. Using multiple xenograft mouse models, artesunate is active against chemo-sensitive and -resistant ATC cells in vivo at doses that do not cause toxicity in mice. Our mechanism analysis reveals that artesunate acts on ATC cells through suppressing mitochondrial functions without affecting glycolysis, leading to oxidative stress and damage, regardless of whether they are sensitive or resistant to chemotherapy. Interestingly, KAT-4-r cells demonstrate decreased glycolysis, increased mitochondrial membrane potential and mitochondrial respiration compared to KAT-4 cells whereas such phenomenon is not observed between 8505C and 8505C-r cells. This suggests that some but not all ATC cells gain enhanced mitochondrial biogenesis after prolonged exposure to chemotherapy drug, which may explain the different sensitivities of 8505C-r and KAT-4-r to artesunate. Our work demonstrates that artesunate is a potential addition to the treatment armamentarium for ATC, particularly those with chemoresistance. Our findings also highlight the therapeutic value of targeting mitochondria in chemoresistant ATC.
Collapse
Affiliation(s)
- Ling Ma
- Department of Endocrinology, First Affiliated Hospital, First Clinical Medical College, Yangtze University, Jingzhou, Hubei, 434000, China
| | - Honghua Fei
- Department of Endocrinology, People's Hospital of Rizhao, No.126, Taian Road, Rizhao, 276826, Shandong, China.
| |
Collapse
|
26
|
Artesunate targets oral tongue squamous cell carcinoma via mitochondrial dysfunction-dependent oxidative damage and Akt/AMPK/mTOR inhibition. J Bioenerg Biomembr 2020; 52:113-121. [PMID: 31965456 DOI: 10.1007/s10863-020-09823-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 01/13/2020] [Indexed: 12/12/2022]
Abstract
Although mitochondrial metabolism has recently gained attention as a promising therapeutic strategy in cancer, little is known on the impact of mitochondrial respiration inhibition on oral tongue squamous cell carcinoma (OTSCC). Using in vitro and in vivo OTSCC models, our work demonstrates that inducing mitochondrial dysfunction by anti-malarial drug artesunate is effective in targeting OTSCC stem-cell like and bulk cells. Artesunate inhibits anchorage-independent colony formation, proliferation and survival in all tested OTSCC cell lines although with varying efficacy. Artesunate displays preferential anti-OTSCC activity by sparing normal cells. Mechanism analysis indicates that artesunate inhibits mitochondrial respiration via suppressing mitochondrial complex I and II but not IV or V, resulting in oxidative stress and damage. Interestingly, OTSCC cells that are more sensitive to artesunate have higher level of basal mitochondrial respiration and reversed respiratory capacity compared to those with less sensitivity to artesunate, suggesting the varying dependence on mitochondrial respiration among OTSCC cell lines. In addition, artesunate induces oxidative stress and damage in cells with low sensitivity to a less extent than in those with high sensitivity. We confirm that mitochondrial respiration inhibition is required for the action of artesunate in OTSCC. Mitochondrial dysfunction by artesunate further activates AMPK and suppresses Akt/mTOR. Importantly, the in vitro observations are reproducible in vivo OTSCC xenograft mouse model. Our findings provide pre-clinical evidence on the efficacy of artesunate and emphasize the therapeutic value of targeting mitochondrial respiration in OTSCC.
Collapse
|
27
|
Ding X, Yue W, Chen H. Effect of artesunate on apoptosis and autophagy in tamoxifen resistant breast cancer cells (TAM-R). Transl Cancer Res 2019; 8:1863-1872. [PMID: 35116937 PMCID: PMC8797964 DOI: 10.21037/tcr.2019.08.41] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/12/2019] [Indexed: 11/24/2022]
Abstract
Background The antitumor effect of artesunate (ART) is well-recognized. To investigate the effect of ART on tamoxifen-resistant breast cancer cells (TAM-R) proliferation, apoptosis, and autophagy with TAM-R cells of breast cancer as objects of study, and to investigate whether ART could re-sensitize TAM-R cells to TAM therapy. Methods Experiments were performed using TAM-R cell lines. Cell Death Detection ELISA kit was used to detect the level of apoptosis. Western blot and immunofluorescent staining analysis were conducted to detect autophagy and apoptosis related proteins in TAM-R cells. Results After treated with ART, the proliferation activity of TAM-R cells was inhibited in a concentration-dependent manner. Increased apoptosis activity was detected in TAM-R cells when treated with ART. Compared with 10−6 M TAM monotherapy group, treatment group with ART and TAM in combination caused significant reduction in the levels of Bcl-2, XIAP, and Survivin proteins, and elevation of caspase-7. However, increased p53 proteins was not detected after ART treatment. No significant change was observed in autophagy proteins LC-3 and Beclin-1 among control, ART, TAM, and ART combined with TAM groups. Conclusions The intervention of ART could not inhibit protective autophagy in TAM-R cells, however, possess potential in inducing apoptosis. In addition, as ART inhibit TAM-R cells growth in a dose-dependent manner, co-administration of 1 or 3 µM of ART with 10−6 M TAM might not be enough to re-sensitize TAM-R cells to TAM therapy.
Collapse
Affiliation(s)
- Xiaoqing Ding
- Department of Hematology, Dongfang Hospital Affiliated to Beijing University of Traditional Chinese Medicine, Beijing 100078, China
| | - Wei Yue
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA 22908, USA
| | - Haiyan Chen
- Department of Hematology, Dongfang Hospital Affiliated to Beijing University of Traditional Chinese Medicine, Beijing 100078, China
| |
Collapse
|
28
|
Fangchinoline, a Bisbenzylisoquinoline Alkaloid can Modulate Cytokine-Impelled Apoptosis via the Dual Regulation of NF-κB and AP-1 Pathways. Molecules 2019; 24:molecules24173127. [PMID: 31466313 PMCID: PMC6749215 DOI: 10.3390/molecules24173127] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 08/24/2019] [Accepted: 08/27/2019] [Indexed: 12/25/2022] Open
Abstract
Fangchinoline (FCN) derived from Stephaniae tetrandrine S. Moore can be employed to treat fever, inflammation, rheumatism arthralgia, edema, dysuria, athlete’s foot, and swollen wet sores. FCN can exhibit a plethora of anti-neoplastic effects although its precise mode of action still remains to be deciphered. Nuclear factor-κB (NF-κB) and activator protein-1 (AP-1) can closely regulate carcinogenesis and thus we analyzed the possible action of FCN may have on these two signaling cascades in tumor cells. The effect of FCN on NF-κB and AP-1 signaling cascades and its downstream functions was deciphered using diverse assays in both human chronic myeloid leukemia (KBM5) and multiple myeloma (U266). FCN attenuated growth of both leukemic and multiple myeloma cells and repressed NF-κB, and AP-1 activation through diverse mechanisms, including attenuation of phosphorylation of IκB kinase (IKK) and p65. Furthermore, FCN could also cause significant enhancement in TNFα-driven apoptosis as studied by various molecular techniques. Thus, FCN may exhibit potent anti-neoplastic effects by affecting diverse oncogenic pathways and may be employed as pro-apoptotic agent against various malignancies.
Collapse
|
29
|
Mota TC, Garcia TB, Bonfim LT, Portilho AJS, Pinto CA, Burbano RMR, Bahia M. Markers of oxidative‐nitrosative stress induced by artesunate are followed by clastogenic and aneugenic effects and apoptosis in human lymphocytes. J Appl Toxicol 2019; 39:1405-1412. [DOI: 10.1002/jat.3826] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 05/02/2019] [Accepted: 05/02/2019] [Indexed: 12/23/2022]
Affiliation(s)
- Tatiane C. Mota
- Laboratory of Human Cytogenetic and Genetic Toxicology, Institute of Biological SciencesFederal University of Pará (UFPA) Belém‐ Pará Brazil
| | - Tarcyane B. Garcia
- Laboratory of Human Cytogenetic and Genetic Toxicology, Institute of Biological SciencesFederal University of Pará (UFPA) Belém‐ Pará Brazil
| | - Laís T. Bonfim
- Laboratory of Human Cytogenetic and Genetic Toxicology, Institute of Biological SciencesFederal University of Pará (UFPA) Belém‐ Pará Brazil
| | - Adrhyann J. S. Portilho
- Laboratory of Human Cytogenetic and Genetic Toxicology, Institute of Biological SciencesFederal University of Pará (UFPA) Belém‐ Pará Brazil
| | - Camila A. Pinto
- Laboratory of Human Cytogenetic and Genetic Toxicology, Institute of Biological SciencesFederal University of Pará (UFPA) Belém‐ Pará Brazil
| | - Rommel M. R. Burbano
- Laboratory of Human Cytogenetic and Genetic Toxicology, Institute of Biological SciencesFederal University of Pará (UFPA) Belém‐ Pará Brazil
| | - Marcelo Bahia
- Laboratory of Human Cytogenetic and Genetic Toxicology, Institute of Biological SciencesFederal University of Pará (UFPA) Belém‐ Pará Brazil
| |
Collapse
|
30
|
Jung YY, Shanmugam MK, Narula AS, Kim C, Lee JH, Namjoshi OA, Blough BE, Sethi G, Ahn KS. Oxymatrine Attenuates Tumor Growth and Deactivates STAT5 Signaling in a Lung Cancer Xenograft Model. Cancers (Basel) 2019; 11:cancers11010049. [PMID: 30621055 PMCID: PMC6356594 DOI: 10.3390/cancers11010049] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/28/2018] [Accepted: 12/28/2018] [Indexed: 12/24/2022] Open
Abstract
Oxymatrine (OMT) is a major alkaloid found in radix Sophorae flavescentis extract and has been reported to exhibit various pharmacological activities. We elucidated the detailed molecular mechanism(s) underlying the therapeutic actions of OMT in non-small cell lung cancer (NSCLC) cells and a xenograft mouse model. Because the STAT5 signaling cascade has a significant role in regulating cell proliferation and survival in tumor cells, we hypothesized that OMT may disrupt this signaling cascade to exert its anticancer effects. We found that OMT can inhibit the constitutive activation of STAT5 by suppressing the activation of JAK1/2 and c-Src, nuclear localization, as well as STAT5 binding to DNA in A549 cells and abrogated IL-6-induced STAT5 phosphorylation in H1299 cells. We also report that a sub-optimal concentration of OMT when used in combination with a low dose of paclitaxel produced significant anti-cancer effects by inhibiting cell proliferation and causing substantial apoptosis. In a preclinical lung cancer mouse model, OMT when used in combination with paclitaxel produced a significant reduction in tumor volume. These results suggest that OMT in combination with paclitaxel can cause an attenuation of lung cancer growth both in vitro and in vivo.
Collapse
Affiliation(s)
- Young Yun Jung
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | | | - Chulwon Kim
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
- Comorbidity Research Institute, College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Jong Hyun Lee
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
- Comorbidity Research Institute, College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Ojas A Namjoshi
- Center for Drug Discovery, RTI International, Research Triangle Park, Durham, NC 27616, USA.
| | - Bruce E Blough
- Center for Drug Discovery, RTI International, Research Triangle Park, Durham, NC 27616, USA.
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
- Comorbidity Research Institute, College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
- Department of Korean Pathology, College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| |
Collapse
|
31
|
Chen L, Wang C, Hu N, Zhao H. Artesunate enhances adriamycin cytotoxicity by inhibiting glycolysis in adriamycin-resistant chronic myeloid leukemia K562/ADR cells. RSC Adv 2019; 9:1004-1014. [PMID: 35547242 PMCID: PMC9087932 DOI: 10.1039/c8ra08041k] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/26/2018] [Indexed: 01/16/2023] Open
Abstract
Adriamycin (ADR) is a widely used drug in multiple cancers including leukemia.
Collapse
Affiliation(s)
- Li Chen
- Department of Hematology
- Huaihe Hospital of Henan University
- Kaifeng 475000
- China
| | - Chao Wang
- Department of Hematology
- Huaihe Hospital of Henan University
- Kaifeng 475000
- China
| | - Ning Hu
- Department of Hematology
- Huaihe Hospital of Henan University
- Kaifeng 475000
- China
| | - Hongmian Zhao
- Department of Hematology
- Huaihe Hospital of Henan University
- Kaifeng 475000
- China
| |
Collapse
|
32
|
Lee JH, Kim C, Ko JH, Jung YY, Jung SH, Kim E, Kong M, Chinnathambi A, Alahmadi TA, Alharbi SA, Sethi G, Ahn KS. Casticin inhibits growth and enhances ionizing radiation-induced apoptosis through the suppression of STAT3 signaling cascade. J Cell Biochem 2018; 120:9787-9798. [PMID: 30520154 DOI: 10.1002/jcb.28259] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 10/22/2018] [Indexed: 12/21/2022]
Abstract
Casticin (CTC), one of the major components of Vitex rotundifolia L., has been reported to exert significant beneficial pharmacological activities and can function as an antiprolactin, anticancer, anti-inflammatory, neuroprotective, analgesic, and immunomodulatory agent. This study aimed at investigating whether the proapoptotic effects of CTC may be mediated through the abrogation of signal transducers and activators of transcription-3 (STAT3) signaling pathway in a variety of human tumor cells. We found that CTC significantly decreased cell viability in a concentration-dependent manner and suppressed cell proliferation in 786-O, YD-8, and HN-9 cells. CTC also induced programmed cell death that was found to be mediated via caspase-3 activation and induction of poly(ADP-ribose) polymerase cleavage. Interestingly, CTC repressed both constitutive and interleukin-6-induced STAT3 activation in 786-O and YD-8 cells but only affected constitutive STAT3 phosphorylation in HN-9 cells. Moreover, CTC could potentiate ionizing radiation-induced apoptotic effects leading to the downregulation of STAT3 activation and thus may be used in combination with radiation against diverse malignancies.
Collapse
Affiliation(s)
- Jong Hyun Lee
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Chulwon Kim
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jeong-Hyeon Ko
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Young Yun Jung
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Sang Hoon Jung
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, Republic of Korea
| | - Eunok Kim
- Korean Medicine Clinical Trial Center, Korean Medicine, Hospital, Kyung Hee University, Seoul, Republic of Korea.,Department of Science in Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Moonkyoo Kong
- Department of Radiation Oncology, Kyung Hee University Medical Center, Kyung Hee University School of Medicine, Seoul, Republic of Korea
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Tahani Awad Alahmadi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia.,Department of Emergency Medicine, Pediatric Emergency Unit, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea.,KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
33
|
Two decades of research in discovery of anticancer drugs targeting STAT3, how close are we? Pharmacol Ther 2018; 191:74-91. [DOI: 10.1016/j.pharmthera.2018.06.006] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
34
|
Yang H, Liu Z, Li X, Zhang Z, Chen D, Lian H. Artesunate-Loaded and Near-Infrared Dye-Conjugated Albumin Nanoparticles as High-Efficiency Tumor-Targeted Photo-Chemo Theranostic Agent. NANOSCALE RESEARCH LETTERS 2018; 13:319. [PMID: 30311065 PMCID: PMC6181830 DOI: 10.1186/s11671-018-2700-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 08/30/2018] [Indexed: 05/20/2023]
Abstract
Herein, a tumor-targeted multifunctional theranostic agent was synthetized using a facile method, combining four clinically approved materials: artesunate (Arte), human serum albumin (HSA), folic acid (FA), and indocyanine green (ICG). The obtained nanocomposites (FA-IHA NPs) showed an excellent photo- and physiological stability. The ICG in the FA-IHA NPs was used not only for near infrared (NIR) fluorescence imaging, but also for photothermal and photodynamic (PTT-PDT) therapy under a single NIR irradiation. In addition, the NIR irradiation (808 nm, 1 W/cm2) could trigger Arte release that showed enhanced chemotherapeutic effect. Through fluorescence imaging, the cell uptake and tumor accumulation of FA-IHA NPs were observed in vitro and in vivo, analyzed by confocal microscopy and NIR fluorescence imaging in tumor xenograft mice. Based on the diagnostic results, FA-IHA NPs at 24 h post injection and combined with NIR irradiation (808 nm, 1 W/cm2) could efficiently suppress tumor growth through a photo-chemo combination therapy, with no tumor recurrence in vitro and in vivo. The obtained results suggested that FA-IHA NPs are promising photo-chemo theranostic agents for future clinical translation.
Collapse
Affiliation(s)
- Hainan Yang
- Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260 China
| | - Zaijia Liu
- Department of Medical Image, Ezhou Central Hospital, Ezhou, 436000 China
| | - Xufeng Li
- Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260 China
| | - Zhenfeng Zhang
- Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260 China
| | - Deji Chen
- Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260 China
| | - Hui Lian
- Department of Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260 China
| |
Collapse
|
35
|
Xu Z, Wang H, Wei S, Wang Z, Ji G. Inhibition of ER stress-related IRE1α/CREB/NLRP1 pathway promotes the apoptosis of human chronic myelogenous leukemia cell. Mol Immunol 2018; 101:377-385. [PMID: 30055408 DOI: 10.1016/j.molimm.2018.07.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 06/15/2018] [Accepted: 07/01/2018] [Indexed: 01/08/2023]
Abstract
Endoplasmic reticulum (ER) stress is induced in chronic myelogenous leukemia (CML) cells. As an important sensor of ER stress, inositol-requiring protein-1α (IRE1α) promotes the survival of acute myeloid leukemia. NLRP1 inflammasome activation promotes metastatic melanoma growth and that IRE1α can increase NLRP1 inflammasome gene expression. This study aimed to investigate the role and molecular mechanism of IRE1α in CML cell growth. We found that overexpression of IRE1α or NLRP1 significantly promoted the proliferation and decreased the apoptosis of CML cells, whereas downregulation of these two genes showed the opposite effects. 4-phenylbutyric acid (4-PBA), an ER stress inhibitor, reduced the expression of IRE1α and NLRP1. IRE1α elevated NLRP1 expression via cAMP responsive element binding protein (CREB) phosphorylation. NLRP1 inflammasome was activated in CML cells and its activation partly reversed ER stress inhibitor-induced cell apoptosis. Furthermore, inhibition of IRE1α/NLRP1 pathway sensitized CML cells to imatinib-mediated apoptosis. Additionally, IRE1α expression was elevated and NLRP1 inflammasome was activated in primary cells from CML patients. Downregulation of IRE1α or NLRP1 suppressed the proliferation and elevated the apoptosis of primary CML cells. Collectively, this study demonstrated that the IRE1α/CREB/NLRP1 pathway contributes to the progression of CML and the development of imatinib resistance. Hence, targeting ER stress-related IRE1α expression or NLRP1 inflammasome activation may block CML development.
Collapse
Affiliation(s)
- Zheng Xu
- Department of Hematology, Xinyang Central Hospital of Henan Province, Xinyang, Henan 464000, PR China
| | - Huirui Wang
- Department of Hematology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, Henan 471009, PR China
| | - Suhua Wei
- Department of Hematology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, PR China
| | - Zhencheng Wang
- Department of Hematopathology, Zibo Central Hospital, Zibo, Shandong 255036, PR China
| | - Guanghou Ji
- Department of Clinical Laboratory, Sishui People's Hospital, Jining, Shandong 273200, PR China.
| |
Collapse
|
36
|
Jiang F, Zhou JY, Zhang D, Liu MH, Chen YG. Artesunate induces apoptosis and autophagy in HCT116 colon cancer cells, and autophagy inhibition enhances the artesunate‑induced apoptosis. Int J Mol Med 2018; 42:1295-1304. [PMID: 29901098 PMCID: PMC6089754 DOI: 10.3892/ijmm.2018.3712] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 05/30/2018] [Indexed: 12/16/2022] Open
Abstract
The present study assessed the antitumor effect of artesunate (ART) in vitro and in vivo, as well as its underlying mechanism of action in HCT116 colon cancer cells. An MTT assay, DAPI staining, flow cytometry, western blotting, immunohistochemistry, transmission electron microscopy and TUNEL assay were performed to study the molecular mechanism underlying the antitumor effects of ART in HCT116 colon cancer cells. ART was observed to inhibit proliferation by inducing the apoptosis of HCT116 cells both in vitro and in vivo. Flow cytometry analysis demonstrated that treatment with 2 and 4 µg/ml ART for 48 h induced early apoptosis in 22.7 and 33.8% of cells, respectively. In the xenograft tumors of BALB/c nude mice, TUNEL-positive cells increased in the ART group compared with that in the normal saline group. Furthermore,the associated mitochondrial cleaved-caspase 3, poly-ADP ribose polymerase (PARP), caspase 9 and Bcl-2-associated X protein levels increased while B-cell lymphoma-2 (Bcl-2) decreased both in the cell and animal ART-treated group. ART-treated cells also exhibited autophagy induction, as evidenced by increased protein expression levels of light chain 3 (LC3) and beclin-1, and the presence of autophagosomes. Notably, pharmacological blockade of autophagy activation using hydroxychloroquine markedly enhanced ART-induced apoptosis and increased the protein levels of cleaved caspase 3 and PARP, while decreasing the levels of LC3 and beclin-1. These findings suggested that the ART-induced autophagy may have a cytoprotective effect by suppressing apoptosis. In conclusion, ART may be a potentially clinically useful anticancer drug for human colon cancer. In addition, co-treatment with ART and an autophagy inhibitor may be an effective anticancer therapy.
Collapse
Affiliation(s)
- Feng Jiang
- Department of Colorectal Surgery, The Affiliated Hospital of Nanjing University of TCM, Nanjing, Jiangsu 210029, P.R. China
| | - Jin-Yong Zhou
- Department of Central Laboratory, The Affiliated Hospital of Nanjing University of TCM, Nanjing, Jiangsu 210029, P.R. China
| | - Dan Zhang
- Department of Colorectal Surgery, The Affiliated Hospital of Nanjing University of TCM, Nanjing, Jiangsu 210029, P.R. China
| | - Ming-Hao Liu
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210023, P.R. China
| | - Yu-Gen Chen
- Department of Colorectal Surgery, The Affiliated Hospital of Nanjing University of TCM, Nanjing, Jiangsu 210029, P.R. China
| |
Collapse
|
37
|
Pei T, Meng Q, Han J, Sun H, Li L, Song R, Sun B, Pan S, Liang D, Liu L. (-)-Oleocanthal inhibits growth and metastasis by blocking activation of STAT3 in human hepatocellular carcinoma. Oncotarget 2017; 7:43475-43491. [PMID: 27259268 PMCID: PMC5190038 DOI: 10.18632/oncotarget.9782] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 05/12/2016] [Indexed: 01/07/2023] Open
Abstract
We explored the anti-cancer capacity of (-)-oleocanthal in human hepatocellular carcinoma (HCC). (-)-Oleocanthal inhibited proliferation and cell cycle progression and induced apoptosis in HCC cells in vitro and suppressed tumor growth in an orthotopic HCC model. (-)-Oleocanthal also inhibited HCC cell migration and invasion in vitro and impeded HCC metastasis in an in vivo lung metastasis model. ( )-Oleocanthal acted by inhibiting epithelial-mesenchymal transition (EMT) through downregulation Twist, which is a direct target of STAT3. (-)-Oleocanthal also reduced STAT3 nuclear translocation and DNA binding activity, ultimately downregulating its downstream effectors, including the cell cycle protein Cyclin D1, the anti-apoptotic proteins Bcl-2 and survivin, and the invasion-related protein MMP 2. Overexpression of constitutively active STAT3 partly reversed the anti cancer effects of (-)-oleocanthal, which inhibited STAT3 activation by decreasing the activities of JAK1 and JAK2 and increasing the activity of SHP-1. These data suggest that (-)-oleocanthal may be a promising candidate for HCC treatment.
Collapse
Affiliation(s)
- Tiemin Pei
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Qinghui Meng
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jihua Han
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Haobo Sun
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Long Li
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Ruipeng Song
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Boshi Sun
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shangha Pan
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Desen Liang
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Lianxin Liu
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Department of General Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
38
|
Liu R, Yu X, Su C, Shi Y, Zhao L. Nanoparticle Delivery of Artesunate Enhances the Anti-tumor Efficiency by Activating Mitochondria-Mediated Cell Apoptosis. NANOSCALE RESEARCH LETTERS 2017; 12:403. [PMID: 28610396 PMCID: PMC5468175 DOI: 10.1186/s11671-017-2169-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 05/29/2017] [Indexed: 05/18/2023]
Abstract
Artemisinin and its derivatives were considered to exert a broad spectrum of anti-cancer activities, and they induced significant anti-cancer effects in tumor cells. Artemisinin and its derivatives could be absorbed quickly, and they were widely distributed, selectively killing tumor cells. Since low concentrations of artesunate primarily depended on oncosis to induce cell death in tumor cells, its anti-tumor effects were undesirable and limited. To obtain better anti-tumor effects, in this study, we took advantage of a new nanotechnology to design novel artesunate-loaded bovine serum albumin nanoparticles to achieve the mitochondrial accumulation of artesunate and induce mitochondrial-mediated apoptosis. The results showed that when compared with free artesunate's reliance on oncotic death, artesunate-loaded bovine serum albumin nanoparticles showed higher cytotoxicity and their significant apoptotic effects were induced through the distribution of artesunate in the mitochondria. This finding indicated that artesunate-loaded bovine serum albumin nanoparticles damaged the mitochondrial integrity and activated mitochondrial-mediated cell apoptosis by upregulating apoptosis-related proteins and facilitating the rapid release of cytochrome C.
Collapse
Affiliation(s)
- Rui Liu
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000 Liaoning People’s Republic of China
| | - Xiwei Yu
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000 Liaoning People’s Republic of China
| | - Chang Su
- School of Veterinary Medicine, Jinzhou Medical University, Jinzhou, 121000 Liaoning People’s Republic of China
| | - Yijie Shi
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000 Liaoning People’s Republic of China
| | - Liang Zhao
- School of Pharmacy, Jinzhou Medical University, Jinzhou, 121000 Liaoning People’s Republic of China
| |
Collapse
|
39
|
Tan M, Rong Y, Su Q, Chen Y. Artesunate induces apoptosis via inhibition of STAT3 in THP-1 cells. Leuk Res 2017; 62:98-103. [PMID: 29031126 DOI: 10.1016/j.leukres.2017.09.022] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 09/26/2017] [Accepted: 09/29/2017] [Indexed: 10/18/2022]
Abstract
OBJECTIVE Our objective was to explore STAT3 expression in patients with acute myeloid leukaemia (AML), assess the anti-proliferative effects of artesunate (ART) on THP-1 cells in vivo and in vitro, and investigate the underlying mechanisms. METHODS In this study, we examined 30 patients with acute myeloid leukaemia diagnosed in our hospital from January 2015 to January 2016. The 20 control group patients had non-haematological diseases and were hospitalized for the same period. We extracted 2ml bone marrow, separated the mononuclear cells, obtained total proteins, and detected STAT3 protein levels with Western blot analyses. The THP-1 cells were treated with different concentrations of ART(0, 10, 25, 50, 100, 200μM). Then, THP-1 cell viability was detected with CCK-8 assays, apoptosis was measured with flow cytometry, and the STAT3, caspase-3 and caspase-8 protein levels were assessed using Western blot analyses. THP-1 cells in logarithmic growth phase were subcutaneously injected into the necks of 5-week-old nude mice. The control group was subcutaneously injected with 0.1ml PBS. After the nude mouse tumours grew, the mice were divided into the control group and drug intervention groups (ART 100μM group, ART 200μM group). The mice in the intervention groups were intraperitoneally injected with ART, and the control group was injected with the same amount of normal saline. Then, changes in the tumours were observed. After the drug intervention, the total protein was extracted, and STAT3 expression was detected by Western blot analysis. RESULTS Compared with the control group, the AML patients had significantly increased STAT3 protein levels (P<0.01). ART significantly inhibited the proliferation of THP-1 cells in a dose-dependent and time-dependent manner. ART also increased THP-1cell apoptosis. After treatment with ART, STAT3 protein was significantly down-regulated, and apoptosis of the cells was induced by the activation of caspase-3 and caspse-8. CONCLUSION AML patients had higher expression of STAT3 than that of the controls. ART induced apoptosis in THP-1 cells and inhibited the growth of xenografts in nude mice, and we also observed that ART down-regulated the expression of STAT3 and activated the caspase-3 and caspase-8. We speculated that the effect of ART on THP-1 cells may be related to inhibition of STAT3 and activation of caspase3 and caspase-8.
Collapse
Affiliation(s)
- Mei Tan
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| | - Ying Rong
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| | - Qiong Su
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China
| | - Yan Chen
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical College, Zunyi, Guizhou, China.
| |
Collapse
|
40
|
Capsazepine inhibits JAK/STAT3 signaling, tumor growth, and cell survival in prostate cancer. Oncotarget 2017; 8:17700-17711. [PMID: 27458171 PMCID: PMC5392279 DOI: 10.18632/oncotarget.10775] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 07/14/2016] [Indexed: 11/25/2022] Open
Abstract
Persistent STAT3 activation is seen in many tumor cells and promotes malignant transformation. Here, we investigated whether capsazepine (Capz), a synthetic analogue of capsaicin, exerts anticancer effects by inhibiting STAT3 activation in prostate cancer cells. Capz inhibited both constitutive and induced STAT3 activation in human prostate carcinoma cells. Capz also inhibited activation of the upstream kinases JAK1/2 and c-Src. The phosphatase inhibitor pervanadate reversed Capz-induced STAT3 inhibition, indicating that the effect of Capz depends on a protein tyrosine phosphatase. Capz treatment increased PTPε protein and mRNA levels. Moreover, siRNA-mediated knockdown of PTPε reversed the Capz-induced induction of PTPε and inhibition of STAT3 activation, indicating that PTPε is crucial for Capz-dependent STAT3 dephosphorylation. Capz also decreased levels of the protein products of various oncogenes, which in turn inhibited proliferation and invasion and induced apoptosis. Finally, intraperitoneal Capz administration decreased tumor growth in a xenograft mouse prostate cancer model and reduced p-STAT3 and Ki-67 expression. These data suggest that Capz is a novel pharmacological inhibitor of STAT3 activation with several anticancer effects in prostate cancer cells.
Collapse
|
41
|
Wang X, Du Q, Mao Z, Fan X, Hu B, Wang Z, Chen Z, Jiang X, Wang Z, Lei N, Wang H, Zhu Y. Combined treatment with artesunate and bromocriptine has synergistic anticancer effects in pituitary adenoma cell lines. Oncotarget 2017; 8:45874-45887. [PMID: 28501857 PMCID: PMC5542234 DOI: 10.18632/oncotarget.17437] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 04/11/2017] [Indexed: 12/25/2022] Open
Abstract
Prolactinomas are the most prevalent functional pituitary adenomas. The preferred treatments for prolactinomas are dopamine agonists (DAs) such as bromocriptine (BRC), but DAs still have the challenges of tumor recurrence and drug resistance. This study demonstrates that the synergy of function and mechanism between artesunate (ART) and BRC inhibits prolactinoma cell growth in vitro. We found that low-dose ART combined with BRC synergistically inhibited the growth of GH3 and MMQ cell lines, caused cell death, attenuated cell migration and invasion, and suppressed the expression of extracellular prolactin. The induction of apoptosis after co-treatment was confirmed by immunofluorescent staining, assessment of caspase-3 protein expression, and flow cytometry. Expression of miR-200c, a carcinogenic factor in pituitary adenoma, was reduced following co-treatment with ART and BRC. This was accompanied by increased expression of the antitumor factor Pten. Transfection experiments with miR-200c analogs and inhibitors confirmed that miR-200c expression was inversely associated with Pten expression. We suggest that ART and BRC used in combination exert synergistic apoptotic and antitumor effects by suppressing miR-200c and stimulating Pten expression.
Collapse
Affiliation(s)
- Xin Wang
- Department of Histology and Embryology, Medical School of Sun Yat-Sen University, Guangzhou, China
- Department of Neurosurgery and Pituitary Tumour Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Qiu Du
- Department of Histology and Embryology, Medical School of Sun Yat-Sen University, Guangzhou, China
| | - Zhigang Mao
- Department of Neurosurgery and Pituitary Tumour Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiang Fan
- Department of Neurosurgery, The Fifth Affiliated Hospital of Sun Yat-Sen University, Zhuhai, China
| | - Bin Hu
- Department of Neurosurgery and Pituitary Tumour Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zhen Wang
- Department of Histology and Embryology, Medical School of Sun Yat-Sen University, Guangzhou, China
| | - Zhiyong Chen
- Department of Neurosurgery and Pituitary Tumour Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaobing Jiang
- Department of Neurosurgery and Pituitary Tumour Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Zongming Wang
- Department of Neurosurgery and Pituitary Tumour Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Ni Lei
- Department of Histology and Embryology, Medical School of Sun Yat-Sen University, Guangzhou, China
| | - Haijun Wang
- Department of Neurosurgery and Pituitary Tumour Center, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yonghong Zhu
- Department of Histology and Embryology, Medical School of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
42
|
Wong YK, Xu C, Kalesh KA, He Y, Lin Q, Wong WSF, Shen HM, Wang J. Artemisinin as an anticancer drug: Recent advances in target profiling and mechanisms of action. Med Res Rev 2017. [PMID: 28643446 DOI: 10.1002/med.21446] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Artemisinin and its derivatives (collectively termed as artemisinins) are among the most important and effective antimalarial drugs, with proven safety and efficacy in clinical use. Beyond their antimalarial effects, artemisinins have also been shown to possess selective anticancer properties, demonstrating cytotoxic effects against a wide range of cancer types both in vitro and in vivo. These effects appear to be mediated by artemisinin-induced changes in multiple signaling pathways, interfering simultaneously with multiple hallmarks of cancer. Great strides have been taken to characterize these pathways and to reveal their anticancer mechanisms of action of artemisinin. Moreover, encouraging data have also been obtained from a limited number of clinical trials to support their anticancer property. However, there are several key gaps in knowledge that continue to serve as significant barriers to the repurposing of artemisinins as effective anticancer agents. This review focuses on important and emerging aspects of this field, highlighting breakthroughs in unresolved questions as well as novel techniques and approaches that have been taken in recent studies. We discuss the mechanism of artemisinin activation in cancer, novel and significant findings with regards to artemisinin target proteins and pathways, new understandings in artemisinin-induced cell death mechanisms, as well as the practical issues of repurposing artemisinin. We believe these will be important topics in realizing the potential of artemisinin and its derivatives as safe and potent anticancer agents.
Collapse
Affiliation(s)
- Yin Kwan Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chengchao Xu
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Karunakaran A Kalesh
- Department of Chemical Engineering, Imperial College London, London, United Kingdom
| | - Yingke He
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Qingsong Lin
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jigang Wang
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
43
|
Kumar B, Kalvala A, Chu S, Rosen S, Forman SJ, Marcucci G, Chen CC, Pullarkat V. Antileukemic activity and cellular effects of the antimalarial agent artesunate in acute myeloid leukemia. Leuk Res 2017. [PMID: 28646646 DOI: 10.1016/j.leukres.2017.05.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The artimisinins are a class of antimalarial compounds whose antiparasitic activity is mediated by induction of reactive oxygen species (ROS). Herein, we report that among the artimisinins, artesunate (ARTS), an orally bioavailable compound has the most potent antileukemic activity in AML models and primary patients' blasts. ARTS was most cytotoxic to the FLT3-ITD+ AML MV4-11 and MOLM-13 cells (IC50 values of 1.1 and 0.82μM respectively), inhibited colony formation in primary AML and MDS cells and augmented cytotoxicity of chemotherapeutics. ARTS lowered cellular BCL-2 level via ROS induction and increased the cytotoxicity of the BCL-2 inhibitor venetoclax (ABT-199). ARTS treatment led to cellular and mitochondrial ROS accumulation, double stranded DNA damage, loss of mitochondrial membrane potential and induction of the intrinsic mitochondrial apoptotic cascade in AML cell lines. The antileukemic activity of ARTS was further confirmed in MV4-11 and FLT3-ITD+ primary AML cell xenografts as well as MLL-AF9 syngeneic murine AML model where ARTS treatment resulted in significant survival prolongation of treated mice compared to control. Our results demonstrate the potent preclinical antileukemic activity of ARTS as well as its potential for a rapid transition to a clinical trial either alone or in combination with conventional chemotherapy or BCL-2 inhibitor, for treatment of AML.
Collapse
Affiliation(s)
- Bijender Kumar
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA; Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute, Duarte, CA, USA; Gehr Family Center for Leukemia Research City of Hope Medical Center, Duarte, CA 91010, USA
| | - Arjun Kalvala
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA; Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute, Duarte, CA, USA; Gehr Family Center for Leukemia Research City of Hope Medical Center, Duarte, CA 91010, USA
| | - Su Chu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA; Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute, Duarte, CA, USA; Gehr Family Center for Leukemia Research City of Hope Medical Center, Duarte, CA 91010, USA
| | - Steven Rosen
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA; Gehr Family Center for Leukemia Research City of Hope Medical Center, Duarte, CA 91010, USA
| | - Stephen J Forman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA; Gehr Family Center for Leukemia Research City of Hope Medical Center, Duarte, CA 91010, USA
| | - Guido Marcucci
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA; Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute, Duarte, CA, USA; Gehr Family Center for Leukemia Research City of Hope Medical Center, Duarte, CA 91010, USA
| | - Ching-Cheng Chen
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA; Division of Hematopoietic Stem Cell and Leukemia Research, Beckman Research Institute, Duarte, CA, USA; Gehr Family Center for Leukemia Research City of Hope Medical Center, Duarte, CA 91010, USA
| | - Vinod Pullarkat
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Duarte, CA 91010, USA; Gehr Family Center for Leukemia Research City of Hope Medical Center, Duarte, CA 91010, USA.
| |
Collapse
|
44
|
Li Q, Ni W, Deng Z, Liu M, She L, Xie Q. Targeting nasopharyngeal carcinoma by artesunate through inhibiting Akt/mTOR and inducing oxidative stress. Fundam Clin Pharmacol 2017; 31:301-310. [PMID: 28078787 DOI: 10.1111/fcp.12266] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2016] [Revised: 12/29/2016] [Accepted: 01/09/2017] [Indexed: 01/03/2023]
Abstract
Drug repurposing has become an alternative therapeutic strategy for cancer treatment given the known pharmacokinetics and toxicity. The inhibitory effects of artesunate have been reported in various cancers. In this work, we investigated the effects of artesunate in nasopharyngeal carcinoma (NPC). We demonstrate that artesunate significantly inhibits proliferation via arresting NPC cells at G2/M phase. It also induces apoptosis through caspase-dependent and mitochondria-independent pathways in multiple NPC cell lines. The combination of artesunate and cisplatin is synergistic in targeting NPC cells in in vitro cellular culture system and in vivo xenograft tumor models. Artesunate inhibits phosphorylation of essential molecules involved in Akt/mTOR pathway in NPC cells, such as Akt, mTOR, and 4EBP1, and its inhibitory effects are partially abolished by overexpression of constitutively active Akt. In addition, artesunate also induces mitochondrial dysfunction and oxidative stress via inhibiting mitochondrial respiration, increasing levels of mitochondrial superoxide and cellular reactive oxygen species (ROS), leading to decreased ATP levels. Two ROS scavengers partially abolish the inhibitory effects of artesunate in NPC cells. These data suggest that both inhibition of Akt/mTOR pathway and induction of ROS are required for the action of artesunate in NPC cells. Our work demonstrates that artesunate is a potential candidate for NPC treatment. Our work also highlights the critical roles of Akt/mTOR pathway and mitochondrial function in NPC cells.
Collapse
Affiliation(s)
- Qin Li
- Otolaryngology-Head and Neck Surgery, The Second Clinical Medical College, Yangtze University, Renmin Road 1, Jing Zhou, 434020, China
| | - Wei Ni
- Otolaryngology-Head and Neck Surgery, The Second Clinical Medical College, Yangtze University, Renmin Road 1, Jing Zhou, 434020, China
| | - Zhifeng Deng
- Otolaryngology-Head and Neck Surgery, The Second Clinical Medical College, Yangtze University, Renmin Road 1, Jing Zhou, 434020, China
| | - Minghe Liu
- Otolaryngology-Head and Neck Surgery, The Second Clinical Medical College, Yangtze University, Renmin Road 1, Jing Zhou, 434020, China
| | - Lazhi She
- Otolaryngology-Head and Neck Surgery, The Second Clinical Medical College, Yangtze University, Renmin Road 1, Jing Zhou, 434020, China
| | - Qiong Xie
- Otolaryngology-Head and Neck Surgery, The Second Clinical Medical College, Yangtze University, Renmin Road 1, Jing Zhou, 434020, China
| |
Collapse
|
45
|
Li Q, Wu Y, Zhang Y, Sun H, Lu Z, Du K, Fang S, Li W. miR-125b regulates cell progression in chronic myeloid leukemia via targeting BAK1. Am J Transl Res 2016; 8:447-459. [PMID: 27158338 PMCID: PMC4846895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 12/29/2015] [Indexed: 06/05/2023]
Abstract
Chronic myeloid leukemia (CML) is a type of malignant tumor characterized by the accumulation of a large number of immature white blood cells in the blood and bone marrow. BAK1 was predicted to be the target gene of microRNA-451 (miR-125b). The present study was designed to illustrate the mechanism of miR-125b in regulating CML via targeting BAK1. In this study, we found that the expression of miR-125b increased strongly, whereas the expression of BAK1 decreased significantly in CML patients and CML cell lines compared with healthy controls. Moreover, the luciferase report assay confirmed the interaction between miR-125b and BAK1 mRNA. After transfection of the miR-125b mimic or miR-125b inhibitor into CML cells, we found that the inhibition of miR-125b decreased the proliferation rates and promoted apoptosis with cell cycle arrest at the G0/G1 phase in both K562 and NB-4 cells, increased the expression of BAK1 and Caspase-3, and decreased the expression of Bcl-2 and c-myc; the miR-125b mimic yielded the opposite results. In addition, siBAK1 offset the suppression effect of the miR-125b inhibitor in K562 cells, indicating that miR-125b promotes these cellular processes by inhibiting the expression of BAK1. Further in vivo experiments supported these findings because miR-125b suppression reduced CML growth in mice. Taken together, our study suggests that the down-regulation of miR-125b affects the expression of BAK1, promotes cell apoptosis and inhibits cell proliferation, leading to up-regulated expression of pro-apoptosis factors, down-regulated expression of anti-apoptosis factors in the mitochondrial apoptotic pathway, and decreased tumor size and weight of CML in vivo. These results provide a potential therapeutic strategy for CML.
Collapse
Affiliation(s)
- Quan Li
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and ScienceJinzhou Road 136, Xiangyang 441053, China
| | - Yaohui Wu
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology1277 Jiefang Avenue, Wuhan 430022, China
| | - Yongkang Zhang
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and ScienceJinzhou Road 136, Xiangyang 441053, China
| | - Huapeng Sun
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and ScienceJinzhou Road 136, Xiangyang 441053, China
| | - Zhaoli Lu
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and ScienceJinzhou Road 136, Xiangyang 441053, China
| | - Ke Du
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and ScienceJinzhou Road 136, Xiangyang 441053, China
| | - Shanshan Fang
- Department of Oncology, Xiangyang Central Hospital, Hubei University of Arts and ScienceJinzhou Road 136, Xiangyang 441053, China
| | - Weiming Li
- Department of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology1277 Jiefang Avenue, Wuhan 430022, China
| |
Collapse
|
46
|
Tong H, Wang Y, Lu X, Wang P, Zhao S, Chang H, Yu X. On the preparation of transferrin modified artesunate nanoliposomes and their glioma-targeting treatment in-vitro and in-vivo. Int J Clin Exp Med 2015; 8:22045-22052. [PMID: 26885179 PMCID: PMC4729965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Accepted: 12/08/2015] [Indexed: 06/05/2023]
Abstract
OBJECTIVE To prepare transferrin modified artesunate nanoliposomes (Tf-ART-LPs) and study their glioma U87 cells-targeting treatment in-vitro and in-vivo. METHODS Ammonium sulfate transmembrane gradient method was used to prepare Tf-ART-LPs, whose size and stability was detected by a Nanosizer. Besides, the encapsulation efficiency and release rate of artesunate (ART) were tested by a ultraviolet spectrophotometer. Further, isothiocyanate (FITC) was used to label nanoliposomes and the cell-targeting property of Tf-ART-LP in-vitro was observed under a fluorescence microscope. In addition, CCK-8 method was used to detect the effect of single nanoliposomes and Tf-ART-LPs on the viability of glioma U87 cells. At last, a subcutaneously implanted tumor model in nude mouse was established for studying the in-vivo anti-tumor effect of Tf-ART-LPs by caudal vein injection. The tumor volume and mice weight were monitored and pathological sections of their major organs were analyzed. RESULTS Tf-ART-LPs were spherical with an average diameter of 94.2 nm. They showed no aggregation after being stored in a refrigerator for 14 days at 4°C. The encapsulation efficiency and highest releasing rate (48 hours after being placed in normal saline under 37°C) of ART was 85.9% and 58.7±2.9%, respectively. The uptake rate of U87 cells was 59.8±3.8% for Tf-ART-LPs and only 18.7±4.5% for ART-LPs. While single liposomes almost showed no toxicity, Tf-ART-LP had a concentration-dependent killing effect on U87 cells. Within 32 days of treatment, the growth of U87 cells was well inhibited by Tf-ART-LPs without significant toxicity. CONCLUSION In this study, transferrin modified artesunate liposomes we prepared have a good targeting property to glioma U87 cells and good effect on glioma both in-vitro and in-vivo.
Collapse
Affiliation(s)
- Huaiyu Tong
- Department of Neurosurgery, PLA General HospitalBeijing 100853, China
| | - Yan Wang
- Department of Neurosurgery, The First Affiliated Hospital of PLA General HospitalBeijing 100853, China
| | - Xuechun Lu
- Department of Hematology, PLA General HospitalBeijing 100853, China
| | - Peng Wang
- Department of Neurosurgery, PLA General HospitalBeijing 100853, China
| | - Shupeng Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical UniversityWeihui 453100, Henan Province, China
| | - Haigang Chang
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical UniversityWeihui 453100, Henan Province, China
| | - Xinguang Yu
- Department of Neurosurgery, PLA General HospitalBeijing 100853, China
| |
Collapse
|
47
|
Anusha S, Mohan CD, Ananda H, Baburajeev CP, Rangappa S, Mathai J, Fuchs JE, Li F, Shanmugam MK, Bender A, Sethi G, Basappa, Rangappa KS. Adamantyl-tethered-biphenylic compounds induce apoptosis in cancer cells by targeting Bcl homologs. Bioorg Med Chem Lett 2015; 26:1056-1060. [PMID: 26725030 DOI: 10.1016/j.bmcl.2015.12.026] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 11/12/2015] [Accepted: 12/10/2015] [Indexed: 02/05/2023]
Abstract
Bcl homologs prominently contribute to apoptotic resistance in cancer cells and serve as molecular targets in treatment of various cancers. Herein, we report the synthesis of biphenyl-adamantane derivatives by a ligand free palladium on carbon based Suzuki reaction using diisopropylamine as a base for the coupling of adamantane based aryl chloride with a variety of aryl boronic acids. Among the biphenyl derivatives synthesized, compound 3'-(adamantan-1-yl)-4'-methoxy-[1,1'-biphenyl]-3-ol (AMB) displayed cytotoxic activity against hepatocellular carcinoma cell lines without significantly affecting the normal cell lines. Further, AMB caused increased accumulation of the HCC cells in subG1 phase, decreased the expression of Bcl-2, Bcl-xL, cyclin D1, caspase-3, survivin and increased the cleavage of PARP in a time-dependent manner. In silico molecular interaction studies between Bcl homologs and AMB showed that the biphenyl scaffold is predicted to form π-π interactions with Phe-101 and Tyr-105 and the adamantyl fragment is predicted to occupy another hydrophobic region in the kink region of the binding groove. In summary, we report on the synthesis and biological characterization of adamantyl-tethered biphenylic compounds that induce apoptosis in tumor cells most likely by targeting Bcl homologs.
Collapse
Affiliation(s)
- Sebastian Anusha
- Laboratory of Chemical Biology, Department of Chemistry, Bangalore University, Central College Campus, Palace Road, Bangalore 560001, India
| | | | - Hanumappa Ananda
- Department of Studies in Chemistry, Manasagangotri, University of Mysore, Mysore 570006, India
| | - C P Baburajeev
- Laboratory of Chemical Biology, Department of Chemistry, Bangalore University, Central College Campus, Palace Road, Bangalore 560001, India
| | - Shobith Rangappa
- Frontier Research Center for Post-Genome Science and Technology, Hokkaido University, Sapporo 060-0808, Japan
| | - Jessin Mathai
- Gulf Medical University, Ajman, United Arab Emirates
| | - Julian E Fuchs
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom; Institute of General, Inorganic and Theoretical Chemistry, Department of Chemistry, University of Innsbruck, Innrain 82, 6020 Innsbruck, Austria
| | - Feng Li
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600
| | - Andreas Bender
- Centre for Molecular Informatics, Department of Chemistry, University of Cambridge, Lensfield Road, CB2 1EW Cambridge, United Kingdom
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600
| | - Basappa
- Laboratory of Chemical Biology, Department of Chemistry, Bangalore University, Central College Campus, Palace Road, Bangalore 560001, India.
| | | |
Collapse
|