1
|
Neale DA, Morris JC, Verrills NM, Ammit AJ. Understanding the regulatory landscape of protein phosphatase 2A (PP2A): Pharmacological modulators and potential therapeutics. Pharmacol Ther 2025; 269:108834. [PMID: 40023321 DOI: 10.1016/j.pharmthera.2025.108834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/20/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
Protein phosphatase 2A (PP2A) is a ubiquitously expressed serine/threonine phosphatase with a diverse and integral role in cellular signalling pathways. Consequently, its dysfunction is frequently observed in disease states such as cancer, inflammation and Alzheimer's disease. A growing understanding of both PP2A and its endogenous regulatory proteins has presented numerous targets for therapeutic intervention. This provides important context for the dynamic control and dysregulation of PP2A function in disease states. Understanding the intricate regulation of PP2A signalling in disease has resulted in the development of novel pharmacological agents aimed at restoring cellular homeostasis. Herein we review the structure and function of PP2A together with pharmacological modulators, both endogenous (proteins) and exogenous (small molecules and peptides), with relevance to targeting PP2A as a future pharmacotherapeutic strategy.
Collapse
Affiliation(s)
- David A Neale
- School of Chemistry, UNSW Sydney, NSW 2052, Australia
| | | | - Nicole M Verrills
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, NSW 2308, Australia; Precision Medicine Program, Hunter Medical Research Institute, New Lambton, NSW 2305, Australia
| | - Alaina J Ammit
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, Macquarie University, NSW, Australia; School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW, Australia.
| |
Collapse
|
2
|
Bidula S, Piyasirananda W, Bielecka H, Bibič L, Beekman A, Stokes L. Screening herbal and natural product libraries to aid discovery of novel allosteric modulators of human P2X7. Purinergic Signal 2025; 21:365-379. [PMID: 39436616 PMCID: PMC12062478 DOI: 10.1007/s11302-024-10055-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 09/30/2024] [Indexed: 10/23/2024] Open
Abstract
P2X7 is an emerging therapeutic target for several disorders and diseases due to its role in inflammatory signalling. This study aimed to exploit the unique chemical libraries of plants used in traditional medicinal practices to discover novel allosteric modulators from natural sources. We identified several compounds from the NCI Natural Product library as P2X7 antagonists including confertifolin and digallic acid (IC50 values 3.86 µM and 4.05 µM). We also identified scopafungin as a novel positive allosteric modulator of hP2X7. Screening a traditional medicinal plant extract library revealed 39 plant species with inhibitory action at hP2X7 and 17 plant species with positive allosteric modulator activity. Using computational docking to filter identified components from these plant species and determine potential antagonists, we investigated nine purified chemicals including flavonoids quercetin, kaempferol, ECG, and EGCG. These were shown to inhibit ATP-induced YO-PRO-1 uptake into HEK-hP2X7 cells; however, we also showed that all four flavonoids demonstrated significant assay interference using a cell-free DNA YO-PRO-1 fluorescence test. One plant extract, Dioscorea nipponica, demonstrating positive modulator activity was investigated, and dioscin was identified as a glycoside with PAM activity in ATP-induced YO-PRO-1 uptake assay and whole-cell patch-clamp recordings. However, membrane permeabilisation was observed following application > 10 min limiting the use of dioscin as a pharmacological tool. This work describes a useful workflow with multiple assays for the identification of novel allosteric modulators for human P2X7.
Collapse
Affiliation(s)
- Stefan Bidula
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Waraporn Piyasirananda
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Hanna Bielecka
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Lučka Bibič
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Andrew Beekman
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK
| | - Leanne Stokes
- School of Pharmacy, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| |
Collapse
|
3
|
Chen M, Qian C, Jin B, Hu C, Zhang L, Wang M, Zhou B, Zuo W, Huang L, Wang Y. Curcumin analog WZ26 induces ROS and cell death via inhibition of STAT3 in cholangiocarcinoma. Cancer Biol Ther 2023; 24:2162807. [PMID: 36647192 PMCID: PMC9851268 DOI: 10.1080/15384047.2022.2162807] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Cholangiocarcinoma (CCA) is an aggressive biliary epithelial tumor with limited therapeutic options and poor prognosis. Curcumin is a promising active natural compound with several anti-cancer properties, though its clinical uses remain hindered due to its poor bioavailability. We recently synthesized curcumin analogs with multifunctional pharmacological and bioactivities with enhanced bioavailability. Among these novel curcumin analogs, WZ26 is a representative molecule. However, the anti-tumor effect of WZ26 against CCA is unclear. In this study, we evaluated the anti-tumor effect of WZ26 in both CCA cells and CCA xenograft mouse model. The underlying molecular anti-cancer mechanism of WZ26 was also studied. Our results show that WZ26 significantly inhibited cell growth and induced mitochondrial apoptosis in CCA cell lines, leading to significant inhibition of tumor growth in xenograft tumor mouse model. Treatment of WZ26 increased reactive oxygen species (ROS) generation, subsequently decreased mitochondrial membrane potential and inhibited the phosphorylation of signal transducer and activator of transcription 3 (STAT3), thereby inducing G2/M cell cycle arrest and cell apoptosis. Pretreatment of N-acetyl cysteine (NAC), an antioxidant agent, could fully reverse the WZ26-induced ROS-mediated changes in CCA cells. Our findings provide experimental evidence that curcumin analog WZ26 could be a potential candidate against CCA via enhancing ROS induction and inhibition of STAT3 activation.
Collapse
Affiliation(s)
- Minxiao Chen
- Department of Gastroenterology, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, China,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Chenchen Qian
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Bo Jin
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Chenghong Hu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Lingxi Zhang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Minshan Wang
- Department of Pharmacy, the First Hospital of Xiangshan, Ningbo, China
| | - Bin Zhou
- Department of Hepatopancreatobiliary Surgery, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wei Zuo
- Department of Gastroenterology, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, China
| | - Lijiang Huang
- Department of Gastroenterology, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, China,Lijiang HuangThe Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, China
| | - Yi Wang
- Department of Gastroenterology, the Affiliated Xiangshan Hospital of Wenzhou Medical University, Ningbo, China,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China,CONTACT Yi Wang
| |
Collapse
|
4
|
Li Y, Li Y, Yao Y, Li H, Gao C, Sun C, Zhuang J. Potential of cucurbitacin as an anticancer drug. Biomed Pharmacother 2023; 168:115707. [PMID: 37862969 DOI: 10.1016/j.biopha.2023.115707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 10/03/2023] [Accepted: 10/11/2023] [Indexed: 10/22/2023] Open
Abstract
In Chinese medicine, the Cucurbitaceae family contains many compounds known as cucurbitacins, which have been categorized into 12 classes ranging from A to T and more than 200 derivatives. Cucurbitacins are a class of highly oxidized tetracyclic triterpenoids with potent anticancer properties. The eight components of cucurbitacins with the strongest anticancer activity are cucurbitacins B, D, E, I, IIa, L-glucoside, Q, and R. Cucurbitacins have also been reported to suppress JAK-STAT 3, mTOR, VEGFR, Wnt/β-catenin, and MAPK signaling pathways, all of which are crucial for the survival and demise of cancer cells. In this paper, we review the progress in research on cucurbitacin-induced apoptosis, autophagy, cytoskeleton disruption, cell cycle arrest, inhibition of cell proliferation, inhibition of invasion and migration, inhibition of angiogenesis, epigenetic alterations, and synergistic anticancer effects in tumor cells. Recent studies have identified cucurbitacins as promising molecules for therapeutic innovation with broad versatility in immune response. Thus, cucurbitacin is a promising class of anticancer agents that can be used alone or in combination with chemotherapy and radiotherapy for the treatment of many types of cancer.Therefore, based on the research reports in the past five years at home and abroad, we further summarize and review the structural characteristics, chemical and biological activities, and studies of cucurbitacins based on the previous studies to provide a reference for further development and utilization of cucurbitacins.
Collapse
Affiliation(s)
- Yan Li
- College of Chinese Medicine, Weifang Medical University, Weifang, China
| | - Yingrui Li
- College of Chinese Medicine, Weifang Medical University, Weifang, China
| | - Yan Yao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250022, China
| | - Huayao Li
- College of Chinese Medicine, Weifang Medical University, Weifang, China
| | - Chundi Gao
- College of Chinese Medicine, Weifang Medical University, Weifang, China
| | - Changgang Sun
- College of Chinese Medicine, Weifang Medical University, Weifang, China; Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261000, China
| | - Jing Zhuang
- Department of Oncology, Weifang Traditional Chinese Hospital, Weifang 261000, China.
| |
Collapse
|
5
|
Zieniuk B, Pawełkowicz M. Recent Advances in the Application of Cucurbitacins as Anticancer Agents. Metabolites 2023; 13:1081. [PMID: 37887406 PMCID: PMC10608718 DOI: 10.3390/metabo13101081] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 10/08/2023] [Accepted: 10/12/2023] [Indexed: 10/28/2023] Open
Abstract
Cucurbitacins are tetracyclic triterpenoid secondary metabolites, widely distributed in the Cucurbitaceae family. These bitter-tasting compounds act primarily as defense mechanisms against external injuries, and thus against herbivores, and furthermore, they have also found use in folk medicine in the treatment of various diseases. Many studies have acknowledged significant biological activities of cucurbitacins, such as antioxidant and anti-inflammatory activities, antimicrobial properties, or antitumor potential. Overall, cucurbitacins have the ability to inhibit cell proliferation and induce apoptosis in various cancer cell lines. Both in vitro and in vivo studies were performed to evaluate the anticancer activity of varied cucurbitacins. Cucurbitacins offer a promising avenue for future cancer treatment strategies, and their diverse mechanisms of action make them attractive candidates for further investigation. The aim of the present study is to shed light on the chemical diversity of this group of compounds by providing the sources of origin of selected compounds and their chemical structure, as well as insight into their anticancer potential. In addition, within this paper molecular targets for cucurbitacins and signalling pathways important for cancer cell proliferation and/or survival that are affected by the described class of compounds have been presented.
Collapse
Affiliation(s)
- Bartłomiej Zieniuk
- Department of Chemistry, Institute of Food Sciences, Warsaw University of Life Sciences, 02-776 Warsaw, Poland;
| | - Magdalena Pawełkowicz
- Department of Plant Genetics, Breeding and Biotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-776 Warsaw, Poland
| |
Collapse
|
6
|
Kumar A, Sharma B, Sharma U, Parashar G, Parashar NC, Rani I, Ramniwas S, Kaur S, Haque S, Tuli HS. Apoptotic and antimetastatic effect of cucurbitacins in cancer: recent trends and advancement. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:1867-1878. [PMID: 37010571 DOI: 10.1007/s00210-023-02471-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 03/21/2023] [Indexed: 04/04/2023]
Abstract
The Cucurbitaceae family produces a class of secondary metabolites known as cucurbitacins. The eight cucurbitacin subunits are cucurbitacin B, D, E, I, IIa, L glucoside, Q, and R with the most significant anticancer activity. They are reported to inhibit cell proliferation, invasion, and migration; induce apoptosis; and encourage cell cycle arrest, as some of their modes of action. The JAK-STAT3, Wnt, PI3K/Akt, and MAPK signaling pathways, which are essential for the survival and apoptosis of cancer cells, have also been shown to be suppressed by cucurbitacins. The goal of the current study is to summarize potential molecular targets that cucurbitacins could inhibit in order to suppress various malignant processes. The review is noteworthy since it presents all putative molecular targets for cucurbitacins in cancer on a single podium.
Collapse
Affiliation(s)
- Ajay Kumar
- Punjab Biotechnology Incubator (PBTI), Phase VIII, Mohali, 160071, India
| | - Bunty Sharma
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India
| | - Ujjawal Sharma
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Gaurav Parashar
- Division of Biomedical and Life Sciences, School of Science, Navrachana University Vadodara, Gujrat, 391410, India
| | - Nidarshana Chaturvedi Parashar
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India
| | - Isha Rani
- Department of Biochemistry, maharishi markendashwar college of medical sciences and Research (MMCMSR), Sadopur, Ambala, 134007, India
| | - Seema Ramniwas
- University Centre for Research and Development, University Institute of Pharmaceutical Sciences, Chandigarh University, Gharuan, Mohali, 140413, India
| | - Satwinderjeet Kaur
- Department of Botanical and Environmental Sciences, Guru Nanak Dev University, Amritsar, 143005, India
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, 45142, Saudi Arabia
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, 133207, India.
| |
Collapse
|
7
|
Standing D, Feess E, Kodiyalam S, Kuehn M, Hamel Z, Johnson J, Thomas SM, Anant S. The Role of STATs in Ovarian Cancer: Exploring Their Potential for Therapy. Cancers (Basel) 2023; 15:2485. [PMID: 37173951 PMCID: PMC10177275 DOI: 10.3390/cancers15092485] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/12/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Ovarian cancer (OvCa) is a deadly gynecologic malignancy that presents many clinical challenges due to late-stage diagnoses and the development of acquired resistance to standard-of-care treatment protocols. There is an increasing body of evidence suggesting that STATs may play a critical role in OvCa progression, resistance, and disease recurrence, and thus we sought to compile a comprehensive review to summarize the current state of knowledge on the topic. We have examined peer reviewed literature to delineate the role of STATs in both cancer cells and cells within the tumor microenvironment. In addition to summarizing the current knowledge of STAT biology in OvCa, we have also examined the capacity of small molecule inhibitor development to target specific STATs and progress toward clinical applications. From our research, the best studied and targeted factors are STAT3 and STAT5, which has resulted in the development of several inhibitors that are under current evaluation in clinical trials. There remain gaps in understanding the role of STAT1, STAT2, STAT4, and STAT6, due to limited reports in the current literature; as such, further studies to establish their implications in OvCa are necessitated. Moreover, due to the deficiency in our understanding of these STATs, selective inhibitors also remain elusive, and therefore present opportunities for discovery.
Collapse
Affiliation(s)
- David Standing
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Emma Feess
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Satvik Kodiyalam
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Michael Kuehn
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Zachary Hamel
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Jaimie Johnson
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| | - Sufi Mary Thomas
- Department of Otolaryngology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Shrikant Anant
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS 66103, USA
| |
Collapse
|
8
|
Tuli HS, Rath P, Chauhan A, Ranjan A, Ramniwas S, Sak K, Aggarwal D, Kumar M, Dhama K, Lee EHC, Yap KCY, Capinpin SM, Kumar AP. Cucurbitacins as Potent Chemo-Preventive Agents: Mechanistic Insight and Recent Trends. Biomolecules 2022; 13:57. [PMID: 36671442 PMCID: PMC9855938 DOI: 10.3390/biom13010057] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 12/29/2022] Open
Abstract
Cucurbitacins constitute a group of cucumber-derived dietary lipids, highly oxidized tetracyclic triterpenoids, with potential medical uses. These compounds are known to interact with a variety of recognized cellular targets to impede the growth of cancer cells. Accumulating evidence has suggested that inhibition of tumor cell growth via induction of apoptosis, cell-cycle arrest, anti-metastasis and anti-angiogenesis are major promising chemo-preventive actions of cucurbitacins. Cucurbitacins may be a potential choice for investigations of synergism with other drugs to reverse cancer cells' treatment resistance. The detailed molecular mechanisms underlying these effects include interactions between cucurbitacins and numerous cellular targets (Bcl-2/Bax, caspases, STAT3, cyclins, NF-κB, COX-2, MMP-9, VEGF/R, etc.) as well as control of a variety of intracellular signal transduction pathways. The current study is focused on the efforts undertaken to find possible molecular targets for cucurbitacins in suppressing diverse malignant processes. The review is distinctive since it presents all potential molecular targets of cucurbitacins in cancer on one common podium.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala 133207, India
| | - Prangya Rath
- Amity Institute of Environmental Sciences, Amity University, Noida 201303, India
| | - Abhishek Chauhan
- Amity Institute of Environmental Toxicology, Safety and Management, Amity University, Noida 201303, India
| | - Anuj Ranjan
- Academy of Biology and Biotechnology, Southern Federal University, 344090 Rostov-on-Don, Russia
| | - Seema Ramniwas
- University Centre for Research and Development, University Institute of Pharmaceutical Sciences, Chandigarh University, Mohali 140413, India
| | | | - Diwakar Aggarwal
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala 133207, India
| | - Manoj Kumar
- Department of Chemistry, Maharishi Markandeshwar University Sadopur, Ambala 134007, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly 243122, India
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Kenneth Chun-Yong Yap
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Sharah Mae Capinpin
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
| |
Collapse
|
9
|
A comprehensive review on the botany, traditional uses, phytochemistry, pharmacology and toxicity of Anagallis arvensis (L).: A wild edible medicinal food plant. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.102328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
10
|
Nisar S, Masoodi T, Prabhu KS, Kuttikrishnan S, Zarif L, Khatoon S, Ali S, Uddin S, Akil AAS, Singh M, Macha MA, Bhat AA. Natural products as chemo-radiation therapy sensitizers in cancers. Biomed Pharmacother 2022; 154:113610. [PMID: 36030591 DOI: 10.1016/j.biopha.2022.113610] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/24/2022] [Accepted: 08/24/2022] [Indexed: 11/02/2022] Open
Abstract
Cancer is a devastating disease and is the second leading cause of death worldwide. Surgery, chemotherapy (CT), and/or radiation therapy (RT) are the treatment of choice for most advanced tumors. Unfortunately, treatment failure due to intrinsic and acquired resistance to the current CT and RT is a significant challenge associated with poor patient prognosis. There is an urgent need to develop and identify agents that can sensitize tumor cells to chemo-radiation therapy (CRT) with minimal cytotoxicity to the healthy tissues. While many recent studies have identified the underlying molecular mechanisms and therapeutic targets for CRT failure, using small molecule inhibitors to chemo/radio sensitize tumors is associated with high toxicity and increased morbidity. Natural products have long been used as chemopreventive agents in many cancers. Combining many of these compounds with the standard chemotherapeutic agents or with RT has shown synergistic effects on cancer cell death and overall improvement in patient survival. Based on the available data, there is strong evidence that natural products have a robust therapeutic potential along with CRT and their well-known chemopreventive effects in many solid tumors. This review article reports updated literature on different natural products used as CT or RT sensitizers in many solid tumors. This is the first review discussing CT and RT sensitizers together in cancer.
Collapse
Affiliation(s)
- Sabah Nisar
- Depertment of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Tariq Masoodi
- Laboratory of Cancer immunology and genetics, Sidra Medicine, Qatar
| | - Kirti S Prabhu
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Qatar
| | - Shilpa Kuttikrishnan
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Qatar
| | - Lubna Zarif
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Qatar
| | - Summaiya Khatoon
- Depertment of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Shahid Ali
- International Potato Center (CIP), Shillong, Meghalaya, India
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Qatar; Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Ammira Al-Shabeeb Akil
- Depertment of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar
| | - Mayank Singh
- Department of Medical Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, AIIMS, New Delhi, India.
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora, Jammu & Kashmir, India.
| | - Ajaz A Bhat
- Depertment of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, Doha, Qatar.
| |
Collapse
|
11
|
Cucurbitacin B exhibits antitumor effects on CD133+ HepG2 liver cancer stem cells by inhibiting JAK2/STAT3 signaling pathway. Anticancer Drugs 2021; 32:548-557. [PMID: 33675610 DOI: 10.1097/cad.0000000000001062] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Cancer stem cells (CSCs), a crucial cancer cell subpopulation, possess stemness phenotypic characteristics. Cucurbitacin B (CuB), a tetracyclic triterpenoid isolated from Cucurbitaceae, exerts widely pharmacological activities in many diseases. The aim of this study was to enrich, identify liver CSCs and investigate antitumor effects of CuB as well as explore the underlying molecular mechanisms in these liver CSCs. HepG2 cell lines were used for the enrichment of liver CSCs by serum-free medium culture and magnetic-activated cell sorting. The CSC characteristics were analyzed by immunofluorescent staining, sphere-forming, western blot and xenograft tumorigenicity assay. CuB' antitumor effects and underlying molecular mechanism were measured by cell counting kit-8, colony formation, sphere-forming, cell cycle, xenograft and western blot assay. Our results showed that we could enrich 97.29% CD133+ HepG2 cells, which possessed CSC characteristics including re-renewal capacity, proliferative ability, sorafenib resistance, overexpressed stemness-related molecules and enhanced tumorigenic potential. Furthermore, we also found that CuB inhibited cell viability, sphere formation, colony formation and arrested cell cycle at G2/M phase as well as sensitized CD133+ HepG2 cells to sorafenib in vitro and in vivo. Western blot assay indicated that CuB inhibited expression levels of cyclin B1, CDK1, CD133, p-JAK2 and p-STAT3. In conclusion, our findings indicated that CuB could exhibit antitumor effects on CD133+ HepG2 CSCs by inhibiting the Janus kinase 2/signal transducers and activators of transcription-3 signaling pathway, expanding basic and preclinical investigations on liver CSCs.
Collapse
|
12
|
Han W, Niu L, Wang L, Liu J, Li H. Downregulation of long non-coding RNA B-Raf proto-oncogene-activated non-coding RNA reverses cisplatin resistance in laryngeal squamous cell carcinoma. Arch Med Sci 2021; 17:1164-1174. [PMID: 34522245 PMCID: PMC8425235 DOI: 10.5114/aoms.2019.91352] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 10/14/2018] [Indexed: 12/30/2022] Open
Abstract
INTRODUCTION This study was performed to explore the function of B-Raf proto-oncogene-activated non-coding RNA (BANCR) in laryngeal squamous cell carcinoma (LSCC) and cisplatin resistance. MATERIAL AND METHODS The relative expression level of long non-coding RNA (lncRNA) BANCR was examined by qRT-PCR in tumor tissues and adjacent tissues, normal laryngeal cells (Het-1A) and laryngeal squamous carcinoma cells (TU686, TU177). Cisplatin-resistant laryngeal squamous carcinoma cell lines (TU686-DDP-R, TU177-DDP-R) were established. Next, we inhibited BANCR expression by transfecting siRNA-BANCR and enhanced BANCR expression by transfecting pcDNA3.1-BANCR into TU686, TU177, TU686-DDP-R and TU177-DDP-R cells. The CCK-8 assay and clone formation assay were performed to detect colony proliferation ability and formation ability of cells. Further, to investigate through which BANCR cell viability/formation is regulated, we detected the expression of MRP1, Bcl-2, p-PKB, and Bax by western blot. RESULTS BANCR was highly expressed in laryngeal squamous carcinoma tissues and cells. Chemoresistance was generated in TU686-DDP-R and TU177-DDP-R compared with TU686 and TU177 cells after cisplatin treatment. In addition, upregulated lncRNA BANCR reduced or postponed cell sensitivity to cisplatin by enhancing cell proliferation in TU686 and TU177 cells. Meanwhile, the expression of MRP1, Bcl-2, and p-PKB was increased, while Bax was reduced. After cisplatin treatment, down-regulation of BANCR could consequently attenuate TU686-DDP-R and TU177-DDP-R cell proliferation, and the expression of MRP1, Bcl-2, and p-PKB was decreased and Bax was increased. CONCLUSIONS Down-regulation of BANCR reverses cisplatin resistance of cisplatin-resistant LSCC cell lines.
Collapse
Affiliation(s)
- Weiwei Han
- Department of Otolaryngology Head and Neck Surgery, Tianjin Union Medicine Centre, Tianjin, China
| | - Lin Niu
- Department of Otolaryngology Head and Neck Surgery, Tianjin Union Medicine Centre, Tianjin, China
| | - Lin Wang
- Department of Otolaryngology Head and Neck Surgery, Tianjin Union Medicine Centre, Tianjin, China
| | - Jixiang Liu
- Department of Otolaryngology Head and Neck Surgery, Tianjin Union Medicine Centre, Tianjin, China
| | - Huanying Li
- Department of Otolaryngology Head and Neck Surgery, Tianjin Union Medicine Centre, Tianjin, China
| |
Collapse
|
13
|
İbiloglu İ, Alabalik U, Keles AN, Aydogdu G, Basuguy E, Buyukbayram H. Ecballium elaterium extract reduces fibrosis during wound healing in rats. BIOTECHNOL BIOTEC EQ 2021. [DOI: 10.1080/13102818.2021.1920847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- İbrahim İbiloglu
- Department of Pathology, School of Medicine, Dicle University, Diyarbakir, Turkey
| | - Ulas Alabalik
- Department of Pathology, School of Medicine, Dicle University, Diyarbakir, Turkey
| | - Ayse Nur Keles
- Department of Pathology, School of Medicine, Dicle University, Diyarbakir, Turkey
| | - Gulay Aydogdu
- Department of Pathology, School of Medicine, Dicle University, Diyarbakir, Turkey
| | - Erol Basuguy
- Department of Pediatric Surgery, School of Medicine, Dicle University, Diyarbakir, Turkey
| | - Huseyin Buyukbayram
- Department of Pathology, School of Medicine, Dicle University, Diyarbakir, Turkey
| |
Collapse
|
14
|
Wang X, Li H, Li D, Bai Y, Zhang Y, Yan X, Li J, Zhao R, Liu J, Liu W, Shi M, Xu C, Yang T, Zhang T. Sorafenib and CuB exert synergistic antitumor effects against hepatocellular carcinoma cells via inhibition of STAT3 phosphorylation. FEBS Open Bio 2020; 11:133-145. [PMID: 33176070 PMCID: PMC7780105 DOI: 10.1002/2211-5463.13035] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 09/27/2020] [Accepted: 10/26/2020] [Indexed: 12/18/2022] Open
Abstract
Sorafenib, the first‐line agent for treatment of advanced hepatocellular carcinoma (HCC), improves median overall survival by approximately 3 months. In the present study, we investigated whether sorafenib combined with cucurbitacin B (CuB), a natural tetracyclic triterpenoid isolated from Cucurbitaceae, exerts enhanced antitumor effects against HCC. Cell viability and colony formation ability were detected by cell‐counting kit‐8 and colony formation assays. Cell cycle and apoptosis were analyzed by flow cytometry. Protein expression was detected by western blotting. HepG2 xenografts in nude mice were used to evaluate in vivo antitumor effects. We report that sorafenib and CuB exhibited synergistic effects on cellular proliferation inhibition and cell apoptosis induction, but not on cell cycle arrest. Furthermore, combination treatment enhanced levels of cleaved caspase 3 and cleaved caspase 9, but suppressed phosphorylation of STAT3. Epidermal growth factor, a potent stimulator of signal transducer and activator of transcription‐3 (STAT3), promoted cell viability and colony formation ability, whereas combination treatment exerted inhibitory effects on epidermal growth factor‐induced STAT3 phosphorylation. Finally, HepG2 xenograft mice cotreated with sorafenib and CuB exhibited reduced tumor progression without notable weight loss. In conclusion, sorafenib and CuB exert synergistic antitumor effects through a pathway that may involve STAT3 phosphorylation, and this may represent a promising therapeutic approach for treatment of HCC.
Collapse
Affiliation(s)
- Xiaoli Wang
- Department of Cancer, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hua Li
- Cancer Center, The General Hospital of Western Theater Command, Chengdu, China
| | - Dong Li
- Cancer Center, The General Hospital of Western Theater Command, Chengdu, China
| | - Yudi Bai
- Basic School of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Yao Zhang
- Basic School of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Xue Yan
- College of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Jin Li
- College of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Ri Zhao
- Scientific Research Center, Chengdu Medical College, Chengdu, China
| | - Jiahui Liu
- Basic School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wei Liu
- Clinical School of Medicine, Southwest Medical University, Luzhou, China
| | - Maolin Shi
- Clinical School of Medicine, Southwest Medical University, Luzhou, China
| | - Cheng Xu
- Department of Cancer, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.,Clinical School of Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tai Yang
- College of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Tao Zhang
- Cancer Center, The General Hospital of Western Theater Command, Chengdu, China
| |
Collapse
|
15
|
Combined treatment with acetazolamide and cisplatin enhances the chemosensitivity of human head and neck squamous cell carcinoma TU868 cells. Arch Oral Biol 2020; 119:104905. [PMID: 32947166 DOI: 10.1016/j.archoralbio.2020.104905] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 05/21/2020] [Accepted: 08/31/2020] [Indexed: 12/17/2022]
Abstract
AIMS To investigate whether combination of acetazolamide and cisplatin can enhance the chemosensitivity of human head and neck squamous cell carcinoma (HNSCC) cell line TU868. METHODS MTT assay was performed to determine the effect of acetazolamide, cisplatin and their combination on the proliferation of TU868 cells. Then the effect of these 2 drugs on the expression of proliferation-related and apoptosis-related proteins was detected by Western blot. Moreover, the effect of acetazolamide and cisplatin on the expression of aquaporin-1 was detected by RT-qPCR. Loss-of-function assays was performed to assess whether the effect of acetazolamide and cisplatin on TU868 cells was mediated by aquaporin-1. The effect of acetazolamide and cisplatin on tumor cell growth was confirmed in mice by testing the tumor growth size. RESULTS Acetazolamide and cisplatin treatment displayed synergistic effects on the inhibition of TU868 cell growth compared with the drugs used alone. Moreover, the acetazolamide/cisplatin combination could decrease the level of PCNA but increase the level of p53; decrease the ratio of Bcl-2/Bax and increase the expression of caspase-3 compared with the single drug treated group. Moreover, we found that the combination also significantly inhibits aquaporin-1 expression. Loss-of-function assays suggested that the anti-tumor effect of these 2 drugs was achieved via affecting aquaporin-1. Consistent with the in vitro assays, combined treatment with acetazolamide and cisplatin significantly inhibits the tumor growth in mice compared with the single drug treated group. CONCLUSION These results demonstrated that combined treatment with acetazolamide and cisplatin could synergistically inhibit the malignant development of HNSCC cells.
Collapse
|
16
|
Jing S, Zou H, Wu Z, Ren L, Zhang T, Zhang J, Wei Z. Cucurbitacins: Bioactivities and synergistic effect with small-molecule drugs. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.104042] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
17
|
Erchen Plus Huiyanzhuyu Decoction Inhibits the Growth of Laryngeal Carcinoma in a Mouse Model of Phlegm-Coagulation-Blood-Stasis Syndrome via the STAT3/Cyclin D1 Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:2803496. [PMID: 32382281 PMCID: PMC7195639 DOI: 10.1155/2020/2803496] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 02/15/2020] [Accepted: 02/19/2020] [Indexed: 01/05/2023]
Abstract
Erchen plus Huiyanzhuyu decoction (EHD), a Chinese herbal medicine (CHM) formula that consists of Erchen decoction and Huiyanzhuyu decoction, has achieved satisfactory results in the clinic. The main function of EHD is to remove phlegm and blood stasis, and EHD is suitable for phlegm-coagulation-blood-stasis (PCBS) syndrome in laryngeal cancer (LC). In this study, a xenograft mouse model of LC with PCBS syndrome was constructed by feeding a high-fat diet, swimming in ice water, and subcutaneously injecting epinephrine hydrochloride for 2 weeks. Based on the successful Chinese medicine syndrome model, Hep2-luciferase-GFP cells were injected subcutaneously under the armpit of the right upper limb in mice to form tumours. A mouse model of LC with PCBS syndrome was established via heterotopic transplantation. Then, the mice received intragastric administration of different concentrations of EHD daily, and cisplatin (DDP) was intraperitoneally injected every week for 21 days. Tumour fluorescence in mice was measured with a living animal imager on days 7, 14, 21, and 28 during treatment. The results of this experiment confirmed that a mouse model of Chinese medicine syndrome was successfully constructed. Moreover, EHD slowed the growth of xenograft tumours in nude mice; decreased the expression levels of STAT3, p-STAT3, and cyclin D1; and upregulated the expression level of P27. In brief, EHD inhibited laryngeal tumour growth in a xenograft mouse model of PCBS syndrome and regulated the STAT3/cyclin D1 signalling pathway. This study was the first to construct a Chinese medicine xenograft mouse model of LC with PCBS syndrome; in addition, this study clarified that EHD regulated the STAT3/cyclin D1 signalling pathway to inhibit the growth of LC and that EHD may be a promising novel therapeutic compound for the treatment of patients with LC.
Collapse
|
18
|
Gacem MA, Ould El Hadj-Khelil A, Boudjemaa B, Gacem H. Phytochemistry, Toxicity and Pharmacology of Pistacia lentiscus, Artemisia herba-alba and Citrullus colocynthis. ACTA ACUST UNITED AC 2020. [DOI: 10.1007/978-3-030-38881-2_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
19
|
Kurman Y, Kiliccioglu I, Dikmen AU, Esendagli G, Bilen CY, Sozen S, Konac E. Cucurbitacin B and cisplatin induce the cell death pathways in MB49 mouse bladder cancer model. Exp Biol Med (Maywood) 2020; 245:805-814. [PMID: 32252554 DOI: 10.1177/1535370220917367] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
IMPACT STATEMENT Alternative agents that will increase the effectiveness of cisplatin, which are widely used in the advanced stage and metastatic bladder cancer, are being investigated. In previous studies, Cucurbitacin B (CuB), which is a natural compound from the Cucurbitaceae family has been shown to inhibit the proliferation of tumor cells and create synergistic effects with cisplatin. In this study, we investigated the synergistic effect of CuB with cisplatin for the first time in bladder cancer in vitro and in vivo models. Our findings showed that CuB treatment with cisplatin reduced cell proliferation, and reduced tumor development through activating apoptosis and autophagy via PI3K/AKT/mTOR signaling pathway. Our results showed that CuB may be a new agent that can support conventional treatment in bladder cancer. Our study is important in terms of enlightening new pathways and developing new treatment methods in the treatment of bladder cancer.
Collapse
Affiliation(s)
- Yener Kurman
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Ilker Kiliccioglu
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Ankara 06510, Turkey.,Department of Medical Biology, Faculty of Medicine, Duzce University, Duzce 81620, Turkey
| | - Asiye U Dikmen
- Department of Public Health, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Guldal Esendagli
- Department of Pathology, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Cenk Y Bilen
- Department of Urology, Faculty of Medicine, Hacettepe University, Ankara 06100, Turkey
| | - Sinan Sozen
- Department of Urology, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| | - Ece Konac
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Ankara 06510, Turkey
| |
Collapse
|
20
|
Sun CY, Nie J, Huang JP, Zheng GJ, Feng B. Targeting STAT3 inhibition to reverse cisplatin resistance. Biomed Pharmacother 2019; 117:109135. [DOI: 10.1016/j.biopha.2019.109135] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/12/2019] [Accepted: 06/13/2019] [Indexed: 12/21/2022] Open
|
21
|
Small-molecule compounds targeting the STAT3 DNA-binding domain suppress survival of cisplatin-resistant human ovarian cancer cells by inducing apoptosis. Eur J Med Chem 2018; 157:887-897. [DOI: 10.1016/j.ejmech.2018.08.037] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 06/19/2018] [Accepted: 08/12/2018] [Indexed: 12/24/2022]
|
22
|
Gao H, Dong H, Li G, Jin H. Combined treatment with acetazolamide and cisplatin enhances chemosensitivity in laryngeal carcinoma Hep-2 cells. Oncol Lett 2018; 15:9299-9306. [PMID: 29928333 PMCID: PMC6004654 DOI: 10.3892/ol.2018.8529] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 01/17/2018] [Indexed: 12/14/2022] Open
Abstract
The aim of the present study was to determine whether acetazolamide (Ace) treatment enhances the chemosensitivity of Hep-2 laryngeal cells to cisplatin (Cis). At the logarithmic growth phase, Hep-2 cells were treated with Ace, Cis or both, and cell viability was detected using an MTT assay. The degree of apoptosis was detected using flow cytometry. Expression levels of apoptosis-related proteins, including BCL2 apoptosis regulator (bcl-2), BCL2 associated X (bax) and caspase-3, and of proliferation-related proteins, including proliferating cell nuclear antigen (PCNA) and tumor protein p53 (P53), were detected using western blotting. mRNA expression levels of aquaporin-1 (AQP1) in each group were detected using reverse transcription-polymerase chain reaction. Compared with the drugs used alone, treatment with both Ace and Cis displayed synergistic effects on the growth inhibition and apoptosis induction in Hep-2 cells. The Ace/Cis combination decreased the expression of PCNA but increased the expression of p53. In addition, the combination treatment decreased the ratio of bcl-2/bax and increased the expression of caspase-3, as well as decreased the expression of AQP1. These results demonstrated that the combined use of Ace and Cis enhanced the chemosensitivity of laryngeal carcinoma cells.
Collapse
Affiliation(s)
- Hong Gao
- Department of Head and Neck Surgery, Tumor Hospital of Jilin Province, Changchun, Jilin 130012, P.R. China
| | - Hai Dong
- Tonghua Mining Group Limited Liability Company General Hospital, Baishan, Jilin 134300, P.R. China
| | - Guijun Li
- Tonghua Mining Group Limited Liability Company General Hospital, Baishan, Jilin 134300, P.R. China
| | - Hui Jin
- Department of Head and Neck Surgery, Tumor Hospital of Jilin Province, Changchun, Jilin 130012, P.R. China
| |
Collapse
|
23
|
Yang T, Liu J, Yang M, Huang N, Zhong Y, Zeng T, Wei R, Wu Z, Xiao C, Cao X, Li M, Li L, Han B, Yu X, Li H, Zou Q. Cucurbitacin B exerts anti-cancer activities in human multiple myeloma cells in vitro and in vivo by modulating multiple cellular pathways. Oncotarget 2018; 8:5800-5813. [PMID: 27418139 PMCID: PMC5351590 DOI: 10.18632/oncotarget.10584] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 06/30/2016] [Indexed: 02/05/2023] Open
Abstract
Cucurbitacin B (CuB), a triterpenoid compound isolated from the stems of Cucumis melo, has long been used to treat hepatitis and hepatoma in China. Although its remarkable anti-cancer activities have been reported, the mechanism by which it achieves this therapeutic activity remains unclear. This study was designed to investigate the molecular mechanisms by which CuB inhibits cancer cell proliferation. Our results indicate that CuB is a novel inhibitor of Aurora A in multiple myeloma (MM) cells, arresting cells in the G2/M phase. CuB also inhibited IL-10-induced STAT3 phosphorylation, synergistically increasing the anti-tumor activity of Adriamycin in vitro. CuB induced dephosphorylation of cofilin, resulting in the loss of mitochondrial membrane potential, release of cytochrome c, and activation of caspase-8. CuB inhibited MM tumor growth in a murine MM model, without host toxicity. In conclusion, these results indicate that CuB interferes with multiple cellular pathways in MM cells. CuB thus represents a promising therapeutic tool for the treatment of MM.
Collapse
Affiliation(s)
- Tai Yang
- School of Pharmacy, Chengdu Medical College, Chengdu, China.,Department of Immunology, Chengdu Medical College, Chengdu, China
| | - Jin Liu
- Department of Immunology, Chengdu Medical College, Chengdu, China
| | - Mali Yang
- Department of Immunology, Chengdu Medical College, Chengdu, China
| | - Ning Huang
- Laboratory for Aging Research, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Yueling Zhong
- School of Pharmacy, Chengdu Medical College, Chengdu, China
| | - Ting Zeng
- Department of Immunology, Chengdu Medical College, Chengdu, China
| | - Rong Wei
- Department of Immunology, Chengdu Medical College, Chengdu, China
| | - Zhongjun Wu
- Department of Immunology, Chengdu Medical College, Chengdu, China
| | - Cui Xiao
- Department of Immunology, Chengdu Medical College, Chengdu, China
| | - Xiaohua Cao
- Department of Immunology, Chengdu Medical College, Chengdu, China
| | - Minhui Li
- Department of Immunology, Chengdu Medical College, Chengdu, China
| | - Limei Li
- Department of Immunology, Chengdu Medical College, Chengdu, China
| | - Bin Han
- Department of Public Health, Chengdu Medical College, Chengdu, China
| | - Xiaoping Yu
- Department of Public Health, Chengdu Medical College, Chengdu, China
| | - Hua Li
- Cancer Center, Chengdu Military General Hospital, Chengdu, China
| | - Qiang Zou
- Department of Immunology, Chengdu Medical College, Chengdu, China
| |
Collapse
|
24
|
LUO WW, ZHAO WW, LU JJ, WANG YT, CHEN XP. Cucurbitacin B suppresses metastasis mediated by reactive oxygen species (ROS) via focal adhesion kinase (FAK) in breast cancer MDA-MB-231 cells. Chin J Nat Med 2018; 16:10-19. [DOI: 10.1016/s1875-5364(18)30025-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Indexed: 12/15/2022]
|
25
|
Garg S, Kaul SC, Wadhwa R. Cucurbitacin B and cancer intervention: Chemistry, biology and mechanisms (Review). Int J Oncol 2017; 52:19-37. [PMID: 29138804 DOI: 10.3892/ijo.2017.4203] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 10/23/2017] [Indexed: 11/06/2022] Open
Abstract
Cancer is one of the most important healthcare matters, with the worst prognosis but the best possibilities for scientific development. It is likely to increase in the future and cause global havoc designating it as an epidemic. Cancer development requires urgent intervention. Past few decades have witnessed extensive research to challenge carcinogenesis. Treatment involving synthetic discipline is often associated with severe adverse effects, or even worsened prognosis. Accordingly, newer economic and patient friendly molecules are warranted. Many natural substances have proved their potential so far. Cucurbitacin B against cancer and other diseases has achieved towering popularity among the researchers around the world, as detailed in the below sections with summarized tables. In line with the fascinating role of cucurbitacin B against various types of cancers, through various molecular signaling pathways, it is justifiable to propose cucurbitacin B as a mainline chemotherapy before the onset and after the diagnosis of cancer.
Collapse
Affiliation(s)
- Sukant Garg
- Drug Discovery and Assets Innovation Lab, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Japan
| | - Sunil C Kaul
- Drug Discovery and Assets Innovation Lab, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Japan
| | - Renu Wadhwa
- Drug Discovery and Assets Innovation Lab, DBT-AIST International Laboratory for Advanced Biomedicine (DAILAB), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba 305-8565, Japan
| |
Collapse
|
26
|
Prophetic medicine as potential functional food elements in the intervention of cancer: A review. Biomed Pharmacother 2017; 95:614-648. [DOI: 10.1016/j.biopha.2017.08.043] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2017] [Revised: 08/05/2017] [Accepted: 08/07/2017] [Indexed: 01/01/2023] Open
|
27
|
Wang Z, Zhu W, Gao M, Wu C, Yang C, Yang J, Wu G, Yang B, Kuang H. Simultaneous determination of cucurbitacin B and cucurbitacin E in rat plasma by UHPLC-MS/MS: A pharmacokinetics study after oral administration of cucurbitacin tablets. J Chromatogr B Analyt Technol Biomed Life Sci 2017; 1065-1066:63-69. [DOI: 10.1016/j.jchromb.2017.09.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 09/05/2017] [Accepted: 09/16/2017] [Indexed: 12/19/2022]
|
28
|
Marostica LL, de Barros ALB, Oliveira J, Salgado BS, Cassali GD, Leite EA, Cardoso VN, Lang KL, Caro MSB, Durán FJ, Schenkel EP, de Oliveira MC, Simões CMO. Antitumor effectiveness of a combined therapy with a new cucurbitacin B derivative and paclitaxel on a human lung cancer xenograft model. Toxicol Appl Pharmacol 2017; 329:272-281. [DOI: 10.1016/j.taap.2017.06.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 06/06/2017] [Accepted: 06/09/2017] [Indexed: 02/07/2023]
|
29
|
Liu X, Duan C, Ji J, Zhang T, Yuan X, Zhang Y, Ma W, Yang J, Yang L, Jiang Z, Yu H, Liu Y. Cucurbitacin B induces autophagy and apoptosis by suppressing CIP2A/PP2A/mTORC1 signaling axis in human cisplatin resistant gastric cancer cells. Oncol Rep 2017; 38:271-278. [DOI: 10.3892/or.2017.5648] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2016] [Accepted: 05/08/2017] [Indexed: 11/06/2022] Open
|
30
|
Hu MH, Chen LJ, Chen YL, Tsai MS, Shiau CW, Chao TI, Liu CY, Kao JH, Chen KF. Targeting SHP-1-STAT3 signaling: A promising therapeutic approach for the treatment of cholangiocarcinoma. Oncotarget 2017; 8:65077-65089. [PMID: 29029413 PMCID: PMC5630313 DOI: 10.18632/oncotarget.17779] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 04/26/2017] [Indexed: 01/04/2023] Open
Abstract
Sorafenib is a multiple kinase inhibitor which targets Raf kinases, VEGFR, and PDGFR and is approved for the treatment of hepatocellular carcinoma (HCC). Previously, we found that p-STAT3 is a major target of SC-43, a sorafenib derivative. In this study, we report that SC-43-induced apoptosis in cholangiocarcinoma (CCA) via a novel mechanism. Three CCA cell lines (HuCCT-1, KKU-100 and CGCCA) were treated with SC-43 to determine their sensitivity to SC-43-induced cell death and apoptosis. We found that SC-43 activated SH2 domain-containing phosphatase 1 (SHP-1) activity, leading to p-STAT3 and downstream cyclin B1 and Cdc2 downregulation, which induced G2-M arrest and apoptotic cell death. Importantly, SC-43 augmented SHP-1 activity by direct binding to N-SH2 and relief of its autoinhibition. Deletion of the N-SH2 domain (dN1) or point mutation (D61A) of SHP-1 counteracted the effect of SC-43-induced SHP-1 phosphatase activation and antiproliferation ability in CCA cells. In vivo assay revealed that SC-43 exhibited xenograft tumor growth inhibition, p-STAT3 reduction and SHP-1 activity elevation. In conclusion, SC-43 induced apoptosis in CCA cells through the SHP-1/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Ming-Hung Hu
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Division of Hematology and Oncology, Department of Medicine, Cardinal Tien Hospital, New Taipei City, Taiwan.,School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Li-Ju Chen
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Yen-Lin Chen
- Department of Pathology, Cardinal Tien Hospital, New Taipei City, Taiwan
| | - Ming-Shen Tsai
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Chung-Wai Shiau
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Tzu-I Chao
- Transplant Medicine and Surgery Research Centre, Changhua Christian Hospital, Changhua, Taiwan
| | - Chun-Yu Liu
- Department of Oncology, Taipei Veterans General Hospital, Taipei City, Taiwan.,School of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Jia-Horng Kao
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan.,Division of Gastroenterology and Hepatology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Kuen-Feng Chen
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan.,National Taiwan University College of Medicine, Taipei, Taiwan
| |
Collapse
|
31
|
Cucurbitacin B purified from Ecballium elaterium (L.) A. Rich from Tunisia inhibits α5β1 integrin-mediated adhesion, migration, proliferation of human glioblastoma cell line and angiogenesis. Eur J Pharmacol 2017; 797:153-161. [DOI: 10.1016/j.ejphar.2017.01.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 01/04/2017] [Accepted: 01/11/2017] [Indexed: 11/20/2022]
|
32
|
Lv X, Song DM, Niu YH, Wang BS. Inhibition of heme oxygenase-1 enhances the chemosensitivity of laryngeal squamous cell cancer Hep-2 cells to cisplatin. Apoptosis 2016; 21:489-501. [PMID: 26801320 DOI: 10.1007/s10495-016-1216-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
It has been previously reported that cisplatin is a well-known anticancer drug being used against a wide range of malignancies including head and neck, ovarian and non-small cell lung carcinoma, and demonstrated its anticancer activity by reacting with DNA or changing cell structure, immune response, reactive oxygen species level (ROS). In this research we proved that cisplatin induced cell injuries and heme oxygenase-1 (HO-1) expression in laryngeal squamous cell cancer Hep-2 cells through ROS generation. The induction of HO-1 clearly protected Hep-2 cells from cisplatin-induced cell death and ROS reaction, and the inhibitor of HO-1 enhanced the cell death and ROS generation induced by cisplatin. Furthermore, the HO-1 expression induced by cisplatin was strongly inhibited by the knockdown of nuclear factor-erythroid-2-related factor-2 (Nrf-2), and the oxidative damages induced by cisplatin were significantly enhanced. Therefore, it may be concluded that the inhibition of HO-1 or the knockdown of Nrf-2 significantly enhanced cisplatin's anticancer effects on Hep-2 cells. In clinic, with the overexpression of HO-1 in laryngeal squamous cancer tissues, the combination of cisplatin with the inhibitor of HO-1 or Nrf-2 siRNA may act as a new method to the treatment of laryngeal squamous cancer.
Collapse
Affiliation(s)
- Xin Lv
- Department of Otolaryngology and Allergy, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China.
| | - Dong-mei Song
- Department of Otolaryngology and Allergy, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China
| | - Ying-hao Niu
- Department of Otolaryngology and Allergy, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China
| | - Bao-shan Wang
- Department of Otolaryngology and Allergy, The First Hospital of Hebei Medical University, Shijiazhuang, 050031, Hebei, People's Republic of China. .,Hebei Medical University, No. 361, Zhongshan East Road, Shijiazhuang, 050017, Hebei, People's Republic of China.
| |
Collapse
|
33
|
Bharadwaj U, Kasembeli MM, Tweardy DJ. STAT3 Inhibitors in Cancer: A Comprehensive Update. ACTA ACUST UNITED AC 2016. [DOI: 10.1007/978-3-319-42949-6_5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
34
|
SUMOylation and SENP3 regulate STAT3 activation in head and neck cancer. Oncogene 2016; 35:5826-5838. [PMID: 27181202 PMCID: PMC5116054 DOI: 10.1038/onc.2016.124] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 03/01/2016] [Accepted: 03/11/2016] [Indexed: 12/19/2022]
Abstract
Hyperphosphorylation of signal transducer and activator of transcription 3 (STAT3) has been found in various types of human cancers, including head and neck cancer (HNC). Although smoking is critical in the development and progression of HNC, how tobacco components activate STAT3 is unclear. We demonstrated that exposure of HNC cell lines to a tobacco extract induced a rapid Y705 phosphorylation of STAT3 and a rapid increase in the SUMO protease SENP3 that depended on a simultaneous increase in reactive oxygen species. We identified that SUMOylation at the lysine 451 site facilitated STAT3 binding to the phosphatase TC45 through an SUMO-interacting motif of TC45. SENP3 could thus enhance STAT3 phosphorylation by de-conjugating the SUMO2/3 modification of STAT3. Knocking-down of SENP3 greatly impaired basal and induced STAT3 phosphorylation by tobacco extract or interleukin 6. A correlation between SENP3 protein levels and STAT3 Y705 phosphorylation levels in human laryngeal carcinoma specimens was found, which was more significant in the specimens derived from the smoker patients and with poor clinicopathological parameters. Our data identified SUMOylation as a previously undescribed post-translational modification of STAT3 and SENP3 as a critical positive modulator of tobacco- or cytokine-induced STAT3 activation. These findings provide novel insights into the hyperphosphorylation of STAT3 in development of HNC.
Collapse
|
35
|
Zhao W, Xu D, Yan W, Wang Y, Zhang N. Development and validation of a UPLC-MS/MS method for the determination of cucurbitacin B in rat plasma and application to a pharmacokinetic study. Biomed Chromatogr 2016; 30:503-507. [PMID: 26207321 DOI: 10.1002/bmc.3571] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/25/2015] [Accepted: 07/20/2015] [Indexed: 01/11/2023]
Abstract
Cucurbitacin B (CuB), one of the most abundant forms of cucurbitacins, is a promising natural anticancer drug candidate. Although the anticancer activity of CuB has been well demonstrated, information regarding the pharmacokinetics is limited. A rapid, selective and sensitive UPLC-MS/MS for CuB was developed and validated using hemslecin A (HeA) as internal standard (IS). Plasma samples were pre-treated by liquid-liquid extraction with dichloromethane. Separation was achieved on a reversed-phase C18 column (50 × 4.6 mm, 5 µm) at 35°C using isocratic elution with water-methanol (25:75, v/v) at a flow rate of 0.3 mL/min. The analytes were monitored by a triple quadrupole tandem mass spectrometer with positive electrospray ionization mode. The calibration curve was linear (r > 0.995) in a concentration range of 0.3-100 ng/mL with a limit of quantification of 0.3 ng/mL. Intra- and inter-day accuracy and precision were validated by percentage relative error and relative standard deviation, respectively, which were both lower than the limit of 15%. This assay was successfully applied to a pharmacokinetic study of CuB in Wistar rats.
Collapse
Affiliation(s)
- Waiou Zhao
- Cardiology Department, the First Hospital of Jilin University, Changchun, 130021, China
| | - Dahai Xu
- Emergency Department, the First Hospital of Jilin University, Changchun, 130021, China
| | - Weiwei Yan
- Emergency Department, the First Hospital of Jilin University, Changchun, 130021, China
| | - Yushi Wang
- Cardiology Department, the First Hospital of Jilin University, Changchun, 130021, China
| | - Nan Zhang
- Emergency Department, the First Hospital of Jilin University, Changchun, 130021, China
| |
Collapse
|
36
|
Cai Y, Fang X, He C, Li P, Xiao F, Wang Y, Chen M. Cucurbitacins: A Systematic Review of the Phytochemistry and Anticancer Activity. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2015; 43:1331-50. [PMID: 26503558 DOI: 10.1142/s0192415x15500755] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Cucurbitacins are highly oxidized tetracyclic triterpenoids that are widely present in traditional Chinese medicines (Cucurbitaceae family), possess strong anticancer activity, and are divided into 12 classes from A to T with over 200 derivatives. The eight most active cucurbitacin components against cancer are cucurbitacin B, D, E, I, IIa, L glucoside, Q, and R. Their mechanisms of action include antiproliferation, inhibition of migration and invasion, proapoptosis, and cell cycle arrest promotion. Cucurbitacins are also found to be the inhibitors of JAK-STAT3, Wnt, PI3K/Akt, and MAPK signaling pathways, which play important roles in the apoptosis and survival of cancer cells. Recently, new studies have discovered synergistic anticancer effects by using cucurbitacins together with clinically approved chemotherapeutic drugs, such as docetaxel and methotrexate. This paper provides a summary of recent research progress on the anticancer property of cucurbitacins and the various intracellular signaling pathways involved in the regulation of cancer cell proliferation, death, invasion, and migration. Therefore, cucurbitacins are a class of promising anticancer drugs to be used alone or be intergraded in current chemotherapies and radiotherapies to treat many types of cancers.
Collapse
Affiliation(s)
- Yuee Cai
- * State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P.R. China
| | - Xiefan Fang
- † Department of Pediatrics, College of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Chengwei He
- * State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P.R. China
| | - Peng Li
- * State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P.R. China
| | - Fei Xiao
- * State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P.R. China.,‡ Department of Pharmacology, School of Medicine, Jinan University, Guangzhou 510632, P.R. China
| | - Yitao Wang
- * State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P.R. China
| | - Meiwan Chen
- * State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau 999078, P.R. China
| |
Collapse
|
37
|
Ranjan A, Fofaria NM, Kim SH, Srivastava SK. Modulation of signal transduction pathways by natural compounds in cancer. Chin J Nat Med 2015; 13:730-742. [PMID: 26481373 DOI: 10.1016/s1875-5364(15)30073-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Indexed: 02/07/2023]
Abstract
Cancer is generally regarded as the result of abnormal growth of cells. According to World Health Organization, cancer is the leading cause of mortality worldwide. Mother nature provides a large source of bioactive compounds with excellent therapeutic efficacy. Numerous phytochemicals from nature have been investigated for anticancer properties. In this review article, we discuss several natural compounds, which have shown anti-cancer activity. Natural compounds induce cell cycle arrest, activate intrinsic and extrinsic apoptosis pathways, generate Reactive Oxygen Species (ROS), and down-regulate activated signaling pathways, resulting in inhibition of cell proliferation, progression and metastasis of cancer. Several preclinical studies have suggested that natural compounds can also increase the sensitivity of resistant cancers to available chemotherapy agents. Furthermore, combining FDA approved anti-cancer drugs with natural compounds results in improved efficacy. On the basis of these exciting outcomes of natural compounds against several cancer types, several agents have already advanced to clinical trials. In conclusion, preclinical results and clinical outcomes against cancer suggest promising anticancer efficacy of agents from natural sources.
Collapse
Affiliation(s)
- Alok Ranjan
- Department of Biomedical Sciences and Cancer Biology Center, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Neel M Fofaria
- Department of Biomedical Sciences and Cancer Biology Center, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA
| | - Sung-Hoon Kim
- Cancer Preventive Material Development Research Center, College of Korean Medicine, Department of Pathology, Kyunghee University, Seoul 131-701, South Korea.
| | - Sanjay K Srivastava
- Department of Biomedical Sciences and Cancer Biology Center, Texas Tech University Health Sciences Center, Amarillo, TX 79106, USA; Cancer Preventive Material Development Research Center, College of Korean Medicine, Department of Pathology, Kyunghee University, Seoul 131-701, South Korea.
| |
Collapse
|
38
|
Marostica LL, Silva IT, Kratz JM, Persich L, Geller FC, Lang KL, Caro MSB, Durán FJ, Schenkel EP, Simões CMO. Synergistic Antiproliferative Effects of a New Cucurbitacin B Derivative and Chemotherapy Drugs on Lung Cancer Cell Line A549. Chem Res Toxicol 2015; 28:1949-60. [PMID: 26372186 DOI: 10.1021/acs.chemrestox.5b00153] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Nonsmall cell lung cancer (NSCLC) represents an important cause of mortality worldwide due to its aggressiveness and growing resistance to currently available therapy. Cucurbitacins have emerged as novel potential anticancer agents showing strong antiproliferative effects and can be promising candidates for combined treatments with clinically used anticancer agents. This study investigates the synergistic antiproliferative effects of a new semisynthetic derivative of cucurbitacin B (DACE) with three chemotherapy drugs: cisplatin (CIS), irinotecan (IRI), and paclitaxel (PAC) on A549 cells. The most effective combinations were selected for studies of the mechanism of action. Using an in silico tool, DACE seems to act by a different mechanism of action when compared with that of different classes of drugs already used in clinical settings. DACE also showed potent synergic effects with drugs, and the most potent combinations induced G2/M cell cycle arrest by modulating survivin and p53 expression, disruption of F-actin cytoskeleton, and cell death by apoptosis. These treatments completely inhibited the clonogenic potential and did not reduce the proliferation of nontumoral lung cells (MRC-5). DACE also showed relevant antimigratory and anti-invasive effects, and combined treatments modulated cell migration signaling pathways evolved with metastasis progression. The effects of DACE associated with drugs was potentiated by the oxidant agent l-buthionine-sulfoximine (BSO), and attenuated by N-acetilcysteine (NAC), an antioxidant agent. The antiproliferative effects induced by combined treatments were attenuated by a pan-caspase inhibitor, indicating that the effects of these treatments are dependent on caspase activity. Our data highlight the therapeutic potential of DACE used in combination with known chemotherapy drugs and offer important insights for the development of more effective and selective therapies against lung cancer.
Collapse
Affiliation(s)
- Lucas Lourenço Marostica
- Departamento de Ciências Farmacêuticas, Universidade Federal de Santa Catarina , Campus Trindade, CEP 88040-900, Florianópolis, Santa Catarina, Brazil
| | - Izabella Thaís Silva
- Departamento de Ciências Farmacêuticas, Universidade Federal de Santa Catarina , Campus Trindade, CEP 88040-900, Florianópolis, Santa Catarina, Brazil
| | - Jadel Müller Kratz
- Departamento de Ciências Farmacêuticas, Universidade Federal de Santa Catarina , Campus Trindade, CEP 88040-900, Florianópolis, Santa Catarina, Brazil
| | - Lara Persich
- Departamento de Ciências Farmacêuticas, Universidade Federal de Santa Catarina , Campus Trindade, CEP 88040-900, Florianópolis, Santa Catarina, Brazil
| | - Fabiana Cristina Geller
- Departamento de Ciências Farmacêuticas, Universidade Federal de Santa Catarina , Campus Trindade, CEP 88040-900, Florianópolis, Santa Catarina, Brazil
| | - Karen Luise Lang
- Departamento de Ciências Farmacêuticas, Universidade Federal de Santa Catarina , Campus Trindade, CEP 88040-900, Florianópolis, Santa Catarina, Brazil
| | - Miguel Soriano Balparda Caro
- Departamento de Química, Universidade Federal de Santa Catarina , Campus Trindade, CEP 88040-900, Florianópolis, Santa Catarina, Brazil
| | - Fernando Javier Durán
- UMYMFOR-CONICET, Departamento de Química Orgánica, Universidad de Buenos Aires , Buenos Aires, Argentina
| | - Eloir Paulo Schenkel
- Departamento de Ciências Farmacêuticas, Universidade Federal de Santa Catarina , Campus Trindade, CEP 88040-900, Florianópolis, Santa Catarina, Brazil
| | - Cláudia Maria Oliveira Simões
- Departamento de Ciências Farmacêuticas, Universidade Federal de Santa Catarina , Campus Trindade, CEP 88040-900, Florianópolis, Santa Catarina, Brazil
| |
Collapse
|
39
|
El-Senduny FF, Badria FA, EL-Waseef AM, Chauhan SC, Halaweish F. Approach for chemosensitization of cisplatin-resistant ovarian cancer by cucurbitacin B. Tumour Biol 2015; 37:685-98. [DOI: 10.1007/s13277-015-3773-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 07/07/2015] [Indexed: 12/22/2022] Open
|
40
|
Yar Saglam AS, Alp E, Elmazoglu Z, Menevse S. Treatment with cucurbitacin B alone and in combination with gefitinib induces cell cycle inhibition and apoptosis via EGFR and JAK/STAT pathway in human colorectal cancer cell lines. Hum Exp Toxicol 2015; 35:526-43. [PMID: 26183715 DOI: 10.1177/0960327115595686] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The epidermal growth factor receptor (EGFR) associated with signaling pathways, such as Janus kinase (JAK)/signal transducer and activator of transcription (STAT), plays an important role in colorectal cancers (CRCs). Gefitinib (Gef) is an orally active inhibitor targeting the adenosine tri phosphate-binding domain of EGFR, and cucurbitacin B (CuB) is a selective inhibitor of JAK/STAT signaling with potent antitumor activity via suppression of STAT3 phosphorylation, but the underlying mechanism is not clear. We aimed to investigate the apoptotic and antiproliferative effects of CuB as a single agent and in combination with Gef on both HT-29 and HCT-116 cell lines. Cell proliferation, cell cycle distribution, and apoptosis were evaluated using viability assay, fluorescent microscopy, cytotoxicity assay, proliferation, DNA fragmentation, and cleaved caspase 3 levels. Real-time polymerase chain reaction and Western blot analyses were performed to determine the expression of relevant genes and proteins including antiapoptotic, proapoptotic, and cell cycle regulation. EGFR, phosphorylated EGFR (pEGFR), STAT3, and pSTAT3 proteins were evalutaed with Western blot analysis. Our results showed that, compared to CuB alone, CuB plus Gef treatment caused a significant growth and cell cycle inhibition and induced apoptosis in both cell lines. Also CuB plus Gef treatment decreased DNA synthesis rate more effectively than CuB alone. Treatment with CuB alone and in combination with Gef decreased the expression levels of B-Cell CLL/Lymphoma 2 (Bcl-2), BCL2-like 1 (BCL2L1), cyclin D1, pSTAT3, and pEGFR and increased the expression levels of Bcl-2-like protein 4, Bcl-2 homologous antagonist/killer, Bcl-2-associated death promoter, Bcl-2-like protein 11, and p27kip1 levels. Our results suggest that treatment with CuB alone and more likely in combination with Gef may be a considerable alternative therapeutic approach for CRC, at least in vitro.
Collapse
Affiliation(s)
- A S Yar Saglam
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Besevler, Ankara, Turkey
| | - E Alp
- Department of Medical Biology, Faculty of Medicine, Giresun University, Giresun, Turkey
| | - Z Elmazoglu
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Besevler, Ankara, Turkey
| | - S Menevse
- Department of Medical Biology and Genetics, Faculty of Medicine, Gazi University, Besevler, Ankara, Turkey
| |
Collapse
|
41
|
Ren G, Sha T, Guo J, Li W, Lu J, Chen X. Cucurbitacin B induces DNA damage and autophagy mediated by reactive oxygen species (ROS) in MCF-7 breast cancer cells. J Nat Med 2015; 69:522-30. [PMID: 26018422 DOI: 10.1007/s11418-015-0918-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 05/06/2015] [Indexed: 01/11/2023]
Abstract
Cucurbitacin B (Cuc B), a natural compound extracted from cucurbitaceous plants, demonstrated potent anticancer activities, while the underlying mechanisms remain unclear. We investigated the anticancer effect of Cuc B on MCF-7 breast cancer cells. Cuc B drastically decreased cell viability in a concentration-dependent manner. Cuc B treatment caused DNA damage, as shown by long tails in the comet assay and increased γH2AX protein expression. Immunofluorescence staining showed that Cuc B treatment induced nuclear γH2AX foci. Cuc B activated DNA damage pathways by phosphorylation of ATM/ATR [two large phosphatidylinositol-3-kinase-like kinase family (PIKKs) members]. Furthermore, it also induced autophagy, as evidenced by monodansylcadaverine (MDC) staining and autophagic protein expression. In addition, Cuc B treatment led to increased reactive oxygen species (ROS) formation, which was inhibited by N-acetyl-L-cysteine (NAC) pretreatment. NAC pretreatment inhibited Cuc-B-induced DNA damage and autophagy. Taken together, these results suggest that ROS-mediated Cuc-B-induced DNA damage and autophagy in MCF-7 cells, which provides new insights into the anticancer molecular mechanism of Cuc B.
Collapse
Affiliation(s)
- Guowen Ren
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | | | | | | | | | | |
Collapse
|
42
|
Bailon-Moscoso N, Romero-Benavides JC, Tinitana-Imaicela F, Ostrosky-Wegman P. Medicinal plants of Ecuador: a review of plants with anticancer potential and their chemical composition. Med Chem Res 2015. [DOI: 10.1007/s00044-015-1335-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
43
|
Discovery of a small-molecule inhibitor of STAT3 by ligand-based pharmacophore screening. Methods 2014; 71:38-43. [PMID: 25160651 DOI: 10.1016/j.ymeth.2014.07.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 07/19/2014] [Accepted: 07/21/2014] [Indexed: 12/16/2022] Open
Abstract
STAT3 modulates the transcription of a wide variety of regulatory genes involved in cell proliferation, differentiation, migration, apoptosis, and other critical cellular functions. Constitutive activation of STAT3 has been detected in a wide spectrum of human malignancies. A pharmacophore model constructed from a training set of STAT3 inhibitors binding to the SH2 domain was used to screen an in-house database of compounds, from which azepine 1 emerged as a top candidate. Compound 1 inhibited STAT3 DNA-binding activity in vitro and attenuated STAT3-directed transcription in cellulo with comparable potency to the well-known STAT3 inhibitor S3I-201. A fluorescence polarization assay revealed that compound 1 targeted the SH2 domain of STAT3. Furthermore, compound 1 inhibited STAT3 phosphorylation in cells without affecting the total expression of STAT3. This study also validates the use of pharmacophore modeling to identify inhibitors of protein-protein interactions.
Collapse
|
44
|
ZHENG QIAN, LIU YUNYI, LIU WEIWEI, MA FENGYUN, ZHOU YI, CHEN MINGJIE, CHANG JUNLI, WANG YUESHENG, YANG GUANGXIAO, HE GUANGYUAN. Cucurbitacin B inhibits growth and induces apoptosis through the JAK2/STAT3 and MAPK pathways in SH-SY5Y human neuroblastoma cells. Mol Med Rep 2014; 10:89-94. [DOI: 10.3892/mmr.2014.2175] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2013] [Accepted: 03/21/2014] [Indexed: 11/05/2022] Open
|
45
|
The Multifaceted Roles of STAT3 Signaling in the Progression of Prostate Cancer. Cancers (Basel) 2014; 6:829-59. [PMID: 24722453 PMCID: PMC4074806 DOI: 10.3390/cancers6020829] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2014] [Revised: 03/11/2014] [Accepted: 03/17/2014] [Indexed: 01/09/2023] Open
Abstract
The signal transducer and activator of transcription (STAT)3 governs essential functions of epithelial and hematopoietic cells that are often dysregulated in cancer. While the role for STAT3 in promoting the progression of many solid and hematopoietic malignancies is well established, this review will focus on the importance of STAT3 in prostate cancer progression to the incurable metastatic castration-resistant prostate cancer (mCRPC). Indeed, STAT3 integrates different signaling pathways involved in the reactivation of androgen receptor pathway, stem like cells and the epithelial to mesenchymal transition that drive progression to mCRPC. As equally important, STAT3 regulates interactions between tumor cells and the microenvironment as well as immune cell activation. This makes it a major factor in facilitating prostate cancer escape from detection of the immune response, promoting an immunosuppressive environment that allows growth and metastasis. Based on the multifaceted nature of STAT3 signaling in the progression to mCRPC, the promise of STAT3 as a therapeutic target to prevent prostate cancer progression and the variety of STAT3 inhibitors used in cancer therapies is discussed.
Collapse
|
46
|
Gao Y, Islam MS, Tian J, Lui VWY, Xiao D. Inactivation of ATP citrate lyase by Cucurbitacin B: A bioactive compound from cucumber, inhibits prostate cancer growth. Cancer Lett 2014; 349:15-25. [PMID: 24690568 DOI: 10.1016/j.canlet.2014.03.015] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 03/12/2014] [Accepted: 03/13/2014] [Indexed: 01/03/2023]
Abstract
Prostate cancer, a leading cause of cancer-related deaths in males, is well recognized as having late disease on-set (mostly at age 60-70) and showing slow/latent disease development, and strategies to prevent cancer formation in late manhood may have significant health impacts. Cucurbitacin B (CuB) is a naturally occurring compound that is found abundantly in cucumbers and other vegetables, and it is known to exert anti-cancer activities (primarily via apoptosis-induction) in several human cancers. However, its chemopreventive potential for prostate cancer has not yet been investigated. Here, we reported that CuB significantly and specifically inhibited prostate cancer cell growth with low IC50 (~0.3 μM; PC-3 and LNCaP), accompanied by marked apoptosis (Caspase 3/7 activation, PARP cleavage, increase of Annexin V-Alexa Fluor 488 (Alexa488)+ cells and accumulation of Sub-G0/G1 population), whereas normal human prostate epithelial cells (PrEC) were CuB-insensitive. Using a chemopreventive model, pre-treatment of mice with CuB (2 weeks before PC-3 prostate cancer cell implantation) significantly reduced the rate of in vivo tumor-formation. A 79% reduction in tumor size (accompanied by marked in situ apoptosis) was observed in the CuB-treated group (with no noticeable toxicity) vs. controls at day 31. Strikingly, mechanistic investigations demonstrated that CuB drove dose-dependent inhibition of ATP citrate lyase phosphorylation (ACLY; an important enzyme for cancer metabolism) both in vitro and in the CuB-chemopreventive mouse model. Importantly, ACLY over-expression abrogated CuB's apoptotic effects in prostate cancer cells, confirming ACLY as a direct target of CuB. Thus, CuB harbors potent chemopreventive activity for prostate cancer, and we revealed a novel anti-tumor mechanism of CuB via inhibition of ACYL signaling in human cancer.
Collapse
Affiliation(s)
- Yajuan Gao
- Department of Urology, University of Pittsburgh Cancer Institute, University of Pittsburgh Medical College, University of Pittsburgh, Shadyside Medical Center, Suit G37, 5200 Centre Avenue, Pittsburgh, PA 15232, USA
| | - Mohammad Shyful Islam
- Department of Urology, University of Pittsburgh Cancer Institute, University of Pittsburgh Medical College, University of Pittsburgh, Shadyside Medical Center, Suit G37, 5200 Centre Avenue, Pittsburgh, PA 15232, USA
| | - Jiang Tian
- Department of Urology, University of Pittsburgh Cancer Institute, University of Pittsburgh Medical College, University of Pittsburgh, Shadyside Medical Center, Suit G37, 5200 Centre Avenue, Pittsburgh, PA 15232, USA
| | - Vivian Wai Yan Lui
- Department of Pharmacology and Pharmacy, Li Ka Faculty of Medicine, University of Hong Kong, Hong Kong Special Administrative Region
| | - Dong Xiao
- Department of Urology, University of Pittsburgh Cancer Institute, University of Pittsburgh Medical College, University of Pittsburgh, Shadyside Medical Center, Suit G37, 5200 Centre Avenue, Pittsburgh, PA 15232, USA.
| |
Collapse
|
47
|
Tundis R, Menichini F, Loizzo MR. Recent Insights into the Emerging Role of Triterpenoids in Cancer Therapy. ACTA ACUST UNITED AC 2014. [DOI: 10.1016/b978-0-444-63294-4.00001-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
|
48
|
Therapeutic modulators of STAT signalling for human diseases. Nat Rev Drug Discov 2013; 12:611-29. [PMID: 23903221 DOI: 10.1038/nrd4088] [Citation(s) in RCA: 342] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The signal transducer and activator of transcription (STAT) proteins have important roles in biological processes. The abnormal activation of STAT signalling pathways is also implicated in many human diseases, including cancer, autoimmune diseases, rheumatoid arthritis, asthma and diabetes. Over a decade has passed since the first inhibitor of a STAT protein was reported and efforts to discover modulators of STAT signalling as therapeutics continue. This Review discusses the outcomes of the ongoing drug discovery research endeavours against STAT proteins, provides perspectives on new directions for accelerating the discovery of drug candidates, and highlights the noteworthy candidate therapeutics that have progressed to clinical trials.
Collapse
|
49
|
Habib L, Jraij A, Khreich N, Fessi H, Charcosset C, Greige-Gerges H. Morphological and physicochemical characterization of liposomes loading cucurbitacin E, an anti-proliferative natural tetracyclic triterpene. Chem Phys Lipids 2013; 177:64-70. [PMID: 24291009 DOI: 10.1016/j.chemphyslip.2013.11.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Revised: 11/19/2013] [Accepted: 11/20/2013] [Indexed: 11/18/2022]
Abstract
Cucurbitacin E (Cuc E), an oxygenated triterpene molecule, has demonstrated anti-proliferative effect on various cancer cells. Here, we examined the effect of Cuc E on the membrane morphology and properties using differential scanning calorimetry, transmission electron microscopy and atomic force microscopy techniques. Dipalmitoylphosphatidylcholine vesicles were prepared by the thin film hydration method in the absence and presence of Cuc E at molar ratios 100:12 and 100:20. The loading efficiency of Cuc E was found to be higher than 98% upon HPLC analysis. The thermodynamic parameters suggest that Cuc E does not penetrate into the bilayers and interacts with the polar/apolar interface of the lipid membranes. Blank and Cuc E loaded liposomes prepared from a mixture of DPPC/DPPE/DPPG/Cho were imaged by TEM and AFM. Images obtained by TEM revealed unilamellar liposomes for blank and Cuc E loaded liposomes. AFM images showed that the size and the height of Cuc E loaded liposomes were respectively smaller and higher than blank ones. Results suggest that Cuc E produces modifications in the lipid membrane structures.
Collapse
Affiliation(s)
- Lamice Habib
- Bioactive Molecules Research Group, Doctoral School of Sciences and Technologies, Department of Chemistry and Biochemistry, Faculty of Sciences-2, Lebanese University, Lebanon; Laboratoire d'Automatique et de Génie des Procédés (LAGEP), UMR 5007, CNRS, CPE, 43 bd du 11 Novembre, 691622 Villeurbanne Cedex, France
| | - Alia Jraij
- Bioactive Molecules Research Group, Doctoral School of Sciences and Technologies, Department of Chemistry and Biochemistry, Faculty of Sciences-2, Lebanese University, Lebanon
| | - Nathalie Khreich
- Bioactive Molecules Research Group, Doctoral School of Sciences and Technologies, Department of Chemistry and Biochemistry, Faculty of Sciences-2, Lebanese University, Lebanon
| | - Hatem Fessi
- Laboratoire d'Automatique et de Génie des Procédés (LAGEP), UMR 5007, CNRS, CPE, 43 bd du 11 Novembre, 691622 Villeurbanne Cedex, France
| | - Catherine Charcosset
- Laboratoire d'Automatique et de Génie des Procédés (LAGEP), UMR 5007, CNRS, CPE, 43 bd du 11 Novembre, 691622 Villeurbanne Cedex, France
| | - Hélène Greige-Gerges
- Bioactive Molecules Research Group, Doctoral School of Sciences and Technologies, Department of Chemistry and Biochemistry, Faculty of Sciences-2, Lebanese University, Lebanon.
| |
Collapse
|
50
|
Yan S, Li Z, Thiele CJ. Inhibition of STAT3 with orally active JAK inhibitor, AZD1480, decreases tumor growth in Neuroblastoma and Pediatric Sarcomas In vitro and In vivo. Oncotarget 2013; 4:433-45. [PMID: 23531921 PMCID: PMC3717306 DOI: 10.18632/oncotarget.930] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The IL-6/JAK/STAT pathway is a key signal transduction pathway implicated in the pathogenesis of many human cancers, suggesting that kinase inhibitors targeting JAK/STAT3 may have a broad spectrum of antitumor activity. AZD1480, a pharmacological JAK1/2 inhibitor, exhibits anti-tumor potency in multiple adult malignancies. To evaluate the efficacy of inhibition of JAK/STAT3 signal transduction pathway we assessed the activity of AZD1480 in pediatric malignancies using preclinical models of three highly malignant pediatric solid tumors: neuroblastoma (NB), rhabdomyosarcoma (RMS) and the Ewing Sarcoma Family Tumors (ESFT). In this study, we employed panels of biomedical and biological experiments to evaluate the in vitro and in vivo activity of AZD1480 in NB, RMS and ESFT. Our data indicate that AZD1480 blocks endogenous as well as IL-6 induced STAT3 activation. AZD1480 decreases cell viability in 7/7NB, 7/7RMS and 2/2 ESFT cell lines (median EC50 is 1.5 μM, ranging from 0.36-5.37μM). AZD1480 induces cell growth inhibition and caspase-dependent apoptosis in vitro and decreases expression of STAT3 target genes, including cell cycle regulators CyclinD1, 3 and CDC25A, anti-apoptotic genes Bcl-2 and survivin, the metastasis-related factor TIMP-1 and c-Myc. In vivo studies showed AZD1480 significantly decreased tumor growth and prolonged overall survival in tumor-bearing mice. Tumors from AZD1480-treated mice showed inhibition of activated STAT3 as well as decreased expression of STAT3 downstream targets. Our study provides strong evidence of the anti-tumor growth potency of JAK inhibitor AZD1480 in pediatric solid tumors, providing proof-of principle that inhibition of the JAK/STAT3 signal transduction could be a promising therapeutic target for high-risk pediatric solid tumors.
Collapse
Affiliation(s)
- Shuang Yan
- Cell and Molecular Biology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, USA
| | | | | |
Collapse
|