1
|
Coppola C, Greco M, Munir A, Musarò D, Quarta S, Massaro M, Lionetto MG, Maffia M. Osteoarthritis: Insights into Diagnosis, Pathophysiology, Therapeutic Avenues, and the Potential of Natural Extracts. Curr Issues Mol Biol 2024; 46:4063-4105. [PMID: 38785519 PMCID: PMC11119992 DOI: 10.3390/cimb46050251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 04/05/2024] [Accepted: 04/18/2024] [Indexed: 05/25/2024] Open
Abstract
Osteoarthritis (OA) stands as a prevalent and progressively debilitating clinical condition globally, impacting joint structures and leading to their gradual deterioration through inflammatory mechanisms. While both non-modifiable and modifiable factors contribute to its onset, numerous aspects of OA pathophysiology remain elusive despite considerable research strides. Presently, diagnosis heavily relies on clinician expertise and meticulous differential diagnosis to exclude other joint-affecting conditions. Therapeutic approaches for OA predominantly focus on patient education for self-management alongside tailored exercise regimens, often complemented by various pharmacological interventions primarily targeting pain alleviation. However, pharmacological treatments typically exhibit short-term efficacy and local and/or systemic side effects, with prosthetic surgery being the ultimate resolution in severe cases. Thus, exploring the potential integration or substitution of conventional drug therapies with natural compounds and extracts emerges as a promising frontier in enhancing OA management. These alternatives offer improved safety profiles and possess the potential to target specific dysregulated pathways implicated in OA pathogenesis, thereby presenting a holistic approach to address the condition's complexities.
Collapse
Affiliation(s)
- Chiara Coppola
- Department of Mathematics and Physics “E. De Giorgi”, University of Salento, Via Lecce-Arnesano, 73100 Lecce, Italy; (C.C.); (A.M.)
| | - Marco Greco
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy; (M.G.); (D.M.); (S.Q.); (M.G.L.)
| | - Anas Munir
- Department of Mathematics and Physics “E. De Giorgi”, University of Salento, Via Lecce-Arnesano, 73100 Lecce, Italy; (C.C.); (A.M.)
| | - Debora Musarò
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy; (M.G.); (D.M.); (S.Q.); (M.G.L.)
| | - Stefano Quarta
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy; (M.G.); (D.M.); (S.Q.); (M.G.L.)
| | - Marika Massaro
- Institute of Clinical Physiology (IFC), National Research Council (CNR), 73100 Lecce, Italy;
| | - Maria Giulia Lionetto
- Department of Biological and Environmental Science and Technology, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy; (M.G.); (D.M.); (S.Q.); (M.G.L.)
| | - Michele Maffia
- Department of Experimental Medicine, University of Salento, Via Lecce-Monteroni, 73100 Lecce, Italy
| |
Collapse
|
2
|
Cheng C, Liu Z, Liu D, Chen H, Wang Y, Sun B. LncRNA CCAT1 participates in pancreatic ductal adenocarcinoma progression by forming a positive feedback loop with c-Myc. Carcinogenesis 2024; 45:69-82. [PMID: 37936306 DOI: 10.1093/carcin/bgad076] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/29/2023] [Accepted: 11/02/2023] [Indexed: 11/09/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) play fundamental roles in cancer development; however, the underlying mechanisms for a large proportion of lncRNAs in pancreatic ductal adenocarcinoma (PDAC) have not been elucidated. The expression of colon cancer-associated transcript-1 (CCAT1) in PDAC specimens and cell lines was measured by quantitative real-time polymerase chain reaction (qRT-PCR). The function of CCAT1 was examined in vitro and in vivo. The interactions among CCAT1, miR-24-3p and c-Myc were determined by bioinformatics analysis, RNA immunoprecipitation (RIP), dual-luciferase reporter assay, and rescue experiments. CCAT1 was significantly increased in PDAC, positively correlated with PDAC progression and predicted a worse prognosis. Furthermore, CCAT1 enhanced Adenosine triphosphate (ATP) production to facilitate PDAC cell proliferation, colony formation and motility in vitro and tumor growth in vivo. CCAT1 may serve as an miR-24-3p sponge, thereby counteracting its repression by c-Myc expression. Reciprocally, c-Myc may act as a transcription factor to alter CCAT1 expression by directly targeting its promoter region, thus forming a positive feedback loop with CCAT1. Collectively, these results demonstrate that a positive feedback loop of CCAT1/miR-24-3p/c-Myc is involved in PDAC development, which may serve as a biomarker and therapeutic target for PDAC.
Collapse
Affiliation(s)
- Chundong Cheng
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, China
| | - Zonglin Liu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, China
| | - Danxi Liu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, China
| | - Hua Chen
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, China
| | - Yongwei Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, China
- Key Laboratory of Hepatosplenic Surgery, Ministry of Education, 23 Youzheng Street, Nangang District, Harbin 150001, Heilongjiang, China
| |
Collapse
|
3
|
Uner B, Ergin AD. Enhanced mitochondrial co-localization of β-escin micelle and pancreatic tumor accumulation relation. J Drug Deliv Sci Technol 2023; 89:104994. [DOI: 13.https:/doi.org/10.1016/j.jddst.2023.104994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2025]
|
4
|
Li C, He Z, Yao F, Liao S, Sun K, Sun S, Li Z, Wang Z. Role of Escin in breast cancer therapy: potential mechanism for inducing ferroptosis and synergistic antitumor activity with cisplatin. Apoptosis 2023:10.1007/s10495-023-01849-x. [PMID: 37149513 DOI: 10.1007/s10495-023-01849-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/17/2023] [Indexed: 05/08/2023]
Abstract
Breast cancer (BC) has threatened women worldwide for a long time, and novel treatments are needed. Ferroptosis is a new form of regulated cell death that is a potential therapeutic target for BC. In this study, we identified Escin, a traditional Chinese medicine, as a possible supplement for existing chemotherapy strategies. Escin inhibited BC cell growth in vitro and in vivo, and ferroptosis is probable to be the main cause for Escin-induced cell death. Mechanistically, Escin significantly downregulated the protein level of GPX4, while overexpression of GPX4 could reverse the ferroptosis triggered by Escin. Further study revealed that Escin could promote G6PD ubiquitination and degradation, thus inhibiting the expression of GPX4 and contributing to the ferroptosis. Moreover, proteasome inhibitor MG132 or G6PD overexpression could partially reverse Escin-induced ferroptosis, when G6PD knockdown aggravated that. In vivo study also supported that downregulation of G6PD exacerbated tumor growth inhibition by Escin. Finally, our data showed that cell apoptosis was dramatically elevated by Escin combined with cisplatin in BC cells. Taken together, these results suggest that Escin inhibits tumor growth in vivo and in vitro via regulating the ferroptosis mediated by G6PD/GPX4 axis. Our findings provide a promising therapeutic strategy for BC.
Collapse
Affiliation(s)
- Chenyuan Li
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Ziqi He
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, People's Republic of China
| | - Feng Yao
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Shichong Liao
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Kai Sun
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China
| | - Shengrong Sun
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China.
| | - Zhiyu Li
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China.
| | - Zhong Wang
- Department of Breast & Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
5
|
Fazliev S, Tursunov K, Razzokov J, Sharipov A. Escin's Multifaceted Therapeutic Profile in Treatment and Post-Treatment of Various Cancers: A Comprehensive Review. Biomolecules 2023; 13:biom13020315. [PMID: 36830684 PMCID: PMC9952945 DOI: 10.3390/biom13020315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/30/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
Although modern medicine is advancing at an unprecedented rate, basic challenges in cancer treatment and drug resistance remain. Exploiting natural-product-based drugs is a strategy that has been proven over time to provide diverse and efficient approaches in patient care during treatment and post-treatment periods of various diseases, including cancer. Escin-a plant-derived triterpenoid saponin-is one example of natural products with a broad therapeutic scope. Initially, escin was proven to manifest potent anti-inflammatory and anti-oedematous effects. However, in the last two decades, other novel activities of escin relevant to cancer treatment have been reported. Recent studies demonstrated escin's efficacy in compositions with other approved drugs to accomplish synergy and increased bioavailability to broaden their apoptotic, anti-metastasis, and anti-angiogenetic effects. Here, we comprehensively discuss and present an overview of escin's chemistry and bioavailability, and highlight its biological activities against various cancer types. We conclude the review by presenting possible future directions of research involving escin for medical and pharmaceutical applications as well as for basic research.
Collapse
Affiliation(s)
- Sunnatullo Fazliev
- Max Planck School Matter to Life, Jahnstrasse 29, 69120 Heidelberg, Germany
- Faculty of Engineering Sciences, Heidelberg University, Im Neuenheimer Feld 205, 69120 Heidelberg, Germany
| | - Khurshid Tursunov
- Department of Inorganic, Physical and Colloidal Chemistry, Tashkent Pharmaceutical Institute, Oybek Street 45, Tashkent 100015, Uzbekistan
- State Center for Expertise and Standardization of Medicines, Medical Devices and Medical Equipment, Agency for the Development of the Pharmaceutical Industry under the Ministry of Health of the Republic of Uzbekistan, Ozod Street 16, Tashkent 100002, Uzbekistan
| | - Jamoliddin Razzokov
- Institute of Fundamental and Applied Research, National Research University TIIAME, Kori Niyoziy 39, Tashkent 100000, Uzbekistan
- College of Engineering, Akfa University, Milliy Bog Street 264, Tashkent 111221, Uzbekistan
- Department of Physics, National University of Uzbekistan, Universitet 4, Tashkent 100174, Uzbekistan
- Laboratory of Experimental Biophysics, Centre for Advanced Technologies, Universitet 7, Tashkent 100174, Uzbekistan
| | - Avez Sharipov
- Department of Inorganic, Physical and Colloidal Chemistry, Tashkent Pharmaceutical Institute, Oybek Street 45, Tashkent 100015, Uzbekistan
- Department of Analytical and Pharmaceutical Chemistry, Institute of Pharmaceutical Education and Research, Yunusota Street 46, Tashkent 100114, Uzbekistan
- Correspondence:
| |
Collapse
|
6
|
Cao HN, Ruan JY, Han Y, Zhao W, Zhang Y, Gao C, Wu HH, Ma L, Gao XM, Zhang Y, Wang T. NO Release Inhibitory Activity of Flavonoids from Aesculus Wilsonii Seeds through MAPK (P38), NF-κB, and STAT3 Cross-Talk Signaling Pathways. PLANTA MEDICA 2023; 89:46-61. [PMID: 35253147 DOI: 10.1055/a-1789-2983] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The flavonoid constituents of Aesculus wilsonii, a source of the Chinese medicinal drug Suo Luo Zi, and their in vitro anti-inflammatory effects were investigated. Fifteen flavonoids, including aeswilflavonosides IA-IC (1: - 3: ) and aeswilflavonosides IIA-IIE (4: - 8: ), along with seven known derivatives were isolated from a seed extract. Their structures were elucidated by extensive spectroscopic methods, acid and alkaline hydrolysis, and calculated electronic circular dichroism spectra. Among them, compounds 3: and 7: possess a 5-[2-(carboxymethyl)-5-oxocyclopent-yl]pent-3-enylate or oleuropeoylate substituent, respectively, which are rarely reported in flavonoids. Compounds 2, 3, 7: , and 12: - 15: were found to inhibit lipopolysaccharide-induced nitric oxide production in RAW 264.7 cell lines. In a mechanistic assay, the flavonoid glycosides 2, 3: , and 7: reduced the expressions of interleukin-6 and tumor necrosis factor-alpha induced by lipopolysaccharide. Further investigations suggest that 2: and 3: downregulated the protein expression of tumor necrosis factor-alpha and interleukin-6 by inhibiting the phosphorylation of p38. Compound 7: was found to reduce the production of inducible nitric oxide synthase, and the secretion of tumor necrosis factor-alpha and interleukin-6 through inhibiting nuclear factor kappa-light-chain-enhancer of activated B signaling pathway. Compounds 2, 3: , and 7: possessed moderate inhibitory activity on the expression of signal transducer and activator of transcription-3. Taken together, the data indicate that the flavonoid glycosides of A. wilsonii seeds exhibit nitric oxide release inhibitory activity through mitogen-activated protein kinase (p38), nuclear factor kappa-light-chain-enhancer of activated B, and signal transducer and activator of transcription-3 cross-talk signaling pathways.
Collapse
Affiliation(s)
- Hui Na Cao
- Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, West Area, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Jing Ya Ruan
- Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, West Area, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Yu Han
- Institute of TCM, Tianjin University of Traditional Chinese Medicine, West Area, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Wei Zhao
- Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, West Area, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Ying Zhang
- Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, West Area, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Chang Gao
- Institute of TCM, Tianjin University of Traditional Chinese Medicine, West Area, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Hong Hua Wu
- Institute of TCM, Tianjin University of Traditional Chinese Medicine, West Area, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Lin Ma
- Institute of TCM, Tianjin University of Traditional Chinese Medicine, West Area, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Xiu Mei Gao
- Institute of TCM, Tianjin University of Traditional Chinese Medicine, West Area, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Yi Zhang
- Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, West Area, Tuanbo New Town, Jinghai District, Tianjin, China
- Institute of TCM, Tianjin University of Traditional Chinese Medicine, West Area, Tuanbo New Town, Jinghai District, Tianjin, China
| | - Tao Wang
- Tianjin Key Laboratory of TCM Chemistry and Analysis, Tianjin University of Traditional Chinese Medicine, West Area, Tuanbo New Town, Jinghai District, Tianjin, China
- Institute of TCM, Tianjin University of Traditional Chinese Medicine, West Area, Tuanbo New Town, Jinghai District, Tianjin, China
| |
Collapse
|
7
|
Okuno K, Xu C, Pascual-Sabater S, Tokunaga M, Han H, Fillat C, Kinugasa Y, Goel A. Berberine Overcomes Gemcitabine-Associated Chemoresistance through Regulation of Rap1/PI3K-Akt Signaling in Pancreatic Ductal Adenocarcinoma. Pharmaceuticals (Basel) 2022; 15:1199. [PMID: 36297310 PMCID: PMC9611392 DOI: 10.3390/ph15101199] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 09/22/2022] [Accepted: 09/23/2022] [Indexed: 11/17/2022] Open
Abstract
Gemcitabine (Gem)-based chemotherapy is one of the first-line treatments for pancreatic ductal adenocarcinoma (PDAC). However, its clinical effect is limited due to development of chemoresistance. Various naturally occurring compounds, including Berberine (BBR), provide an anti-cancer efficacy with time-tested safety, individually and in combination with chemotherapeutic drugs. Accordingly, we hypothesized that BBR might enhance the chemosensitivity to Gem in PDAC. In this study, cell culture studies using MIA PaCa-2 and BxPC-3 cells, followed by analysis in patient-derived organoids were performed to evaluate the anti-cancer effects of BBR in PDAC. Considering that cancer is a significant manifestation of increased chronic inflammatory stress, systems biology approaches are prudent for the identification of molecular pathways and networks responsible for phytochemical-induced anti-cancer activity, we used these approaches for BBR-mediated chemosensitization to Gem. Firstly, Gem-resistant (Gem-R) PDAC cells were established, and the combination of BBR and Gem revealed superior anti-cancer efficacy in Gem-R cells. Furthermore, the combination treatment induced cell cycle arrest and apoptosis in Gem-R PDAC cells. Transcriptomic profiling investigated the Rap1 and PI3K-Akt signaling pathway as a key regulator of Gem-resistance and was a key mediator for BBR-mediated chemosensitization in PDAC cells. All cell culture-based findings were successfully validated in patient-derived organoids. In conclusion, we demonstrate that BBR-mediated reversal of chemoresistance to Gem manifests through Rap1/PI3K-Akt signaling in PDAC.
Collapse
Affiliation(s)
- Keisuke Okuno
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, CA 91016, USA
- Department of Gastrointestinal Surgery, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Caiming Xu
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, CA 91016, USA
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116004, China
| | - Silvia Pascual-Sabater
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Masanori Tokunaga
- Department of Gastrointestinal Surgery, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Haiyong Han
- Molecular Medicine Division, The Translational Genomics Research Institute, Phoenix, AZ 85004, USA
| | - Cristina Fillat
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Yusuke Kinugasa
- Department of Gastrointestinal Surgery, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Ajay Goel
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Monrovia, CA 91016, USA
- City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
8
|
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] [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
|
9
|
Nishimoto A. Effective combinations of anti-cancer and targeted drugs for pancreatic cancer treatment. World J Gastroenterol 2022; 28:3637-3643. [PMID: 36161054 PMCID: PMC9372808 DOI: 10.3748/wjg.v28.i28.3637] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/06/2022] [Accepted: 06/30/2022] [Indexed: 02/06/2023] Open
Abstract
Pancreatic cancer is highly aggressive and lethal. Due to the lack of effective methods for detecting the disease at an early stage, pancreatic cancer is frequently diagnosed late. Gemcitabine has been the standard chemotherapy drug for patients with pancreatic cancer for over 20 years, but its anti-tumor effect is limited. Therefore, FOLFIRINOX (leucovorin, fluorouracil, irinotecan, oxaliplatin) as well as combination therapies using gemcitabine and conventional agents, such as cisplatin and capecitabine, has also been administered; however, these have not resulted in complete remission. Therefore, there is a need to develop novel and effective therapies for pancreatic cancer. Recently, some studies have reported that combinations of gemcitabine and targeted drugs have had significant anti-tumor effects on pancreatic cancer cells. As gemcitabine induced DNA damage response, the proteins related to DNA damage response can be suitable additional targets for novel gemcitabine-based combination therapy. Furthermore, KRAS/ RAF/MEK/ERK signaling triggered by oncogenic mutated KRAS and autophagy are frequently activated in pancreatic cancer. Therefore, these characteristics of pancreatic cancer are potential targets for developing effective novel therapies. In this minireview, combinations of gemcitabine and targeted drugs to these characteristics, combinations of targeted drugs, combinations of natural products and anti-cancer agents, including gemcitabine, and combinations among natural products are discussed.
Collapse
Affiliation(s)
- Arata Nishimoto
- Division of Basic Pharmaceutical Science, Department of Pharmacy, Faculty of Pharmaceutical Sciences, Sanyo-Onoda City University, Sanyo-Onoda City 756-0884, Yamaguchi, Japan
| |
Collapse
|
10
|
β-Escin reduces cancer progression in aggressive MDA-MB-231 cells by inhibiting glutamine metabolism through downregulation of c-myc oncogene. Mol Biol Rep 2022; 49:7409-7415. [PMID: 35655054 DOI: 10.1007/s11033-022-07536-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 04/22/2022] [Accepted: 04/27/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND The c-myc oncogene, which causes glutamine dependence in triple negative breast cancers (TNBC), is also the target of one of the signaling pathways affected by β-Escin. METHODS AND RESULTS We sought to determine how c-myc protein affects glutamine metabolism and the proteins, glutamine transporter alanine-serine-cysteine 2 (ASCT2) and glutaminase (GLS1), in β-Escin-treated MDA-MB-231 cells using glutamine uptake and western blot analysis. Cell viability, colony formation, migration and apoptosis were also evaluated in MDA-MB-231 cells in response to β-Escin treatment using MTS, colony forming, wound healing, and Annexin-V assay. We determined that β-Escin decreased glutamine uptake and reduced c-myc and GLS1 protein expressions and increased the expression of ASCT2. In addition, this inhibition of glutamine metabolism decreased cell proliferation, colony formation and migration, and induced apoptosis. CONCLUSIONS In this study, it was suggested that β-Escin inhibits glutamine metabolism via c-myc in MDA-MB-231 cells, and it is thought that as a result of interrupting the energy supply in these cells via c-myc, it results in a decrease in the carcinogenic properties of the cells. Consequently, β-Escin may be promising as a therapeutic agent for glutamine-dependent cancers.
Collapse
|
11
|
Abstract
Horse chestnut is known as a venotonizing agent of plant origin. The main active ingredient of chestnut common extract is aescin. It has anti-edema, anti-inflammatory and venotonizing properties. The aescin medicinal agent should be used for chronic vein disease, hemorrhoidal disease and post-traumatic edema. The pharmacological properties of chestnut horse extract allow the inclusion of medications based on it in the rehabilitation program of patients who have suffered a new coronavirus infection (COVID-19).
Collapse
|
12
|
Kenny HA, Hart PC, Kordylewicz K, Lal M, Shen M, Kara B, Chen YJ, Grassl N, Alharbi Y, Pattnaik BR, Watters KM, Patankar MS, Ferrer M, Lengyel E. The Natural Product β-Escin Targets Cancer and Stromal Cells of the Tumor Microenvironment to Inhibit Ovarian Cancer Metastasis. Cancers (Basel) 2021; 13:cancers13163931. [PMID: 34439084 PMCID: PMC8394501 DOI: 10.3390/cancers13163931] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/30/2021] [Accepted: 07/31/2021] [Indexed: 01/11/2023] Open
Abstract
Simple Summary β-escin, a component of horse chestnut seed extract, was first identified as an inhibitor of ovarian cancer (OvCa) adhesion/invasion in our high-throughput screening program using a three-dimensional organotypic model assembled from primary human cells and extracellular matrix. The goal of the study presented here is to determine if β-escin and structurally-similar compounds have a therapeutic potential against OvCa metastasis. β-escin and cardiac glycosides inhibit ovarian cancer adhesion/invasion to the omental microenvironment in vivo, and β-escin inhibits ovarian cancer metastasis in the prevention and intervention setting. Additionally, β-escin was found to decrease the stemness of ovarian cancer cells, inhibit extracellular matrix production in the tumor microenvironment, and inhibit HIF1α stability in ovarian cancer cells and the tumor microenvironment. This study reveals that the natural compound β-escin has therapeutic potential because of its ability to prevent OvCa dissemination by targeting both cancer and stromal cells in the OvCa tumor microenvironment. Abstract The high mortality of OvCa is caused by the wide dissemination of cancer within the abdominal cavity. OvCa cells metastasize to the peritoneum, which is covered by mesothelial cells, and invade into the underlying stroma, composed of extracellular matrices (ECM) and stromal cells. In a study using a three-dimensional quantitative high-throughput screening platform (3D-qHTS), we found that β-escin, a component of horse chestnut seed extract, inhibited OvCa adhesion/invasion. Here, we determine whether β-escin and structurally similar compounds have a therapeutic potential against OvCa metastasis. Different sources of β-escin and horse chestnut seed extract inhibited OvCa cell adhesion/invasion, both in vitro and in vivo. From a collection of 160 structurally similar compounds to β-escin, we found that cardiac glycosides inhibited OvCa cell adhesion/invasion and proliferation in vitro, and inhibited adhesion/invasion and metastasis in vivo. Mechanistically, β-escin and the cardiac glycosides inhibited ECM production in mesothelial cells and fibroblasts. The oral administration of β-escin inhibited metastasis in both OvCa prevention and intervention mouse models. Specifically, β-escin inhibited ECM production in the omental tumors. Additionally, the production of HIF1α-targeted proteins, lactate dehydrogenase A, and hexokinase 2 in omental tumors was blocked by β-escin. This study reveals that the natural compound β-escin has a therapeutic potential because of its ability to prevent OvCa dissemination by targeting both cancer and stromal cells in the OvCa tumor microenvironment.
Collapse
Affiliation(s)
- Hilary A. Kenny
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, University of Chicago, Chicago, IL 60637, USA; (P.C.H.); (K.K.); (B.K.); (Y.-J.C.); (K.M.W.); (E.L.)
- Correspondence:
| | - Peter C. Hart
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, University of Chicago, Chicago, IL 60637, USA; (P.C.H.); (K.K.); (B.K.); (Y.-J.C.); (K.M.W.); (E.L.)
| | - Kasjusz Kordylewicz
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, University of Chicago, Chicago, IL 60637, USA; (P.C.H.); (K.K.); (B.K.); (Y.-J.C.); (K.M.W.); (E.L.)
| | - Madhu Lal
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20852, USA; (M.L.); (M.S.); (M.F.)
| | - Min Shen
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20852, USA; (M.L.); (M.S.); (M.F.)
| | - Betul Kara
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, University of Chicago, Chicago, IL 60637, USA; (P.C.H.); (K.K.); (B.K.); (Y.-J.C.); (K.M.W.); (E.L.)
| | - Yen-Ju Chen
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, University of Chicago, Chicago, IL 60637, USA; (P.C.H.); (K.K.); (B.K.); (Y.-J.C.); (K.M.W.); (E.L.)
| | - Niklas Grassl
- Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany;
| | - Yousef Alharbi
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53792, USA; (Y.A.); (M.S.P.)
| | - Bikash R. Pattnaik
- Department of Pediatrics and Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA;
| | - Karen M. Watters
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, University of Chicago, Chicago, IL 60637, USA; (P.C.H.); (K.K.); (B.K.); (Y.-J.C.); (K.M.W.); (E.L.)
| | - Manish S. Patankar
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53792, USA; (Y.A.); (M.S.P.)
| | - Marc Ferrer
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD 20852, USA; (M.L.); (M.S.); (M.F.)
| | - Ernst Lengyel
- Department of Obstetrics and Gynecology, Section of Gynecologic Oncology, University of Chicago, Chicago, IL 60637, USA; (P.C.H.); (K.K.); (B.K.); (Y.-J.C.); (K.M.W.); (E.L.)
| |
Collapse
|
13
|
Omi K, Matsuo Y, Ueda G, Aoyama Y, Kato T, Hayashi Y, Imafuji H, Saito K, Tsuboi K, Morimoto M, Ogawa R, Takahashi H, Takiguchi S. Escin inhibits angiogenesis by suppressing interleukin‑8 and vascular endothelial growth factor production by blocking nuclear factor‑κB activation in pancreatic cancer cell lines. Oncol Rep 2021; 45:55. [PMID: 33760162 PMCID: PMC7962110 DOI: 10.3892/or.2021.8006] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 12/21/2020] [Indexed: 12/20/2022] Open
Abstract
Pancreatic cancer (PaCa) is one of the most aggressive types of cancer. Thus, the development of new and more effective therapies is urgently required. Escin, a pentacyclic triterpenoid from the horse chestnut, has been reported to exhibit antitumor potential by reducing cell proliferation and blocking the nuclear factor‑κB (NF‑κB) signaling pathway in several types of cancer. Our previous study reported that NF‑κB enhanced the secretion of interleukin (IL)‑8 and vascular endothelial growth factor (VEGF), thereby inducing angiogenesis in PaCa cell lines. In the present study, it was examined whether escin inhibited angiogenesis by blocking NF‑κB activation in PaCa. It was initially confirmed that escin, at concentrations >10 µM, significantly inhibited the proliferation of several PaCa cell lines. Next, using immunocytochemical staining, it was found that escin inhibited the nuclear translocation of NF‑κB. Furthermore, ELISA confirmed that NF‑κB activity in the escin‑treated PaCa cells was significantly inhibited and reverse transcription‑quantitative PCR showed that the mRNA expression levels of tumor necrosis factor‑α‑induced IL‑8 and VEGF were significantly suppressed following escin treatment in the PaCa cell lines. ELISA also showed that escin decreased the secretion of IL‑8 and VEGF from the PaCa cells. Furthermore, tube formation in immortalized human endothelial cells was inhibited following incubation with the supernatants from escin‑treated PaCa cells. These results indicated that escin inhibited angiogenesis by reducing the secretion of IL‑8 and VEGF by blocking NF‑κB activity in PaCa. In conclusion, escin could be used as a novel molecular therapy for PaCa.
Collapse
Affiliation(s)
- Kan Omi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Yoichi Matsuo
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Goro Ueda
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Yoshinaga Aoyama
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Tomokatsu Kato
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Yuichi Hayashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Hiroyuki Imafuji
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Kenta Saito
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Ken Tsuboi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Mamoru Morimoto
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Ryo Ogawa
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Hiroki Takahashi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Shuji Takiguchi
- Department of Gastroenterological Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| |
Collapse
|
14
|
Yang Y, Wang L, Yuan M, Yu Q, Fu F. Anti-Inflammatory and Gastroprotective Effects of Escin. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20982111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Escin is a triterpenoid saponin extracted from the fruit of Aesculus wilsonii Rehd. and Aesculus hippocastanum (Hippocastanaceae). Clinically, it is widely used in the treatment of edema induced by either trauma or surgery, as well as treating chronic venous insufficiency. The anti-inflammatory and antiedema effects of escin have been extensively investigated. This article systematically reviews the effects of escin on inflammation and gastrointestinal diseases, including its role in inflammation, as an antioxidant, and in inhibiting gastric acid secretion and promoting gastrointestinal movement, especially, the molecular mechanism. The advantages and potential uses of escin have also been discussed.
Collapse
Affiliation(s)
- Yunqi Yang
- Department of Pharmacology, School of Pharmacy, University of Yantai, P. R. China
| | - Linlin Wang
- Department of Pharmacology, School of Pharmacy, University of Yantai, P. R. China
| | - Man Yuan
- Department of Pharmacology, School of Pharmacy, University of Yantai, P. R. China
| | - Qi Yu
- Department of Pharmacology, School of Pharmacy, University of Yantai, P. R. China
| | - Fenghua Fu
- Department of Pharmacology, School of Pharmacy, University of Yantai, P. R. China
| |
Collapse
|
15
|
Yan Y, Lin HW, Zhuang ZN, Li M, Guo S. Interleukin-1 receptor antagonist enhances chemosensitivity to fluorouracil in treatment of Kras mutant colon cancer. World J Gastrointest Oncol 2020; 12:877-892. [PMID: 32879665 PMCID: PMC7443842 DOI: 10.4251/wjgo.v12.i8.877] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/20/2020] [Accepted: 07/26/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Kras mutant colon cancer shows abnormal activation of the nuclear factor kappa-B (NF-κB) pathway, resulting in the proliferation of tumor cells. Treatment with fluorouracil (5-FU) might not achieve the expected inhibition of proliferation of malignant cells based on the fluorouracil-induced activation of the NF-κB pathway.
AIM To detect whether interleukin (IL)-1 receptor antagonist (IL-1RA) could increase the chemosensitivity to 5-FU by decreasing the activation of the NF-κB pathway and reducing the proliferation of colon cancer cells.
METHODS Western blot analysis was performed to detect the persistent activation of the NF-κB pathway in colon cancer cell lines. Reverse transcription-polymerase chain reaction was used to detect the IL-1RA-reduced expression levels of IL-6, IL-8, IL-17, IL-21 and TLR4 in colon cancer cell lines. We used a xenograft nude mouse model to demonstrate the downregulation of the NF-κB pathway by blocking the NF-κB-regulated IL-1α feedforward loop, which could increase the efficacy of chemotherapeutic agents in inhibiting tumor cell growth.
RESULTS IL-1 receptor antagonist could decrease the expression of IL-1α and IL-1β and downregulate the activity of the NF-κB pathway in Kras mutant colon cancer cells. Treatment with 5-FU combined with IL-1RA could increase the chemosensitivity of the SW620 cell line, and decreased expression of the TAK1/NF-κB and MEK pathways resulted in limited proliferation in the SW620 cell line.
CONCLUSION Adjuvant chemotherapy with IL-1RA and 5-FU has a stronger effect than single chemotherapeutic drugs. IL-1RA combined with fluorouracil could be a potential neoadjuvant chemotherapy in the clinic.
Collapse
Affiliation(s)
- Yan Yan
- Department of Operating Room, Qilu Hospital, Shandong University, Jinan 250012, Shandong Province, China
| | - Hong-Wei Lin
- Department of Gastrointestinal Surgery, Beijing Tsinghua Changgung Hospital School of Clinical Medicine, Tsinghua University, Beijing 102200, China
| | - Zhuo-Nan Zhuang
- Department of Gastrointestinal Surgery, Beijing Tsinghua Changgung Hospital School of Clinical Medicine, Tsinghua University, Beijing 102200, China
| | - Ming Li
- Department of General Surgery, Zouping Traditional Chinese Medicine Hospital, Zhouping 256200, Shandong Province, China
| | - Sen Guo
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan 250012, Shandong Province, China
| |
Collapse
|
16
|
Khatoon E, Banik K, Harsha C, Sailo BL, Thakur KK, Khwairakpam AD, Vikkurthi R, Devi TB, Gupta SC, Kunnumakkara AB. Phytochemicals in cancer cell chemosensitization: Current knowledge and future perspectives. Semin Cancer Biol 2020; 80:306-339. [DOI: 10.1016/j.semcancer.2020.06.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 06/11/2020] [Accepted: 06/12/2020] [Indexed: 02/07/2023]
|
17
|
New Indole Glycosides from Aesculus chinensis var. chekiangensis and Their Neuroprotective Activities. Molecules 2019; 24:molecules24224063. [PMID: 31717579 PMCID: PMC6891264 DOI: 10.3390/molecules24224063] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/04/2019] [Accepted: 11/07/2019] [Indexed: 11/27/2022] Open
Abstract
The dried seeds of Aesculus chinensis Bge. var. chekiangensis (Hu et Fang) Fang, called “Suo Luo Zi”, have been used in traditional Chinese medicine. Nevertheless, most studies have been focused on components of less polarity fractions. In this research, twelve indoles, including six new indole glycosides (1–6) as well as six known analogs were isolated from the polar portion which has been seldom studied. This is the first description of N-glucosylated indoles obtained from the genus of Aesculus. Structures of the new compounds (1–6) were elucidated based on comprehensive interpretation of HRESIMS, 1D and 2D NMR. Additionally, the neuroprotective activities of the N-glucosylated indoles were evaluated for the first time indicating that compounds 1–5 and 9–10 exhibited moderate neuroprotective activities. Further cytotoxicity tests of isolates 1–10 on three human tumor cell lines suggested that none of these compounds were cytotoxic (IC50 > 50 μM).
Collapse
|
18
|
Mazrouei R, Raeisi E, Lemoigne Y, Heidarian E. Activation of p53 Gene Expression and Synergistic Antiproliferative Effects of 5-Fluorouracil and β-escin on MCF7 Cells. JOURNAL OF MEDICAL SIGNALS & SENSORS 2019; 9:196-203. [PMID: 31544060 PMCID: PMC6743244 DOI: 10.4103/jmss.jmss_44_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
One of the most common malignancies in women is breast cancer. β-escin has pharmacological anticancer effects. 5-fluorouracil (5-FU) has antimetabolite and antiproliferative properties. The purpose of this study was to investigate the combined effects of 5-FU and β-escin on apoptosis, colony formation, Bcl-2 signaling protein, and p53 gene expression in MCF7 breast cancer cell line. The cytotoxic effects, the number of colonies, apoptosis, p53 gene expression, and Bcl-2 signaling protein of the combined 5-FU and β-escin on MCF7 cells were determined using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide, clonogenic assay, flow cytometry, real-time quantitative polymerase chain reaction, and western blotting methods, respectively. Half-maximal inhibitory concentration values of β-escin and 5-FU were 80 μg/ml and 2 μM, respectively. The combination of 5-FU and β-escin on MCF7 cell viability showed a combination index equal to 0.5. The expression of p53 and apoptosis increased in the combination of 5-FU and β-escin on MCF7 cells compared to that of control group (P < 0.05). In addition, the number of colonies and Bcl-2 signaling protein in combination of 5-FU and β-escin decreased with respect to untreated control cells or single treatment of 5-FU and β-escin. The combination of 5-FU and β-escin not only has synergistic effects by increasing cell apoptosis and p53 gene expression but also decreases Bcl-2 signaling protein in MCF7 cell lines.
Collapse
Affiliation(s)
- Raziyeh Mazrouei
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Elham Raeisi
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.,Department of Medical Physics and Radiology, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | | - Esfandiar Heidarian
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
19
|
Satari A, Amini SA, Raeisi E, Lemoigne Y, Heidarian E. Synergetic Impact of Combined 5-Fluorouracil and Rutin on Apoptosis in PC3 Cancer Cells through the Modulation of P53 Gene Expression. Adv Pharm Bull 2019; 9:462-469. [PMID: 31592435 PMCID: PMC6773939 DOI: 10.15171/apb.2019.055] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 02/15/2019] [Accepted: 03/06/2019] [Indexed: 12/22/2022] Open
Abstract
Purpose: Prostate cancer is as far the most prevalent male cancer. Rutin (a glycoside from
quercetin flavonoid) displays antioxidant activity leading to cell apoptosis. Combined effects of
rutin with the widely used anti-cancer drug, 5-fluorouracil (5-FU), on prostate cancer cell line
(PC3) was investigated herein.
Methods: Different concentrations of combined 5-FU and rutin were applied to PC3 cells
compared to separate treatment for 48 hours. Cell viability, as well p53 gene expression
respectively were assessed by MTT assay and real-time quantitative polymerase chain reaction
(qPCR). Changes of Bcl-2 signal protein and apoptosis were determined using western blot
and flow cytometry procedures, respectively. Clonogenic assay was used to colony counts
assessment.
Results: 50% inhibitory concentration (IC50) of separate cell treatment with either rutin and
5-FU respectively were 900 μM and 3Mm, while combination index (CI) of combined 5-FU
/rutin application reached a level of synergistic effects (0.33). Combination of 5-FU/rutin
enhanced apoptosis and p53 gene expression in PC3 cells. PC3 cell colony counts and Bcl-2
signaling protein were decreased by 5-FU/rutin combination.
Conclusion: Synergistic effects of 5-FU/rutin combination on PC3 cells line enhanced apoptosis,
p53 gene expression, and down-regulation of Bcl-2 protein, compared to control separate
application. 5-FU/rutin combination does seem an interesting therapeutic pathway to be further
investigated.
Collapse
Affiliation(s)
- Atefeh Satari
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Sayed Asadollah Amini
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Elham Raeisi
- Department of Medical Physics & Radiology, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | | - Esfandiar Heidarian
- Clinical Biochemistry Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
20
|
Mao Y, Zhang W, Zhang R, Zuo J. Alkannin restrains oral squamous carcinoma cell growth, migration and invasion by regulating microRNA-9/RECK axis. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2019; 47:3153-3162. [PMID: 31349748 DOI: 10.1080/21691401.2019.1642206] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Yulong Mao
- Department of Oral and Maxillofacial Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Weiwei Zhang
- Department of Orthodontics, Binzhou Medical University Hospital, Binzhou, China
| | - Ronghe Zhang
- Department of Orthodontics, Binzhou Medical University Hospital, Binzhou, China
| | - Jinhua Zuo
- Department of Oral and Maxillofacial Surgery, Binzhou Medical University Hospital, Binzhou, China
| |
Collapse
|
21
|
Sun JM, Xu HT, Zhao L, Zhang YB, Kang PC, Song ZF, Liu HS, Cui YF. Induction of cell-cycle arrest and apoptosis in human cholangiocarcinoma cells by pristimerin. J Cell Biochem 2019; 120:12002-12009. [PMID: 30825242 DOI: 10.1002/jcb.28485] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 12/12/2018] [Accepted: 12/14/2018] [Indexed: 01/24/2023]
Abstract
Pristimerin, a triterpenoid isolated from Celastraceae and Hippocrateaceae, is known to induce cytotoxicity in several cancer cell lines. However, whether pristimerin can induce apoptosis in cholangiocarcinoma cells and the underlying mechanism remain unexplored. We assessed the function of human cholangiocarcinoma QBC and RBE cell lines using various experimental methods such as the cell viability assay to elucidate the viability of cells, flow cytometry to detect the death rate of cells, and Western blot analysis to evaluate the expression of cell cycle-related proteins and autophagy-related proteins. Human cholangiocarcinoma QBC cells were transplanted to nude mice to establish an animal model, and the effect of pristimerin on tumor growth in this model was observed. QBC and RBE cell lines treated with pristimerin (0, 5, 10, and 20 μmol/L) demonstrated the induction of apoptosis in a dose-dependent manner. The cell viability assay revealed a reduction in the cell viability with an increase in the pristimerin concentration. Similarly, flow cytometry revealed a gradual increase in the cell death rate with an increase in the pristimerin concentration. In addition, pristimerin significantly lowered the expression of apoptosis-related proteins (Bcl-2, Bcl-xL, and procaspase-3), but increased the Bax expression. Furthermore, pristimerin resulted in the G0/G1 cell-cycle arrest, reducing the expression of cell cycle-related proteins (cyclin E, CDK2, and CDK4), and increased the expression of autophagy-related proteins (LC3) in QBC cell line. Treatment with pristimerin could inhibit tumor growth in the nude mouse model. Overall, this study suggests the potential effect of pristimerin on the cell-cycle arrest and apoptosis in human cholangiocarcinoma cells.
Collapse
Affiliation(s)
- Jian-Min Sun
- Department of Hepatobiliary Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Hai-Tao Xu
- Department of Hepatobiliary Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Liang Zhao
- Department of Hepatobiliary Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yu-Bao Zhang
- Department of Hepatobiliary Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Peng-Cheng Kang
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zeng-Fu Song
- Department of Hepatobiliary Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Hai-Shi Liu
- Department of Hepatobiliary Surgery, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Yun-Fu Cui
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
22
|
Zhang X, Zhang S, Yang Y, Wang D, Gao H. Natural barrigenol-like triterpenoids: A comprehensive review of their contributions to medicinal chemistry. PHYTOCHEMISTRY 2019; 161:41-74. [PMID: 30818173 DOI: 10.1016/j.phytochem.2019.01.017] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 01/03/2019] [Accepted: 01/30/2019] [Indexed: 05/07/2023]
Abstract
Barrigenol-like triterpenoids (BATs), which contain an unusual oleanane substituted by many hydroxyl groups as the skeleton, are subdivided into five subtypes: barrigenol A1, barrigenol A2, barrigenol R1, barringtogenol C, and 16-deoxybarringtogenol C. The variations in acyl derivatives, hydroxyl groups, and carbohydrate chains in their structures have enhanced the diversity of BATs. Moreover, the stable polyhydroxy-replaced pentacyclic skeleton provides an ideal platform for structural modifications. To date, more than 500 BAT derivatives have been isolated from plants. Synchronously, BATs possess anti-tumour, anti-Alzheimer's disease, anti-inflammatory, anti-microbial, anti-obesity and anti-allergic activities by regulating numerous cellular molecules. Some BAT derivatives, such as escin obtained from Aesculus hippocastanum L. and xanthoceraside isolated from Xanthoceras sorbifolia Bunge, have been used to treat encephaloedema or inflammatory diseases. This review aims to provide comprehensive information about the chemistry, sources, bioavailability, and anti-tumour effects of BATs, with a particular emphasis on the molecular mechanisms of action. The pharmacokinetics and clinical progress are also concerned. More than 300 structures identified over past 25 years are summarized here (249 compounds) and in the supplementary information (114 compounds). Accordingly, the pharmaceutical activity of barrigenol triterpenoids suggests that some compounds should be developed as promising anti-tumour or anti- Alzheimer's disease agents in future.
Collapse
Affiliation(s)
- Xinxin Zhang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Song Zhang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Yiren Yang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Da Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China.
| | - Huiyuan Gao
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China.
| |
Collapse
|
23
|
TIGAR knockdown enhanced the anticancer effect of aescin via regulating autophagy and apoptosis in colorectal cancer cells. Acta Pharmacol Sin 2019; 40:111-121. [PMID: 29769743 DOI: 10.1038/s41401-018-0001-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2017] [Accepted: 01/15/2018] [Indexed: 12/31/2022]
Abstract
Our previous study showed that TP53-induced glycolysis and apoptosis regulator (TIGAR) regulated ROS, autophagy, and apoptosis in response to hypoxia and chemotherapeutic drugs. Aescin, a triterpene saponin, exerts anticancer effects and increases ROS levels. The ROS is a key upstream signaling to activate autophagy. Whether there is a crosstalk between TIGAR and aescin in regulating ROS, autophagy, and apoptosis is unknown. In this study, we found that aescin inhibited cell viability and colony formation, and induced DNA damage, cell cycle arrest, and apoptosis in cancer cell lines HCT-116 and HCT-8 cells. Concurrently, aescin increased the expression of TIGAR, ROS levels, and autophagy activation. Knockdown of TIGAR enhanced the anticancer effects of aescin in vitro and in vivo, whereas overexpression of TIGAR or replenishing TIGAR downstream products, NADPH and ribose, attenuated aescin-induced apoptosis. Furthermore, aescin-induced ROS elevation and autophagy activation were further strengthened by TIGAR knockdown in HCT-116 cells. However, autophagy inhibition by knockdown of autophagy-related gene ATG5 or 3-methyladenine (3-MA) exaggerated aescin-induced apoptosis when TIGAR was knocked down. In conclusion, TIGAR plays a dual role in determining cancer cell fate via inhibiting both apoptosis and autophagy in response to aescin, which indicated that inhibition of TIGAR and/or autophagy may be a junctional therapeutic target in treatment of cancers with aescin.
Collapse
|
24
|
Cheng CL, Chao WT, Li YH, Ou YC, Wang SS, Chiu KY, Yuan SY. Escin induces apoptosis in human bladder cancer cells: An in vitro and in vivo study. Eur J Pharmacol 2018; 840:79-88. [PMID: 30287153 DOI: 10.1016/j.ejphar.2018.09.033] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 09/27/2018] [Accepted: 09/27/2018] [Indexed: 12/24/2022]
Abstract
Escin (β-escin) is used as traditional folk medicine. The anti-tumour effects of escin have been demonstrated in vitro in certain cell lines, but its effect on bladder cancer has not been well investigated. In this study, the apoptotic activity of escin dissolved in dimethyl sulfoxide (DMSO) in bladder cancer cells and normal peripheral blood mononuclear cells (PBMC) and SV-HUC1 cells (controls) was determined. Cell cytotoxicity was assessed using the MTT assay. Cell cycle, Reactive oxygen species (ROS) generation, annexin V-FITC staining (for detecting early apoptosis), and changes in mitochondrial membrane potential were evaluated using flow cytometry. Expression of apoptosis-related proteins such as Fas (CD95) death receptor/FADD (Fas-associated protein with death domain) and BCL2 family of proteins was assessed using immunoblotting. Escin dose-dependently inhibited the growth of human bladder cancer cells, and showed IC50 of ~40 μM. The cell population in the sub-G1 phase, annexin-V staining, Fas expression, ratio of BAX/BCL2, cleavage of activated caspase-3/-8/-9, increase in poly (ADP-ribose) polymerase (PARP) levels, and suppression of nuclear factor kappa B (NF-κB) were observed after 24 h of escin treatment. Escin decreased mitochondrial membrane potential and increased cytochrome C release via generation of reactive oxygen species, which led to apoptosis of bladder cancer cells. Furthermore, escin effectively inhibited bladder tumour growth in a xenograft mouse model. Together, these results demonstrate that escin induces apoptosis in human bladder cancer cells through the Fas death receptor and mitochondrial pathways and inhibits bladder tumour growth. Escin is a potential chemotherapeutic agent for bladder cancer.
Collapse
Affiliation(s)
- Chen-Li Cheng
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| | - Wei-Ting Chao
- Department of Life Science, Tunghai University, Taichung 40704, Taiwan
| | - Yu-Hsuan Li
- Department of Life Science, Tunghai University, Taichung 40704, Taiwan
| | - Yen-Chuan Ou
- Division of Urology, Department of Surgery, Tungs' Taichung Metro Harbor Hospital, Taichung 43503, Taiwan
| | - Shian-Shiang Wang
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung 40705, Taiwan; School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan; Department of Applied Chemistry, National Chi Nan University, Nantou 54561, Taiwan
| | - Kun-Yuan Chiu
- Division of Urology, Department of Surgery, Taichung Veterans General Hospital, Taichung 40705, Taiwan; Department of Applied Chemistry, National Chi Nan University, Nantou 54561, Taiwan
| | - Sheau-Yun Yuan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40705,Taiwan; Department of Nursing, Hung Kuang University, Taichung 43302, Taiwan.
| |
Collapse
|
25
|
Wang Z, Chen Q, Li B, Xie JM, Yang XD, Zhao K, Wu Y, Ye ZY, Chen ZR, Qin ZH, Xing CG. Escin-induced DNA damage promotes escin-induced apoptosis in human colorectal cancer cells via p62 regulation of the ATM/γH2AX pathway. Acta Pharmacol Sin 2018; 39:1645-1660. [PMID: 29849127 PMCID: PMC6289333 DOI: 10.1038/aps.2017.192] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 12/29/2017] [Indexed: 12/18/2022]
Abstract
Escin, a triterpene saponin isolated from horse chestnut seed, has been used to treat encephaledema, tissue swelling and chronic venous insufficiency. Recent studies show that escin induces cell cycle arrest, tumor proliferation inhibition and tumor cell apoptosis. But the relationship between escin-induced DNA damage and cell apoptosis in tumor cells remains unclear. In this study, we investigated whether and how escin-induced DNA damage contributed to escin-induced apoptosis in human colorectal cancer cells. Escin (5-80 μg/mL) dose-dependently inhibited the cell viability and colony formation in HCT116 and HCT8 cells. Escin treatment induced DNA damage, leading to p-ATM and γH2AX upregulation. Meanwhile, escin treatment increased the expression of p62, an adaptor protein, which played a crucial role in controlling cell survival and tumorigenesis, and had a protective effect against escin-induced DNA damage: knockdown of p62 apparently enhanced escin-induced DNA damage, whereas overexpression of p62 reduced escin-induced DNA damage. In addition, escin treatment induced concentration- and time-dependent apoptosis. Similarly, knockdown of p62 significantly increased escin-induced apoptosis in vitro and produced en escin-like antitumor effect in vivo. Overexpression of p62 decreased the rate of apoptosis. Further studies revealed that the functions of p62 in escin-induced DNA damage were associated with escin-induced apoptosis, and p62 knockdown combined with the ATM inhibitor KU55933 augmented escin-induced DNA damage and further increased escin-induced apoptosis. In conclusion, our results demonstrate that p62 regulates ATM/γH2AX pathway-mediated escin-induced DNA damage and apoptosis.
Collapse
Affiliation(s)
- Zhong Wang
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou, 215007, China
| | - Qiang Chen
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou, 215007, China
| | - Bin Li
- Department of General Surgery, the First People's Hospital of Wu Jiang, Suzhou, 215200, China
| | - Jia-Ming Xie
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou, 215007, China
| | - Xiao-Dong Yang
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou, 215007, China
| | - Kui Zhao
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou, 215007, China
| | - Yong Wu
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou, 215007, China
| | - Zhen-Yu Ye
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou, 215007, China
| | - Zheng-Rong Chen
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou, 215007, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China.
| | - Chun-Gen Xing
- Department of General Surgery, Second Affiliated Hospital of Soochow University, Suzhou, 215007, China.
| |
Collapse
|
26
|
de Oliveira Júnior RG, Christiane Adrielly AF, da Silva Almeida JRG, Grougnet R, Thiéry V, Picot L. Sensitization of tumor cells to chemotherapy by natural products: A systematic review of preclinical data and molecular mechanisms. Fitoterapia 2018; 129:383-400. [DOI: 10.1016/j.fitote.2018.02.025] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 02/19/2018] [Accepted: 02/20/2018] [Indexed: 12/13/2022]
|
27
|
Wang Y, Wang S, Liu J, Lu Y, Li D. Licoricidin enhances gemcitabine-induced cytotoxicity in osteosarcoma cells by suppressing the Akt and NF-κB signal pathways. Chem Biol Interact 2018; 290:44-51. [PMID: 29782821 DOI: 10.1016/j.cbi.2018.05.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 04/13/2018] [Accepted: 05/17/2018] [Indexed: 10/16/2022]
Abstract
Osteosarcoma (OS) is the most common bone malignancy in children and adolescents. Combined treatments of anti-cancer drugs can remarkably improve chemotherapeutic outcomes. Gemcitabine and licoricidin both have potential anti-tumor activity in several cancers. However, the combined therapeutic efficiency of gemcitabine and licoricidin for OS has not been explored. Here, we found that licoricidin or gemcitabine inhibited OS cell viability in a dose-dependent manner. Cotreatment with licoricidin and gemcitabine enhanced gemcitabine-induced cytotoxicity in OS cells. Licoricidin suppressed activation of the Akt and nuclear factor-kappa B (NF-κB) pathways. Gemcitabine had no effect on Akt signal, but facilitated the activation of NF-κB signal in OS cells. Moreover, combined treatment of licoricidin and gemcitabine markedly curbed the activation of Akt and NF-κB pathways in OS cells. Inhibition of the Akt and NF-κB pathways enhanced gemcitabine-induced cytotoxicity in OS cells. In vivo assay further manifested that licoricidin enhanced gemcitabine-induced cytotoxicity in tumor xenograft models of OS via inactivation of the Akt and NF-κB pathways. In conclusion, licoricidin enhanced gemcitabine-induced cytotoxicity in OS cells by inactivation of the Akt and NF-κB pathways in vitro and in vivo.
Collapse
Affiliation(s)
- Yifei Wang
- Department of Orthopedics, The First Affiliated Hospital of Henan University, Kaifeng 475001, China
| | - Shengli Wang
- Department of Orthopedics, The First Affiliated Hospital of Henan University, Kaifeng 475001, China.
| | - Jianhua Liu
- Department of Orthopedics, The First Affiliated Hospital of Henan University, Kaifeng 475001, China
| | - Yanxiao Lu
- Department of Orthopedics, The First Affiliated Hospital of Henan University, Kaifeng 475001, China
| | - Donghui Li
- Department of Orthopedics, The First Affiliated Hospital of Henan University, Kaifeng 475001, China
| |
Collapse
|
28
|
Dihydroartemisinin suppresses pancreatic cancer cells via a microRNA-mRNA regulatory network. Oncotarget 2018; 7:62460-62473. [PMID: 27613829 PMCID: PMC5308739 DOI: 10.18632/oncotarget.11517] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 08/08/2016] [Indexed: 01/10/2023] Open
Abstract
Despite improvements in surgical procedures and chemotherapy, pancreatic cancer remains one of the most aggressive and fatal human malignancies, with a low 5-year survival rate of only 8%. Therefore, novel strategies for prevention and treatment are urgently needed. Here, we investigated the mechanisms underlying the anti-pancreatic cancer effects dihydroartemisinin (DHA). Microarray and systematic analysis showed that DHA suppressed proliferation, inhibited angiogenesis and promoted apoptosis in two different human pancreatic cancer cell lines, and that 5 DHA-regulated microRNAs and 11 of their target mRNAs were involved in these effects via 19 microRNA-mRNA interactions. Four of these microRNAs, 9 of the mRNAs and 17 of the interactions were experimentally verified. Furthermore, we found that the anti-pancreatic caner effects of DHA in vivo involved 4 microRNAs, 9 mRNAs and 17 microRNA-mRNA interactions. These results improve the understanding of the mechanisms by which DHA suppresses proliferation and angiogenesis and promotes apoptosis in pancreatic cancer cells and indicate that DHA, an effective antimalarial drug, might improve pancreatic cancer treatments.
Collapse
|
29
|
Cheong DHJ, Arfuso F, Sethi G, Wang L, Hui KM, Kumar AP, Tran T. Molecular targets and anti-cancer potential of escin. Cancer Lett 2018; 422:1-8. [PMID: 29474858 DOI: 10.1016/j.canlet.2018.02.027] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/06/2018] [Accepted: 02/15/2018] [Indexed: 02/06/2023]
Abstract
Escin is a mixture of triterpenoid saponins extracted from the horse chestnut tree, Aesculus hippocastanum. Its potent anti-inflammatory and anti-odematous properties makes it a choice of therapy against chronic venous insufficiency and odema. More recently, escin is being actively investigated for its potential activity against diverse cancers. It exhibits anti-cancer effects in many cancer cell models including lung adenocarcinoma, hepatocellular carcinoma and leukemia. Escin also attenuates tumor growth and metastases in various in vivo models. Importantly, escin augments the effects of existing chemotherapeutic drugs, thereby supporting the role of escin as an adjunct or alternative anti-cancer therapy. The beneficial effects of escin can be attributed to its inhibition of proliferation and induction of cell cycle arrest. By regulating transcription factors/growth factors mediated oncogenic pathways, escin also potentially mitigates chronic inflammatory processes that are linked to cancer survival and resistance. This review provides a comprehensive overview of the current knowledge of escin and its potential as an anti-cancer therapy through its anti-proliferative, pro-apoptotic, and anti-inflammatory effects.
Collapse
Affiliation(s)
- Dorothy H J Cheong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117593, Singapore
| | - Frank Arfuso
- Stem Cell and Cancer Biology Laboratory, School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth WA 6009, Australia
| | - Gautam Sethi
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City 700000, Viet Nam; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Viet Nam; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore.
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Kam Man Hui
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre, Singapore
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore; Medical Science Cluster, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Curtin Medical School, Faculty of Health Sciences, Curtin University, Perth WA, Australia; National University Cancer Institute, National University Health System, Singapore.
| | - Thai Tran
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 117593, Singapore.
| |
Collapse
|
30
|
Varinská L, Fáber L, Kello M, Petrovová E, Balážová Ľ, Solár P, Čoma M, Urdzík P, Mojžiš J, Švajdlenka E, Mučaji P, Gál P. β-Escin Effectively Modulates HUVECS Proliferation and Tube Formation. Molecules 2018; 23:E197. [PMID: 29342121 PMCID: PMC6017140 DOI: 10.3390/molecules23010197] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 01/11/2018] [Accepted: 01/12/2018] [Indexed: 12/28/2022] Open
Abstract
In the present study we evaluated the anti-angiogenic activities of β-escin (the major active compound of Aesculus hippocastanum L. seeds). Human umbilical-vein endothelial cells (HUVECs) were used as an in vitro model for studying the molecular mechanism underlying the anti-angiogenic effect of β-escin. We investigated the in vitro effects on proliferation, migration, and tube formation of HUVECs and in vivo anti-angiogenic activity was evaluated in a chick chorioallantoic membrane (CAM) angiogenesis assay. Moreover, the effect on gene expressions was determined by the RT2 ProfilerTM human angiogenesis PCR Array. It was found that β-escin exerts inhibitory effect on the basic fibroblast growth factor (bFGF)-induced proliferation, migration and tube formation, as well as CAM angiogenesis in vivo. The inhibition of critical steps of angiogenic process observed with β-escin could be partially explained by suppression of Akt activation in response to bFGF. Moreover, the anti-angiogenic effects of β-escin could also be mediated via inhibition of EFNB2 and FGF-1 gene expressions in endothelial cells. In conclusion, β-escin affects endothelial cells as a negative mediator of angiogenesis in vitro and in vivo and may therefore be considered as a promising candidate for further research elucidating its underlying mechanism of action.
Collapse
Affiliation(s)
- Lenka Varinská
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Košice, Slovakia.
- Department of Biomedical Research, East-Slovak Institute of Cardiovascular Diseases, Inc., 040 11 Košice, Slovakia.
| | - Lenka Fáber
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Košice, Slovakia.
| | - Martin Kello
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Košice, Slovakia.
| | - Eva Petrovová
- Department of Anatomy, Histology and Physiology, University of Veterinary Medicine and Pharmacy, 040 11 Košice, Slovakia.
| | - Ľudmila Balážová
- Department of Pharmacognosy and Botany, University of Veterinary Medicine and Pharmacy, 041 81 Košice, Slovakia.
| | - Peter Solár
- Department of Medical Biology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Košice, Slovakia.
| | - Matúš Čoma
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Košice, Slovakia.
| | - Peter Urdzík
- Department of Gynaecology and Obstetrics, Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Košice, Slovakia.
| | - Ján Mojžiš
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Košice, Slovakia.
| | - Emil Švajdlenka
- Department of Chemical Theory of Drugs, Faculty of Pharmacy, Comenius University, 831 04 Bratislava, Slovakia.
- Eurofins SK, Testing Laboratory Bratislava, 811 07 Bratislava, Slovakia.
| | - Pavel Mučaji
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University, 831 04 Bratislava, Slovakia.
| | - Peter Gál
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University, 040 11 Košice, Slovakia.
- Department of Biomedical Research, East-Slovak Institute of Cardiovascular Diseases, Inc., 040 11 Košice, Slovakia.
- Department of Pharmacognosy and Botany, Faculty of Pharmacy, Comenius University, 831 04 Bratislava, Slovakia.
| |
Collapse
|
31
|
Zhu M, Ying J, Lin C, Wang Y, Huang K, Zhou Y, Teng H. β-Escin inhibits the proliferation of osteosarcoma cells via blocking the PI3K/Akt pathway. RSC Adv 2018; 8:29637-29644. [PMID: 35547316 PMCID: PMC9085260 DOI: 10.1039/c8ra03578d] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/04/2018] [Indexed: 12/05/2022] Open
Abstract
β-Escin exhibits anticancer effects on a panel of established cancer cells. However, the effects of β-escin on human osteosarcoma (OS) are still unknown. The aim of the present study was to investigate whether β-escin was effective against OS both in vivo and in vitro. Our results showed that β-escin induced dose- and time-dependent effects against MG-63, OS732, U-2OS, HOS and SAOS-2 cell proliferation. β-Escin also exhibited excellent anti-proliferative and pro-apoptotic effects in an established OS xenograft model. β-Escin and cytotoxic drugs, including cisplatin, methotrexate (MTX), doxorubicin (Dox) and ifosfamide (Ifos), synergistically inhibited proliferation of MG-63 and OS732 cells in vitro. Moreover, β-escin induced apoptotic death, activated caspase-3, caspase-8 and caspase-9, and regulated expression of Bax and Bcl-2 in MG-63 cells. In addition, our results showed that β-escin treatment reduced expression of p-PI3K, p-Akt and p-mTOR both in MG-63 cells and in an MG-63 xenograft OS model. Interestingly, SC79, which is an Akt activator, inhibited the anti-proliferative effects of β-escin on MG-63 cells. Taken together, our data support the conclusion that β-escin effectively inhibits OS proliferation both in vivo and in vitro. The inhibitory effect of β-escin, at least in part, is due to the inactivation of the PI3K/Akt signalling pathway. β-Escin exhibits anticancer effects on a panel of established cancer cells. However, the effects of β-escin on human osteosarcoma (OS) are still unknown.![]()
Collapse
Affiliation(s)
- Minyu Zhu
- Department of Spine Surgery
- The First Affiliated Hospital of Wenzhou Medical University
- Wenzhou 325000
- China
| | - Jinwei Ying
- Department of Spine Surgery
- The First Affiliated Hospital of Wenzhou Medical University
- Wenzhou 325000
- China
| | - Chaowei Lin
- Department of Spine Surgery
- The First Affiliated Hospital of Wenzhou Medical University
- Wenzhou 325000
- China
| | - Yu Wang
- Department of Spine Surgery
- The First Affiliated Hospital of Wenzhou Medical University
- Wenzhou 325000
- China
| | - Kelun Huang
- Department of Spine Surgery
- The First Affiliated Hospital of Wenzhou Medical University
- Wenzhou 325000
- China
| | - Yang Zhou
- Department of Spine Surgery
- The First Affiliated Hospital of Wenzhou Medical University
- Wenzhou 325000
- China
| | - Honglin Teng
- Department of Spine Surgery
- The First Affiliated Hospital of Wenzhou Medical University
- Wenzhou 325000
- China
| |
Collapse
|
32
|
Lai ZQ, Ip SP, Liao HJ, Lu Z, Xie JH, Su ZR, Chen YL, Xian YF, Leung PS, Lin ZX. Brucein D, a Naturally Occurring Tetracyclic Triterpene Quassinoid, Induces Apoptosis in Pancreatic Cancer through ROS-Associated PI3K/Akt Signaling Pathway. Front Pharmacol 2017; 8:936. [PMID: 29311937 PMCID: PMC5744014 DOI: 10.3389/fphar.2017.00936] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Accepted: 12/11/2017] [Indexed: 12/14/2022] Open
Abstract
Brucein D (BD), a major active quassinoid in Brucea javanica, has exhibited pronounced anticancer activities. However, the biologic mechanisms have not been fully explored. In this study, BD exhibited more potent cytotoxic effect on pancreatic cancer (PanCa) cell lines, while exerted weaker cytotoxic effects on GES-1 cells (non-tumorigenic). BD was shown to elicit apoptosis through inducing both the intrinsic and extrinsic mitochondria-mediated caspase activations. Furthermore, the BD-induced apoptotic effects were dependent on the accumulated reactive oxygen species (ROS) and inactivation of PI3K/Akt signaling pathway. Pretreatment with tempol completely prevented the cellular apoptosis induced by BD, and recovered the inactivation of AKT, which suggested ROS essentially involved in BD-elicited apoptosis and down-regulation of PI3K/Akt pathway. In addition, the results obtained from orthotopic xenograft in nude mice were congruent with those of the in vitro investigations. These results support the notion that BD held good potential to be further developed into an effective pharmaceutical agent for the treatment of PanCa.
Collapse
Affiliation(s)
- Zheng-Quan Lai
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Siu-Po Ip
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Hui-Jun Liao
- Department of Clinical Pharmacy and Pharmaceutical Services, Shenzhen Sixth People's Hospital - Nanshan Hospital, Shenzhen, China
| | - Zheng Lu
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong.,Liver Cirrhosis Diagnosis and Treatment Center, Beijing 302 Hospital, Beijing, China
| | - Jian-Hui Xie
- Guangdong Provincial Key Laboratory of Clinical Research on Traditional Chinese Medicine Syndrome, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zi-Ren Su
- Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yun-Long Chen
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yan-Fang Xian
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Po-Sing Leung
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Zhi-Xiu Lin
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
33
|
Skorkina MY, Shamray EA, Salo VA, Buchelnikov AS, Evstigneev MP. Study of the properties of doxorubicin-resistant cells affected by acute leucosis. J Bioenerg Biomembr 2017; 50:53-58. [PMID: 29260425 DOI: 10.1007/s10863-017-9738-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 11/28/2017] [Indexed: 12/20/2022]
Abstract
The stiffness of cell membrane was found to be one of the factors determining resistance of a cell in vitro to antibiotic doxorubicin action. Membranes of surviving cells are negatively charged (-35 - -30 mV) and have high values of stiffness (2.2-5.1 μРа) at the doxorubicin concentrations in the medium of 1-500 μg/ml. If the drug concentration and exposure time are being increased, only cells with 'soft' membrane (0.25-1 μРа) and positive surface potential (15-29 mV) survive. The data obtained have important prognostic value in studying drug resistance of tumour blood cells and can be used as objective markers of efficiency of the antitumor therapy.
Collapse
Affiliation(s)
- Marina Yu Skorkina
- Department of Biology, Belgorod State University, 85 Pobedy str, 308015, Belgorod, Russia.
| | - Elena A Shamray
- Department of Biology, Belgorod State University, 85 Pobedy str, 308015, Belgorod, Russia
| | - Victoria A Salo
- Department of Physics, Sevastopol State University, 33 Universitetskaya str, 299053, Sevastopol, Russia
| | - Anatoly S Buchelnikov
- Theodosius Dobzhansky Center for Genome Bioinformatics, Saint Petersburg State University, 41 Sredniy Avenue, 199034, Saint Petersburg, Russia.
| | - Maxim P Evstigneev
- Department of Physics, Sevastopol State University, 33 Universitetskaya str, 299053, Sevastopol, Russia
| |
Collapse
|
34
|
Shikonin causes apoptosis by disrupting intracellular calcium homeostasis and mitochondrial function in human hepatoma cells. Exp Ther Med 2017; 15:1484-1492. [PMID: 29434733 PMCID: PMC5776525 DOI: 10.3892/etm.2017.5591] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 10/27/2017] [Indexed: 11/09/2022] Open
Abstract
Shikonin is known to suppress proliferation and induce apoptosis in a variety of cancer cell lines. In the present study, SMMC-7721 human hepatocellular carcinoma cells were treated with shikonin (1, 2 or 4 µM) for 12–48 h. Cell morphological alterations and DNA damage were determined. Furthermore, changes in cell cycle, mitochondrial transmembrane potential, calcium homeostasis and levels of reactive oxygen species were measured. Shikonin-treated SMMC-7721 cells exhibited morphological changes and DNA damage. Shikonin inhibited cell proliferation causing cell cycle arrest at the G0/G1 phase and induced apoptosis in a dose- and time-dependent manner. Shikonin-induced apoptosis was associated with activation of caspases-3, −8 and −9, elevated levels of intracellular Ca2+ and reactive oxygen species, reduced mitochondrial membrane potential and enhanced efflux of Ca2+ and K+. Gene expression B-cell lymphoma 2 (Bcl-2)-associated X protein (Bax), p53 and caspase-3 was up-regulated, whereas Bcl-2 expression was downregulated. Shikonin caused apoptosis by inhibiting cell cycle progression, disrupting Ca2+ homeostasis, inducing oxidative stress and triggering mitochondrial dysfunction. Activation of caspases-3, −8 and −9, K+ efflux, and regulation of Bax, Bcl-2, p53 and caspase-3 expression are involved in the process. These results provide in-depth insight into the mechanisms of action of shikonin in the treatment of cancer.
Collapse
|
35
|
Zhu J, Yu W, Liu B, Wang Y, Shao J, Wang J, Xia K, Liang C, Fang W, Zhou C, Tao H. Escin induces caspase-dependent apoptosis and autophagy through the ROS/p38 MAPK signalling pathway in human osteosarcoma cells in vitro and in vivo. Cell Death Dis 2017; 8:e3113. [PMID: 29022891 PMCID: PMC5682655 DOI: 10.1038/cddis.2017.488] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 08/05/2017] [Accepted: 08/09/2017] [Indexed: 02/06/2023]
Abstract
Osteosarcoma is one of the most malignant neoplasms in adolescents, and it generally develops multidrug resistance. Escin, a natural mixture of triterpene saponins isolated from Aesculus hippocastanum (horse chestnut), has demonstrated potent anti-tumour potential in vitro and in vivo. In the present study, we found that escin inhibited osteosarcoma proliferation in a dose- and time-dependent manner. Additionally, escin-induced apoptosis was evidenced by the increased expression of caspase-related proteins and the formation of apoptotic bodies. Escin also induced autophagy, with elevated LC3, ATG5, ATG12 and Beclin expression as well as autophagosome formation. Inhibition of escin-induced autophagy promoted apoptosis. Moreover, p38 mitogen-activated protein kinases (MAPKs) and reactive oxygen species (ROS) were activated by escin. A p38 MAPK inhibitor partially attenuated the autophagy and apoptosis triggered by escin, but a ROS scavenger showed a greater inhibitory effect. Finally, the therapeutic efficacy of escin against osteosarcoma was demonstrated in an orthotopic model. Overall, escin counteracted osteosarcoma by inducing autophagy and apoptosis via the activation of the ROS/p38 MAPK signalling pathway; these findings provide evidence for escin as a novel and potent therapeutic for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Jian Zhu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| | - Wei Yu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| | - Bing Liu
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| | - Yitian Wang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| | - Jianlin Shao
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Junjie Wang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| | - Kaishun Xia
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| | - Chengzhen Liang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| | - Weijing Fang
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| | - Chenhe Zhou
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| | - Huimin Tao
- Department of Orthopedics, 2nd Affiliated Hospital, School of Medicine, Zhejiang University, #88 Jie Fang Road, Hangzhou, Zhejiang 310009, PR China.,Orthopedics Research Institute of Zhejiang University, #88, Jiefang Road, Hangzhou 310009, PR China
| |
Collapse
|
36
|
Li Y, Li L, Zhang G, Wang Y, Chen H, Kong R, Pan S, Sun B. Crucial microRNAs and genes in metformin’s anti-pancreatic cancer effect explored by microRNA-mRNA integrated analysis. Invest New Drugs 2017; 36:20-27. [DOI: 10.1007/s10637-017-0508-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Accepted: 08/29/2017] [Indexed: 02/07/2023]
|
37
|
Mei JY, Zhang MJ, Wang YY, Liu YH. The positive clinical therapeutically effects of Escin on advanced thyroid cancer. Cancer Med 2017; 6:937-943. [PMID: 28378396 PMCID: PMC5430090 DOI: 10.1002/cam4.1031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 01/02/2017] [Accepted: 01/16/2017] [Indexed: 01/05/2023] Open
Abstract
The incidences of thyroid cancer keep rising worldwide over the past few decades. Although most thyroid cancers are indolent and highly curable, the treatment for advanced thyroid cancer remains challengeable in clinical practice. We performed two separate cohorts to evaluate the safety and efficiency of Escin in patients with advanced thyroid cancer . In cohort 1, 120 patients were divided into four groups equally and were administrated with placebo or different dosages of Escin. The pharmacokinetics of Escin and the side effects were evaluated. In cohort 2, 120 patients were treated with Escin. Several biomarkers related to the progression of thyroid cancer were evaluated. Kaplan–Meier (KM) analyses were performed to evaluate progression‐free survival (PFS) and overall survival (OS). The serum Escin concentrations were stable during the treatment. Escin (0.6 mg/kg/day for 9 days, intravenous injection) was tolerable for patients with thyroid cancer . Escin significantly reduced the serum levels of TSH, TgAb, Tg, and calcitonin and prolonged the PFS and OS for patients with advanced thyroid cancer. This study showed Escin is efficient and well tolerated in patients with advanced thyroid cancer. Future studies are needed to investigate the mechanism of Escin on thyroid cancer and the proper dosage of Escin clinically.
Collapse
Affiliation(s)
- Jin-Yu Mei
- Department of Otolaryngology, The First Hospital of Anhui Medical University, Hefei, 230031, China.,Department of Otolaryngology, The Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Ming-Jun Zhang
- Department of Oncology, The Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yuan-Yuan Wang
- Department of Pharmacy, The Second Hospital of Anhui Medical University, Hefei, 230601, China
| | - Ye-Hai Liu
- Department of Otolaryngology, The First Hospital of Anhui Medical University, Hefei, 230031, China
| |
Collapse
|
38
|
Natural Products as Adjunctive Treatment for Pancreatic Cancer: Recent Trends and Advancements. BIOMED RESEARCH INTERNATIONAL 2017; 2017:8412508. [PMID: 28232946 PMCID: PMC5292383 DOI: 10.1155/2017/8412508] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 12/09/2016] [Accepted: 12/27/2016] [Indexed: 12/17/2022]
Abstract
Pancreatic cancer is a type of common malignant tumors with high occurrence in the world. Most patients presented in clinic had pancreatic cancer at advanced stages. Furthermore, chemotherapy or radiotherapy had very limited success in treating pancreatic cancer. Complementary and alternative medicines, such as natural products/herbal medicines, represent exciting adjunctive therapies. In this review, we summarize the recent advances of using natural products/herbal medicines, such as Chinese herbal medicine, in combination with conventional chemotherapeutic agents to treat pancreatic cancer in preclinical and clinical trials.
Collapse
|
39
|
Yuan SY, Cheng CL, Wang SS, Ho HC, Chiu KY, Chen CS, Chen CC, Shiau MY, Ou YC. Escin induces apoptosis in human renal cancer cells through G2/M arrest and reactive oxygen species-modulated mitochondrial pathways. Oncol Rep 2017; 37:1002-1010. [DOI: 10.3892/or.2017.5348] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 07/01/2016] [Indexed: 11/06/2022] Open
|
40
|
Yang L, Wang Y, Guo H, Guo M. Synergistic Anti-Cancer Effects of Icariin and Temozolomide in Glioblastoma. Cell Biochem Biophys 2016; 71:1379-85. [PMID: 25384619 DOI: 10.1007/s12013-014-0360-3] [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: 12/19/2022]
Abstract
Glioblastoma is an aggressive malignancy, which is associated with poor prognosis. Temozolomide (TMZ) has been showed to be an effective chemotherapeutic agent for glioblastoma treatment; however, the response rate is not satisfactory. Icariin is a natural compound with anti-cancer activity against a variety of cancers. This study is designed to determine whether icariin could potentiate the antitumor activity of TMZ in glioblastoma. Cell proliferation and apoptosis were measured using MTT assay and flow cytometry, respectively. Expression of apoptosis and proliferation-related molecules was detected by Western blotting while NF-κB activity was detected by ELISA. Icariin dose-dependently inhibited proliferation and induced apoptosis in tested glioblastoma cell lines. Icariin enhanced the anti-tumor activity of TMZ in vitro. The anti-tumor activity of icariin and the enhanced anti-tumor activity of TMZ by icariin correlated with suppression of NF-κB activity. Our results showed that icariin exhibited anti-tumor activity and potentiated the anti-tumor activity of TMZ in glioblastoma, at least in part, by inhibiting NF-κB activity. Although more studies including clinical trials are needed, this study provides insight for using icariin as a chemosensitizing agent in clinic settings.
Collapse
Affiliation(s)
- Lijuan Yang
- Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, No.324, JingWu Road, Ji'nan, 250021, China
| | - Yuexun Wang
- CT Department, First People's Hospital of Jining, Shandong, 272011, China
| | - Hua Guo
- Shandong Provincial Hospital Affiliated to Shandong University, Shandong University, No.324, JingWu Road, Ji'nan, 250021, China
| | - Meiling Guo
- CT Department, First People's Hospital of Jining, Shandong, 272011, China.
| |
Collapse
|
41
|
Wang Y, Wu X, Zhou Y, Jiang H, Pan S, Sun B. Piperlongumine Suppresses Growth and Sensitizes Pancreatic Tumors to Gemcitabine in a Xenograft Mouse Model by Modulating the NF-kappa B Pathway. Cancer Prev Res (Phila) 2016; 9:234-44. [PMID: 26667450 DOI: 10.1158/1940-6207.capr-15-0306] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Accepted: 11/24/2015] [Indexed: 11/16/2022]
Abstract
Pancreatic cancer is an aggressive malignancy, which generally respond poorly to chemotherapy. Hence, novel agents that are safe and effective are highly needed. The aim of this study was to investigate whether piperlongumine, a natural product isolated from the fruit of the pepper Piper longum, has any efficacy against human pancreatic cancer when used either alone or in combination with gemcitabine in vitro and in a xenograft mouse model. In vitro, piperlongumine inhibited the proliferation of pancreatic cancer cell lines, potentiated the apoptotic effects of gemcitabine, inhibited the constitutive and inducible activation of NF-κB, and suppressed the NF-κB-regulated expression of c-Myc, cyclin D1, Bcl-2, Bcl-xL, Survivin, XIAP, VEGF, and matrix metalloproteinase-9 (MMP-9). Furthermore, in an in vivo xenograft model, we found piperlongumine alone significantly suppressed tumor growth and enhanced the antitumor properties of gemcitabine. These results were consistent with the downregulation of NF-κB activity and its target genes, decreased proliferation (PCNA and Ki-67), decreased microvessel density (CD31), and increased apoptosis (TUNEL) in tumor remnants. Collectively, our results suggest that piperlongumine alone exhibits significant antitumor effects against human pancreatic cancer and it further enhances the therapeutic effects of gemcitabine, possibly through the modulation of NF-κB- and NF-κB-regulated gene products.
Collapse
Affiliation(s)
- Yongwei Wang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiangsong Wu
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yinan Zhou
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Hongchi Jiang
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Shangha Pan
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bei Sun
- Department of Pancreatic and Biliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
42
|
Hyperoside induces apoptosis and inhibits growth in pancreatic cancer via Bcl-2 family and NF-κB signaling pathway both in vitro and in vivo. Tumour Biol 2015; 37:7345-55. [PMID: 26676634 DOI: 10.1007/s13277-015-4552-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 11/30/2015] [Indexed: 01/27/2023] Open
Abstract
Although advanced surgical operation and chemotherapy have been under taken, pancreatic cancer remains one of the most aggressive and fatal human malignancies with a low 5-year survival rate of less than 5 %. Therefore, novel therapeutic strategies for prevention and remedy are urgently needed in pancreatic cancer. This present research aimed to investigate the anti-cancer effects of hyperoside in human pancreatic cancer cells. Our in vitro results showed that hyperoside suppressed the proliferation and promoted apoptosis of two different human pancreatic cancer cell lines, which correlated with up-regulation of the ratios of Bax/Bcl-2 and Bcl-xL and down-regulation of levels of nuclear factor-κB (NF-κB) and NF-κB's downstream gene products. What's more, using an orthotopic model of human pancreatic cancer, we found that hyperoside also inhibited the tumor growth significantly. Mechanically, these outcomes could also be associated with the up-regulation of the ratios of Bax/Bcl-2 and Bcl-xL and down-regulation of levels of NF-κB and NF-κB's downstream gene products. Collectively, our experiments indicate that hyperoside may be a promising candidate agent for the treatment of pancreatic cancer.
Collapse
|
43
|
Bosio C, Tomasoni G, Martínez R, Olea AF, Carrasco H, Villena J. Cytotoxic and apoptotic effects of leptocarpin, a plant-derived sesquiterpene lactone, on human cancer cell lines. Chem Biol Interact 2015; 242:415-21. [PMID: 26562779 DOI: 10.1016/j.cbi.2015.11.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 10/18/2015] [Accepted: 11/04/2015] [Indexed: 12/25/2022]
Abstract
Sesquiterpene lactones have attracted much attention in drug research because they present a series of biological activities such as anticancer, antifungal, anti-inflammatory, antimicrobial and antioxidant. Leptocarpin (LTC) is a sesquiterpene lactone isolated from a native Chilean plant, Leptocarpha rivularis, which has been widely used in traditional medicine by Mapuche people. Previous work has demonstrated that LTC decreases cell viability of cancer cell lines. In this contribution, we analyze the mechanism of LTC cytotoxicity on different cancer cell lines. The results show that in all cases LTC induces an apoptotic process and inhibition of NF-κB. Apoptosis has been confirmed by observing condensation of chromatin, nuclear fragmentation, release of cytochrome c into the cytosol, and increasing of caspase-3 activity. It has also been found that LTC is an effective inhibitor of NF-κB, which suggests that leptocarpin-induced cytotoxicity involves in some degree the inhibition of NF-κB signaling pathway. The concentration at which LTC inhibits NF-κB activity to the control level is similar or even lower than that found for parthenolide and others sesquiterpene lactones. These results indicate that leptocarpine is a very interesting molecule that could be considered as therapeutic agent for cancer treatment.
Collapse
Affiliation(s)
- Claudia Bosio
- Departamento de Química, Facultad de Ciencias Exactas, Universidad Andrés Bello, Quillota 910, Viña del Mar, 2520000, Chile.
| | - Giacomo Tomasoni
- Departamento de Química, Facultad de Ciencias Exactas, Universidad Andrés Bello, Quillota 910, Viña del Mar, 2520000, Chile.
| | - Rolando Martínez
- Departamento de Química, Facultad de Ciencias Exactas, Universidad Andrés Bello, Quillota 910, Viña del Mar, 2520000, Chile.
| | - Andrés F Olea
- Instituto de Ciencias Químicas Aplicadas, Facultad de Ingeniería, Universidad Autónoma de Chile, Llano Subercaseaux 2801, San Miguel, Santiago, Chile.
| | - Héctor Carrasco
- Instituto de Ciencias Químicas Aplicadas, Facultad de Ingeniería, Universidad Autónoma de Chile, Llano Subercaseaux 2801, San Miguel, Santiago, Chile.
| | - Joan Villena
- Centro de Investigaciones Biomédicas (CIB), Escuela de Medicina, Universidad de Valparaíso, Av. Hontaneda 2664, Valparaíso, 234000, Chile.
| |
Collapse
|
44
|
Wang K, Jiang Y, Wang W, Ma J, Chen M. Escin activates AKT-Nrf2 signaling to protect retinal pigment epithelium cells from oxidative stress. Biochem Biophys Res Commun 2015; 468:541-7. [PMID: 26505797 DOI: 10.1016/j.bbrc.2015.10.117] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 10/21/2015] [Indexed: 12/19/2022]
Abstract
Here we explored the anti-oxidative and cytoprotective potentials of escin, a natural triterpene-saponin, against hydrogen peroxide (H2O2) in retinal pigment epithelium (RPE) cells. We showed that escin remarkably attenuated H2O2-induced death and apoptosis of established (ARPE-19) and primary murine RPE cells. Meanwhile, ROS production and lipid peroxidation by H2O2 were remarkably inhibited by escin. Escin treatment in RPE cells resulted in NF-E2-related factor 2 (Nrf2) signaling activation, evidenced by transcription of anti-oxidant-responsive element (ARE)-regulated genes, including HO-1, NQO-1 and SRXN-1. Knockdown of Nrf2 through targeted shRNAs/siRNAs alleviated escin-mediated ARE gene transcription, and almost abolished escin-mediated anti-oxidant activity and RPE cytoprotection against H2O2. Reversely, escin was more potent against H2O2 damages in Nrf2-over-expressed ARPE-19 cells. Further studies showed that escin-induced Nrf2 activation in RPE cells required AKT signaling. AKT inhibitors (LY294002 and perifosine) blocked escin-induced AKT activation, and dramatically inhibited Nrf2 phosphorylation, its cytosol accumulation and nuclear translocation in RPE cells. Escin-induced RPE cytoprotection against H2O2 was also alleviated by the AKT inhibitors. Together, these results demonstrate that escin protects RPE cells from oxidative stress possibly through activating AKT-Nrf2 signaling.
Collapse
Affiliation(s)
- Kaijun Wang
- Eye Center, The 2nd Affiliated Hospital, Medical College of Zhejiang University, Hangzhou, China; Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, China
| | - Yiqian Jiang
- The First People Hospital of Xiaoshan, Hangzhou, China
| | - Wei Wang
- Eye Center, The 2nd Affiliated Hospital, Medical College of Zhejiang University, Hangzhou, China; Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, China
| | - Jian Ma
- Eye Center, The 2nd Affiliated Hospital, Medical College of Zhejiang University, Hangzhou, China; Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, China
| | - Min Chen
- Eye Center, The 2nd Affiliated Hospital, Medical College of Zhejiang University, Hangzhou, China; Zhejiang Provincial Key Lab of Ophthalmology, Hangzhou, China.
| |
Collapse
|
45
|
Shi DB, Li XX, Zheng HT, Li DW, Cai GX, Peng JJ, Gu WL, Guan ZQ, Xu Y, Cai SJ. Icariin-mediated inhibition of NF-κB activity enhances the in vitro and in vivo antitumour effect of 5-fluorouracil in colorectal cancer. Cell Biochem Biophys 2015; 69:523-30. [PMID: 24435883 DOI: 10.1007/s12013-014-9827-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Colorectal cancer (CRC) is an aggressive malignancy that has a poor prognosis. 5-Fluorouracil (5-FU) is a first line chemotherapeutic medication used in the treatment of gallbladder cancer; however, the efficacy is below satisfactory. Icariin is a natural compound that is conventionally reported to have activity against a variety of cancers. This study was carried out to investigate the anti-cancer effect of icariin in CRC cells and to determine whether the compound can enhance the antitumour activity of 5-FU. Cell proliferation and apoptosis were measured using an MTT assay and flow cytometry, respectively. The activity of transcription factor NF-κB was determined by EMSA method. The expression of apoptosis- and proliferation-related proteins was determined by western blotting. The in vivo antitumour effect of combination treatment with icariin and 5-FU on CRC was also assessed using a murine model of CRC. Icariin sensitized the CRC cells to 5-FU both in vitro and in vivo. The antitumour activity of icariin and its potentiating effect on the antitumour activity of 5-FU implicated the suppression of NF-κB activity and consequent down-regulation of the gene products regulated by NF-κB. Our results showed that icariin, suppressed tumour growth and enhanced the antitumour activity of 5-FU in CRC by inhibiting NF-κB activity. Therefore, we suggest that combination of icariin with 5-FU might offer a therapeutic benefit to the patients with CRC; however, further studies are required to ascertain this proposition.
Collapse
Affiliation(s)
- De-Bing Shi
- Department of Colorectal Surgery, Cancer Hospital, Fudan University, Shanghai, 200032, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
KIM SELIM, LIU YUCHUAN, SEO SEUNGYOUNG, KIM SEONGHUN, KIM INHEE, LEE SEUNGOK, LEE SOOTEIK, KIM DAEGHON, KIM SANGWOOK. Parthenolide induces apoptosis in colitis-associated colon cancer, inhibiting NF-κB signaling. Oncol Lett 2015; 9:2135-2142. [PMID: 26137027 PMCID: PMC4467311 DOI: 10.3892/ol.2015.3017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 01/13/2015] [Indexed: 12/15/2022] Open
Abstract
Recently, the nuclear factor (NF)-κB inhibitor parthenolide (PT) was identified as a promising anticancer agent for the promotion of cancer cell apoptosis. Additionally, our previous study demonstrated that PT administration suppresses tumor growth in a xenograft model of colorectal cancer cells via regulation of the B-cell lymphoma-2 (Bcl-2) family. However, the role of PT in the development of colitis-associated colon cancer (CAC) is poorly understood. Therefore, the aim of the present study was to investigate the effects of PT administration on CAC using a murine model. Azoxymethane (AOM) and dextran sulfate sodium (DSS) were administered to induce experimental CAC in the following three groups of treated mice: i) AOM and DSS plus vehicle; ii) AOM, DSS and 2 mg/kg PT; and iii) AOM, DSS and 4 mg/kg PT. It was demonstrated that the histological acuteness of AOM/DSS-induced CAC was significantly reduced following the administration of PT, resulting in decreased NF-κB p65 expression levels via a blockade of phosphorylation and subsequent degradation of inhibitor of κB-α (IκBα). Furthermore, PT administration appeared to enhance the process of carcinogenesis via the downregulation of the antiapoptotic proteins Bcl-2 and Bcl-extra large, mediated by inhibition of NF-κB activation. Apoptosis and caspase-3 expression were markedly increased in the PT-treated group. These findings indicate that PT inhibits IκBα phosphorylation and NF-κB activation, resulting in the initiation of apoptosis and the eventual suppression of CAC development. The beneficial effects of PT treatment observed in the experimental CAC model indicate the potential chemopreventive and therapeutic role of PT in CAC.
Collapse
Affiliation(s)
- SE LIM KIM
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
- Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
| | - YU CHUAN LIU
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
- Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
| | - SEUNG YOUNG SEO
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
- Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
| | - SEONG HUN KIM
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
- Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
| | - IN HEE KIM
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
- Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
| | - SEUNG OK LEE
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
- Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
| | - SOO TEIK LEE
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
- Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
| | - DAE-GHON KIM
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
- Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
| | - SANG WOOK KIM
- Department of Internal Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
- Research Institute of Clinical Medicine, Chonbuk National University Hospital, Chonbuk National University, Jeonju, Jeollabuk-do 561-712, Republic of Korea
| |
Collapse
|
47
|
Patlolla JMR, Rao CV. Anti-inflammatory and Anti-cancer Properties of β-Escin, a Triterpene Saponin. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/s40495-015-0019-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
48
|
Huang GL, Shen DY, Cai CF, Zhang QY, Ren HY, Chen QX. β-escin reverses multidrug resistance through inhibition of the GSK3β/β-catenin pathway in cholangiocarcinoma. World J Gastroenterol 2015; 21:1148-1157. [PMID: 25632187 PMCID: PMC4306158 DOI: 10.3748/wjg.v21.i4.1148] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Revised: 09/01/2014] [Accepted: 09/30/2014] [Indexed: 02/06/2023] Open
Abstract
AIM: To develop a safe and effective agent for cholangiocarcinoma (CCA) chemotherapy.
METHODS: A drug combination experiment was conducted to determine the effects of β-escin in combination with chemotherapy on CCA cells. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay was performed to determine the effects of β-escin and common chemotherapeutics on the proliferation of human CCA cells (QBC939, Sk-ChA-1, and MZ-ChA-1). Immunocytochemistry was used to detect the expression of P-glycoprotein (P-gp) protein. Luciferase reporter assay was used to detect the activation of the Wnt/β-catenin pathway. The protein levels of P-gp, pS9-GSK3β, pT216-GSK3β, GSK3β, β-catenin, and p-β-catenin were further confirmed by western blotting.
RESULTS: The drug sensitivity of QBC939 and QBC939/5-fluorouracil (5-FU) cells to 5-FU, vincristine sulfate (VCR), or mitomycin C was significantly enhanced by β-escin compared with either agent alone (P < 0.05). In addition, the combination of β-escin (20 μmol/L) with 5-FU and VCR was synergic with a combination index < 1. Further investigation found that the mRNA and protein expression of P-gp was down-regulated by β-escin. Moreover, β-escin induced GSK3β phosphorylation at Tyr-216 and dephosphorylation at Ser-9, resulting in phosphorylation and degradation of β-catenin. Interestingly, activation of the GSK3β/β-catenin pathway induced by Wnt3a resulted in up-regulation of P-gp, which was effectively abolished by β-escin, indicating that β-escin down-regulated P-gp expression in a GSK3β-dependent manner.
CONCLUSION: β-escin was a potent reverser of P-gp-dependent multidrug resistance, with said effect likely being achieved via inhibition of the GSK3β/β-catenin pathway and thus suggesting a promising strategy of developing combination drugs for CCA.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B/genetics
- ATP Binding Cassette Transporter, Subfamily B/metabolism
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Bile Duct Neoplasms/enzymology
- Bile Duct Neoplasms/genetics
- Bile Duct Neoplasms/pathology
- Bile Ducts, Intrahepatic/drug effects
- Bile Ducts, Intrahepatic/enzymology
- Bile Ducts, Intrahepatic/pathology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cholangiocarcinoma/enzymology
- Cholangiocarcinoma/genetics
- Cholangiocarcinoma/pathology
- Dose-Response Relationship, Drug
- Drug Resistance, Multiple/drug effects
- Drug Resistance, Neoplasm/drug effects
- Escin/pharmacology
- Fluorouracil/pharmacology
- Gene Expression Regulation, Neoplastic
- Genes, Reporter
- Glycogen Synthase Kinase 3/genetics
- Glycogen Synthase Kinase 3/metabolism
- Glycogen Synthase Kinase 3 beta
- Humans
- Mitomycin/pharmacology
- Phosphorylation
- RNA, Messenger/metabolism
- Signal Transduction/drug effects
- Transfection
- Vincristine/pharmacology
- beta Catenin/genetics
- beta Catenin/metabolism
Collapse
|
49
|
Abstract
OBJECTIVE This study aimed to evaluate the ability of luteolin (Lut), gemcitabine (Gem), and their combination (Lut + Gem) to prevent the growth of pancreatic tumors in vivo. METHODS The antitumor effect of intraperitoneally administered Lut, Gem, and Lut + Gem was evaluated using an orthotopic mouse model for 6 weeks. Tumor growth after injection of human pancreatic cancer cells was assessed by measuring pancreatic tumor mass. The mechanism of action of antitumor effect was assessed by immunohistochemistry and Western blot procedures. RESULTS Luteolin + Gem significantly lowered (P = 0.048) the pancreatic tumor mass compared with control. Luteolin, Gem, and Lut + Gem significantly reduced the proliferating cell nuclear antigen expression (25%, 37%, and 37%, respectively). Luteolin + Gem treatment led to a significant reduction in the expressions of K-ras (46%, P = 0.0006), GSK-3β (34%, P = 0.014), P(Tyr216)GSK-3β (16%, P = 0.033), P(Ser311)NF-κB p65 (27%, P = 0.036), and bcl-2/bax ratio (68%, P = 0.006) while significantly increasing the expressions of cytochrome c (44%, P = 0.035) and caspase 3 (417%, P = 0.003). CONCLUSIONS Luteolin + Gem promoted apoptotic cell death in pancreatic tumor cells in vivo through inhibition of the K-ras/GSK-3β/NF-κB signaling pathway, leading to a reduction in the Bcl-2/Bax ratio, release of cytochrome c, and activation of caspase 3.
Collapse
|
50
|
Lee HS, Hong JE, Kim EJ, Kim SH. Escin Suppresses Migration and Invasion Involving the Alteration of CXCL16/CXCR6 Axis in Human Gastric Adenocarcinoma AGS Cells. Nutr Cancer 2014; 66:938-45. [DOI: 10.1080/01635581.2014.922202] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|