1
|
Zhou M, Hong J, Qiu X, Xiong Z, Liu X, Qin Z, Luo Z, Chen Q, Lin M, Min L, Yang X, Guo X, Xu B, Mao J. Serum-derived extracellular vesicles mediate acquired multidrug resistance of MCF-7 breast cancer cells induced by chemotherapeutic drugs. Biochem Pharmacol 2025; 237:116923. [PMID: 40194604 DOI: 10.1016/j.bcp.2025.116923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/25/2025] [Accepted: 04/02/2025] [Indexed: 04/09/2025]
Abstract
Multidrug resistance (MDR) in tumor cells presents a significant challenge in cancer therapy. This study investigates the role of serum-derived extracellular vesicles (EVs) in mediating MDR during chemotherapeutic exposure. The findings indicate that short- or long-term co-incubation of doxorubicin (Dox)-pretreated serum derived EVs (EVs(S-PT)) caused drug-sensitive MCF-7 breast cancer cells to develop a MDR phenotype. In addition, serum EVs contain a high concentration of unglycosylated P-glycoprotein (P-gp). Chemotherapy treatment of tumor patients or exposure to chemotherapeutic drugs in vitro activates serum glycosyltransferases, inducing glycosylation of EVs P-gp and giving it drug-pumping activity. Furthermore, damage caused by Dox to the vascular endothelial barrier facilitates the crossing of serum EVs into the tumor microenvironment. These EVs are then taken up by tumor cells, providing them with access to a significant quantity of glycosylated P-gp proteins that possess transporter activity and the ability to evade degradation by the ubiquitin proteasome system. The results indicate that EVs(S-PT) transfers glycosylated P-gp across the damaged vascular endothelial barrier into MCF-7 cells and that these glycosylated P-gp remain intracellular for a long period of time, inducing MDR in the cells. Our study highlights a novel mechanism of acquired MDR and provides a potential avenue for therapeutic interventions targeting the serum EVs pathway in cancer therapy.
Collapse
Affiliation(s)
- Mi Zhou
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jiahuan Hong
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiaofeng Qiu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zixian Xiong
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Xiaoyong Liu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhuan Qin
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zhesi Luo
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qi Chen
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Mianjie Lin
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China; School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ling Min
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou 510095, China
| | - Xiaorong Yang
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510000, China
| | - Xinmin Guo
- Department of Ultrasound, Guangzhou Red Cross Hospital, Medical College, Jinan University, Guangzhou, Guangdong 510220, China.
| | - Bin Xu
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China; School of Biosciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Jianwen Mao
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances and School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
2
|
Zeng H, Zeng X, Wang C, Wang G, Tian Q, Zhao J, Zhao L, Li R, Luo Y, Peng H, Zhang Z, Li X, Wu X. Combination therapy using Cel-CSO/Taxol NPs for reversing drug resistance in breast cancer through inhibiting PI3K/AKT/NF-κB/HIF-1α pathway. Drug Deliv Transl Res 2025; 15:992-1010. [PMID: 38922561 DOI: 10.1007/s13346-024-01653-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2024] [Indexed: 06/27/2024]
Abstract
The resistance of malignant tumors to multiple drugs is a significant obstacle in cancer treatment and prognosis. Accordingly, we synthesized a celastrol (Cel) prodrug (Cel-CSO) by conjugating chitosan oligosaccharides (CSO) to Cel for reversing Taxol resistance in chemotherapy, followed by self-assembly with Taxol into a novel nanoplatform of Cel-CSO/Taxol nanoparticles (termed NPs). NPs showed a suitable size (about 153 nm), excellent stability and prolonged release of Cel and Taxol in a manner that depended on both pH and time. NPs effectively inhibited the overexpression of multidrug resistance-related protein P-gp, hypoxia inducible factor-1α (HIF-1α), and triggered the MCF-7/Taxol cell apoptosis through inhibiting the PI3K/AKT/NF-κB/HIF-1α pathway. In tumor-bearing mice, NPs exhibited significant curative effects in inducing apoptosis of MCF-7/Taxol tumors which showed a low expression level of P-gp, microtubule-related proteins TUBB3 and Tau. The results indicated that NPs may be a promising strategy to overcome drug resistance caused by P-gp, which improve the antitumor effects in drug-resistant breast cancer.
Collapse
Affiliation(s)
- Huahui Zeng
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, China
| | - Xiaohu Zeng
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Can Wang
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, China
| | - Guoqiang Wang
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Qikang Tian
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Junwei Zhao
- Department of Clinical Laboratory, Core Unit of National Clinical Research Center for Laboratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450046, China
| | - Lingzhou Zhao
- Department of Nuclear Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Ruiqin Li
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Ying Luo
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao, Henan Province, Zhengzhou, 450046, China
| | - Haotian Peng
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Zhenqiang Zhang
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Xiaofang Li
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Xiangxiang Wu
- Academy of Chinese Medicine Science, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| |
Collapse
|
3
|
Moreira H, Szyjka A, Bęben D, Siwiela O, Radajewska A, Stankiewicz N, Grzesiak M, Wiatrak B, Emhemmed F, Muller CD, Barg E. Genotoxic and Anti-Migratory Effects of Camptothecin Combined with Celastrol or Resveratrol in Metastatic and Stem-like Cells of Colon Cancer. Cancers (Basel) 2024; 16:3279. [PMID: 39409900 PMCID: PMC11476312 DOI: 10.3390/cancers16193279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Colorectal cancer is one of the leading and most lethal neoplasms. Standard chemotherapy is ineffective, especially in metastatic cancer, and does not target cancer stem cells. A promising approach to improve cancer treatment is the combination therapy of standard cytostatic drugs with natural compounds. Several plant-derived compounds have been proven to possess anticancer properties, including the induction of apoptosis and inhibition of cancer invasion. This study was focused on investigating in vitro the combination of camptothecin (CPT) with celastrol (CEL) or resveratrol (RSV) as a potential strategy to target metastatic (LOVO) and stem-like (LOVO/DX) colon cancer cells. Methods: The genotoxic effects that drive cancer cells into death-inducing pathways and the ability to inhibit the migratory properties of cancer cells were evaluated. The γH2AX+ assay and Fast-Halo Assay (FHA) were used to evaluate genotoxic effects, the annexin-V apoptosis assay to rate the level of apoptosis, and the scratch test to assess antimigratory capacity. Results: The results showed that both combinations CPT-CEL and CPT-RSV improve general genotoxicity of CPT alone on metastatic cells and CSCs. However, the assessment of specific double-stranded breaks (DSBs) indicated a better efficacy of the CPT-CEL combination on LOVO cells and CPT-RSV in LOVO/DX cells. Interestingly, the combinations CPT-CEL and CPT-RSV did not improve the pro-apoptotic effect of CPT alone, with both LOVO and LOVO/DX cells suggesting activation of different cell death mechanisms. Furthermore, it was found that the combinations of CPT-CEL and CPT-RSV improve the inhibitory effect of camptothecin on cell migration. Conclusions: These findings suggest the potential utility of combining camptothecin with celastrol or resveratrol in the treatment of colon cancer, including more aggressive forms of the disease. So far, no studies evaluating the effects of combinations of these compounds have been published in the available medical databases.
Collapse
Affiliation(s)
- Helena Moreira
- Department of Basic Medical Sciences, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
- The Hubert Curien pluridisciplinary Institute-IPHC, UMR 7178, University of Strasbourg, 67401 Illkirch, France
| | - Anna Szyjka
- Department of Basic Medical Sciences, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Dorota Bęben
- Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Oliwia Siwiela
- Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Anna Radajewska
- Division of Clinical Chemistry and Laboratory Hematology, Department of Medical Laboratory Diagnostics, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Nadia Stankiewicz
- Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | | | - Benita Wiatrak
- Department of Basic Medical Sciences, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| | - Fathi Emhemmed
- The Hubert Curien pluridisciplinary Institute-IPHC, UMR 7178, University of Strasbourg, 67401 Illkirch, France
| | - Christian D. Muller
- The Hubert Curien pluridisciplinary Institute-IPHC, UMR 7178, University of Strasbourg, 67401 Illkirch, France
| | - Ewa Barg
- Department of Basic Medical Sciences, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland
| |
Collapse
|
4
|
Wang C, Dai S, Zhao X, Zhang Y, Gong L, Fu K, Ma C, Peng C, Li Y. Celastrol as an emerging anticancer agent: Current status, challenges and therapeutic strategies. Biomed Pharmacother 2023; 163:114882. [PMID: 37196541 DOI: 10.1016/j.biopha.2023.114882] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/10/2023] [Accepted: 05/12/2023] [Indexed: 05/19/2023] Open
Abstract
Celastrol is a pentacyclic triterpenoid extracted from the traditional Chinese medicine Tripterygium wilfordii Hook F., which has multiple pharmacological activities. In particular, modern pharmacological studies have demonstrated that celastrol exhibits significant broad-spectrum anticancer activities in the treatment of a variety of cancers, including lung cancer, liver cancer, colorectal cancer, hematological malignancies, gastric cancer, prostate cancer, renal carcinoma, breast cancer, bone tumor, brain tumor, cervical cancer, and ovarian cancer. Therefore, by searching the databases of PubMed, Web of Science, ScienceDirect and CNKI, this review comprehensively summarizes the molecular mechanisms of the anticancer effects of celastrol. According to the data, the anticancer effects of celastrol can be mediated by inhibiting tumor cell proliferation, migration and invasion, inducing cell apoptosis, suppressing autophagy, hindering angiogenesis and inhibiting tumor metastasis. More importantly, PI3K/Akt/mTOR, Bcl-2/Bax-caspase 9/3, EGFR, ROS/JNK, NF-κB, STAT3, JNK/Nrf2/HO-1, VEGF, AR/miR-101, HSF1-LKB1-AMPKα-YAP, Wnt/β-catenin and CIP2A/c-MYC signaling pathways are considered as important molecular targets for the anticancer effects of celastrol. Subsequently, studies of its toxicity and pharmacokinetic properties showed that celastrol has some adverse effects, low oral bioavailability and a narrow therapeutic window. In addition, the current challenges of celastrol and the corresponding therapeutic strategies are also discussed, thus providing a theoretical basis for the development and application of celastrol in the clinic.
Collapse
Affiliation(s)
- Cheng Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shu Dai
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xingtao Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yafang Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Lihong Gong
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Ke Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Yunxia Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Key Laboratory of Standardization for Chinese Herbal Medicine, Ministry of Education, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| |
Collapse
|
5
|
Song J, He GN, Dai L. A comprehensive review on celastrol, triptolide and triptonide: Insights on their pharmacological activity, toxicity, combination therapy, new dosage form and novel drug delivery routes. Biomed Pharmacother 2023; 162:114705. [PMID: 37062220 DOI: 10.1016/j.biopha.2023.114705] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/01/2023] [Accepted: 04/12/2023] [Indexed: 04/18/2023] Open
Abstract
Celastrol, triptolide and triptonide are the most significant active ingredients of Tripterygium wilfordii Hook F (TWHF). In 2007, the 'Cell' journal ranked celastrol, triptolide, artemisinin, capsaicin and curcumin as the five natural drugs that can be developed into modern medicinal compounds. In this review, we collected relevant data from the Web of Science, PubMed and China Knowledge Resource Integrated databases. Some information was also acquired from government reports and conference papers. Celastrol, triptolide and triptonide have potent pharmacological activity and evident anti-cancer, anti-tumor, anti-obesity and anti-diabetes effects. Because these compounds have demonstrated unique therapeutic potential for acute and chronic inflammation, brain injury, vascular diseases, immune diseases, renal system diseases, bone diseases and cardiac diseases, they can be used as effective drugs in clinical practice in the future. However, celastrol, triptolide and triptonide have certain toxic effects on the liver, kidney, cholangiocyte heart, ear and reproductive system. These shortcomings limit their clinical application. Suitable combination therapy, new dosage forms and new routes of administration can effectively reduce toxicity and increase the effect. In recent years, the development of different targeted drug delivery formulations and administration routes of celastrol and triptolide to overcome their toxic effects and maximise their efficacy has become a major focus of research. However, in-depth investigation is required to elucidate the mechanisms of action of celastrol, triptolide and triptonide, and more clinical trials are required to assess the safety and clinical value of these compounds.
Collapse
Affiliation(s)
- Jing Song
- School of Pharmacy, Binzhou Medical University, Yantai, China; Shandong Yuze Pharmaceutical Industry Technology Research Institute Co., Ltd, Dezhou, China
| | - Guan-Nan He
- Shandong University of Traditional Chinese Medicine, Ji'nan 250014, China
| | - Long Dai
- School of Pharmacy, Binzhou Medical University, Yantai, China.
| |
Collapse
|
6
|
Guo TH, Li YY, Hong SW, Cao QY, Chen H, Xu Y, Dai GL, Shao G. Evidence for Anticancer Effects of Chinese Medicine Monomers on Colorectal Cancer. Chin J Integr Med 2022; 28:939-952. [PMID: 35419728 DOI: 10.1007/s11655-022-3466-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/25/2021] [Indexed: 12/26/2022]
Abstract
Colorectal cancer is one of the most commonly occurring cancers worldwide. Although clinical reports have indicated the anticancer effects of Chinese herbal medicine, the multiple underlying molecular and biochemical mechanisms of action remain to be fully characterized. Chinese medicine (CM) monomers, which are the active components of CM, serve as the material basis of the functional mechanisms of CM. The aim of this review is to summarize the current experimental evidence from in vitro, in vivo, and clinical studies for the effects of CM monomers in colorectal cancer prevention and treatment, providing some useful references for future research.
Collapse
Affiliation(s)
- Tian-Hao Guo
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yuan-Yuan Li
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Sheng-Wei Hong
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qian-Yu Cao
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Heng Chen
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yuan Xu
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Guo-Liang Dai
- Department of Clinical Pharmacology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, China.
| | - Gang Shao
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| |
Collapse
|
7
|
Novel Epoxides of Soloxolone Methyl: An Effect of the Formation of Oxirane Ring and Stereoisomerism on Cytotoxic Profile, Anti-Metastatic and Anti-Inflammatory Activities In Vitro and In Vivo. Int J Mol Sci 2022; 23:ijms23116214. [PMID: 35682893 PMCID: PMC9181525 DOI: 10.3390/ijms23116214] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 02/07/2023] Open
Abstract
It is known that epoxide-bearing compounds display pronounced pharmacological activities, and the epoxidation of natural metabolites can be a promising strategy to improve their bioactivity. Here, we report the design, synthesis and evaluation of biological properties of αO-SM and βO-SM, novel epoxides of soloxolone methyl (SM), a cyanoenone-bearing derivative of 18βH-glycyrrhetinic acid. We demonstrated that the replacement of a double-bound within the cyanoenone pharmacophore group of SM with α- and β-epoxide moieties did not abrogate the high antitumor and anti-inflammatory potentials of the triterpenoid. It was found that novel SM epoxides induced the death of tumor cells at low micromolar concentrations (IC50(24h) = 0.7–4.1 µM) via the induction of mitochondrial-mediated apoptosis, reinforced intracellular accumulation of doxorubicin in B16 melanoma cells, probably by direct interaction with key drug efflux pumps (P-glycoprotein, MRP1, MXR1), and the suppressed pro-metastatic phenotype of B16 cells, effectively inhibiting their metastasis in a murine model. Moreover, αO-SM and βO-SM hampered macrophage functionality in vitro (motility, NO production) and significantly suppressed carrageenan-induced peritonitis in vivo. Furthermore, the effect of the stereoisomerism of SM epoxides on the mentioned bioactivities and toxic profiles of these compounds in vivo were evaluated. Considering the comparable antitumor and anti-inflammatory effects of SM epoxides with SM and reference drugs (dacarbazine, dexamethasone), αO-SM and βO-SM can be considered novel promising antitumor and anti-inflammatory drug candidates.
Collapse
|
8
|
Wastag M, Bieżuńska-Kusiak K, Szewczyk A, Szlasa W, Grimling B, Kulbacka J. Celastrol and Rhynchophylline in the mitigation of simulated muscle atrophy under in vitro. Saudi Pharm J 2022; 30:1387-1395. [PMID: 36387339 PMCID: PMC9649342 DOI: 10.1016/j.jsps.2022.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 06/17/2022] [Indexed: 11/24/2022] Open
Abstract
Muscular atrophy (MA) is a disease of various origins, i.e., genetic or the most common, caused by mechanical injury. So far, there is no universal therapeutic model because this disease is often progressive with numerous manifested symptoms. Moreover, there is no safe and low-risk therapy dedicated to muscle atrophy. For this reason, our research focuses on finding an alternative method using natural compounds to treat MA. This study proposes implementing natural substances such as celastrol and Rhynchophylline on the cellular level, using a simulated and controlled atrophy process. Methods: Celastrol and Rhynchophylline were used as natural compounds against simulated atrophy in C2C12 cells. Skeletal muscle C2C12 cells were stimulated for the differentiation process. Atrophic conditions were obtained by the exposure to the low concertation of doxorubicin and validated by FoxO3 and MAFbx. The protective and regenerative effect of drugs on cell proliferation was determined by the MTT assay and MT-CO1, VDAC1, and prohibitin expression. Results: The obtained results revealed that both natural substances reduced atrophic symptoms. Rhynchophylline and celastrol attenuated atrophic cells in the viability studies, morphology analysis by diameter measurements, modulated prohibitin VDAC, and MT-CO1 expression. Conclusions: The obtained results revealed that celastrol and Rhynchophylline could be effectively used as a supportive treatment in atrophy-related disorders. Thus, natural drugs seem promising for muscle regeneration.
Collapse
Affiliation(s)
- Maksymilian Wastag
- Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556, Wroclaw, Poland
| | - Katarzyna Bieżuńska-Kusiak
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A 50-556, Wroclaw, Poland
| | - Anna Szewczyk
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A 50-556, Wroclaw, Poland
- Department of Animal Developmental Biology, Institute of Experimental Biology, University of Wroclaw, Sienkiewicza 21, 50-335 Wroclaw, Poland
| | - Wojciech Szlasa
- Faculty of Medicine, Wroclaw Medical University, Pasteura 1, 50-367 Wroclaw, Poland
| | - Bożena Grimling
- Department of Drug Form Technology, Wroclaw Medical University, Borowska 211 A, 50-556 Wroclaw, Poland
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A 50-556, Wroclaw, Poland
- Corresponding author at: Borowska 211A, 50-556 Wroclaw, Poland.
| |
Collapse
|
9
|
Cheng G, Li L, Li Q, Lian S, Chu H, Ding Y, Li C, Leng Y. β-elemene suppresses tumor metabolism and stem cell-like properties of non-small cell lung cancer cells by regulating PI3K/AKT/mTOR signaling. Am J Cancer Res 2022; 12:1535-1555. [PMID: 35530288 PMCID: PMC9077083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 03/16/2022] [Indexed: 06/14/2023] Open
Abstract
Multi-drug resistance remains a critical issue in cancer treatment that hinders the effective use of chemotherapeutic drugs. The active components of traditional Chinese medicine have been applied as adjuvants to accentuate the anticancer properties of conventional drugs such as cisplatin. However, their application requires further validation and optimization. This study explored the anticancer activity of β-elemene, a natural component of traditional Chinese medical formulations. The effect of β-elemene on the anticancer properties of cisplatin was evaluated in A549 and NCI-H1650 lung cancer cells. Cell apoptosis, stem-like properties, glucose metabolism, multi-drug resistance, and PI3K/AKT/mTOR activation were assessed via flow cytometry, tumorsphere formation, and western blotting. The target genes of β-elemene were predicted using bioinformatics tools and validated in both cell lines. A xenograft model of lung cancer was established in nude mice to evaluate the combined effects of β-elemene and cisplatin in vivo. We found that β-elemene acted synergistically with cisplatin against non-small cell lung cancer cells by promoting apoptosis and impairing glucose metabolism, multi-drug resistance, and stemness maintenance. These effects were mediated by the inhibition of PI3K/AKT/mTOR activation. Bioinformatics analysis revealed that RB1 and TP53 are common target genes associated with lung cancer and β-elemene. The anti-tumorigenic properties of β-elemene were confirmed in vivo, wherein β-elemene, along with cisplatin, significantly suppressed tumor growth in a mouse xenograft model of non-small cell lung cancer. As such, β-elemene acted as an inhibitor of PI3K/AKT/mTOR signaling and enhanced the anticancer effect of cisplatin by targeting tumor metabolism, chemoresistance, and stem-like behavior. Thus, β-elemene is an effective anticancer adjuvant agent with potential clinical applications.
Collapse
Affiliation(s)
- Guangyu Cheng
- Research Center, Affiliated Hospital to Changchun University of Chinese MedicineJilin 130021, China
| | - Lin Li
- Department of Health Care, Affiliated Hospital to Changchun University of Chinese MedicineJilin 130021, China
| | - Qingjie Li
- Research Center, Affiliated Hospital to Changchun University of Chinese MedicineJilin 130021, China
| | - Shulin Lian
- Department of Surgery, Affiliated Hospital to Changchun University of Chinese MedicineJilin 130021, China
| | - Hongbo Chu
- Research Center, Affiliated Hospital to Changchun University of Chinese MedicineJilin 130021, China
| | - Yunlu Ding
- Research Center, Affiliated Hospital to Changchun University of Chinese MedicineJilin 130021, China
| | - Chikun Li
- Research Center, Affiliated Hospital to Changchun University of Chinese MedicineJilin 130021, China
| | - Yan Leng
- Department of Encephalopathy, Affiliated Hospital to Changchun University of Chinese MedicineJilin 130021, China
| |
Collapse
|
10
|
Celastrol and Resveratrol Modulate SIRT Genes Expression and Exert Anticancer Activity in Colon Cancer Cells and Cancer Stem-like Cells. Cancers (Basel) 2022; 14:cancers14061372. [PMID: 35326523 PMCID: PMC8945991 DOI: 10.3390/cancers14061372] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 02/24/2022] [Accepted: 03/06/2022] [Indexed: 02/07/2023] Open
Abstract
Simple Summary The recovery rate in patients with metastatic colorectal cancer (CRC) remains low and declines with successive lines of treatment. This phenomenon is caused by the development of drug resistance and the presence of colorectal cancer stem cells (CSCs). Phytochemicals, like -celastrol and resveratrol, are very promising for colon cancer therapy, owing to their low or no toxicity and their pleiotropic activity, enabling them to interact with various biological targets. In the present study, the potential anticancer mechanisms of both compounds against metastatic colon cancer cells and the capacity to eradicate CSCs were investigated. Abstract Metastatic colorectal cancer (CRC) remains a hard-to-cure neoplasm worldwide. Its curability declines with successive lines of treatment due to the development of various cancer resistance mechanisms and the presence of colorectal cancer stem cells (CSCs). Celastrol and resveratrol are very promising phytochemicals for colon cancer therapy, owing to their pleiotropic activity that enables them to interact with various biological targets. In the present study, the anticancer activities of both compounds were investigated in metastatic colon cancer cells (LoVo cells) and cancer stem-like cells (LoVo/DX). We showed that celastrol is a very potent anti-tumor compound against metastatic colon cancer, capable of attenuating CSC-like cells at the molecular and cellular levels. In contrast, resveratrol has a much greater effect on colon cancer cells that are expressing standard sensitivity to anticancer drugs, than on CSC-like cells. In addition, both polyphenols have different influences on the expression of SIRT genes, which seems to be at least partly related to their anti-tumor activity.
Collapse
|
11
|
Chabowska G, Moreira H, Tylińska B, Barg E. S16020 pyridocarbazole derivatives display high activity to lung cancer cells. Anticancer Agents Med Chem 2021; 22:2419-2428. [PMID: 34906061 DOI: 10.2174/1871520621666211214104926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 08/11/2021] [Accepted: 11/01/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Despite the dynamic development of medicine, globally cancer diseases remain the second leading cause of death. Therefore, there is a strong necessity to improve chemotherapy regimens and search for new anticancer agents. Pyridocarbazoles are compounds with confirmed antitumor properties based on multimodal mechanisms, i.a. DNA intercalation and topoisomerase II-DNA complex inhibition. One of them, S16020, displayed a wide spectrum of activity. OBJECTIVE The aim of the study was to investigate the antitumor potency of six S16020 derivatives, synthesized according to the SAR (structure-activity relationship) method. METHODS The biological evaluation included influence on cancer cell viability, proliferation, and migration, as well as P-glycoprotein activity. NHDF, A549, MCF-7, LoVo, and LoVo/DX cell lines were used in the study. RESULTS All derivatives displayed low toxicity to normal (NHDF) cells at 1 and 2 µM (≤ 20% of cell growth inhibition). The highest reduction in cell viability was noted in A549 cells which was accompanied by significant disruption of cells proliferation and motility. Compound 1 exhibited the strongest cytotoxic, antiproliferative, and antimigratory effects, higher than the reference olivacine. A significant reduction in P-glycoprotein activity was found for derivatives 6 and 1. CONCLUSION S16020 derivatives could be considered as potential candidates for new anticancer drugs.
Collapse
Affiliation(s)
- Gabriela Chabowska
- Department of Basic Medical Sciences, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw. Poland
| | - Helena Moreira
- Department of Basic Medical Sciences, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw. Poland
| | - Beata Tylińska
- Department of Organic Chemistry, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw. Poland
| | - Ewa Barg
- Department of Basic Medical Sciences, Faculty of Pharmacy, Wroclaw Medical University, Wroclaw. Poland
| |
Collapse
|
12
|
Zhang H, Zhao X, Shang F, Sun H, Zheng X, Zhu J. Celastrol inhibits the proliferation and induces apoptosis of colorectal cancer cells via downregulating NF-κB/COX-2 signaling pathways. Anticancer Agents Med Chem 2021; 22:1921-1932. [PMID: 34732120 DOI: 10.2174/1871520621666211103103530] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 07/18/2021] [Accepted: 08/26/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Colorectal cancer (CRC) is the third-ranked malignant tumor in the world that contributes to the death of a major population of the world. Celastrol, a bioactive natural product isolated from the medicinal plant Tripterygium wilfordii Hook F, has been proved to be an effective anti-tumor inhibitor for multiple tumors. OBJECTIVE To reveal the therapeutic effect and underlying mechanisms of celastrol on CRC cells. METHODS CCK-8 and clonogenic assay were used to analyze the cell proliferation in CRC cells. Flow cytometry analysis was conducted to assess the cell cycle and cell apoptosis. Wound-healing and cell invasion assay were used to evaluate the migrating and invasion capability of CRC cells. The potential antitumor mechanism of celastrol was investigated by qPCR, western blot, and confocal immunofluorescence analyses. RESULTS Celastrol effectively inhibited CRC cell proliferation by activating caspase-dependent cell apoptosis and facilitating G1 cell cycle arrest in a dose-dependent manner, as well as cell migration and invasion by downregulating the MMP2 and MMP9. Mechanistic protein expression revealed that celastrol suppressed the expression of COX-2 by inhibiting the phosphorylation of NF-κB p65 and subsequently leading to cytoplasmic retention of p65 protein, thereby inhibiting its nuclear translocation and transcription activities. CONCLUSION These findings indicate that celastrol is an effective inhibitor for CRC, regulating the NF-κB/COX-2 pathway, leading to the inhibition of cell proliferation characterized by cell cycle arrest and caspase-dependent apoptosis, providing a potential alternative therapeutic agent for CRC patients.
Collapse
Affiliation(s)
- Hua Zhang
- Department of anus & intestine surgery, The Affiliated Renhe Hospital, China Three Gorges University, Yichang 443000. China
| | - Xiaojin Zhao
- Department of Gastroenterology, The Affiliated Renhe Hospital, China Three Gorges University, Yichang 443000. China
| | - Fajun Shang
- Department of Neurosurgery, The Affiliated Renhe Hospital, China Three Gorges University, Yichang 443000. China
| | - Huan Sun
- Department of Neurosurgery, The Affiliated Renhe Hospital, China Three Gorges University, Yichang 443000. China
| | - Xu Zheng
- Department of Neurosurgery, The Affiliated Renhe Hospital, China Three Gorges University, Yichang 443000. China
| | - Jiabin Zhu
- Department of Neurosurgery, The Affiliated Renhe Hospital, China Three Gorges University, Yichang 443000. China
| |
Collapse
|
13
|
Radajewska A, Przybyszewski O, Emhemmed F, Muller CD, Barg E, Moreira H. Three dimensional in vitro culture systems in anticancer drug discovery targeted on cancer stem cells. Am J Cancer Res 2021; 11:4931-4946. [PMID: 34765301 PMCID: PMC8569359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 07/28/2021] [Indexed: 06/13/2023] Open
Abstract
Worldwide, tumors are one of the most common causes of death. Every year 3.7 million new cases occur in Europe and more than 1.9 million patients die (WHO data). Most of the fields of research are focused on developing new therapeutic strategies that will be effective in eliminating the tumor, preventing its remission, and avoiding or reducing the side effects of therapy. In the past, generally classical 2D cell cultures or immunodeficient animal models had been used to cultivate and test drugs on human cancer cell lines. Nowadays, there are increasing interests in three-dimensional (3D) cell cultures, a method with significant differences from flat cultured cells, both considering gene expressions and cell-cell interactions. Various evidence suggests that high tumorigenic properties might be dependent on the occurrence of a small cell population, pointed out to be responsible for metastasis and recurrence. This population is called cancer stem cells (CSCs), hinted to have a lot of similarities with normal stem cells. CSCs are the main reason for chemotherapy failure as well as multi-drug resistance (MDR). CSCs can also interact through the cytokine network, with other cells like the macrophages of the inflammatory system. The big advantage of a 3D culture is the possibility to isolate and investigate the CSCs population surrounded by its environment. This article aims to sum up known 3D cell cultures, especially in the field of CSCs research due to the importance of the tumor's environment on stem cell's markers expression and their development.
Collapse
Affiliation(s)
- Anna Radajewska
- Department of Basic Medical Sciences, Wroclaw Medical UniversityWroclaw, Poland
- Department of Medical Laboratory Diagnostics, Division of Clinical Chemistry and Laboratory Hematology, Wroclaw Medical UniversityWroclaw, Poland
| | - Oskar Przybyszewski
- Department of Basic Medical Sciences, Wroclaw Medical UniversityWroclaw, Poland
| | - Fathi Emhemmed
- IPHC, UMR 7178, University of StrasbourgIllkirch, France
| | | | - Ewa Barg
- Department of Basic Medical Sciences, Wroclaw Medical UniversityWroclaw, Poland
| | - Helena Moreira
- Department of Basic Medical Sciences, Wroclaw Medical UniversityWroclaw, Poland
- IPHC, UMR 7178, University of StrasbourgIllkirch, France
| |
Collapse
|
14
|
Lim HY, Ong PS, Wang L, Goel A, Ding L, Li-Ann Wong A, Ho PCL, Sethi G, Xiang X, Goh BC. Celastrol in cancer therapy: Recent developments, challenges and prospects. Cancer Lett 2021; 521:252-267. [PMID: 34508794 DOI: 10.1016/j.canlet.2021.08.030] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/11/2021] [Accepted: 08/25/2021] [Indexed: 01/05/2023]
Abstract
Cancer is one of the world's biggest healthcare burdens and despite the current advancements made in treatment plans, the outcomes for oncology patients have yet to reach their full potential. Hence, there is a pressing need to develop novel anti-cancer drugs. A popular drug class for research are natural compounds, due to their multi-targeting potential and enhanced safety profile. One such promising natural bioactive compound derived from a vine, Tripterygium wilfordii is celastrol. Pre-clinical studies revolving around the use of celastrol have revealed positive pharmacological activities in various types of cancers, thus suggesting the chemical's potential anti-cancerous effects. However, despite the numerous preclinical studies carried out over the past few decades, celastrol has not reached human trials for cancer. In this review, we summarize the mechanisms and therapeutic potentials of celastrol in treatment for different types of cancer. Subsequently, we also explore the possible reasons hindering its development for human use as cancer therapy, like its narrow therapeutic window and poor pharmacokinetic properties. Additionally, after critically analysing both in vitro and in vivo evidence, we discuss about the key pathways effected by celastrol and the suitable types of cancer that can be targeted by the natural drug, thus giving insight into future directions that can be taken, such as in-depth analysis and research of the druggability of celastrol derivatives, to aid the clinical translation of this promising anti-cancer lead compound.
Collapse
Affiliation(s)
- Hannah Ying Lim
- Department of Pharmacy, National University of Singapore, 117559, Singapore; Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore
| | - Pei Shi Ong
- Department of Pharmacy, National University of Singapore, 117559, Singapore
| | - Lingzhi Wang
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore
| | - Arul Goel
- La Canada High School, La Canada Flintridge, CA, 91011, USA
| | - Lingwen Ding
- Department of Pathology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Andrea Li-Ann Wong
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore; Department of Haematology-Oncology, National University Cancer Institute, 119228, Singapore
| | - Paul Chi-Lui Ho
- Department of Pharmacy, National University of Singapore, 117559, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore.
| | - Xiaoqiang Xiang
- Department of Clinical Pharmacy and Pharmacy Administration, School of Pharmacy, Fudan University, Shanghai, 201203, PR China.
| | - Boon Cher Goh
- Cancer Science Institute of Singapore, National University of Singapore, 117599, Singapore; Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117600, Singapore; Department of Haematology-Oncology, National University Cancer Institute, 119228, Singapore.
| |
Collapse
|
15
|
The Impact of Extracellular Ca 2+ and Nanosecond Electric Pulses on Sensitive and Drug-Resistant Human Breast and Colon Cancer Cells. Cancers (Basel) 2021; 13:cancers13133216. [PMID: 34203184 PMCID: PMC8268418 DOI: 10.3390/cancers13133216] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/15/2021] [Accepted: 06/23/2021] [Indexed: 01/16/2023] Open
Abstract
Simple Summary The drug resistance phenomenon in cancer constantly induces problems in therapeutic protocols. Pulsed electric fields (PEFs) seem to be a promising method in drug molecule delivery. Here we have proved that electroporation supported by calcium ions can alternate the activity of drug resistance proteins. Our results indicated that MDR1 expression is not significantly modified by nanosecond electroporation in multidrug-resistant cells. However, PEF significantly inhibited MDR1 activity and cell viability when combined with calcium ions. Abstract (1) Background: Calcium electroporation (CaEP) is based on the application of electrical pulses to permeabilize cells (electroporation) and allow cytotoxic doses of calcium to enter the cell. (2) Methods: In this work, we have used doxorubicin-resistant (DX) and non-resistant models of human breast cancer (MCF-7/DX, MCF-7/WT) and colon cancer cells (LoVo, LoVo/DX), and investigated the susceptibility of the cells to extracellular Ca2+ and electric fields in the 20 ns–900 ns pulse duration range. (3) Results: We have observed that colon cancer cells were less susceptible to PEF than breast cancer cells. An extracellular Ca2+ (2 mM) with PEF was more disruptive for DX-resistant cells. The expression of glycoprotein P (MDR1, P-gp) as a drug resistance marker was detected by the immunofluorescent (CLSM) method and rhodamine-123 efflux as an MDR1 activity. MDR1 expression was not significantly modified by nanosecond electroporation in multidrug-resistant cells, but a combination with calcium ions significantly inhibited MDR1 activity and cell viability. (4) Conclusions: We believe that PEF with calcium ions can reduce drug resistance by inhibiting drug efflux activity. This phenomenon of MDR mechanism disruption seems promising in anticancer protocols.
Collapse
|
16
|
Celastrol Prevents Oxidative Stress Effects on FSHR, PAPP, and CYP19A1 Gene Expression in Cultured Human Granulosa-Lutein Cells. Int J Mol Sci 2021; 22:ijms22073596. [PMID: 33808393 PMCID: PMC8037896 DOI: 10.3390/ijms22073596] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/26/2021] [Accepted: 03/27/2021] [Indexed: 12/31/2022] Open
Abstract
Regulation of oxidative stress (OS) is important to prevent damage to female reproductive physiology. While normal OS levels may have a regulatory role, high OS levels may negatively affect vital processes such as folliculogenesis or embryogenesis. The aim of this work was to study OS induced by glucose, a reactive oxygen species generator, or peroxynitrite, a reactive nitrogen species generator, in cultured human granulosa-lutein (hGL) cells from oocyte donors, analyzing expression of genes involved in oocyte maturation (FSHR, PAPP, and CYP19A1) and OS damage response (ALDH3A2). We also evaluated the effect of celastrol as an antioxidant. Our results showed that although both glucose and peroxynitrite produce OS increments in hGL cells, only peroxynitrite treatment increases ALDH3A2 and PAPP gene expression levels and decreases FSHR gene expression levels. Celastrol pre-treatment prevents this effect of peroxynitrite. Interestingly, when celastrol alone was added, we observed a reduction of the expression of all genes studied, which was independent of both OS inductors. In conclusion, regulation of OS imbalance by antioxidant substances such as celastrol may prevent negative effects of OS in female fertility. In addition to the antioxidant activity, celastrol may well have an independent role on regulation of gene expression in hGL cells.
Collapse
|
17
|
Chen YJ, You GR, Lai MY, Lu LS, Chen CY, Ting LL, Lee HL, Kanno Y, Chiou JF, Cheng AJ. A Combined Systemic Strategy for Overcoming Cisplatin Resistance in Head and Neck Cancer: From Target Identification to Drug Discovery. Cancers (Basel) 2020; 12:cancers12113482. [PMID: 33238517 PMCID: PMC7700594 DOI: 10.3390/cancers12113482] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/02/2020] [Accepted: 11/21/2020] [Indexed: 12/24/2022] Open
Abstract
Simple Summary The efficiency of cisplatin is limited by drug resistance in head–neck cancer (HNC) patients. In this study, we established a cisplatin resistance (CR) cell model, generated CR related transcriptome profiling, and combined application of bioinformatics methodology to discover a possible way to overcome CR. Analysis of the functional pathway revealed that mitotic division is a novel mechanism significantly contributing to CR. Spindle pole body component 25 (SPC25), a kinetochore protein, was overexpressed in CR cells and significantly correlated with worse HNC patient survival. The silencing of SPC25 increased cisplatin sensitivity and reduced cancer stemness property. Integration of CR transcriptome profiling and drug database discovered a natural extract compound, celastrol, possessing a potent cytotoxic effect in CR cells to reverse CR. Thus, we combined systemic strategies to demonstrated that a novel biological process (mitotic cell division), a hub gene (SPC25), and a natural compound (celastrol) as novel strategies for the treatment of refractory HNC. Abstract Cisplatin is the first-line chemotherapy agent for head and neck cancer (HNC), but its therapeutic effects are hampered by its resistance. In this study, we employed systemic strategies to overcome cisplatin resistance (CR) in HNC. CR cells derived from isogenic HNC cell lines were generated. The CR related hub genes, functional mechanisms, and the sensitizing candidates were globally investigated by transcriptomic and bioinformatic analyses. Clinically, the prognostic significance was assessed by the Kaplan–Meier method. Cellular and molecular techniques, including cell viability assay, tumorsphere formation assay, RT-qPCR, and immunoblot, were used. Results showed that these CR cells possessed highly invasive and stem-like properties. A total of 647 molecules was identified, and the mitotic division exhibited a novel functional mechanism significantly related to CR. A panel of signature molecules, MSRB3, RHEB, ULBP1, and spindle pole body component 25 (SPC25), was found to correlate with poor prognosis in HNC patients. SPC25 was further shown as a prominent molecule, which markedly suppressed cancer stemness and attenuated CR after silencing. Celastrol, a nature extract compound, was demonstrated to effectively inhibit SPC25 expression and reverse CR phenotype. In conclusion, the development of SPC25 inhibitors, such as the application of celastrol, maybe a novel strategy to sensitize cisplatin for the treatment of refractory HNC.
Collapse
Affiliation(s)
- Yin-Ju Chen
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; (Y.-J.C.); (L.-S.L.)
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 11031, Taiwan; (L.-L.T.); (H.-L.L.); (J.-F.C.)
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Guo-Rung You
- Department of Medical Biotechnology, Medical College, Chang Gung University, Taoyuan 33302, Taiwan; (G.-R.Y.); (M.-Y.L.)
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Meng-Yu Lai
- Department of Medical Biotechnology, Medical College, Chang Gung University, Taoyuan 33302, Taiwan; (G.-R.Y.); (M.-Y.L.)
| | - Long-Sheng Lu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; (Y.-J.C.); (L.-S.L.)
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 11031, Taiwan; (L.-L.T.); (H.-L.L.); (J.-F.C.)
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chang-Yu Chen
- Division of Molecular Regulation of Inflammatory and Immune Disease, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan; (C.-Y.C.); (Y.K.)
- Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| | - Lai-Lei Ting
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 11031, Taiwan; (L.-L.T.); (H.-L.L.); (J.-F.C.)
| | - Hsin-Lun Lee
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 11031, Taiwan; (L.-L.T.); (H.-L.L.); (J.-F.C.)
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Taipei Cancer Center, Taipei Medical University, Taipei 11031, Taiwan
| | - Yuzuka Kanno
- Division of Molecular Regulation of Inflammatory and Immune Disease, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan; (C.-Y.C.); (Y.K.)
- Department of Medicinal and Life Sciences, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba 278-0022, Japan
| | - Jeng-Fong Chiou
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei 11031, Taiwan; (L.-L.T.); (H.-L.L.); (J.-F.C.)
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Department of Radiology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Taipei Cancer Center, Taipei Medical University, Taipei 11031, Taiwan
| | - Ann-Joy Cheng
- Department of Medical Biotechnology, Medical College, Chang Gung University, Taoyuan 33302, Taiwan; (G.-R.Y.); (M.-Y.L.)
- Graduate Institute of Biomedical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Radiation Oncology, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan
- Correspondence: ; Tel.: +886-3-211-8800
| |
Collapse
|
18
|
Xie X, Zhan C, Wang J, Zeng F, Wu S. An Activatable Nano-Prodrug for Treating Tyrosine-Kinase-Inhibitor-Resistant Non-Small Cell Lung Cancer and for Optoacoustic and Fluorescent Imaging. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2003451. [PMID: 32815304 DOI: 10.1002/smll.202003451] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/11/2020] [Indexed: 06/11/2023]
Abstract
Non-small cell lung cancer (NSCLC) is the most common type of lung cancer and the cause of high rate of mortality. The epidermal growth factor receptor (EGFR)-targeted tyrosine kinase inhibitors are used to treat NSCLC, yet their curative effects are usually compromised by drug resistance. This study demonstrates a nanodrug for treating tyrosine-kinase-inhibitor-resistant NSCLC through inhibiting upstream and downstream EGFR signaling pathways. The main molecule of the nanodrug is synthesized by linking a tyrosine kinase inhibitor gefitinib and a near-infrared dye (NIR) on each side of a disulfide via carbonate bonds, and the nanodrug is then obtained through nanoparticle formation of the main molecule in aqueous medium and concomitant encapsulation of a serine threonine protein kinase (Akt) inhibitor celastrol. Upon administration, the nanodrug accumulates at the tumor region of NSCLC-bearing mice and releases the drugs for tumor inhibition, and the dye for fluorescence and optoacoustic imaging. Through suppressing the phosphorylation of upstream EGFR and downstream Akt in the EGFR pathway by gefitinib and celastrol, respectively, the nanodrug exhibits high inhibition efficacy against orthotopic NSCLC in mouse models.
Collapse
Affiliation(s)
- Xin Xie
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Chenyue Zhan
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Jie Wang
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Fang Zeng
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| | - Shuizhu Wu
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Wushan Road 381, Guangzhou, 510640, China
| |
Collapse
|
19
|
Lactoferrin-dual drug nanoconjugate: Synergistic anti-tumor efficacy of docetaxel and the NF-κB inhibitor celastrol. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 118:111422. [PMID: 33255023 DOI: 10.1016/j.msec.2020.111422] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/27/2020] [Accepted: 08/19/2020] [Indexed: 12/12/2022]
Abstract
Despite the progress in cancer nanotherapeutics, some obstacles still impede the success of nanocarriers and hinder their clinical translation. Low drug loading, premature drug release, off-target toxicity and multi-drug resistance are among the most difficult challenges. Lactoferrin (LF) has demonstrated a great tumor targeting capacity via its high binding affinity to low density lipoprotein (LDL) and transferrin (Tf) receptors overexpressed by various cancer cells. Herein, docetaxel (DTX) and celastrol (CST) could be successfully conjugated to LF backbone for synergistic breast cancer therapy. Most importantly, the conjugate self-assembled forming nanoparticles of 157.8 nm with elevated loading for both drugs (6.94 and 5.98% for DTX and CST, respectively) without risk of nanocarrier instability. Moreover, the nanoconjugate demonstrated enhanced in vivo anti-tumor efficacy in breast cancer-bearing mice, as reflected by a reduction in tumor volume, prolonged survival rate and significant suppression of NF-κB p65, TNF-α, COX-2 and Ki-67 expression levels compared to the group given free combined DTX/CST therapy and to positive control. This study demonstrated the proof-of-principle for dual drug coupling to LF as a versatile nanoplatform that could augment their synergistic anticancer efficacy.
Collapse
|
20
|
Antitumor Activity of New Olivacine Derivatives. Molecules 2020; 25:molecules25112512. [PMID: 32481577 PMCID: PMC7321363 DOI: 10.3390/molecules25112512] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 05/22/2020] [Accepted: 05/25/2020] [Indexed: 12/24/2022] Open
Abstract
Olivacine is an alkaloid-containing pyridocarbazole structure. It is isolated from the bark of the evergreen timber tree, Aspidosperma olivaceum. Its well-documented anticancer activity led to the synthesis of new derivatives, which are semisynthetic and fully synthetic pyridocarbazoles. This study aimed to evaluate the potential antineoplastic activity of four newly synthesized olivacine derivatives. Multidrug resistance is a common phenomenon causing failure in the chemotherapy of many tumors. It is mainly related to increased function of P-glycoprotein, an efflux pump removing cytostatic out of the cells. The cell lines used in the study were colorectal carcinoma cell lines: LoVo (doxorubicin-sensitive) and LoVo/DX (doxorubicin-resistant). The NHDF cell line was used to assess cell viability. First, the cells were incubated with olivacine derivatives. In the next step, the following assays were performed: DCF-DA assay, MTT assay, rhodamine 123 assay, detection of apoptosis, proliferation inhibition-mitotic index. The tested compounds showed higher antineoplastic potential and lower toxicity than the reference compound ellipticine. The results indicate that the new olivacine derivatives are good candidates for future anticancer drugs.
Collapse
|
21
|
Jiang ZT, Han Y, Liu XY, Lv LY, Pan JH, Liu CD. Tripterine Restrains the Aggressiveness of Hepatocellular Carcinoma Cell via Regulating miRNA-532-5p/CXCL2 Axis. Onco Targets Ther 2020; 13:2973-2985. [PMID: 32308429 PMCID: PMC7152543 DOI: 10.2147/ott.s238074] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 03/23/2020] [Indexed: 12/24/2022] Open
Abstract
Introduction Triterpene has attracted considerable interests because it exhibits anticancer effects. However, the effects of tripterine on hepatocellular carcinoma (HCC) are not well studied. In the current study, the mechanism of tripterine on HCC cells growth and metastasis was examined. Methods The inhibitory effect on the growth and aggressiveness in HCC cells was analyzed by Cell Counting Kit-8 (CCK-8), wound healing and Transwell assay. The levels of microRNA-532-5p (miR-532-5p) in HCC cells and tissues were measured using qRT-PCR. The expression of chemokine (C-X-C Motif) ligand 2 (CXCL2) was determined by Western blotting and immunohistochemistry (IHC). Luciferase reporter gene assay was used to validate the binding between miR-532-5p and CXCL2. The impact of tripterine on the growth and metastasis of HCC cells in vivo was analyzed using transplanted tumor model and experimental lung metastasis model, respectively. Results We found that tripterine inhibited HCC cells proliferation, migration ability and invasion. Under tripterine treatment, the level of miR-532-5p was strikingly raised, and overexpression of miR‑532-5p reduced cell viability and metastatic-related traits. In addition, we identified CXCL2 as a target of miR-532-5p in HCC. Rescue experiments indicated that overexpression of CXCL2 restored the migration and invasive capacity of HCC cells inhibited by miR-532-5p or tripterine treatment. Finally, the tumor growth and metastatic ability of HCC MHCC97H cell in vivo were also significantly restrained by tripterine. The expression of CXCL2 was distinctly decreased and miR-532-5p level was increased by tripterine in vivo. Conclusion In conclusion, tripterine inhibits the growth, migration ability and invasiveness of HCC cells through intervening miR-532-5p/CXCL2.
Collapse
Affiliation(s)
- Zhi Tao Jiang
- Department of Pharmacy Office, Zhangjiagang Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, People's Republic of China
| | - Yi Han
- Department of Pharmacy Office, Zhangjiagang Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, People's Republic of China
| | - Xiao Yan Liu
- Department of Pharmacy Office, Zhangjiagang Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, People's Republic of China
| | - Ling Yan Lv
- Department of Pharmacy Office, Zhangjiagang Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, People's Republic of China
| | - Jin Huo Pan
- College of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People's Republic of China
| | - Chun Di Liu
- Department of Pharmacy Office, Zhangjiagang Hospital of Traditional Chinese Medicine Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, Jiangsu, People's Republic of China
| |
Collapse
|
22
|
Wang K, Huang W, Sang X, Wu X, Shan Q, Tang D, Xu X, Cao G. Atractylenolide I inhibits colorectal cancer cell proliferation by affecting metabolism and stemness via AKT/mTOR signaling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2020; 68:153191. [PMID: 32135457 DOI: 10.1016/j.phymed.2020.153191] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 01/16/2020] [Accepted: 02/12/2020] [Indexed: 06/10/2023]
Abstract
BACKGROUND Atractylenolide I (ATL-1) is a natural herbal compound used in traditional Chinese medicine that has exhibited anti-cancer properties. The anti-tumorigenic activity of ATL-1 against colorectal cancer (CRC) and the underlying signaling pathways involved in its mechanisms are examined here. HYPOTHESIS ATL-1 exerts therapeutic effect against CRC by disrupting glucose metabolism and cancer stem cell maintenance via AKT/mTOR pathway regulation. STUDY DESIGN In vitro studies were performed in COLO205 and HCT116 CRC cell lines and in vivo studies were conducted in a mouse xenograft model of CRC tumor. METHODS CRC cells were treated with ATL-1 at various concentrations, with or without inhibitors of AKT or mTOR. Cell proliferation, apoptosis, invasion, stemness maintenance, glucose metabolism, and AKT/mTOR signaling were evaluated. CRC tumor-xenografted mice were treated with an AKT inhibitor and/or ATL-1, and glucose metabolism and stemness maintenance were examined in tumor tissues. RESULTS ATL-1 significantly inhibited the invasion of CRC cells by inducing their apoptosis, possibly via the excessive production of reactive oxygen species. Glucose metabolism (Warburg effect) was also altered and stem-like traits were suppressed by ATL-1. In addition, ATL-1 effectively acted as an inhibitor or AKT/mTOR by downregulating the phosphorylation of proteins related to the AKT/mTOR pathway. In vivo studies showed that tumor weight and volume were reduced by ATL-1 and that aerobic glycolysis, stemness maintenance, and AKT/mTOR activation were impaired by ATL-1 in colorectal tumors. CONCLUSIONS ATL-1 acts as an effective agent to suppress colorectal tumor progression, mainly by inhibiting CRC cell proliferation through altering apoptosis, glucose metabolism, and stem-like behavior. These processes were mediated by the AKT/mTOR signaling pathway both in vitro and in vivo. ATL-1 may be a potential agent to be used in molecular-targeted strategies for cancer treatment.
Collapse
Affiliation(s)
- Kuilong Wang
- School of Pharmacy, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
| | - Wei Huang
- First Affiliated Hospital of Guiyang College of Traditional Chinese Medicine (TCM), Guiyang, China
| | - Xianan Sang
- School of Pharmacy, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
| | - Xin Wu
- School of Pharmacy, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
| | - Qiyuan Shan
- School of Pharmacy, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
| | - Dongxin Tang
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofen Xu
- School of Pharmacy, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China
| | - Gang Cao
- School of Pharmacy, Zhejiang Chinese Medical University, 548 Binwen Road, Hangzhou 310053, China.
| |
Collapse
|
23
|
Garcia-Mayea Y, Mir C, Masson F, Paciucci R, LLeonart ME. Insights into new mechanisms and models of cancer stem cell multidrug resistance. Semin Cancer Biol 2020; 60:166-180. [PMID: 31369817 DOI: 10.1016/j.semcancer.2019.07.022] [Citation(s) in RCA: 202] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 07/25/2019] [Accepted: 07/26/2019] [Indexed: 12/24/2022]
Abstract
The acquisition of genetic alterations, clonal evolution, and the tumor microenvironment promote cancer progression, metastasis and therapy resistance. These events correspond to the establishment of the great phenotypic heterogeneity and plasticity of cancer cells that contribute to tumor progression and resistant disease. Targeting resistant cancers is a major challenge in oncology; however, the underlying processes are not yet fully understood. Even though current treatments can reduce tumor size and increase life expectancy, relapse and multidrug resistance (MDR) ultimately remain the second cause of death in developed countries. Recent evidence points toward stem-like phenotypes in cancer cells, promoted by cancer stem cells (CSCs), as the main culprit of cancer relapse, resistance (radiotherapy, hormone therapy, and/or chemotherapy) and metastasis. Many mechanisms have been proposed for CSC resistance, such as drug efflux through ABC transporters, overactivation of the DNA damage response (DDR), apoptosis evasion, prosurvival pathways activation, cell cycle promotion and/or cell metabolic alterations. Nonetheless, targeted therapy toward these specific CSC mechanisms is only partially effective to prevent or abolish resistance, suggesting underlying additional causes for CSC resilience. This article aims to provide an integrated picture of the MDR mechanisms that operate in CSCs' behavior and to propose a novel model of tumor evolution during chemotherapy. Targeting the pathways mentioned here might hold promise and reveal new strategies for future clinical therapeutic approaches.
Collapse
Affiliation(s)
- Y Garcia-Mayea
- Biomedical Research in Cancer Stem Cells, Vall d´Hebron Research Institute (VHIR), Passeig Vall d´Hebron 119-129, 08035 Barcelona, Spain
| | - C Mir
- Biomedical Research in Cancer Stem Cells, Vall d´Hebron Research Institute (VHIR), Passeig Vall d´Hebron 119-129, 08035 Barcelona, Spain
| | - F Masson
- Biomedical Research in Cancer Stem Cells, Vall d´Hebron Research Institute (VHIR), Passeig Vall d´Hebron 119-129, 08035 Barcelona, Spain
| | - R Paciucci
- Clinical Biochemistry Group, Vall d'Hebron Hospital and Vall d´Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Passeig Vall d´Hebron 119-129, 08035 Barcelona, Spain
| | - M E LLeonart
- Biomedical Research in Cancer Stem Cells, Vall d´Hebron Research Institute (VHIR), Passeig Vall d´Hebron 119-129, 08035 Barcelona, Spain; Spanish Biomedical Research Network Centre in Oncology, CIBERONC, Spain.
| |
Collapse
|
24
|
Prooxidative Activity of Celastrol Induces Apoptosis, DNA Damage, and Cell Cycle Arrest in Drug-Resistant Human Colon Cancer Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6793957. [PMID: 31485297 PMCID: PMC6710751 DOI: 10.1155/2019/6793957] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/12/2019] [Accepted: 07/18/2019] [Indexed: 01/02/2023]
Abstract
Cancer resistance to chemotherapy is closely related to tumor heterogeneity, i.e., the existence of distinct subpopulations of cancer cells in a tumor mass. An important role is assigned to cancer stem cells (CSCs), a small subset of cancer cells with high tumorigenic potential and capacity of self-renewal and differentiation. These properties of CSCs are sustained by the ability of those cells to maintain a low intracellular reactive oxygen species (ROS) levels, via upregulation of ROS scavenging systems. However, the accumulation of ROS over a critical threshold disturbs CSCs—redox homeostasis causing severe cytotoxic consequences. In the present study, we investigated the capacity of celastrol, a natural pentacyclic triterpenoid, to induce the formation of ROS and, consequently, cell death of the colon cancer cells with acquired resistant to cytotoxic drugs (LOVO/DX cell line). LOVO/DX cells express several important stem-like cell features, including a higher frequency of side population (SP) cells, higher expression of multidrug resistant proteins, overexpression of CSC-specific cell surface marker (CD44), increased expression of DNA repair gene (PARP1), and low intracellular ROS level. We found that celastrol, at higher concentrations (above 1 μM), significantly increased ROS amount in LOVO/DX cells at both cytoplasmic and mitochondrial levels. This prooxidant activity was associated with the induction of DNA double-strand breaks (DSBs) and apoptotic/necrotic cell death, as well as with inhibition of cell proliferation by S phase cell cycle arrest. Coincubation with NAC, a ROS scavenger, completely reversed the above effects. In summary, our results provide evidence that celastrol exhibits effective cytotoxic effects via ROS-dependent mechanisms on drug-resistant colon cancer cells. These findings strongly suggest the potential of celastrol to effectively kill cancer stem-like cells, and thus, it is a promising agent to treat severe, resistant to conventional therapy, colon cancers.
Collapse
|
25
|
Li X, Wei W, Zhao Z, Lv S. Tripterine up-regulates miR-223 to alleviate lipopolysaccharide-induced damage in murine chondrogenic ATDC5 cells. Int J Immunopathol Pharmacol 2019; 33:2058738418824521. [PMID: 30791741 PMCID: PMC6350133 DOI: 10.1177/2058738418824521] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Tripterine, also known as celastrol, is a main natural ingredient in
Tripterygium wilfordii. Tripterine has a variety of
pharmacological functions, and the therapeutic potential of tripterine in many
kinds of inflammation-linked diseases has been revealed. However, the function
of tripterine on osteoarthritis still remains unclear. The objective of this
study was to study the function of tripterine (TPR) on lipopolysaccharide
(LPS)-injured chondrocyte. ATDC5 cells were treated with tripterine after LPS
stimulation and then cell survival, the release of pro-inflammatory cytokines,
and the expression of chondrogenic differentiation-associated proteins were
assessed by performing CCK-8, flow cytometry, reverse transcription quantitative
polymerase chain reaction (RT-qPCR), enzyme-linked immunosorbent assay (ELISA),
and Western blot. Moreover, the expression of miR-223 and core factors in
PI3K/AKT and nuclear factor kappa B (NF-κB) signaling was tested by
RT-qPCR/Western blot. LPS stimulation significantly reduced ATDC5 cells
viability, induced apoptosis, and increased the release of interleukin (IL)-6
and tumor necrosis factor (TNF)-α. Tripterine protected ATDC5 cells against
LPS-induced chondrocyte loss and the release of IL-6 and TNF-α. miR-223 was
down-regulated by LPS, while was up-regulated by tripterine. The protective
actions of tripterine were eliminated when miR-223 was silenced. Besides,
tripterine inhibited hypertrophic differentiation induced by LPS, and the
inhibitory effects of tripterine on hypertrophic differentiation could be
abolished when miR-223 was silenced. Furthermore, tripterine activated PI3K/AKT
pathway and deactivated NF-κB pathway. And the regulatory effects of tripterine
on these two pathways were abolished by miR-223 silence. This study revealed
that tripterine protected ATDC5 cells against LPS-induced cell damage possibly
via up-regulation of miR-223 and modulation of NF-κB and PI3K/AKT pathways.
Collapse
Affiliation(s)
- Xuefu Li
- Department of Orthopedics, Liaocheng Third People's Hospital, Liaocheng, China
| | - Wei Wei
- Department of Orthopedics, Liaocheng Third People's Hospital, Liaocheng, China
| | - Zhongquan Zhao
- Department of Orthopedics, Liaocheng Third People's Hospital, Liaocheng, China
| | - Shuzhen Lv
- Department of Orthopedics, Liaocheng Third People's Hospital, Liaocheng, China
| |
Collapse
|
26
|
Xiang Y, Guo Z, Zhu P, Chen J, Huang Y. Traditional Chinese medicine as a cancer treatment: Modern perspectives of ancient but advanced science. Cancer Med 2019; 8:1958-1975. [PMID: 30945475 PMCID: PMC6536969 DOI: 10.1002/cam4.2108] [Citation(s) in RCA: 466] [Impact Index Per Article: 77.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 02/26/2019] [Accepted: 03/07/2019] [Indexed: 12/24/2022] Open
Abstract
Traditional Chinese medicine (TCM) has been practiced for thousands of years and at the present time is widely accepted as an alternative treatment for cancer. In this review, we sought to summarize the molecular and cellular mechanisms underlying the chemopreventive and therapeutic activity of TCM, especially that of the Chinese herbal medicine-derived phytochemicals curcumin, resveratrol, and berberine. Numerous genes have been reported to be involved when using TCM treatments and so we have selectively highlighted the role of a number of oncogene and tumor suppressor genes in TCM therapy. In addition, the impact of TCM treatment on DNA methylation, histone modification, and the regulation of noncoding RNAs is discussed. Furthermore, we have highlighted studies of TCM therapy that modulate the tumor microenvironment and eliminate cancer stem cells. The information compiled in this review will serve as a solid foundation to formulate hypotheses for future studies on TCM-based cancer therapy.
Collapse
Affiliation(s)
- Yuening Xiang
- College of Life and Health SciencesNortheastern UniversityShenyangChina
| | - Zimu Guo
- College of Life and Health SciencesNortheastern UniversityShenyangChina
| | - Pengfei Zhu
- College of Life and Health SciencesNortheastern UniversityShenyangChina
| | - Jia Chen
- College of Life and Health SciencesNortheastern UniversityShenyangChina
| | - Yongye Huang
- College of Life and Health SciencesNortheastern UniversityShenyangChina
| |
Collapse
|
27
|
Wang S, Ma K, Zhou C, Wang Y, Hu G, Chen L, Li Z, Hu C, Xu Q, Zhu H, Liu M, Xu N. LKB1 and YAP phosphorylation play important roles in Celastrol-induced β-catenin degradation in colorectal cancer. Ther Adv Med Oncol 2019; 11:1758835919843736. [PMID: 31040884 PMCID: PMC6477772 DOI: 10.1177/1758835919843736] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 03/18/2019] [Indexed: 02/05/2023] Open
Abstract
Wnt/β-catenin and Hippo pathways play essential roles in the tumorigenesis and
development of colorectal cancer. We found that Celastrol, isolated from
Tripterygium wilfordii plant, exerted a significant
inhibitory effect on colorectal cancer cell growth in vitro and
in vivo, and further unraveled the molecular mechanisms.
Celastrol induced β-catenin degradation through phosphorylation of
Yes-associated protein (YAP), a major downstream effector of Hippo pathway, and
also Celastrol-induced β-catenin degradation was dependent on liver kinase B1
(LKB1). Celastrol increased the transcriptional activation of LKB1, partially
through the heat shock factor 1 (HSF1). Moreover, LKB1 activated AMP-activated
protein kinase α (AMPKα) and further phosphorylated YAP, which eventually
promoted the degradation of β-catenin. In addition, LKB1 deficiency promoted
colorectal cancer cell growth and attenuated the inhibitory effect of Celastrol
on colorectal cancer growth both in vitro and in
vivo. Taken together, Celastrol inhibited colorectal cancer cell
growth by promoting β-catenin degradation via the
HSF1–LKB1–AMPKα–YAP pathway. These results suggested that Celastrol may
potentially serve as a future drug for colorectal cancer treatment.
Collapse
Affiliation(s)
- Shuren Wang
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kai Ma
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Cuiqi Zhou
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yu Wang
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guanghui Hu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lechuang Chen
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhuo Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chenfei Hu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Xu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongxia Zhu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mei Liu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 PanjiayuanNanli, Chaoyang District, P.O. Box 2258, 100021, Beijing, P. R. China
| | - Ningzhi Xu
- Laboratory of Cell and Molecular Biology and State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 PanjiayuanNanli, Chaoyang District, P.O. Box 2258, 100021, Beijing, P. R. China State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, No.17, 3rd Section of People's South Road, Chengdu, 610041, P.R. China
| |
Collapse
|
28
|
Riganti C, Contino M, Guglielmo S, Perrone MG, Salaroglio IC, Milosevic V, Giampietro R, Leonetti F, Rolando B, Lazzarato L, Colabufo NA, Fruttero R. Design, Biological Evaluation, and Molecular Modeling of Tetrahydroisoquinoline Derivatives: Discovery of A Potent P-Glycoprotein Ligand Overcoming Multidrug Resistance in Cancer Stem Cells. J Med Chem 2018; 62:974-986. [DOI: 10.1021/acs.jmedchem.8b01655] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Chiara Riganti
- Dipartimento di Oncologia, Università degli Studi di Torino, Via Santena 5/bis, 10126 Torino, Italy
| | - Marialessandra Contino
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari ALDO MORO, Via Orabona 4, 70125 Bari, Italy
| | - Stefano Guglielmo
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via P. Giuria 9, 10125 Torino, Italy
| | - Maria G. Perrone
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari ALDO MORO, Via Orabona 4, 70125 Bari, Italy
| | - Iris C. Salaroglio
- Dipartimento di Oncologia, Università degli Studi di Torino, Via Santena 5/bis, 10126 Torino, Italy
| | - Vladan Milosevic
- Dipartimento di Oncologia, Università degli Studi di Torino, Via Santena 5/bis, 10126 Torino, Italy
| | - Roberta Giampietro
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari ALDO MORO, Via Orabona 4, 70125 Bari, Italy
| | - Francesco Leonetti
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari ALDO MORO, Via Orabona 4, 70125 Bari, Italy
| | - Barbara Rolando
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via P. Giuria 9, 10125 Torino, Italy
| | - Loretta Lazzarato
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via P. Giuria 9, 10125 Torino, Italy
| | - Nicola A. Colabufo
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari ALDO MORO, Via Orabona 4, 70125 Bari, Italy
- Biofordrug s.r.l., Spin-off dell’Università degli Studi di Bari ALDO MORO, Via Orabona 4, 70125 Bari, Italy
| | - Roberta Fruttero
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, Via P. Giuria 9, 10125 Torino, Italy
| |
Collapse
|