1
|
Wang Y, Dong S, Hu K, Xu L, Feng Q, Li B, Wang G, Chen G, Zhang B, Jia X, Xu Z, Gao X, Zhang H, Xie Y, Lu M, Chang S, Song D, Wu X, Jia Q, Zhu H, Zhou J, Zhu W, Shi J. The novel norcantharidin derivative DCZ5417 suppresses multiple myeloma progression by targeting the TRIP13-MAPK-YWHAE signaling pathway. J Transl Med 2023; 21:858. [PMID: 38012658 PMCID: PMC10680230 DOI: 10.1186/s12967-023-04739-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 11/17/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Multiple myeloma (MM), an incurable disease owing to drug resistance, requires safe and effective therapies. Norcantharidin (NCTD), an active ingredient in traditional Chinese medicines, possesses activity against different cancers. However, its toxicity and narrow treatment window limit its clinical application. In this study, we synthesized a series of derivatives of NCTD to address this. Among these compounds, DCZ5417 demonstrated the greatest anti-MM effect and fewest side effects. Its anti-myeloma effects and the mechanism were further tested. METHODS Molecular docking, pull-down, surface plasmon resonance-binding, cellular thermal shift, and ATPase assays were used to study the targets of DCZ5417. Bioinformatic, genetic, and pharmacological approaches were used to elucidate the mechanisms associated with DCZ5417 activity. RESULTS We confirmed a highly potent interaction between DCZ5417 and TRIP13. DCZ5417 inhibited the ATPase activity of TRIP13, and its anti-MM activity was found to depend on TRIP13. A mechanistic study verified that DCZ5417 suppressed cell proliferation by targeting TRIP13, disturbing the TRIP13/YWHAE complex and inhibiting the ERK/MAPK signaling axis. DCZ5417 also showed a combined lethal effect with traditional anti-MM drugs. Furthermore, the tumor growth-inhibitory effect of DCZ5417 was demonstrated using in vivo tumor xenograft models. CONCLUSIONS DCZ5417 suppresses MM progression in vitro, in vivo, and in primary cells from drug-resistant patients, affecting cell proliferation by targeting TRIP13, destroying the TRIP13/YWHAE complex, and inhibiting ERK/MAPK signaling. These results imply a new and effective therapeutic strategy for MM treatment.
Collapse
Affiliation(s)
- Yingcong Wang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Sanfeng Dong
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research; Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ke Hu
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Li Xu
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Qilin Feng
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Bo Li
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research; Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Guangli Wang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Gege Chen
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Bibo Zhang
- Department of Hematology, The Affiliated People's Hospital of Ningbo University, Ningbo, 315000, China
| | - Xinyan Jia
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Zhijian Xu
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research; Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Xuejie Gao
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Hui Zhang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Yongsheng Xie
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Meiling Lu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Shuaikang Chang
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Dongliang Song
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Xiaosong Wu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Qi Jia
- Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Huabin Zhu
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Jinfeng Zhou
- Department of Hematology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China
| | - Weiliang Zhu
- CAS Key Laboratory of Receptor Research, State Key Laboratory of Drug Research; Drug Discovery and Design Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
| | - Jumei Shi
- Department of Hematology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China.
| |
Collapse
|
2
|
Yang G, Xu H, Yao M, Yan S, Wu M, Zhou C. Norcantharidin ameliorates estrogen deficient-mediated bone loss by attenuating the activation of extracellular signal-regulated kinase/ROS/NLRP3 inflammasome signaling. Front Pharmacol 2022; 13:1019478. [DOI: 10.3389/fphar.2022.1019478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
Osteoporosis, characterized by reduced bone mass, aberrant bone architecture, and elevated bone fragility, is driven by a disruption of bone homeostasis between bone resorption and bone formation. However, up to now, no drugs are perfect for osteoporosis treatment due to different defects. In this study, we demonstrated that norcantharidin (NCTD) could inhibit osteoclast formation and bone resorption by attenuating the ERK, ROS and NLRP3 inflammasomes pathways in vitro. Moreover, our in vivo study further confirms its preventive effects on estrogen-deficiency bone loss by inhibiting osteoclast formation and functions. Therefore, we could conclude that NCTD might be a potential candidates for the prevention and treatment of osteoporosis.
Collapse
|
3
|
Sheng J, Liu K, Sun D, Nie P, Mu Z, Chen H, Zhang Z. Association of RDM1 with osteosarcoma progression via cell cycle and MEK/ERK signalling pathway regulation. J Cell Mol Med 2021; 25:8039-8046. [PMID: 34264012 PMCID: PMC8358872 DOI: 10.1111/jcmm.16735] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 05/12/2021] [Accepted: 05/18/2021] [Indexed: 12/11/2022] Open
Abstract
RAD52 motif‐containing 1 (RDM1), a key regulator of DNA double‐strand break repair and recombination, has been reported to play an important role in the development of various human cancers, such as papillary thyroid carcinoma, neuroblastoma and lung cancer. However, the effect of RDM1 on osteosarcoma (OS) progression remains unclear. Here, this study mainly explored the connection between RDM1 and OS progression, as well as the underlying mechanism. It was found that RDM1 was highly expressed in OS cells compared with human osteoblast cells. Knockdown of RDM1 caused OS cell proliferation inhibition, cell apoptosis promotion and cell cycle arrest at G1 stage, whereas RDM1 overexpression resulted in the opposite phenotypes. Furthermore, RDM1 silencing leads to a significant decrease in tumour growth in xenograft mouse model. RDM1 also increased the protein levels of MEK 1/2 and ERK 1/2. All these findings suggest that RDM1 plays an oncogenic role in OS via stimulating cell cycle transition from G1 to S stage, and regulating MEK/ERK signalling pathway, providing a promising therapeutic factor for the treatment of OS.
Collapse
Affiliation(s)
- Jun Sheng
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Kun Liu
- Department of Orthopedic Surgery, PLA Rocket Force Characteristic Medical Center, Beijing, China
| | - Dawei Sun
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Piming Nie
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Zhiping Mu
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Hui Chen
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Zhengfeng Zhang
- Department of Orthopedics, Xinqiao Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
4
|
Zhang S, Yang Y, Hua Y, Hu C, Zhong Y. NCTD elicits proapoptotic and antiglycolytic effects on colorectal cancer cells via modulation of Fam46c expression and inhibition of ERK1/2 signaling. Mol Med Rep 2020; 22:774-782. [PMID: 32468032 PMCID: PMC7339822 DOI: 10.3892/mmr.2020.11151] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 11/01/2019] [Indexed: 12/19/2022] Open
Abstract
Colorectal cancer is a digestive tract malignancy and the third leading cause of cancer‑related mortality worldwide. Norcantharidin (NCTD), the demethylated form of cantharidin, has been reported to possess anticancer properties. Family‑with‑sequence‑similarity‑46c (Fam46c), a non‑canonical poly(A) polymerase, has been reported to be critical in NCTD‑mediated effects in numerous types of cancer, including hepatoma. In the current study, it was found that Fam46c expression was reduced in colorectal cancer tissues and cells. Treatment with NCTD was observed to significantly enhance apoptosis and inhibit glycolysis in colorectal cancer cells. In addition, Fam46c and cleaved caspase 3 expression levels were found to be increased in response to NCTD treatment, in contrast to tumor‑specific pyruvate kinase M2 and phosphorylated ERK expression, which was reduced. Importantly, overexpression of Fam46c exerted similar effects as NCTD treatment on the apoptosis and glycolysis of colorectal cancer cells, whereas Fam46c knockdown strongly attenuated the effect of NCTD. Moreover, epidermal growth factor, which acts as an agonist of ERK1/2 signaling, weakened the effects of NCTD on colorectal cancer cells. Taken together, the results indicated that NCTD promotes apoptosis and suppresses glycolysis in colorectal cancer cells by possibly targeting Fam46c and inhibiting ERK1/2 signaling, hence suggesting that Fam46c may act as a tumor suppressor in colorectal cancer. Thus, the present study identified a novel therapeutic target of NCTD in the clinical treatment of colorectal cancer.
Collapse
Affiliation(s)
- Shiqiang Zhang
- Shanghai University of Traditional Chinese Medicine, Shanghai 200082, P.R. China
- Department of Oncology, Shanghai Traditional Chinese Medicine-Integrated Hospital, Shanghai 200082, P.R. China
| | - Yun Yang
- Shanghai University of Traditional Chinese Medicine, Shanghai 200082, P.R. China
- Department of Oncology, Shanghai Traditional Chinese Medicine-Integrated Hospital, Shanghai 200082, P.R. China
| | - Yunwei Hua
- Department of Gastroenterology, Shanghai Traditional Chinese Medicine-Integrated Hospital, Shanghai 200082, P.R. China
| | - Chen Hu
- School of Life Sciences and Technology, Tongji University, Shanghai 200082, P.R. China
| | - Yi Zhong
- Department of Oncology, Shanghai Traditional Chinese Medicine-Integrated Hospital, Shanghai 200082, P.R. China
| |
Collapse
|
5
|
Pan MS, Cao J, Fan YZ. Insight into norcantharidin, a small-molecule synthetic compound with potential multi-target anticancer activities. Chin Med 2020; 15:55. [PMID: 32514288 PMCID: PMC7260769 DOI: 10.1186/s13020-020-00338-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 05/25/2020] [Indexed: 02/07/2023] Open
Abstract
Norcantharidin (NCTD) is a demethylated derivative of cantharidin, which is an anticancer active ingredient of traditional Chinese medicine, and is currently used clinically as a routine anti-cancer drug in China. Clarifying the anticancer effect and molecular mechanism of NCTD is critical for its clinical application. Here, we summarized the physiological, chemical, pharmacokinetic characteristics and clinical applications of NCTD. Besides, we mainly focus on its potential multi-target anticancer activities and underlying mechanisms, and discuss the problems existing in clinical application and scientific research of NCTD, so as to provide a potential anticancer therapeutic agent for human malignant tumors.
Collapse
Affiliation(s)
- Mu-Su Pan
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Tongji University, 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| | - Jin Cao
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Tongji University, 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| | - Yue-Zu Fan
- Department of Surgery, Tongji Hospital, Tongji University School of Medicine, Tongji University, 389 Xincun Road, Shanghai, 200065 People’s Republic of China
| |
Collapse
|
6
|
Cheng M, Liu L. MUC15 promotes growth and invasion of glioma cells by activating Raf/MEK/ERK pathway. Clin Exp Pharmacol Physiol 2020; 47:1041-1048. [PMID: 32031702 DOI: 10.1111/1440-1681.13277] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 02/01/2020] [Accepted: 02/05/2020] [Indexed: 12/27/2022]
Abstract
MUC15 is a novel mucin associated with the cell membrane that is overexpressed in human gliomas. Its function in glioma is unclear. In this study, high MUC15 levels were detected in glioma tissues and cells. We found that transfection with MUC15 siRNA in U251 and T98G cells reduced MUC15 expression and decreased cell proliferation, invasion, and migration (P < .05). After transfecting U251 and T98G cells with pcDNA3.1-myc-His-MUC15 plasmid to overexpress MUC15, MUC15 expression was significantly upregulated and cell proliferation, invasion, and migration were increased (P < .05). MUC15 activated the Raf/MEK/ERK signalling pathway and the ERK inhibitor PD98059 partly reversed MUC15-enhanced proliferation, invasion, and migration of glioma cells (P < .05). The results indicate that MUC15 plays a part in glioma tumorigenesis, and the Raf/MEK/ERK signalling is involved in the regulation of MUC15 on glioma cell activity.
Collapse
Affiliation(s)
- Meixiong Cheng
- Department of Neurosurgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| | - Ling Liu
- Department of Neurosurgery, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, Chengdu, China
| |
Collapse
|
7
|
Chen AWG, Tseng YS, Lin CC, Hsi YT, Lo YS, Chuang YC, Lin SH, Yu CY, Hsieh MJ, Chen MK. Norcantharidin induce apoptosis in human nasopharyngeal carcinoma through caspase and mitochondrial pathway. ENVIRONMENTAL TOXICOLOGY 2018; 33:343-350. [PMID: 29193574 DOI: 10.1002/tox.22521] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 11/17/2017] [Accepted: 11/17/2017] [Indexed: 05/26/2023]
Abstract
While Nasopharyngeal carcinoma (NPC) is uncommon in western countries, it is endemic in Southeast Asia and Southern China. Previous study of norcantharidin (NCTD), isolated from blister beetles, has proved its anticancer effect on various tumors. However, the effect of NCTD in NPC has never been studied. The purpose of this study is to inspect the suppression activity of NCTD on NPC, along with the underlying mechanism. NPC cell line NPC-BM was treated with NCTD. NCTD remarkably inhibited proliferation and induce apoptosis in NPC-BM cell. Activation of caspase-3, -8, -9 was observed through western blotting. The expression of antiapoptotic protein Bcl-XL was significantly reduced, but expression of proapoptotic protein Bak was increased after treatment of NCTD. The cytotoxic effect of NCTD on NPC-BM cell is mainly due to apoptosis, mediated by caspase and mitochondrial pathway. These results suggested that NCTD could be a potential anticancer agent for NPC.
Collapse
Affiliation(s)
- Andy Wei-Ge Chen
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Taiwan
| | - Yen-Shuo Tseng
- Department of Dermatology, Changhua Christian Hospital, Taiwan
| | - Chia-Chieh Lin
- Cancer Research Center, Changhua Christian Hospital, Taiwan
| | - Yi-Ting Hsi
- Cancer Research Center, Changhua Christian Hospital, Taiwan
| | - Yu-Sheng Lo
- Cancer Research Center, Changhua Christian Hospital, Taiwan
| | | | - Shu-Hui Lin
- Department of Surgical Pathology, Changhua Christian Hospital, Taiwan
| | - Chia-Yun Yu
- Senior High School, Mingdao High School, Taichung, Taiwan
| | - Ming-Ju Hsieh
- Cancer Research Center, Changhua Christian Hospital, Taiwan
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Mu-Kuan Chen
- Department of Otorhinolaryngology, Head and Neck Surgery, Changhua Christian Hospital, Taiwan
| |
Collapse
|
8
|
Wang D, Yang C, Wang Z, Yang Y, Li D, Ding X, Xu W, Zheng Q. Norcantharidin combined with Coix seed oil synergistically induces apoptosis and inhibits hepatocellular carcinoma growth by downregulating regulatory T cells accumulation. Sci Rep 2017; 7:9373. [PMID: 28839202 PMCID: PMC5571147 DOI: 10.1038/s41598-017-09668-2] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 07/27/2017] [Indexed: 12/30/2022] Open
Abstract
The immune system plays a critical role in exerts effects in the growth and progression of hepatocellular carcinoma (HCC), which needs interacting approaches for effective therapy. In this study, we have found that the Norcantharidin (NCTD) + Coix lacryma-jobi seed oil (CLSO) combination exhibited more potent antitumor effects in an terms of cytotoxicity and apoptotic induction in human HepG2 and HepG2/ADM cells than NCTD or CLSO alone. In vivo, administration of NCTD+CLSO combinations significantly suppressed the formation of tumor in Hepal-1 hepatoma-bearing mice. Furthermore, we found that the in vitro co-cultures of HepG2 or HepG2/ADM cells with PBMCs from healthy donors led to an increase in the number of CD4 + CD25 + T cells. This increase was down-regulated by the combination effectively. Down-regulation of FoxP3 mRNA and protein expression occurred during the combination in the co-cultures. The amount of Tregs of Hepal-1 hepatoma-bearing mice was significantly decreased in the combination treated group. The combination down-regulated the expression of FoxP3, CTLA-4 and Tregs related cytokine (TGF-β and IL-10) in the serum of tumor bearing mice. Taken together, these results suggest that the most valuable aspect of the NCTD+CLSO combined use improves the anti-tumor activity and regulates tumor infiltrating Tregs.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Biomarkers
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/metabolism
- Carcinoma, Hepatocellular/pathology
- Cell Cycle Checkpoints/drug effects
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Cell Survival/drug effects
- Coix/chemistry
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Drug Synergism
- Humans
- Immunophenotyping
- Liver Neoplasms/drug therapy
- Liver Neoplasms/immunology
- Liver Neoplasms/metabolism
- Liver Neoplasms/pathology
- Mice
- Plant Oils/pharmacology
- Seeds/chemistry
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Dan Wang
- Binzhou Medical University, Yantai, 264003, China
| | - Chendong Yang
- Yantai Hospital of Traditional Chinese Medicine, Yantai, 264003, China
| | - Zhuien Wang
- Binzhou Medical University, Yantai, 264003, China
| | - Yi Yang
- Binzhou Medical University, Yantai, 264003, China
| | - Defang Li
- Binzhou Medical University, Yantai, 264003, China
| | - Xiaojie Ding
- Binzhou Medical University, Yantai, 264003, China
| | - Wenjuan Xu
- Binzhou Medical University, Yantai, 264003, China.
| | | |
Collapse
|
9
|
Zhang QY, Yue XQ, Jiang YP, Han T, Xin HL. FAM46C is critical for the anti-proliferation and pro-apoptotic effects of norcantharidin in hepatocellular carcinoma cells. Sci Rep 2017; 7:396. [PMID: 28341836 PMCID: PMC5428258 DOI: 10.1038/s41598-017-00313-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Accepted: 02/20/2017] [Indexed: 02/08/2023] Open
Abstract
Norcantharidin (NCTD), a demethylated analog of cantharidin derived from Chinese traditional medicine blister beetle, has been currently used as an anticancer drug for various cancers including hepatocellular carcinoma (HCC). In this study, for a more comprehensive understanding of the targets of NCTD in HCC, next-generation RNA-Seq was utilized. We revealed that the expression of FAM46C, which has been reported as a tumor suppressor for multiple myeloma, was enhanced after NCTD treatment. Re-analysis of TCGA (The Cancer Genome Atlas) LIHC (liver hepatocellular carcinoma) dataset demonstrated that FAM46C expression was significantly lower in HCC tissues than in normal liver tissues. NCTD injection or FAM46C overexpression could mitigate diethylnitrosamine (DEN)-initiated HCC in mice. Ectopic expression of FAM46C in two HCC cell lines, SMCC-7721 and SK-Hep-1, significantly repressed cell proliferation, and increased cells population in G2/M phase and cell apoptotic rate. We also found that FAM46C overexpression caused a notable decrease in Ras expression, MEK1/2 phosphorylation and ERK1/2 phosphorylation. More importantly, FAM46C knockdown significantly weakened the biological effects of NCTD on HCC cells, which suggested NCTD exerted the anticancer functions partially through up-regulating FAM46C. In conclusion, FAM46C, a tumor suppressor for HCC, is important for the anti-proliferation and proapoptotic effects of NCTD.
Collapse
Affiliation(s)
- Qiao-Yan Zhang
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, P. R. China
| | - Xiao-Qiang Yue
- Department of Traditional Chinese Medicine, Changzheng Hospital, Second Military Medical University, Shanghai, 200433, P. R. China
| | - Yi-Ping Jiang
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, P. R. China
| | - Ting Han
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, P. R. China
| | - Hai-Liang Xin
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical University, Shanghai, 200433, P. R. China.
| |
Collapse
|
10
|
Propylene Glycol Alginate Sodium Sulfate Alleviates Cerulein-Induced Acute Pancreatitis by Modulating the MEK/ERK Pathway in Mice. Mar Drugs 2017; 15:md15020045. [PMID: 28218693 PMCID: PMC5334625 DOI: 10.3390/md15020045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 01/17/2017] [Indexed: 12/13/2022] Open
Abstract
Previous studies have focused on the effects of propylene glycol alginate sodium sulfate (PSS) against thrombosis, but the anti-inflammatory potential is unknown. Therefore, we specifically focused on the protective effects of PSS on cerulein-induced acute pancreatitis (AP) using a mouse model, and investigated the mechanism of PSS on autophagy and apoptosis via the Mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway. Cerulein (100 ug/kg) was used to induce AP by ten intraperitoneal injections at hourly intervals in Balb/C mice. Pretreatment with vehicle or PSS was carried out 1 h before the first cerulein injection and two doses (25 mg/kg and 50 mg/kg) of PSS were injected intraperitoneally. The severity of AP was assessed by pathological score, biochemistry, pro-inflammatory cytokine levels, myeloperoxidase (MPO) activity and MEK/ERK activity. Furthermore, pancreatic histological scores, serum amylase and lipase activities, tumor necrosis factor-α (TNF-α), interleukin (IL)-1β interleukin (IL)-6 levels, and MPO activity were significantly reduced by PSS via up-regulated MEK/ERK activity. The representative molecules of apoptosis and autophagy, such as Bcl-2, Bax, Lc-3, Beclin-1, P62, were remarkably reduced. Taken together, these results indicate that PSS attenuates pancreas injury by inhibiting autophagy and apoptosis through a mechanism involving the MEK/ERK signaling pathway.
Collapse
|
11
|
Wan XY, Zhai XF, Jiang YP, Han T, Zhang QY, Xin HL. Antimetastatic effects of norcantharidin on hepatocellular carcinoma cells by up-regulating FAM46C expression. Am J Transl Res 2017; 9:155-166. [PMID: 28123642 PMCID: PMC5250712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 09/12/2016] [Indexed: 06/06/2023]
Abstract
Hepatocellular carcinoma (HCC) is the third leading cause of cancer-related mortality worldwide. Norcantharidin (NCTD), a demethylated analog of cantharidin, possesses antimetastatic effects on HCC cells. The aim of this study was to identify target proteins of NCTD. In this study, we confirmed the antimetastatic effects of NCTD on SMMC-7721 and MHCC-97H cells. Through RNA sequencing, we found a non-canonical poly (A) polymerase, Family-with-sequence-similarity-46C (FAM46C) was up-regulated in response to NCTD exposure. Gene set enrichment analysis on The Cancer Genome Atlas liver HCC (LIHC) dataset revealed that metastasis down pathway was strongly associated with FAM46C expression. Overexpression of FAM46C in HCC cells suppressed cell migration and invasion via suppressing transforming growth factor-β (TGF-β)/Smad signaling and epithelial-mesenchymal transition (EMT) process. Additionally, the antimetastatic effects of NCTD on HCC cells were partially rescued by FAM46C knockdown. Collectively, our results suggested that FAM46C, up-regulated by NCTD treatment, played a critical role in promoting the migration and invasion of HCC cells via TGF-β/Smad signaling. We identified a new therapeutic target of NCTD.
Collapse
Affiliation(s)
- Xu-Ying Wan
- The integrated TCM & Western Medicine Department, Eastern Hepatobiliary Surgery Hospital, Second Military Medical UniversityShanghai 200433, P. R. China
| | - Xiao-Feng Zhai
- Department of Traditional Chinese Medicine, Changhai Hospital, Second Military Medical UniversityShanghai 200433, P. R. China
| | - Yi-Ping Jiang
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical UniversityShanghai 200433, P. R. China
| | - Ting Han
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical UniversityShanghai 200433, P. R. China
| | - Qiao-Yan Zhang
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical UniversityShanghai 200433, P. R. China
| | - Hai-Liang Xin
- Department of Pharmacognosy, School of Pharmacy, Second Military Medical UniversityShanghai 200433, P. R. China
| |
Collapse
|
12
|
Skała E, Sitarek P, Toma M, Szemraj J, Radek M, Nieborowska-Skorska M, Skorski T, Wysokińska H, Śliwiński T. Inhibition of human glioma cell proliferation by altered Bax/Bcl-2-p53 expression and apoptosis induction by Rhaponticum carthamoides extracts from transformed and normal roots. ACTA ACUST UNITED AC 2016; 68:1454-1464. [PMID: 27696406 DOI: 10.1111/jphp.12619] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 07/26/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVE The objective of this study was to determine the cytotoxic effect and apoptotic activity of Rhaponticum carthamoides transformed root (TR) and root of soil-grown plant (NR) extracts in a human glioma primary cells. The effect of these root extracts on cell cycle arrest, mitochondrial membrane potential (ΔΨm) and expression levels of apoptosis-related genes (Bcl-2, Bax and p53) were also examined. METHODS Cytotoxic activity of root extracts was evaluated by MTT assay. Apoptosis and cell cycle were determined by flow cytometry. Expression levels of apoptosis-related gene were analysed by RT-PCR and Western blotting. ΔΨm was examined by the use of JC-1 reagent. KEY FINDINGS Rhaponticum carthamoides root extracts inhibit cell growth and induce apoptosis in a dose-dependent manner in human glioma cells. The root extracts were found to up-regulate the pro-apoptotic Bax protein and down-regulate the anti-apoptotic Bcl-2 protein, consequently increasing the ratios of Bax/Bcl-2 protein levels. Moreover, an increase of the p53 protein level and reduction of ΔΨm in glioma cells were observed after treatment with NR and TR extracts. CONCLUSION The results of this study may offer a new insight into the potential anticancer activity of R. carthamoides root extracts.
Collapse
Affiliation(s)
- Ewa Skała
- Department of Biology and Pharmaceutical Botany, Medical University of Łódź, Poland.
| | - Przemysław Sitarek
- Department of Biology and Pharmaceutical Botany, Medical University of Łódź, Poland
| | - Monika Toma
- Department of Molecular Genetics, University of Łódź, Poland
| | - Janusz Szemraj
- Department of Medical Biochemistry, Medical University of Łódź, Poland
| | - Maciej Radek
- Department of Neurosurgery and Peripheral Nerve Surgery, Medical University of Łódź, University Hospital WAM-CSW, Łódź, Poland
| | - Malgorzata Nieborowska-Skorska
- Department of Microbiology and Immunology, Fels Institute for Cancer Research and Molecular Biology, Temple University, School of Medicine, Philadelphia, PA, USA
| | - Tomasz Skorski
- Department of Microbiology and Immunology, Fels Institute for Cancer Research and Molecular Biology, Temple University, School of Medicine, Philadelphia, PA, USA
| | - Halina Wysokińska
- Department of Biology and Pharmaceutical Botany, Medical University of Łódź, Poland
| | | |
Collapse
|
13
|
Natural compounds for pediatric cancer treatment. Naunyn Schmiedebergs Arch Pharmacol 2015; 389:131-49. [DOI: 10.1007/s00210-015-1191-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 11/08/2015] [Indexed: 12/13/2022]
|
14
|
Gao B, Huang Q, Jie Q, Zhang HY, Wang L, Guo YS, Sun Z, Wei BY, Han YH, Liu J, Yang L, Luo ZJ. Ginsenoside-Rb2 inhibits dexamethasone-induced apoptosis through promotion of GPR120 induction in bone marrow-derived mesenchymal stem cells. Stem Cells Dev 2014; 24:781-90. [PMID: 25314926 DOI: 10.1089/scd.2014.0367] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Apoptosis of bone marrow-derived mesenchymal stem cells (BMMSCs) is an essential pathogenic factor of osteoporosis. Ginsenoside-Rb2 (Rb2), a 20(S)-protopanaxadiol glycoside extracted from ginseng, is a potent treatment for bone loss, which raises interest regarding the bone metabolism area. In the present study, we found that dose-response Rb2 inhibited high dosage of dexamethasone (Dex)-induced apoptosis in primary murine BMMSCs. Interestingly, Rb2 promoted GPR120 induction, which is the unsaturated long-chain fatty acid receptor. We further confirmed that GPR120-specific ShRNA reversed the inhibition of Rb2 on Dex-induced apoptosis by activating caspase-3 and reducing cell viability. In addition, Rb2 notably increased phosphorylated ERK1/2 levels and Ras kinase activity dependently through the GPR120. The ERK1/2 activity-specific inhibitor U0126 remarkably blocked the Rb2-induced antiapoptotic effect in response to Dex-induced apoptosis. Together, dose-response Rb2 protected BMMSCs against Dex-induced apoptosis dependently by inducing GPR120 promoted Ras-ERK1/2 signaling pathway. Therefore, in the prevalence of the abuse of Dex in the clinic, our findings suggest for the first time that Rb2 is not only a key to understand the link between Chinese medicine and the pathology of osteoporosis but also an underlying target for the treatment of bone complications in the foreseeable future.
Collapse
Affiliation(s)
- Bo Gao
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University , Xi'an, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|