1
|
Valadares JMM, Azalim-Neto P, Liu X, Carrozza NC, O'Doherty GA, Quintas LEM, Barbosa LA. Pharmacodynamic characterization and evaluation of oxidative stress effects of digitoxigenin derivatives on HeLa cells. J Membr Biol 2025; 258:63-73. [PMID: 39812699 DOI: 10.1007/s00232-024-00334-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 12/18/2024] [Indexed: 01/16/2025]
Abstract
Cancer is a leading cause of death worldwide and its treatment is hampered by the lack of specificity and side effects of current drugs. Cardiotonic steroids (CTS) interact with Na+/K+-ATPase (NKA) and induce antineoplastic effects, but their narrow therapeutic window is key limiting factor. The synthesis of digitoxigenin derivatives with glycosidic unit modifications is a promising approach to develop more selective and effective antitumor agents. This study aimed to compare the pharmacological properties as well as the cytotoxic effects of digitoxigenin-α-L-amiceto-pyranoside and digitoxigenin-α-L-rhamno-pyranoside and to evaluate the mechanism of these derivatives in oxidative conditions in HeLa cells. The rhamnose derivative increased the binding affinity and inhibitory effect of digitoxigenin by approximately 5-15 times, unlike the amicetose derivative. Despite this difference, both compounds similarly increased H2O2 levels, induced membrane lipid peroxidation, and reduced GSH levels and SOD activity at nanomolar concentrations. This study highlights the importance of the sugar moiety in CTS structure for NKA binding and demonstrates that a primary mechanism of cytotoxicity of digitoxigenin derivatives may involve cellular oxidative stress, underscoring their potential as therapeutic agents for cancer treatment.
Collapse
Affiliation(s)
- Jessica M M Valadares
- Laboratório de Bioquímica Celular, Universidade Federal de São João del-Rei (UFSJ), Divinópolis, Brazil
| | - Pedro Azalim-Neto
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biomédicas, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil
| | - Xiaofan Liu
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115, USA
| | | | - George A O'Doherty
- Department of Chemistry and Chemical Biology, Northeastern University, Boston, MA, 02115, USA
| | - Luis Eduardo M Quintas
- Laboratório de Farmacologia Bioquímica e Molecular, Instituto de Ciências Biomédicas, Universidade Federal Do Rio de Janeiro, Rio de Janeiro, 21941-902, Brazil.
| | - Leandro A Barbosa
- Laboratório de Bioquímica Celular, Universidade Federal de São João del-Rei (UFSJ), Divinópolis, Brazil
| |
Collapse
|
2
|
Chen N, Wu Y, Wei H, Zhi S, Liu L. The advancement of structure, bioactivity, mechanism, and synthesis of bufotalin. Steroids 2025; 214:109555. [PMID: 39709107 DOI: 10.1016/j.steroids.2024.109555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
Toad venom, a family of toxic yet pharmacologically valuable biotoxins, has long been utilized in traditional medicine and holds significant promise in modern drug development. Bufotalin, a prominent bufotoxin, has demonstrated potent cytotoxic properties through mechanisms such as apoptosis induction, cell cycle arrest, endoplasmic reticulum stress activation, and inhibition of metastasis by modulating key pathways including Akt, p53, and STAT3/EMT signaling-these multi-target mechanisms position bufotalin as a promising agent to combat multidrug resistance in cancer therapy. Additionally, advances in bufotalin synthesis, including chemical and biocatalytic methods, have streamlined production, with strategies such as C14α-hydroxylation and novel coupling techniques enhancing yield and reducing environmental impact. This review consolidates recent progress on bufotalin's structure, activity, cytotoxic mechanisms, and synthetic methodologies, offering a foundation for further development as an innovative chemotherapy agent.
Collapse
Affiliation(s)
- Nuo Chen
- Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China; Department of Marine Pharmacy, College of Food Science and Engineering, Ningbo University, Ningbo, Zhejiang 315832, China
| | - Yunqiang Wu
- Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China; Department of Marine Pharmacy, College of Food Science and Engineering, Ningbo University, Ningbo, Zhejiang 315832, China
| | - Huamao Wei
- Department of Marine Pharmacy, College of Food Science and Engineering, Ningbo University, Ningbo, Zhejiang 315832, China
| | - Shuai Zhi
- School of Public Health, Ningbo University, Ningbo, Zhejiang 315000, China.
| | - Liwei Liu
- Health Science Center, Ningbo University, Ningbo, Zhejiang 315211, China.
| |
Collapse
|
3
|
Wu Q, Ge XL, Geng ZK, Wu H, Yang JY, Cao SR, Yang AL. HuaChanSu suppresses the growth of hepatocellular carcinoma cells by interfering with pentose phosphate pathway through down-regulation of G6PD enzyme activity and expression. Heliyon 2024; 10:e25144. [PMID: 38322888 PMCID: PMC10844274 DOI: 10.1016/j.heliyon.2024.e25144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 02/08/2024] Open
Abstract
HuaChanSu is active water extracts from the skin of Bufo bufo gargarizans Cantor. It has been already used to treat clinical cancers including HCC (Hepatocellular carcinoma, HCC), however, the molecular mechanisms under HuaChanSu's anti-cancer effects remain unclear. PPP (Pentose phosphate pathway, PPP), the major source of ribose and NADPH (Nicotinamide adenine dinucleotide phosphate, NADPH), is always over-activated and particularly critical for tumor cells growth. In this study, firstly, we illustrate that HuaChanSu restrains the growth of human hepatoma cells. More importantly, we demonstrate that the expression of G6PD (Glucose-6-phosphate dehydrogenase, G6PD), the first rate-limiting enzyme of the PPP, is restrained in human hepatoma cells after treatment with HuaChanSu. Additionally, our results show that G6PD enzyme activity and dimer formation are inhibited by HuaChanSu. Furthermore, we find that HuaChanSu could inhibit NADPH production and nucleotide level. In addition, we identify that expression of PLK1 (Polo-like kinase 1, PLK1) is also reduced in response to HuaChanSu, and knockdown of PLK1 restrains enzyme activity and dimer formation of G6PD, but has no effect on G6PD protein level. Subsequently, we demonstrate that inhibition of G6PD could restrain the proliferation of tumor cells and enhance the inhibitory effect of HuaChanSu on cell proliferation of human hepatoma cells. In conclusion, for the first time, our study reveals that HuaChanSu interferes with PPP via suppression of G6PD expression and enzyme activity to restrain growth of tumor cells, and these results provide a novel insight for the anti-hepatoma mechanisms of HuaChanSu and promote the innovation of the research model of TCM. Moreover, the development of drugs targeting abnormal tumor metabolism is currently a hot topic, our works provide theoretical support for further drug development from HuaChanSu, meanwhile, the revelation of the new molecular mechanism also provides a new perspective for the study of the pathogenesis of liver cancer. Short abstract HuaChanSu suppresses expression of G6PD, the first rate-limiting enzyme of the PPP, restrains G6PD enzyme activity and dimer formation via inhibition of PLK1, knockdown of G6PD could impair the growth of human hepatoma cells and increase the blocking effect of HuaChanSu on cell proliferation of cancer cells. In addition, HuaChanSu restrains NADPH production and nucleotide level, implying the suppression of PPP flux. Our study suggests that HuaChanSu interferes with PPP via G6PD inhibition to exert anti-hepatoma effects.
Collapse
Affiliation(s)
| | | | | | - Hao Wu
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Jing-yi Yang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Shi-rong Cao
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| | - Ai-lin Yang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China
| |
Collapse
|
4
|
Wang PF, Yang Y, Patel V, Neiner A, Kharasch ED. Natural Products Inhibition of Cytochrome P450 2B6 Activity and Methadone Metabolism. Drug Metab Dispos 2024; 52:252-265. [PMID: 38135504 PMCID: PMC10877711 DOI: 10.1124/dmd.123.001578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/16/2023] [Accepted: 12/20/2023] [Indexed: 12/24/2023] Open
Abstract
Methadone is cleared predominately by hepatic cytochrome P450 (CYP) 2B6-catalyzed metabolism to inactive metabolites. CYP2B6 also catalyzes the metabolism of several other drugs. Methadone and CYP2B6 are susceptible to pharmacokinetic drug-drug interactions. Use of natural products such as herbals and other botanicals is substantial and growing, and concomitant use of prescription medicines and non-prescription herbals is common and may result in interactions, often precipitated by CYP inhibition. Little is known about herbal product effects on CYP2B6 activity, and CYP2B6-catalyzed methadone metabolism. We screened a family of natural product compounds used in traditional medicines, herbal teas, and synthetic analogs of compounds found in plants, including kavalactones, flavokavains, chalcones and gambogic acid, for inhibition of expressed CYP2B6 activity and specifically inhibition of CYP2B6-mediated methadone metabolism. An initial screen evaluated inhibition of CYP2B6-catalyzed 7-ethoxy-4-(trifluoromethyl) coumarin O-deethylation. Hits were further evaluated for inhibition of racemic methadone metabolism, including mechanism of inhibition and kinetic constants. In order of decreasing potency, the most effective inhibitors of methadone metabolism were dihydromethysticin (competitive, K i 0.074 µM), gambogic acid (noncompetitive, K i 6 µM), and 2,2'-dihydroxychalcone (noncompetitive, K i 16 µM). Molecular modeling of CYP2B6-methadone and inhibitor binding showed substrate and inhibitor binding position and orientation and their interactions with CYP2B6 residues. These results show that CYP2B6 and CYP2B6-catalyzed methadone metabolism are inhibited by certain natural products, at concentrations which may be clinically relevant. SIGNIFICANCE STATEMENT: This investigation identified several natural product constituents which inhibit in vitro human recombinant CYP2B6 and CYP2B6-catalyzed N-demethylation of the opioid methadone. The most potent inhibitors (K i) were dihydromethysticin (0.074 µM), gambogic acid (6 µM) and 2,2'-dihydroxychalcone (16 µM). Molecular modeling of ligand interactions with CYP2B6 found that dihydromethysticin and 2,2'-dihydroxychalcone bound at the active site, while gambogic acid interacted with an allosteric site on the CYP2B6 surface. Natural product constituents may inhibit CYP2B6 and methadone metabolism at clinically relevant concentrations.
Collapse
Affiliation(s)
- Pan-Fen Wang
- Department of Anesthesiology, Duke University, Durham, North Carolina (P.-F.W., E.D.K.) and Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri (Y.Y., V.P., A.N.)
| | - Yanming Yang
- Department of Anesthesiology, Duke University, Durham, North Carolina (P.-F.W., E.D.K.) and Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri (Y.Y., V.P., A.N.)
| | - Vishal Patel
- Department of Anesthesiology, Duke University, Durham, North Carolina (P.-F.W., E.D.K.) and Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri (Y.Y., V.P., A.N.)
| | - Alicia Neiner
- Department of Anesthesiology, Duke University, Durham, North Carolina (P.-F.W., E.D.K.) and Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri (Y.Y., V.P., A.N.)
| | - Evan D Kharasch
- Department of Anesthesiology, Duke University, Durham, North Carolina (P.-F.W., E.D.K.) and Department of Anesthesiology, Washington University in St. Louis, St. Louis, Missouri (Y.Y., V.P., A.N.)
| |
Collapse
|
5
|
Pei X, Tian M, Wang Y, Xin Y, Jiang J, Wang Y, Gong Y. Advances in the knowledge on the role of apoptosis repressor with caspase recruitment domain in hemorrhagic stroke. JOURNAL OF INTENSIVE MEDICINE 2023; 3:138-143. [PMID: 37188117 PMCID: PMC10175730 DOI: 10.1016/j.jointm.2022.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/03/2022] [Accepted: 11/23/2022] [Indexed: 05/17/2023]
Abstract
The apoptosis repressor with caspase recruitment domain (ARC) plays a critical role in extrinsic apoptosis initiation via death receptor ligands, physiological stress, infection response in a tissue-dependent manner, endoplasmic reticulum (ER) stress, genotoxic drugs, ionizing radiation, oxidative stress, and hypoxia. Recent studies have suggested that regulating apoptosis-related pathways can improve outcomes for patients with neurological diseases, such as hemorrhagic stroke. ARC expression is significantly correlated with acute cerebral hemorrhage. However, the mechanism by which it mediates the anti-apoptosis pathway remains poorly known. Here, we discuss the function of ARC in hemorrhagic stroke and argue that it could serve as an effective target for the treatment of hemorrhagic stroke.
Collapse
Affiliation(s)
- Xu Pei
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Mi Tian
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yao Wang
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yuewen Xin
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Junliang Jiang
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Yunyun Wang
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Ye Gong
- Department of Critical Care Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200040, China
- Corresponding author: Ye Gong, Department of Critical Care Medicine, Huashan Hospital, Fudan University, 12 Urumqi Road, Shanghai 200040, China.
| |
Collapse
|
6
|
NOTCH Signaling in Osteosarcoma. Curr Issues Mol Biol 2023; 45:2266-2283. [PMID: 36975516 PMCID: PMC10047431 DOI: 10.3390/cimb45030146] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/05/2023] [Accepted: 03/06/2023] [Indexed: 03/11/2023] Open
Abstract
The combination of neoadjuvant chemotherapy and surgery has been promoted for the treatment of osteosarcoma; however, the local recurrence and lung metastasis rates remain high. Therefore, it is crucial to explore new therapeutic targets and strategies that are more effective. The NOTCH pathway is not only involved in normal embryonic development but also plays an important role in the development of cancers. The expression level and signaling functional status of the NOTCH pathway vary in different histological types of cancer as well as in the same type of cancer from different patients, reflecting the distinct roles of the Notch pathway in tumorigenesis. Studies have reported abnormal activation of the NOTCH signaling pathway in most clinical specimens of osteosarcoma, which is closely related to a poor prognosis. Similarly, studies have reported that NOTCH signaling affected the biological behavior of osteosarcoma through various molecular mechanisms. NOTCH-targeted therapy has shown potential for the treatment of osteosarcoma in clinical research. After the introduction of the composition and biological functions of the NOTCH signaling pathway, the review paper discussed the clinical significance of dysfunction in osteosarcoma. Then the paper reviewed the recent relevant research progress made both in the cell lines and in the animal models of osteosarcoma. Finally, the paper explored the potential of the clinical application of NOTCH-targeted therapy for the treatment of osteosarcoma.
Collapse
|
7
|
Xia Z, Li M, Hu M, Lin Y, Atteh LL, Fu W, Gao L, Bai M, Huang C, Yue P, Liu Y, Meng W. Phosphoproteomics reveals that cinobufotalin promotes intrahepatic cholangiocarcinoma cell apoptosis by activating the ATM/CHK2/p53 signaling pathway. Front Oncol 2022; 12:982961. [PMID: 36185307 PMCID: PMC9523695 DOI: 10.3389/fonc.2022.982961] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 08/31/2022] [Indexed: 11/30/2022] Open
Abstract
Intrahepatic cholangiocarcinoma (ICC) is a malignant tumor that originates from bile duct’s epithelial cells and is usually characterized by insidious symptoms and poor prognosis. Cinobufotalin (CB), an active ingredient obtained from the Traditional Chinese Medicine ChanSu, is purported to exhibit a wide range of antitumorigenic activities. However, the mechanism by which it achieves such pharmacological effects remains elusive. Here, we disclosed the mechanism of action by which CB inhibits ICC cells. Initial experiments revealed that the proliferation of RBE and HCCC-9810 cells was significantly inhibited by CB with IC50 values of 0.342 μM and 0.421 μM respectively. CB induced the expression of caspase-3 subsequently leading to the apoptosis of ICC cells. Phosphoproteomics revealed that the phosphorylation of many proteins associated with DNA damage response increased. Kinase-substrate enrichment analysis revealed that ATM was activated after CB treatment, while CDK1 was inactivated. Activated ATM increased p-CHK2-T68 and p-p53-S15, which promoted the expression of FAS, DR4 and DR5 and triggered cell apoptosis. In summary, this work reveals the role of CB in inducing DNA damage and cell apoptosis involved in the activation of the ATM/CHK2/p53 signaling pathway, and indicates that CB may serve as a chemotherapeutic drug candidate for ICC treatment.
Collapse
Affiliation(s)
- Zhili Xia
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Minzhen Li
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Meng Hu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Yanyan Lin
- The Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Institute of Hepatopancreatobiliary Surgery, Lanzhou, China
| | | | - Wenkang Fu
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Long Gao
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Mingzhen Bai
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Chongfei Huang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Ping Yue
- The Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Institute of Hepatopancreatobiliary Surgery, Lanzhou, China
| | - Yu Liu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, China
- *Correspondence: Wenbo Meng, ; Yu Liu,
| | - Wenbo Meng
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- The Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Institute of Hepatopancreatobiliary Surgery, Lanzhou, China
- *Correspondence: Wenbo Meng, ; Yu Liu,
| |
Collapse
|
8
|
Wang J, Chang H, Su M, Zhao H, Qiao Y, Wang Y, Shang L, Shan C, Zhang S. The Potential Mechanisms of Cinobufotalin Treating Colon Adenocarcinoma by Network Pharmacology. Front Pharmacol 2022; 13:934729. [PMID: 35814224 PMCID: PMC9262105 DOI: 10.3389/fphar.2022.934729] [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: 05/03/2022] [Accepted: 05/30/2022] [Indexed: 11/13/2022] Open
Abstract
Network pharmacology, as a novel way using bioinformatics to explore drug targets and interactions in cancer, broadens our understanding of drug action, thereby facilitating drug discovery. Here, we utilized network pharmacology to explore the role and mechanism by which cinobufotalin functions in colon adenocarcinoma (COAD). We found that cinobufotalin represses the growth and proliferation of colon cancer cells, and integrated public databases for targets reported to be associated with COAD, together with those predicted to be targets of cinobufotalin. Targets overlapped between COAD-associated proteins and cinobufotalin target proteins were used to filter candidate targets of cinobufotalin in COAD. The following proteins were thought to occupy a key position in COAD-cinobufotalin target networks: SRC, PIK3R1, MAPK1, PIK3CA, HSP90AA1, CTNNB1, GRB2, RHO1, PTPN11, and EGFR. The networks regulated by cinobufotalin were involved mainly in extracellular signal stimulation and transduction, including MAPK signaling pathway, PI3K-AKT signaling pathway, and JAK-STAT signaling pathway. Besides, transcriptome sequencing results also indicated that cinobufotalin inhibits the response of colon cancer cells to extracellular stimulation and promotes cell apoptosis. Molecular docking results showed that cinobufotalin matches in the pocket of the top candidate cinobufotalin target proteins (SRC, PIK3R1, MAPK1 and PIK3CA). These findings demonstrate cinobufotalin can be developed as potential anti-cancer therapeutics.
Collapse
Affiliation(s)
- Jiyan Wang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
- *Correspondence: Shuai Zhang, ; Changliang Shan, ; Jiyan Wang,
| | - Hongkai Chang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Meng Su
- School of Life Science and Bio-Pharmaceutics, Shenyang Pharmaceutical University, Shenyang, China
| | - Huifang Zhao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yaya Qiao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Yu Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Luqing Shang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
| | - Changliang Shan
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin, China
- *Correspondence: Shuai Zhang, ; Changliang Shan, ; Jiyan Wang,
| | - Shuai Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Shuai Zhang, ; Changliang Shan, ; Jiyan Wang,
| |
Collapse
|
9
|
Combined Treatment of Cinobufotalin and Gefitinib Exhibits Potent Efficacy against Lung Cancer. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6612365. [PMID: 34122599 PMCID: PMC8189783 DOI: 10.1155/2021/6612365] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 03/08/2021] [Accepted: 03/10/2021] [Indexed: 01/22/2023]
Abstract
This study aimed to evaluate the efficacy of cinobufotalin combined with gefitinib in the treatment of lung cancer. A549 cells were treated with gefitinib, cinobufotalin, or cinobufotalin plus gefitinib. MTT assay, annexin-V/PI staining and flow cytometry, TUNEL staining, DCFH-DA staining, Western blot, and real-time RT-PCR were performed to investigate the synergistic inhibitory effect of cinobufotalin combined with gefitinib on the growth of A549 cells. Results showed that cinobufotalin synergized with gefitinib displayed inhibited cell viability and enhanced apoptosis in the combination group. Cinobufotalin combined with gefitinib induced a significant enhancement in reactive oxygen species (ROS) production accompanied by cell cycle arrest in the S phase arrest, characterized by upregulation of p21 and downregulation of cyclin A, cyclin E, and CDK2. Besides, cinobufotalin plus gefitinib downregulated the levels of HGF and c-Met. In summary, cinobufotalin combined with gefitinib impedes viability and facilitates apoptosis of A549 cells, indicating that the combined therapy might be a new promising treatment for lung cancer patients who are resistant to gefitinib.
Collapse
|
10
|
Pan Z, Fan J, Xie Q, Zhang X, Zhang W, Ren Q, Li M, Zheng Q, Lu J, Li D. Novel sulfonamide porphyrin TBPoS-2OH used in photodynamic therapy for malignant melanoma. Biomed Pharmacother 2020; 133:111042. [PMID: 33378950 DOI: 10.1016/j.biopha.2020.111042] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 11/10/2020] [Accepted: 11/15/2020] [Indexed: 12/28/2022] Open
Abstract
The application of photodynamic therapy (PDT) for the treatment of skin diseases has been receiving much attention. Here, we examined the anti-tumor effect of a novel porphyrin-based photosensitizer TBPoS-2OH in the malignant melanoma A375 and B16 cells. TBPoS-2OH has obvious cell photo-cytotoxicity, but it has low cell dark-cytotoxicity. Further research showed that TBPoS-2OH is enriched in lysosomes after being taken up by cells. Subsequently, the apoptotic rates were significantly increased in TBPoS-2OH-treated A375 and B16 cells. The specific mechanism may be that after receiving light stimulation, TBPoS-2OH could effectively increase the level of intracellular reactive oxygen species (ROS), thereby activating mitochondrial apoptosis pathway-related proteins in A375 and B16 cells. We found an increase in the content of cytochrome C in the cytoplasm, and the levels of related proteins, such as cleaved caspase-3, cleaved caspase-9, and cleaved PARP1, were significantly increased in TBPoS-2OH-treated cells. These results indicated that the new compound TBPoS-2OH could be developed and become an alternative drug for the treatment of melanoma. Some reference ideas for the development of new photosensitizers are also provided.
Collapse
Affiliation(s)
- Zhaohai Pan
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, 264003, Yantai, China
| | - Jiaojiao Fan
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, 264003, Yantai, China
| | - Qi Xie
- Jiangsu College of Nursing, 9 Science and Technology Avenue, Huaian, 223005, Jiangsu, China
| | - Xin Zhang
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, 264003, Yantai, China
| | - Wen Zhang
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, 264003, Yantai, China
| | - Qing Ren
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Minjing Li
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, 264003, Yantai, China
| | - Qiusheng Zheng
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, 264003, Yantai, China; Key Laboratory of Xinjiang Endemic Phytomedicine Resources of Ministry of Education, School of Pharmacy, Shihezi University, Shihezi, 832002, Xinjiang, China
| | - Jun Lu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China.
| | - Defang Li
- Yantai Key Laboratory of Pharmacology of Traditional Chinese Medicine in Tumor Metabolism, School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, 264003, Yantai, China.
| |
Collapse
|
11
|
Review of Natural Compounds for the Management and Prevention of Lymphoma. Processes (Basel) 2020. [DOI: 10.3390/pr8091164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Lymphoma is a type of blood cancer that can be categorized into two types-Hodgkin lymphoma (HL) and Non-Hodgkin lymphoma (NHL). A total of 509,590 and 79,990 cases of NHL and HL were newly diagnosed in 2018, respectively. Although conventional therapy has stridden forward over recent decades, its adverse effects are still a hurdle to be solved. Thus, to help researchers develop better lymphoma treatment, this study aims to review the systematic anticancer data for natural products and their compounds. A variety of natural products showed anticancerous effects on lymphoma by regulation of intracellular mechanisms including apoptosis as well as cell cycle arrest. As these results shed light on the potential to substitute conventional therapy with natural products, it may become a promising strategy for lymphoma treatment in the near future.
Collapse
|
12
|
Deng LJ, Li Y, Qi M, Liu JS, Wang S, Hu LJ, Lei YH, Jiang RW, Chen WM, Qi Q, Tian HY, Han WL, Wu BJ, Chen JX, Ye WC, Zhang DM. Molecular mechanisms of bufadienolides and their novel strategies for cancer treatment. Eur J Pharmacol 2020; 887:173379. [PMID: 32758567 DOI: 10.1016/j.ejphar.2020.173379] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 07/05/2020] [Accepted: 07/13/2020] [Indexed: 12/16/2022]
Abstract
Bufadienolides are cardioactive C24 steroids with an α-pyrone ring at position C17. In the last ten years, accumulating studies have revealed the anticancer activities of bufadienolides and their underlying mechanisms, such as induction of autophagy and apoptosis, cell cycle disruption, inhibition of angiogenesis, epithelial-mesenchymal transition (EMT) and stemness, and multidrug resistance reversal. As Na+/K+-ATPase inhibitors, bufadienolides have inevitable cardiotoxicity. Short half-lives, poor stability, low plasma concentration and oral bioavailability in vivo are obstacles for their applications as drugs. To improve the drug potency of bufadienolides and reduce their side effects, prodrug strategies and drug delivery systems such as liposomes and nanoparticles have been applied. Therefore, systematic and recapitulated information about the antitumor activity of bufadienolides, with special emphasis on the molecular or cellular mechanisms, prodrug strategies and drug delivery systems, is of high interest. Here, we systematically review the anticancer effects of bufadienolides and the molecular or cellular mechanisms of action. Research advancements regarding bufadienolide prodrugs and their tumor-targeting delivery strategies are critically summarized. This work highlights recent scientific advances regarding bufadienolides as effective anticancer agents from 2011 to 2019, which will help researchers to understand the molecular pathways involving bufadienolides, resulting in a selective and safe new lead compound or therapeutic strategy with improved therapeutic applications of bufadienolides for cancer therapy.
Collapse
Affiliation(s)
- Li-Juan Deng
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China; School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Yong Li
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Ming Qi
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Jun-Shan Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Sheng Wang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Li-Jun Hu
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Yu-He Lei
- Shenzhen Hospital of Guangzhou University of Chinese Medicine, Shenzhen, 518034, China
| | - Ren-Wang Jiang
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Wei-Min Chen
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Qi Qi
- Clinical Translational Center for Targeted Drug, Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Hai-Yan Tian
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China
| | - Wei-Li Han
- School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Bao-Jian Wu
- College of Pharmacy, Jinan University, Guangzhou, 510632, China
| | - Jia-Xu Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, China
| | - Wen-Cai Ye
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China.
| | - Dong-Mei Zhang
- College of Pharmacy, Jinan University, Guangzhou, 510632, China; Guangdong Province Key Laboratory of Pharmacodynamic Constituents of Traditional Chinese Medicine and New Drugs Research, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
13
|
Epoxide containing molecules: A good or a bad drug design approach. Eur J Med Chem 2020; 201:112327. [PMID: 32526552 DOI: 10.1016/j.ejmech.2020.112327] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 04/06/2020] [Accepted: 04/08/2020] [Indexed: 12/13/2022]
Abstract
Functional group modification is one of the main strategies used in drug discovery and development. Despite the controversy of being identified for many years as a biologically hazardous functional group, the introduction of an epoxide function in a structural backbone is still one of the possible modifications being implemented in drug design. In this manner, it is our intention to prove with this work that epoxides can have significant interest in medicinal chemistry, not only as anticancer agents, but also as important drugs for other pathologies. Thus, this revision paper aims to highlight the biological activity and the proposed mechanisms of action of several epoxide-containing molecules either in preclinical studies or in clinical development or even in clinical use. An overview of the chemistry of epoxides is also reported. Some of the conclusions are that effectively most of the epoxide-containing molecules referred in this work were being studied or are in the market as anticancer drugs. However, some of them in preclinical studies, were also associated with other different activities such as anti-malarial, anti-arthritic, insecticidal, antithrombotic, and selective inhibitory activity of FXIII-A (a transglutaminase). As for the epoxide-containing molecules in clinical trials, some of them are being tested for obesity and schizophrenia. Finally, drugs containing epoxide groups already in the market are mostly used for the treatment of different types of cancer, such as breast cancer and multiple myeloma. Other diseases for which the referred drugs are being used include heart failure, infections and gastrointestinal disturbs. In summary, epoxides can be a suitable option in drug design, particularly in the design of anticancer agents, and deserve to be better explored. However, and despite the promising results, it is imperative to explore the mechanisms of action of these compounds in order to have a better picture of their efficiency and safety.
Collapse
|
14
|
Zhang L, Huang X, Guo T, Wang H, Fan H, Fang L. Study of Cinobufagin as a Promising Anticancer Agent in Uveal Melanoma Through Intrinsic Apoptosis Pathway. Front Oncol 2020; 10:325. [PMID: 32300551 PMCID: PMC7142239 DOI: 10.3389/fonc.2020.00325] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 02/24/2020] [Indexed: 12/20/2022] Open
Abstract
Uveal melanoma (UM) is the most common primary intraocular carcinoma in adults. Cinobufagin, secreted by the Asiatic toad Bufo gargarizans, is a traditional Chinese medicine, widely used in tumor treatment. Here, we explored the potential antitumor function of cinobufagin and investigated its biochemical mechanisms in UM cells. The antitumor potential of cinobufagin was determined via cell viability, cell cycle, and apoptosis assays. Colony formation assays confirmed that cinobufagin exerted potent antitumor activity in a dose-dependent manner. We found that cinobufagin could induce cell apoptosis and upregulate the expression of cleaved caspase-3, cleaved poly(ADP-ribose) polymerase (PARP), and cleaved caspase-9 in vivo and in vitro. In addition, after treatment with increased concentrations of cinobufagin, the intrinsic mitochondrial apoptosis pathway was also activated, which was demonstrated by increased cell apoptosis with increased expression of Bad and Bax, decreased expression of Bcl-2 and Bcl-xl, and reduced mitochondrial membrane potential (MMP) in OCM1 cells. Taken together, the results of this preclinical study suggest that cinobufagin can both inhibit cell survival and induce cell apoptosis in a dose-dependent manner in UM cells, which provides new insights into the biochemical mechanism of cinobufagin and its potential as a future chemotherapeutic agent for UM.
Collapse
Affiliation(s)
- Leilei Zhang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Xiaolin Huang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Tao Guo
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Huixue Wang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Haiyan Fan
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| | - Li Fang
- Department of Ophthalmology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Orbital Diseases and Ocular Oncology, Shanghai, China
| |
Collapse
|
15
|
Liu Y, Jiang Q, Liu X, Lin X, Tang Z, Liu C, Zhou J, Zhao M, Li X, Cheng Z, Li L, Xie Y, Liu Z, Fang W. Cinobufotalin powerfully reversed EBV-miR-BART22-induced cisplatin resistance via stimulating MAP2K4 to antagonize non-muscle myosin heavy chain IIA/glycogen synthase 3β/β-catenin signaling pathway. EBioMedicine 2019; 48:386-404. [PMID: 31594754 PMCID: PMC6838365 DOI: 10.1016/j.ebiom.2019.08.040] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 08/16/2019] [Accepted: 08/19/2019] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is an Epstein-Barr virus (EBV)-related tumor. The role of EBV-encoding miR-BART22 is still unclear in NPC. This study aimed to identify the detailed mechanisms by which EBV-miR-BART22 functions as a tumor-promoting factor and evaluate the action of cinobufotalin in treating EBV-miR-BART22-overexpressing NPC cells. METHODS Using real-time PCR, western blotting, immunohistochemistry, and In situ hybridization, we detected the expression of miR-BART22 and MAP2K4 in tissues and cells, as well as evaluated their clinical relevance in NPC patients. The effects of miR-BART22 on cell metastasis, stemness and DDP chemoresistance were examined by sphere formation assay, side population analysis, transwell, boyden, in vivo xenograft tumor mouse model et al. Western blotting, immunofluorescence staining, luciferase reporter assay, ChIP, EMSA and Co-IP assay et al. were performed to explore the detailed molecular mechanism of EBV-miR-BART22 in NPC. Finally, we estimated the effects and molecular basis of Cinobufotalin on EBV-miR-BART22-overexpressing NPC cells in vitro and in vivo assays. FINDINGS We observed that EBV-miR-BART22 not only promoted tumor stemness and metastasis, but also enhanced the resistance to Cisplatin (DDP) in vitro and in vivo. Mechanistic analysis indicated that EBV-miR-BART22 directly targeted the MAP2K4 and upregulated non-muscle myosin heavy chain IIA (MYH9) expression by PI3K/AKT/c-Jun-induced transcription. Further, MYH9 interacted with glycogen synthase 3β(GSK3β) protein and induced its ubiquitin degradation by activating PI3K/AKT/c-Jun-induced ubiquitin transcription and the latter combined with increased TRAF6 E3 ligase, which further bound to GSK3β protein. Reductions in the GSK3β protein thus promoted β-catenin expression and nuclear translocation, which induced tumor stemness and the epithelial-to-mesenchymal transition (EMT) signals. Furthermore, we observed that cinobufotalin, a new chemically synthesized compound, significantly suppressed EBV-miR-BART22-induced DDP chemoresistance by upregulating MAP2K4 to suppress MYH9/GSK3β/β-catenin and its downstream tumor stemness and EMT signals in NPC. Finally, clinical data revealed that increased miR-BART22 and reduced MAP2K4 expression caused the poor prognoses of NPC patients. INTERPRETATION Our study provides a novel mechanism that cinobufotalin reversed the DDP chemoresistance and EMT induced by EBV-miR-BART22 in NPC.
Collapse
Affiliation(s)
- Yiyi Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Qingping Jiang
- Cancer Institute, School of Basic Medical Science, Southern Medical University, Guangzhou, China; Department of Pathology, Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiong Liu
- Cancer Institute, School of Basic Medical Science, Southern Medical University, Guangzhou, China; Department of Otolaryngology, Head and Neck Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, People's Republic of China
| | - Xian Lin
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - ZiBo Tang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Chen Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jin Zhou
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Mengyang Zhao
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Xin Li
- Shenzhen Key Laboratory of Viral Oncology, the Clinical Innovation & Research Center (CIRC), Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Zhao Cheng
- Shenzhen Key Laboratory of Viral Oncology, the Clinical Innovation & Research Center (CIRC), Shenzhen Hospital, Southern Medical University, Shenzhen, China; Department of Pediatric Otorhinolaryngology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Libo Li
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yingying Xie
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Zhen Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China; Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China.
| | - Weiyi Fang
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
16
|
A high-efficiency strategy integrating offline two-dimensional separation and data post-processing with dereplication: Characterization of bufadienolides in Venenum Bufonis as a case study. J Chromatogr A 2019; 1603:179-189. [DOI: 10.1016/j.chroma.2019.06.037] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 06/12/2019] [Accepted: 06/13/2019] [Indexed: 01/09/2023]
|
17
|
Zhang F, Yin Y, Xu T. Cinobufotalin injection combined with chemotherapy for the treatment of advanced NSCLC in China: A PRISMA-compliant meta-analysis of 29 randomized controlled trials. Medicine (Baltimore) 2019; 98:e16969. [PMID: 31464940 PMCID: PMC6736171 DOI: 10.1097/md.0000000000016969] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVE Cinobufotalin injection (CFI), a kind of Chinese medicine, has been considered as a promising complementary therapy option for advanced non-small cell lung cancer (NSCLC), but their efficacy and safety remain controversial. This study aimed to systematically evaluate the efficacy and safety of CFI and chemotherapy-combined therapy for advanced NSCLC. METHODS Clinical trials were searched from Web of Science, Cochrane Library, PubMed, Embase, China National Knowledge Infrastructure (CNKI), Chinese Biological Medicine Database (CBM), Chinese Medical Citation Index (CMCI), Wanfang database and Chinese Scientific Journal Database (VIP). Main measurements, including therapeutic efficacy, quality of life (QoL) and adverse events, were extracted from the retrieved publications and were systematically evaluated. RESULTS The 29 trials including 2300 advanced NSCLC patients were involved in this study. Compared with chemotherapy alone, its combination with CFI significantly prolonged the patients' 1-, 2- and 3-year overall survival rate (OS) (1-year OS, OR = 1.94, 95% CI = 1.42-2.65, P < .0001; 2-year OS, OR = 2.31, 95% CI = 1.55-3.45, P < .0001; 3-year OS, OR = 4.69, 95% CI = 1.78-12.39, P = .002) and improved patients' overall response (ORR, OR = 1.84, CI = 1.54-2.18, P < .00001), disease control rate (DCR, OR = 2.09, 95% CI = 1.68-2.60, P < .00001) and QoL (quality of life improved rate, QIR, OR = 2.64, 95% CI = 1.98-3.52, P < .00001; karnofsky performance score, KPS, OR = 10.97, 95% CI = 5.48-16.47, P < .0001). Most adverse events caused by chemotherapy were obviously alleviated (P < .05) when CFI was also applied to patients. CONCLUSION The combination of CFI and chemotherapy is safe, and is more effective in treating NSCLC than chemotherapy alone. Therefore, CFI mediated therapy could be recommended as an adjuvant treatment method for NSCLC.
Collapse
Affiliation(s)
| | - Yantong Yin
- Department of Respiratory Medicine, Liaocheng People's Hospital, Liaocheng, Shandong Province, China
| | | |
Collapse
|
18
|
Sun H, Wang W, Bai M, Liu D. Cinobufotalin as an effective adjuvant therapy for advanced gastric cancer: a meta-analysis of randomized controlled trials. Onco Targets Ther 2019; 12:3139-3160. [PMID: 31118669 PMCID: PMC6507077 DOI: 10.2147/ott.s196684] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Accepted: 03/26/2019] [Indexed: 01/20/2023] Open
Abstract
Purpose: This study aimed to investigate the efficacy and safety of combining cinobufotalin and chemotherapy for advanced gastric cancer (GC). Patients and methods: Literature retrieval was performed in Cochrane Library, Web of Science, PubMed, Embase, China National Knowledge Infrastructure (CNKI), Chinese Biological Medicine Database (CBM), Wanfang database and Chinese Scientific Journal Database (VIP) before September 2018. The primary reported outcomes including therapeutic efficacy, quality of life (QoL), and adverse events were systematically evaluated. Results: Data from 27 trials including 1,939 advanced GC patients were included. The results indicated that, compared with chemotherapy alone, the combination of chemotherapy and cinobufotalin significantly improved patients' overall response rate (odds ratio [OR] =1.88, 95% confidence interval [CI] =1.54–2.31, P<0.00001) and disease control rate (OR =2.05, 95% CI =1.63–2.58, P<0.00001). The QoL of patients also evidently improved after chemotherapy and cinobufotalin combined treatment, as indicated by increased QoL improved rate (OR =2.39, 95% CI =1.81–3.15, P<0.00001), Karnofsky Performance Score (OR =7.00, 95% CI =2.25–11.75, P=0.004) and pain relief rate (OR =7.00, 95% CI =2.25–11.75, P=0.004). Adverse events including nausea and vomiting, diarrhea, leukopenia, hand-foot syndrome, anemia, gastrointestinal side effects and peripheral neurotoxicity caused by chemotherapy were evidently alleviated (P<0.05) when cinobufotalin was administered to GC patients. Conclusion: Evidence from the meta-analysis suggested that the combination of chemotherapy and cinobufotalin is more effective in treating GC than chemotherapy alone. It alleviates the adverse effects associated with chemotherapy and improves the QoL of GC patients.
Collapse
Affiliation(s)
- Huiling Sun
- Department of Gastroenterology, Liaocheng People's Hospital, Liaocheng, Shandong 252000, People's Republic of China
| | - Wenxiao Wang
- Department of Gastroenterological Surgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, People's Republic of China
| | - Minghua Bai
- Department of Health, Liaocheng People's Hospital, Liaocheng, Shandong 252000, People's Republic of China
| | - Dongling Liu
- Department of Pharmacy, Liaocheng People's Hospital, Liaocheng, Shandong 252000, People's Republic of China
| |
Collapse
|
19
|
He C, Zou Z, Xia S, Xing X, Hu S, Hu Z, Li Y, Li S, Zhang H, Yang Y, Liu Y, Xu X, Liu B, Wang Y, Xu Y, Du X. Application of highly efficient and lowly toxic bufadienolides screened from toad skin in lymphatic chemotherapy for colorectal cancer through a lymphatic metastatic model. Int Immunopharmacol 2019; 70:241-251. [PMID: 30851704 DOI: 10.1016/j.intimp.2019.02.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/14/2019] [Accepted: 02/22/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Lymph node metastasis (LNM) remains a major obstacle to treat colorectal cancer (CRC). Increasing evidences have suggested that bufadienolides contain several fractions displaying antitumor activity and may be applied in lymphatic chemotherapy. However, effects of the highly efficient and lowly toxic (HELT) bufadienolides on CRC in lymphatic chemotherapy have not been reported. METHODS Adenosine triphosphate tumor chemosensitivity assays (ATP-TCA) was performed to detect the inhibition rate (IR) of fractions of bufadienolides to cytokine-induced killer (CIK) cells and tumor cells. HELT fraction-loaded emulsions of different concentrations were prepared. Nude mouse bearing HCT116 tumors in footpad received high-dose emulsion (HD-E), middle-dose emulsion (MD-E), low-dose emulsion (LD-E), control emulsion (CE), Cinobufacini Injection (CI), or normal saline (NS), respectively. Hematoxylin and eosin (H&E) staining, Flow Cytometry (FCM), enzyme-linked immune sorbent assay (ELISA) and hematological examination were applied to evaluate therapeutic effects and potential toxicity. RESULTS F18 and F19 were screened out as HELT fractions in vivo and F18-loaded emulsions of different concentrations for lymphatic administration were prepared. We confirmed that HD-E and MD-E produced obvious antitumor activities in footpad tumors and LNM compared with other groups in vitro. We also verified the effects of F18-loaded emulsions on activating hematopoietic function, stimulating proliferation of the spleen and natural killer (NK) cells, and promoting the secretion of IFN-γ and IgG1, although HD-E performed mild toxicity on liver. CONCLUSION The present study demonstrated that lymphatic chemotherapy with HELT fraction of bufadienolides could be an effective approach to the treatment of CRC patients with LNM.
Collapse
Affiliation(s)
- Changzheng He
- Department of General Surgery, Chinese General Hospital of People's Liberation Army, Beijing 100853, PR China
| | - Zhenyu Zou
- Department of Hernia and Abdominal Wall Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100043, PR China
| | - Shaoyou Xia
- Department of General Surgery, Chinese General Hospital of People's Liberation Army, Beijing 100853, PR China
| | - Xiaowei Xing
- Department of General Surgery, Chinese General Hospital of People's Liberation Army, Beijing 100853, PR China
| | - Shidong Hu
- Department of General Surgery, Chinese General Hospital of People's Liberation Army, Beijing 100853, PR China
| | - Zilong Hu
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, PR China
| | - Yuxuan Li
- Department of General Surgery, Chinese General Hospital of People's Liberation Army, Beijing 100853, PR China
| | - Songyan Li
- Department of General Surgery, Chinese General Hospital of People's Liberation Army, Beijing 100853, PR China
| | - Hongliang Zhang
- Department of General Surgery, Chinese General Hospital of People's Liberation Army, Beijing 100853, PR China
| | - Yu Yang
- Department of General Surgery, Chinese General Hospital of People's Liberation Army, Beijing 100853, PR China
| | - Yichen Liu
- Department of General Surgery, Chinese General Hospital of People's Liberation Army, Beijing 100853, PR China
| | - Xiaolei Xu
- Department of General Surgery, Chinese General Hospital of People's Liberation Army, Beijing 100853, PR China
| | - Boyan Liu
- Department of General Surgery, Chinese General Hospital of People's Liberation Army, Beijing 100853, PR China
| | - Yufeng Wang
- Department of Patient Admission Management, Chinese General Hospital of People's Liberation Army, Beijing 100853, PR China
| | - Yingxin Xu
- Institute of General Surgery, Chinese General Hospital of People's Liberation Army, Beijing 100853, PR China.
| | - Xiaohui Du
- Department of General Surgery, Chinese General Hospital of People's Liberation Army, Beijing 100853, PR China.
| |
Collapse
|
20
|
Guo N, Miao Y, Sun M. Transcatheter hepatic arterial chemoembolization plus cinobufotalin injection adjuvant therapy for advanced hepatocellular carcinoma: a meta-analysis of 27 trials involving 2,079 patients. Onco Targets Ther 2018; 11:8835-8853. [PMID: 30573972 PMCID: PMC6290874 DOI: 10.2147/ott.s182840] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Objective The aim of this study was to systematically investigate the safety and efficacy of the combination of transcatheter hepatic arterial chemoembolization (TACE) and cinobufotalin injection for advanced hepatocellular carcinoma (HC). Methods Clinical trials were searched from Web of Science, Cochrane Library, PubMed, Embase, Chinese Medical Citation Index (CMCI), China National Knowledge Infrastructure (CNKI), Chinese Scientific Journal Database (VIP), and Wanfang database. Outcome measures including therapeutic efficacy, quality of life, liver function, immune function, and adverse events were extracted and evaluated. Results After final assessment, 27 studies including 2,079 advanced HC patients were involved in this study. Compared with TACE alone, the combination of TACE with cinobufotalin injection adjuvant therapy significantly prolonged the patients’ 1-, 1.5-, 2-, and 3-year overall survival (OS) rate (1-year OS, OR=2.84, 95% CI=2.20–3.67, P<0.00001; 1.5-year OS, OR=3.57, 95% CI=1.92–6.66, P<0.0001; 2-year OS, OR=3.17, 95% CI=2.36–4.25, P<0.00001; 3-year OS, OR=2.88, 95% CI=1.82–4.57, P<0.00001). The combined therapy also improved patients’ overall response rate (ORR; OR=1.86, 95% CI=1.54–2.24, P<0.00001), disease control rate (DCR; OR=2.05, 95% CI=1.59–2.64, P<0.00001), and quality of life improved rate (QIR; OR=3.45, 95% CI=2.52–4.72, P<0.00001). Moreover, the immune function and liver function of HC patients were all significantly enhanced after the combined therapy of TACE and cinobufotalin injection (CD3+, P=0.001; CD4+, P=0.0006; CD4+/CD8+, P=0.03; natural killer [NK] cell, P=0.01; total bilirubin [TBIL], P=0.003; alanine aminotransferase [ALT], P<0.00001; aspartate aminotransferase [AST], P<0.00001). No serious adverse events occurred during cinobufotalin injection-mediated therapy. Conclusion The combination of TACE and cinobufotalin injection adjuvant therapy is safe and more effective for end-stage HC treatment than TACE alone.
Collapse
Affiliation(s)
- Na Guo
- Department of Hepatopathy, The Sixth People's Hospital of Qingdao, Qingdao 266033, Shandong Province, China,
| | - Yanyan Miao
- Department of Hepatopathy, The Sixth People's Hospital of Qingdao, Qingdao 266033, Shandong Province, China,
| | - Mingzhong Sun
- Department of Hepatopathy, The Sixth People's Hospital of Qingdao, Qingdao 266033, Shandong Province, China,
| |
Collapse
|
21
|
Mitsuhashi Y, Furusawa Y, Aradate T, Zhao QL, Moniruzzaman R, Kanamori M, Noguchi K, Kondo T. 3-O-trans-p-coumaroyl-alphitolic acid, a triterpenoid from Zizyphus jujuba, leads to apoptotic cell death in human leukemia cells through reactive oxygen species production and activation of the unfolded protein response. PLoS One 2017; 12:e0183712. [PMID: 28832644 PMCID: PMC5568338 DOI: 10.1371/journal.pone.0183712] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 08/09/2017] [Indexed: 11/23/2022] Open
Abstract
3-O-trans-p-coumaroyl-alphitolic acid (3OTPCA), a triterpenoid isolated from the plant Zizyphus jujuba (ZJ), is known to be cytotoxic to cancer cells; however, the molecular mechanism underlying 3OTPCA-induced cell death remains unknown. Here, we provide novel evidence that 3OTPCA induces apoptotic cell death in human leukemia cells. We found that 3OPTCA induces DNA fragmentation within 24 h after treatment in U937 cells, which was also observed in other leukemia cell lines, including Molt-4 and Jurkat cells. We then investigated other parameters involved in apoptosis, including phosphatidylserine externalization and caspase-3 cleavage in U937 cells treated with 3OTPCA. 3OTPCA caused significant DNA fragmentation, annexin-V binding, and caspase-3 cleavage, indicating that 3OTPCA exerts cytotoxicity through apoptosis induction. RNA-seq analysis revealed that the expression of transcripts associated with the unfolded protein response (UPR), such as spliced XBP-1 and CHOP, were up-regulated by 3OTPCA treatment. 3OTPCA-induced UPR activation may be due to endoplasmic reticulum (ER) stress because both 3OTPCA and thapsigargin, an endoplasmic Ca2+ transport ATPase inhibitor, increased intracellular calcium levels. 3OTPCA down-regulated the expression of Bcl-2, a target of CHOP, and led to the loss of the mitochondrial membrane, indicating that the intrinsic (mitochondrial) apoptotic pathway was triggered by 3OTPCA, likely through UPR activation. Furthermore, we found that 3OTPCA induced superoxide anion generation and, following p38 MAPK phosphorylation, caspase-8 cleavage without affecting Fas expression. It also induced subsequent Bid cleavage, which may enhance the apoptosis triggered by the intrinsic pathway. These findings reveal for the first time that 3OTPCA induces apoptotic cell death through the generation of reactive oxygen species and activation of UPR.
Collapse
Affiliation(s)
- Yohei Mitsuhashi
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani, Toyama, Japan
| | - Yukihiro Furusawa
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani, Toyama, Japan
- Department of Liberal Arts and Sciences, Toyama Prefectural University, Kurokawa, Toyama, Japan
- * E-mail:
| | - Tadashi Aradate
- Department of Medical Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani, Toyama, Japan
| | - Qing-Li Zhao
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani, Toyama, Japan
| | - Rohan Moniruzzaman
- Department of Oral and Maxillofacial Surgery, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani, Toyama, Japan
| | - Masahiko Kanamori
- Department of Human Science 1, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani, Toyama, Japan
| | - Kyo Noguchi
- Department of Radiology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani, Toyama, Japan
| | - Takashi Kondo
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani, Toyama, Japan
| |
Collapse
|
22
|
The synergistic antitumor effect of cinobufagin and cisplatin in human osteosarcoma cell line in vitro and in vivo. Oncotarget 2017; 8:85150-85168. [PMID: 29156710 PMCID: PMC5689600 DOI: 10.18632/oncotarget.19554] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 06/24/2017] [Indexed: 01/01/2023] Open
Abstract
Cisplatin (CDDP) has been shown to be a promising anticancer drug that is effective against many types of cancer, which include osteosarcoma (OS). However, its therapeutic application is restricted by its toxicity in normal tissues and by side effects caused in patients. Reduction of the toxicity of CDDP is necessary to improve cancer treatment. In the present study, we attempted to clarify how cinobufagin, a traditional Chinese medicine, enhances CDDP-induced cytotoxicity in OS cells. OS 143B cells were treated with cinobufagin and CDDP alone or in combination. After low dose combined treatments with cinobufagin and CDDP, the effects of these therapeutics on cell proliferation, apoptosis, cell cycle, migration, invasion, and involvement in Notch pathway, as well as tumor growth and metastatic capability were determined. It was found that the combination of low doses of cinobufagin and CDDP markedly inhibited cell activity, motility, and induced apoptosis and cell cycle arrest in S phase, as well as suppressing tumor growth, metastasis and prolonging longer survival of nude mice in OS xenograft models compared with the actions of either drug alone or vehicle. The results also demonstrated that cinobufagin plus CDDP significantly suppressed the Notch pathway. The anticancer mechanism of these two drugs may involve intervention in the Notch signaling, which may contribute to inhibit tumor growth. All of these results suggest that application of lower concentration cinobufagin plus CDDP could produce a synergistic antitumor effect and this finding warrants further investigation for its potential clinical applications in human OS patients.
Collapse
|
23
|
Emam H, Refaat A, Jawaid P, Ur Rehman M, Li P, Zhao QL, Kondo T. Hyperthermia and radiation reduce the toxic side-effects of bufadienolides for cancer therapy. Oncol Lett 2017; 14:1035-1040. [PMID: 28693270 DOI: 10.3892/ol.2017.6256] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Accepted: 03/30/2017] [Indexed: 11/06/2022] Open
Abstract
Bufadienolides are constituents of the traditional Chinese medicine Chan Su and are found in toad venom. Cardiovascular side-effects are one of the limiting factors towards developing bufadienolides as chemotherapeutic agents. Thus, in the present study, low doses of bufalin and cinobufotalin, prominent members of the bufadienolides, were investigated for their cytotoxic activity in combination with hyperthermia (HT) or radiation (Rad) therapy. In addition, the underlying mechanism involved was investigated. A DNA fragmentation assay, viability assay and microscopic observation were primarily used to assess the effect of low doses of the two drugs in human lymphoma U937 cells. Furthermore, the effects of these drugs on the mitochondrial membrane potential (MMP) and apoptotic-associated protein activation were investigated. HT/bufadienolide- and RT/bufadienolide-treated samples significantly increased the DNA fragmentation percentile and decreased the MMP, as well as increasing the apoptotic features observed microscopically within a relatively short time (6 h) after treatment. The two combinations affected the expression of important apoptotic markers, including caspase-3 and BH3 interacting domain death agonist. The findings of the current study confirm the additive effect of HT with this group of drugs, directing a novel therapeutic avenue for the clinical use of bufadienolides at lower doses with more restrained cardio toxic side-effects.
Collapse
Affiliation(s)
- Heba Emam
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Alaa Refaat
- Department of Cancer Cell Biology, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Paras Jawaid
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Mati Ur Rehman
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Peng Li
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Qing-Li Zhao
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| | - Takashi Kondo
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Toyama 930-0194, Japan
| |
Collapse
|
24
|
Li P, Zhao QL, Jawaid P, Rehman MU, Ahmed K, Sakurai H, Kondo T. Enhancement of hyperthermia-induced apoptosis by 5Z-7-oxozeaenol, a TAK1 inhibitor, in Molt-4 cells. Int J Hyperthermia 2017; 33:411-418. [PMID: 28111999 DOI: 10.1080/02656736.2017.1278629] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
PURPOSE Transforming growth factor-β-activated kinase1 (TAK1) plays an anti-apoptotic role in response to multiple stresses. TAK1 inhibitor, 5Z-7-oxozeaenol (OZ) has been studied for its apoptotic effects. However, the combined effect of OZ with physical stresses remains to be elusive. Therefore, in this study we focussed to determine the combined effects of OZ with hyperthermia (HT) using Molt-4 cell line. MATERIALS AND METHODS Molt-4 cells were pre-treated with OZ for 1 h followed by heat exposure (44 °C, 10 min) and harvested 24 h after incubation at 37 °C, apoptosis was measured by Annexin V-FITC/PI double staining assay using flow cytometry and cell growth was observed by cell counting assay. Further mechanism involved in the combination was investigated by measuring mitochondrial membrane potential (MMP), intracellular ROS generation, expression of apoptosis related protein, intracellular calcium ion level and Fas activity. RESULTS Combination of OZ with HT significantly enhances MMP loss and superoxide generation. Furthermore, OZ pre-treatment promotes caspase-8 cleavage, Fas externalisation, caspase 3 activity and intracellular calcium ion levels. OZ pre-treatment decreased the expression of HT-induced Bcl-2 and increased the expression of pro-apoptotic Bax, while markedly suppressed the phosphorylation of JNK and p38. In addition, increased expression of CHOP following combined treatment indicates that ER stress may also involve in the enhancement of HT-induced apoptosis. CONCLUSION Our data showed for the first time that OZ sensitizes Molt-4 cells to HT-induced apoptosis via extrinsic and intrinsic apoptotic pathways. Furthermore, ROS and ER stress may also play role in the enhancement of HT-induced apoptosis by OZ.
Collapse
Affiliation(s)
- Peng Li
- a Department of Radiological Sciences , Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama , Toyama , Japan
| | - Qing-Li Zhao
- a Department of Radiological Sciences , Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama , Toyama , Japan
| | - Paras Jawaid
- a Department of Radiological Sciences , Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama , Toyama , Japan
| | - Mati Ur Rehman
- a Department of Radiological Sciences , Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama , Toyama , Japan
| | - Kanwal Ahmed
- c Department of Basic Medical Sciences , College of Medicine, King Saud Bin Abdulaziz University of Health Sciences , Jeddah , Kingdom of Saudi Arabia
| | - Hiroaki Sakurai
- b Department of Cancer Cell Biology , Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama , Toyama , Japan
| | - Takashi Kondo
- a Department of Radiological Sciences , Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama , Toyama , Japan
| |
Collapse
|
25
|
Cao Y, Yu L, Dai G, Zhang S, Zhang Z, Gao T, Guo W. Cinobufagin induces apoptosis of osteosarcoma cells through inactivation of Notch signaling. Eur J Pharmacol 2016; 794:77-84. [PMID: 27845066 DOI: 10.1016/j.ejphar.2016.11.016] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2016] [Revised: 11/05/2016] [Accepted: 11/07/2016] [Indexed: 12/21/2022]
Abstract
Osteosarcoma (OS) is a major cause of cancer-related mortality in children and young adults worldwide. Due to preexisting or acquired chemoresistance, the current standard neoadjuvant chemotherapy regimens show only moderate activity against OS. In the current study, we explored the potential anti-OS Cinobufagin in vitro and in vivo, and investigated its underlying mechanisms. The antitumor potential of Cinobufagin was assessed using cell viability assays, and cycle and apoptosis were determined. In a cell-based assay, the mRNA and protein expression of Notch-1, Hes-1, Hes-5 and Hey-1 were determined by quantitative polymerase chain reactions and western blotting. The involvement of Notch signaling in Cinobufagin-induced apoptosis was confirmed using gain and loss-of function assays. A xenograft OS model was established and the antitumor effect and biosafety of Cinobufagin were evaluated. Cinobufagin suppressed OS cells growth in a dose- and time-dependent manner, involving both cell cycle arrest at the S phase and programmed cell death. Cinobufagin treatment decreased the expression of Notch-1, and Hes-1, Hes-5 and Hey-1 gene expression in OS cell lines. Furthermore, Notch activation attenuated the Cinobufagin-induced apoptosis, while Notch inhibition enhanced this effect. Using a mouse xenograft model, we found that Cinobufagin inhibited OS cell growth in vivo. The mice showed excellent tolerance to Cinobufagin treatment. Taken together, our data suggested that Cinobufagin inhibited cell survival and induced apoptosis in OS cells both in vitro and in vivo, and these effects were partly mediated through the Notch pathway.
Collapse
Affiliation(s)
- Yongfei Cao
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ling Yu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Guo Dai
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Shanshan Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zhengpei Zhang
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Tian Gao
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Department of Orthopedic Oncology, Peking University Cancer Hospital & Institute, Beijing, China
| | - Weichun Guo
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
26
|
Afroze SH, Sloan J, Osuji GAC, Drever N, Pilkinton K, Zawieja DC, Kuehl TJ, Nasir Uddin M. Cinobufotalin impedes Sw.71 cytotrophoblast cell line function via cell cycle arrest and apoptotic signaling. Mol Cell Biochem 2016; 422:189-196. [PMID: 27699589 DOI: 10.1007/s11010-016-2820-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 09/08/2016] [Indexed: 10/20/2022]
Abstract
Preeclampsia (preE) is a hypertensive disorder of pregnancy. Cardiotonic steroids (CTS) are endogenous inhibitors of Na+/K+ ATPase, and at least one CTS, marinobufagenin (MBG), is elevated in a rat model of preE prior to the development of the syndrome. MBG and ouabain impair cytotrophoblast (CTB) cell function, which is critical for placental development. We evaluated the effect of a CTS, cinobufotalin (CINO), on CTB cell function in vitro. CINO at ≥1 nM inhibited CTB cell proliferation, migration, and invasion (p < 0.05), but had no effect on cell viability. There was a higher (p < 0.05) percentage of G0/G1 phase cells in groups treated with CINO at ≥1 nM. CINO caused an increase in stress signaling p38 MAPK and a positive annexin-V staining in CTB cells, indicating the activation of apoptotic signaling. However, the CINO-induced apoptotic signaling was prevented by p38 inhibition. These data demonstrate that CINO impairs CTB cell function via cell cycle arrest and apoptotic signaling.
Collapse
Affiliation(s)
- Syeda H Afroze
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Temple, TX, USA
| | - Jenna Sloan
- Department of Obstetrics and Gynecology, Baylor Scott & White Health/Texas A&M Health Science Center College of Medicine, Scott & White Hospital (Building 1), Room #352, 2401 South 31st Street, Temple, TX, 76508, USA
| | - Grace-Ann C Osuji
- Department of Clinical Pathology, Baylor Scott & White Health, Temple, TX, USA
| | - Nathan Drever
- Department of Obstetrics and Gynecology, Baylor Scott & White Health/Texas A&M Health Science Center College of Medicine, Scott & White Hospital (Building 1), Room #352, 2401 South 31st Street, Temple, TX, 76508, USA
| | - Kimberly Pilkinton
- Department of Obstetrics and Gynecology, Baylor Scott & White Health/Texas A&M Health Science Center College of Medicine, Scott & White Hospital (Building 1), Room #352, 2401 South 31st Street, Temple, TX, 76508, USA
| | - David C Zawieja
- Department of Medical Physiology, Texas A&M Health Science Center College of Medicine, Temple, TX, USA
| | - Thomas J Kuehl
- Department of Obstetrics and Gynecology, Baylor Scott & White Health/Texas A&M Health Science Center College of Medicine, Scott & White Hospital (Building 1), Room #352, 2401 South 31st Street, Temple, TX, 76508, USA
- Department of Pediatrics, Baylor Scott & White Health/Texas A&M Health Science Center College of Medicine, Temple, TX, USA
| | - M Nasir Uddin
- Department of Obstetrics and Gynecology, Baylor Scott & White Health/Texas A&M Health Science Center College of Medicine, Scott & White Hospital (Building 1), Room #352, 2401 South 31st Street, Temple, TX, 76508, USA.
- Department of Pediatrics, Baylor Scott & White Health/Texas A&M Health Science Center College of Medicine, Temple, TX, USA.
- Department of Internal Medicine, Baylor Scott & White Health/Texas A&M Health Science Center College of Medicine, Temple, TX, USA.
| |
Collapse
|
27
|
Meng Q, Yau LF, Lu JG, Wu ZZ, Zhang BX, Wang JR, Jiang ZH. Chemical profiling and cytotoxicity assay of bufadienolides in toad venom and toad skin. JOURNAL OF ETHNOPHARMACOLOGY 2016; 187:74-82. [PMID: 27063985 DOI: 10.1016/j.jep.2016.03.062] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Revised: 03/16/2016] [Accepted: 03/30/2016] [Indexed: 06/05/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Toad venom and toad skin have been widely used for treating various cancers in China. Bufadienolides are regarded as the main anticancer components of toad venom, but the difference on composition and anticancer activities of bufadienolides between toad venom and toad skin remains unclear. METHODS Fractions enriched with free and conjugated bufadienolides were prepared from toad venom and toad skin. Bufadienolides in each fraction were comprehensively profiled by using a versatile UHPLC-TOF-MS method. Relative contents of major bufadienolides were determined by using three bufogenins and one bufotoxin as marker compounds with validated UHPLC-TOF-MS method. Furthermore, cytotoxicity of the fractions was examined by MTT assay. RESULTS Two fractions, i.e., bufogenin and bufotoxin fractions (TV-F and TV-C) were isolated from toad venom, and one bufotoxin fraction (TS-C) was isolated from toad skin. Totally 56 bufadienolides in these three fractions were identified, and 29 were quantified or semi-quantified. Bufotoxins were identified in both toad venom and toad skin, whereas bufogenins exist only in toad venom. Bufalin-3-conjugated bufotoxins are major components in toad venom, whereas cinobufotalin and cinobufagin-3-conjugated bufotoxins are main bufotoxins in toad skin. MTT assay revealed potent cytotoxicity of all the fractions in an order of TV-F>TV-C>TS-C. CONCLUSIONS Our study represents the most comprehensive investigation on the chemical profiles of toad venom and toad skin from both qualitative and quantitative aspects. Eight bufotoxins were identified in toad skin responsible for the cytotoxicity for the first time. Our research provides valuable chemical evidence for the appropriate processing method, quality control and rational exploration of toad skin and toad venom for the development of anticancer medicines.
Collapse
Affiliation(s)
- Qiong Meng
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Lee-Fong Yau
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Jing-Guang Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China
| | - Zhen-Zhen Wu
- Beijing Increase Pharm. Co. Ltd., Beijing, China
| | | | - Jing-Rong Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China.
| | - Zhi-Hong Jiang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau Institute for Applied Research in Medicine and Health, Macau University of Science and Technology, Macau, China.
| |
Collapse
|
28
|
Cytotoxic L-amino-acid oxidases from Amanita phalloides and Clitocybe geotropa induce caspase-dependent apoptosis. Cell Death Discov 2016; 2:16021. [PMID: 27551514 PMCID: PMC4979486 DOI: 10.1038/cddiscovery.2016.21] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 02/05/2016] [Accepted: 02/14/2016] [Indexed: 11/26/2022] Open
Abstract
L-amino-acid oxidases (LAO) purified from fungi induce cell death in various mammalian cells including human tumor cell lines. The mechanism, however, remains poorly understood. In this study, we aimed to define a precise mechanism of cell death induced in Jurkat and MCF7 cancer cell lines by ApLAO and CgLAO, LAOs isolated from Amanita phalloides and Clitocybe geotropa, respectively. Cell death induced by both LAOs is shown to be concentration- and time-dependent, with higher toxic effects in Jurkat cells. LAO activity is required for the cytotoxicity. Detailed study on Jurkat cells further demonstrated that ApLAO and CgLAO both induce the intrinsic mitochondrial pathway of apoptosis, accompanied by a time-dependent depolarization of the mitochondrial membrane through the generation of reactive oxygen species. Treatment with the LAOs resulted in an increased ratio of the expression of proapoptotic Bax to that of antiapoptotic Bcl-2, subsequently leading to the activation of caspase-9 and -3. However, the pancaspase inhibitor, Z-VAD-FMK, did not completely abolish the cell death induced by either ApLAO or CgLAO, suggesting an alternative pathway for LAO-induced apoptosis. Indeed, caspase-8 activity in ApLAO- and CgLAO-treated cells was increased. Further, Fas/FasL (Fas ligand) antagonist caused a slight reduction in toxin-induced cell death, supporting the involvement of ApLAO and CgLAO in death-receptor-mediated apoptosis. These results thus provide new evidence that ApLAO and CgLAO induce apoptosis in Jurkat cells via both the intrinsic and extrinsic pathways, although the significantly higher increase of caspase-9 over caspase-8 activity suggests that it is the intrinsic pathway that is the predominant mode of ApLAO- and CgLAO-induced apoptosis.
Collapse
|
29
|
New cytotoxic 19-norbufadienolide and bufogargarizin isolated from Chan Su. Fitoterapia 2015; 104:1-6. [DOI: 10.1016/j.fitote.2015.05.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/12/2015] [Accepted: 05/14/2015] [Indexed: 01/09/2023]
|
30
|
Cheng L, Chen YZ, Peng Y, Yi N, Gu XS, Jin Y, Bai XM. Ceramide production mediates cinobufotalin-induced growth inhibition and apoptosis in cultured hepatocellular carcinoma cells. Tumour Biol 2015; 36:5763-71. [PMID: 25724183 DOI: 10.1007/s13277-015-3245-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 02/10/2015] [Indexed: 12/21/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a highly aggressive and lethal neoplasm with poor prognosis. The aim of this study is to investigate the anticancer activity of cinobufotalin, a bufadienolide isolated from toad venom, in cultured HCC cells, and to study the underlying mechanisms. We found that cinobufotalin (at nmol/L) significantly inhibited HCC cell growth and survival while inducing considerable cell apoptosis. Further, cinobufotalin inhibited sphingosine kinase 1 (SphK1) activity and induced pro-apoptotic ceramide production. Ceramide synthase-1 small hairpin RNA (shRNA)-depletion inhibited cinobufotalin-induced ceramide production and HCC cell apoptosis. On the other hand, the glucosylceramide synthase (GCS) inhibitor 1-phenyl-2-decanoylamino-3-morpholino-1-propanol (PDMP) facilitated cinobufotalin-induced ceramide production and cell apoptosis. SphK1 inhibitor II (SKI-II), similar to cinobufotalin, increased cellular ceramide level and promoted HCC cell apoptosis. Finally, we observed that cinobufotalin inactivated Akt-S6K1 signaling in HepG2 cells, which was again inhibited by ceramide synthase-1 shRNA-depletion. In conclusion, the results of this study suggest that cinobufotalin induces growth inhibition and apoptosis in cultured HCC cells through ceramide production. Cinobufotalin may be investigated as a novel anti-HCC agent.
Collapse
Affiliation(s)
- Long Cheng
- Department of Interventional Radiology, The Second Affiliated Hospital of Soochow University, Soochow University, Suzhou, 215001, China
| | | | | | | | | | | | | |
Collapse
|
31
|
Li P, Zhao QL, Wu LH, Jawaid P, Jiao YF, Kadowaki M, Kondo T. Isofraxidin, a potent reactive oxygen species (ROS) scavenger, protects human leukemia cells from radiation-induced apoptosis via ROS/mitochondria pathway in p53-independent manner. Apoptosis 2014; 19:1043-53. [PMID: 24692054 DOI: 10.1007/s10495-014-0984-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Ionizing radiation (IR) leads to oxidizing events such as excessive reactive oxygen species (ROS) in the exposed cells, resulting in further oxidative damage to lipids, proteins and DNA. To screen the potential radio-protective drug, the intracellular ROS was measured in irradiated U937 cells pretreated with 80 candidate traditional herbal medicine, respectively. Isofraxidin (IF) was one possible radio-protector in these 80 drugs. This study investigated the radio-protective role of IF, a Coumarin compound, in human leukemia cell lines, for the first time. Results indicate that IF protects against IR-induced apoptosis in U937 cells in the time- and concentration- dependent manner. IF decreases IR-induced intracellular ROS generation, especially hydroxyl radicals formation, inhibits IR-induced mitochondrial membrane potential loss and reduces IR-induced high intracellular Ca(2+) levels regardless of ER stress. IF down-regulates the expression of caspase-3, phospho-JNK, phospho-p38 and activates Bax in mitochondria. IF inhibits cytochrome c release from mitochondria to cytosol. IF also moderates IR-induced Fas externalization and caspase-8 activation. IF also exhibits significant protection against IR-induced cell death in other leukemia cell lines such as Molt-4 cells and HL60 cells regardless of p53. Taken together, the data demonstrate that IF protects leukemia cells from radiation-induced apoptosis via ROS/mitochondria pathway in a p53-independent manner.
Collapse
Affiliation(s)
- Peng Li
- Department of Radiological Sciences, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, Sugitani 2630, Toyama, 930-0194, Japan
| | | | | | | | | | | | | |
Collapse
|
32
|
Kai S, Lu JH, Hui PP, Zhao H. Pre-clinical evaluation of cinobufotalin as a potential anti-lung cancer agent. Biochem Biophys Res Commun 2014; 452:768-74. [PMID: 25201730 DOI: 10.1016/j.bbrc.2014.08.147] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Accepted: 08/30/2014] [Indexed: 10/24/2022]
Abstract
Lung cancer is a major cause of cancer-related mortality in the United States and around the world. Due to the pre-existing or acquired chemo-resistance, the current standard chemotherapy regimens only show moderate activity against lung cancer. In the current study, we explored the potential anti-lung cancer activity of cinobufotalin in vivo and in vitro, and studied the underlying mechanisms. We demonstrated that cinobufotalin displayed considerable cytotoxicity against lung cancer cells (A549, H460 and HTB-58 lines) without inducing significant cell apoptosis. Our data suggest that mitochondrial protein cyclophilin D (Cyp-D)-dependent mitochondrial permeability transition pore (mPTP) opening mediates cinobufotalin-induced non-apoptotic death of lung cancer cells. The Cyp-D inhibitor cyclosporine A (CsA), the mPTP blocker sanglifehrin A (SfA), and Cyp-D shRNA-silencing significantly inhibited cinobufotalin-induced mitochondrial membrane potential (MMP) reduction and A549 cell death (but not apoptosis). Using a mice xenograft model, we found that cinobufotalin inhibited A549 lung cancer cell growth in vivo. Thus, cinobufotalin mainly induces Cyp-D-dependent non-apoptotic death in cultured lung cancer cells. The results of this study suggest that cinobufotalin might be further investigated as a novel anti-lung cancer agent.
Collapse
Affiliation(s)
- Sheng Kai
- Department of Geriatrics, Shanghai Changning Center Hospital, Shanghai 200336, China
| | - Jia-Huan Lu
- Department of Geriatrics, Shanghai Changning Center Hospital, Shanghai 200336, China
| | - Ping-Ping Hui
- Department of Geriatrics, Shanghai Changning Center Hospital, Shanghai 200336, China
| | - Hui Zhao
- Department of Geriatrics, Shanghai Changning Center Hospital, Shanghai 200336, China.
| |
Collapse
|
33
|
TRAIL combinations: The new 'trail' for cancer therapy (Review). Oncol Lett 2014; 7:1327-1332. [PMID: 24765133 PMCID: PMC3997674 DOI: 10.3892/ol.2014.1922] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 01/21/2014] [Indexed: 12/11/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) therapy is anticipated to be one of the most effective cancer treatments. However, resistance to TRAIL therapy remains a challenge facing the development of anticancer strategies. To circumvent this problem, TRAIL combinations have been experimented with for over ten years to induce synergism or sensitize resistant cancer cells. By analyzing the signaling pathways triggered by these combinations, this review has defined a set of core targets for novel combinatorial treatments. The review suggests specific pathways to be targeted together with TRAIL for more efficient treatment, including cellular FLICE inhibitory protein and its downstream survival factors, the Bcl-2 family and other prominent targets. The suggested pathways provide new avenues for more effective TRAIL-based cancer therapy.
Collapse
|
34
|
Faião-Flores F, Suarez JAQ, Soto-Cerrato V, Espona-Fiedler M, Pérez-Tomás R, Maria DA. Bcl-2 family proteins and cytoskeleton changes involved in DM-1 cytotoxic effect on melanoma cells. Tumour Biol 2013; 34:1235-43. [PMID: 23341182 DOI: 10.1007/s13277-013-0666-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 01/11/2013] [Indexed: 01/08/2023] Open
Abstract
Melanoma is one of the most aggressive types of skin cancer and its incidence rate is still increasing. All existing treatments are minimally effective. Consequently, new therapeutic agents for melanoma treatment should be developed. The DM-1 compound is a curcumin analog that possesses several curcumin characteristics, such as antiproliferative, antitumor, and anti-metastatic properties. The aim of this study was to evaluate the different signaling pathways involved in the cytotoxic effect of DM-1 on melanoma cells. The apoptotic process and cytoskeletal changes were evaluated by immunoblotting and immunofluorescence, respectively, in melanoma cells. After DM-1 treatment, SK-MEL-5 melanoma cells showed actin filament disorganization with spicule formation throughout the cytoskeleton and significant reduction of focal adhesion as well as they were present only at cell extremities, conferring a poor connection between the cell and the substrate. Besides this, there was significant filopodium retraction and loss of typical cytoskeleton scaffold. These modifications contributed to cell detachment followed by cell death. Furthermore, DM-1-induced apoptosis was triggered by multiple Bcl-2 proteins involved in both the extrinsic and the intrinsic apoptotic pathways. SK-MEL-5 cells showed a death mechanism mainly by Bcl-2/Bax ratio decrease, whereas A375 cells presented apoptosis induction by Mcl-1 and Bcl-xL downregulation. In SK-MEL-5 and A375 melanoma cells, there was a significant increase in the active form of caspase 9, and the inactive form of the effector caspase 3 was decreased in both cell lines. Expression of cleaved poly ADP ribose polymerase was increased after DM-1 treatment in these melanoma cell lines, demonstrating that the apoptotic process occurred. Altogether, these data elucidate the cellular and molecular mechanisms involved in the cytotoxicity induced by the antitumor agent DM-1 in melanoma cells.
Collapse
Affiliation(s)
- Fernanda Faião-Flores
- Laboratory of Biochemistry and Biophysics, Butantan Institute, 1500 Vital Brasil Avenue, São Paulo, 05503-900, Brazil.
| | | | | | | | | | | |
Collapse
|