1
|
He J, Qiu Z, Fan J, Xie X, Sheng Q, Sui X. Drug tolerant persister cell plasticity in cancer: A revolutionary strategy for more effective anticancer therapies. Signal Transduct Target Ther 2024; 9:209. [PMID: 39138145 PMCID: PMC11322379 DOI: 10.1038/s41392-024-01891-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 05/21/2024] [Accepted: 06/03/2024] [Indexed: 08/15/2024] Open
Abstract
Non-genetic mechanisms have recently emerged as important drivers of anticancer drug resistance. Among these, the drug tolerant persister (DTP) cell phenotype is attracting more and more attention and giving a predominant non-genetic role in cancer therapy resistance. The DTP phenotype is characterized by a quiescent or slow-cell-cycle reversible state of the cancer cell subpopulation and inert specialization to stimuli, which tolerates anticancer drug exposure to some extent through the interaction of multiple underlying mechanisms and recovering growth and proliferation after drug withdrawal, ultimately leading to treatment resistance and cancer recurrence. Therefore, targeting DTP cells is anticipated to provide new treatment opportunities for cancer patients, although our current knowledge of these DTP cells in treatment resistance remains limited. In this review, we provide a comprehensive overview of the formation characteristics and underlying drug tolerant mechanisms of DTP cells, investigate the potential drugs for DTP (including preclinical drugs, novel use for old drugs, and natural products) based on different medicine models, and discuss the necessity and feasibility of anti-DTP therapy, related application forms, and future issues that will need to be addressed to advance this emerging field towards clinical applications. Nonetheless, understanding the novel functions of DTP cells may enable us to develop new more effective anticancer therapy and improve clinical outcomes for cancer patients.
Collapse
Affiliation(s)
- Jun He
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Zejing Qiu
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Jingjing Fan
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Xiaohong Xie
- Department of Breast Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| | - Qinsong Sheng
- Department of Colorectal Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Xinbing Sui
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| |
Collapse
|
2
|
Yang S, Li Z, Yi J, Pan M, Cao W, Ma J, Zhang P. Nebivolol, an antihypertensive agent, has new application in inhibiting melanoma. Anticancer Drugs 2024; 35:512-524. [PMID: 38602174 PMCID: PMC11078289 DOI: 10.1097/cad.0000000000001597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 01/04/2024] [Indexed: 04/12/2024]
Abstract
Repurposing existing drugs for cancer therapy has become an important strategy because of its advantages, such as cost reduction, effect and safety. The present study was designed to investigate the antimelanoma effect and possible mechanisms of action of nebivolol, which is an approved and widely prescribed antihypertensive agent. In this study, we explored the effect of nebivolol on cell proliferation and cell activity in melanoma in vitro and the potential antimelanoma mechanism of nebivolol through a series of experiments, including the analysis of the effects with regard to cell apoptosis and metastasis. Furthermore, we evaluated the antimelanoma effect on xenograft tumor models and inspected the antimelanoma mechanism of nebivolol in vivo using immunohistochemical and immunofluorescence staining assays. As results in this work, in vitro , nebivolol possessed a strong activity for suppression proliferation and cell cycle arrest on melanoma. Moreover, nebivolol significantly induced cell apoptosis in melanoma through a mitochondrial-mediated endogenous apoptosis pathway. Additionally, nebivolol inhibited melanoma cell metastasis. More importantly, nebivolol exhibited significantly effective melanoma xenograft models in vivo , which related to the mechanism of apoptosis induction, proliferation inhibition, metastasis blocking and angiogenesis arrest. Overall, the data of the present study recommend that nebivolol holds great potential in application as a novel agent for the treatment of melanoma.
Collapse
Affiliation(s)
- Shuping Yang
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong
| | - Zhi Li
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong
| | - Jiamei Yi
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong
| | - Mingyue Pan
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong
| | - Weiling Cao
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong
| | - Jing Ma
- Department of Pharmacy, South China Hospital, Medical School, Shenzhen University, Shenzhen, China
| | - Peng Zhang
- Department of Pharmacy, Shenzhen Luohu People’s Hospital, Shenzhen, Guangdong
| |
Collapse
|
3
|
Hussain T, Alafnan A, Almazni IA, Helmi N, Moin A, Baeissa HM, Awadelkareem AM, Elkhalifa AO, Bakhsh T, Alzahrani A, Alghamdi RM, Khalid M, Tiwari RK, Rizvi SMD. Aloe-emodin exhibits growth-suppressive effects on androgen-independent human prostate cancer DU145 cells via inhibiting the Wnt/β-catenin signaling pathway: an in vitro and in silico study. Front Pharmacol 2024; 14:1325184. [PMID: 38348349 PMCID: PMC10859413 DOI: 10.3389/fphar.2023.1325184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/28/2023] [Indexed: 02/15/2024] Open
Abstract
At the molecular level, several developmental signaling pathways, such as Wnt/β-catenin, have been associated with the initiation and subsequent progression of prostate carcinomas. The present report elucidated the anti-cancerous attributes of an anthraquinone, aloe-emodin (AE), against androgen-independent human prostate cancer DU145 cells. The cytotoxicity profiling of AE showed that it exerted significant cytotoxic effects and increased lactose dehydrogenase levels in DU145 cells (p < 0.01 and p < 0.001). AE also induced considerable reactive oxygen species (ROS)-mediated oxidative stress, which escalated at higher AE concentrations of 20 and 25 μM. AE also efficiently instigated nuclear fragmentation and condensation concomitantly, followed by the activation of caspase-3 and -9 within DU145 cells. AE further reduced the viability of mitochondria with increased cytosolic cytochrome-c levels (p < 0.01 and p < 0.001) in DU145 cells. Importantly, AE exposure was also correlated with reduced Wnt2 and β-catenin mRNA levels along with their target genes, including cyclin D1 and c-myc. Furthermore, the molecular mechanism of AE was evaluated by performing molecular docking studies with Wnt2 and β-catenin. Evidently, AE exhibited good binding energy scores toward Wnt2 and β-catenin comparable with their respective standards, CCT036477 (Wnt2 inhibitor) and FH535 (β-catenin inhibitor). Thus, it may be considered that AE was competent in exerting anti-growth effects against DU145 androgen-independent prostate cancer cells plausibly by modulating the expression of Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Talib Hussain
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Ha’il, Ha’il, Saudi Arabia
| | - Ahmed Alafnan
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Ha’il, Ha’il, Saudi Arabia
| | - Ibrahim Abdullah Almazni
- Department of Clinical Laboratory Sciences, Faculty of Applied Medical Sciences, Najran University, Najran, Saudi Arabia
| | - Nawal Helmi
- Department of Biochemistry, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Afrasim Moin
- Department of Pharmaceutics, College of Pharmacy, University of Ha’il, Ha’il, Saudi Arabia
| | - Hanadi M. Baeissa
- Department of Biological Science, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Amir Mahgoub Awadelkareem
- Department of Clinical Nutrition, College of Applied Medical Sciences, University of Hail, Ha’il, Saudi Arabia
| | - AbdElmoneim O. Elkhalifa
- Department of Clinical Nutrition, College of Applied Medical Sciences, University of Hail, Ha’il, Saudi Arabia
| | - Tahani Bakhsh
- Department of Biological Science, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Abdulrahman Alzahrani
- Department of Applied Medical Sciences, Applied College, Al-Baha University, Al-Baha, Saudi Arabia
| | - Rashed Mohammed Alghamdi
- Department of Laboratory Medicine, Faculty of Applied College, Al-Baha University, Al-Baha, Saudi Arabia
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Rohit Kumar Tiwari
- Department of Clinical Research, Sharda School of Allied Health Sciences, Sharda University, Gautam Buddh Nagar, India
| | - Syed Mohd Danish Rizvi
- Department of Pharmaceutics, College of Pharmacy, University of Ha’il, Ha’il, Saudi Arabia
| |
Collapse
|
4
|
Semlali A, Beji S, Ajala I, Al-Zharani M, Rouabhia M. Synergistic Effects of New Curcumin Analog (PAC) and Cisplatin on Oral Cancer Therapy. Curr Issues Mol Biol 2023; 45:5018-5035. [PMID: 37367068 DOI: 10.3390/cimb45060319] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/02/2023] [Accepted: 06/05/2023] [Indexed: 06/28/2023] Open
Abstract
Oral cancer has traditionally been treated with surgery, radiotherapy, chemotherapy, or a combination of these therapies. Although cisplatin, a chemotherapy drug, can effectively kill oral cancer cells by forming DNA adducts, its clinical use is limited due to adverse effects and chemo-resistance. Therefore, there is a need to develop new, targeted anticancer drugs to complement chemotherapy, allowing for reduced cisplatin doses and minimizing adverse effects. Recent studies have shown that 3,5-Bis (4-hydroxy-3-methoxybenzylidene)-N-methyl-4-piperidine (PAC), a new curcumin analog, possesses anticancer properties and could be considered a complementary or alternative therapy. In this study, we aimed to assess the potential complementary effects of PAC in combination with cisplatin for treating oral cancer. We conducted experiments using oral cancer cell lines (Ca9-22) treated with different concentrations of cisplatin (ranging from 0.1 μM to 1 μM), either alone or in conjunction with PAC (2.5 and 5 μM). Cell growth was measured using the MTT assay, while cell cytotoxicity was evaluated using an LDH assay. Propidium iodide and annexin V staining were employed to examine the impact on cell apoptosis. Flow cytometry was used to investigate the effects of the PAC/cisplatin combination on cancer cell autophagy, oxidative stress, and DNA damage. Additionally, a Western Blot analysis was performed to assess the influence of this combination on pro-carcinogenic proteins involved in various signaling pathways. The results demonstrated that PAC enhanced the efficacy of cisplatin in a dose-dependent manner, leading to a significant inhibition of oral cancer cell proliferation. Importantly, treatment with PAC (5 μM) alongside different concentrations of cisplatin reduced the IC50 of cisplatin tenfold. Combining these two agents increased apoptosis by further inducing caspase activity. In addition, the concomitant use of PAC and cisplatin enhances oral cancer cell autophagy, ROS, and MitoSOX production. However, combined PAC with cisplatin inhibits the mitochondrial membrane potential (ΔΨm), which is a marker for cell viability. Finally, this combination further enhances the inhibition of oral cancer cell migration via the inhibition of epithelial-to-mesenchymal transition genes, such as E-cadherin. We demonstrated that the combination of PAC and cisplatin markedly enhanced oral cancer cell death by inducing apoptosis, autophagy, and oxidative stress. The data presented indicate that PAC has the potential to serve as a powerful complementary agent to cisplatin in the treatment of gingival squamous cell carcinomas.
Collapse
Affiliation(s)
- Abdelhabib Semlali
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V0A6, Canada
| | - Sarra Beji
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V0A6, Canada
| | - Ikram Ajala
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V0A6, Canada
| | - Mohammed Al-Zharani
- Biology Department, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh 11623, Saudi Arabia
| | - Mahmoud Rouabhia
- Groupe de Recherche en Écologie Buccale, Faculté de Médecine Dentaire, Université Laval, Québec, QC G1V0A6, Canada
| |
Collapse
|
5
|
Li R, Zheng C, Shiu PHT, Rangsinth P, Wang W, Kwan YW, Wong ESW, Zhang Y, Li J, Leung GPH. Garcinone E triggers apoptosis and cell cycle arrest in human colorectal cancer cells by mediating a reactive oxygen species–dependent JNK signaling pathway. Biomed Pharmacother 2023; 162:114617. [PMID: 37001180 DOI: 10.1016/j.biopha.2023.114617] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 03/31/2023] Open
Abstract
Despite various therapeutic approaches, colorectal cancer is among the most fatal diseases globally. Hence, developing novel and more effective methods for colorectal cancer treatment is essential. Recently, reactive oxygen species (ROS)/JNK signaling pathway has been proposed as the potential target for the anticancer drug discovery. The present study investigated the anticancer effects of the bioactive xanthone garcinone E (GAR E) in mangosteen and explored its underlying mechanism of action. HT-29 and Caco-2 cancer cells were used as in vitro models to study the anticancer effect of GAR E. The findings demonstrated that GAR E inhibited colony formation and wound healing, whereas triggered the production of ROS, which induced mitochondrial dysfunction and apoptosis, causing cell cycle arrest at the Sub G1 phase. Additionally, GAR E treatment elevated the ratio of Bax/Bcl-2 and activated PARP, caspases 3 and 9, and JNK1/2. These GAR E-induced cytotoxic activities and expression of signaling proteins were reversed by the antioxidant N-acetyl-L-cysteine and JNK inhibitor SP600125, indicating the involvement of ROS/JNK signaling pathways. In vivo experiments using an HT-29 xenograft nude mouse model also demonstrated the antitumor effect of GAR E. In conclusion, our findings showed that GAR E might be potentially effective in treating colorectal cancer and provided insights into the development of xanthones as novel chemotherapeutic agents.
Collapse
Affiliation(s)
- Renkai Li
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Chengwen Zheng
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Polly Ho-Ting Shiu
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Panthakarn Rangsinth
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Wen Wang
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Yiu-Wa Kwan
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Emily Sze-Wan Wong
- Department of Applied Science, School of Science and Technology, Hong Kong Metropolitan University, Hong Kong Special Administrative Region of China
| | - Yanbo Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China
| | - Jingjing Li
- Department of Rehabilitation Sciences, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region of China.
| | - George Pak-Heng Leung
- Department of Pharmacology and Pharmacy, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region of China.
| |
Collapse
|
6
|
Guo K, Liu R, Jing R, Wang L, Li X, Zhang K, Fu M, Ye J, Hu Z, Zhao W, Xu N. Cryptotanshinone protects skin cells from ultraviolet radiation-induced photoaging via its antioxidant effect and by reducing mitochondrial dysfunction and inhibiting apoptosis. Front Pharmacol 2022; 13:1036013. [PMID: 36386220 PMCID: PMC9640529 DOI: 10.3389/fphar.2022.1036013] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/13/2022] [Indexed: 11/25/2022] Open
Abstract
The integrity of skin tissue structure and function plays an important role in maintaining skin rejuvenation. Ultraviolet (UV) radiation is the main environmental factor that causes skin aging through photodamage of the skin tissue. Cryptotanshinone (CTS), an active ingredient mianly derived from the Salvia plants of Lamiaceae, has many pharmacological effects, such as anti-inflammatory, antioxidant, and anti-tumor effects. In this study, we showed that CTS could ameliorate the photodamage induced by UV radiation in epidermal keratinocytes (HaCaT) and dermal fibroblasts (HFF-1) when applied to the cells before exposure to the radiation, effectively delaying the aging of the cells. CTS exerted its antiaging effect by reducing the level of reactive oxygen species (ROS) in the cells, attenuating DNA damage, activating the nuclear factor E2-related factor 2 (Nrf2) signaling pathway, and reduced mitochondrial dysfunction as well as inhibiting apoptosis. Further, CTS could promote mitochondrial biosynthesis in skin cells by activating the AMP-activated protein kinase (AMPK)/sirtuin-1 (SIRT1)/peroxisome proliferator-activated receptor-γ co-activator-1α (PGC-1α) signaling pathway. These findings demonstrated the protective effects of CTS against UV radiation-induced skin photoaging and provided a theoretical and experimental basis for the application of CTS as an anti-photodamage and anti-aging agent for the skin.
Collapse
Affiliation(s)
- Keke Guo
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Run Liu
- Zhiyuan College, Shanghai Jiao Tong University, Shanghai, China
| | - Rongrong Jing
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Lusheng Wang
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Xuenan Li
- Department of Pharmacy, Zhoupu Hospital, Shanghai, China
| | - Kaini Zhang
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Mengli Fu
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Jiabin Ye
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
| | - Zhenlin Hu
- School of Medicine, Shanghai University, Shanghai, China
| | - Wengang Zhao
- Institute of Life Sciences, Wenzhou University, Wenzhou, China
- *Correspondence: Wengang Zhao, ; Nuo Xu,
| | - Nuo Xu
- College of Life and Environmental Sciences, Wenzhou University, Wenzhou, China
- *Correspondence: Wengang Zhao, ; Nuo Xu,
| |
Collapse
|
7
|
Lu M, Lan X, Wu X, Fang X, Zhang Y, Luo H, Gao W, Wu D. Salvia miltiorrhiza in cancer: Potential role in regulating MicroRNAs and epigenetic enzymes. Front Pharmacol 2022; 13:1008222. [PMID: 36172186 PMCID: PMC9512245 DOI: 10.3389/fphar.2022.1008222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Accepted: 08/24/2022] [Indexed: 11/21/2022] Open
Abstract
MicroRNAs are small non-coding RNAs that play important roles in gene regulation by influencing the translation and longevity of various target mRNAs and the expression of various target genes as well as by modifying histones and DNA methylation of promoter sites. Consequently, when dysregulated, microRNAs are involved in the development and progression of a variety of diseases, including cancer, by affecting cell growth, proliferation, differentiation, migration, and apoptosis. Preparations from the dried root and rhizome of Salvia miltiorrhiza Bge (Lamiaceae), also known as red sage or danshen, are widely used for treating cardiovascular diseases. Accumulating data suggest that certain bioactive constituents of this plant, particularly tanshinones, have broad antitumor effects by interfering with microRNAs and epigenetic enzymes. This paper reviews the evidence for the antineoplastic activities of S. miltiorrhiza constituents by causing or promoting cell cycle arrest, apoptosis, autophagy, epithelial-mesenchymal transition, angiogenesis, and epigenetic changes to provide an outlook on their future roles in the treatment of cancer, both alone and in combination with other modalities.
Collapse
Affiliation(s)
- Meng Lu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Xintian Lan
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Xi Wu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Xiaoxue Fang
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Yegang Zhang
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Haoming Luo
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
- Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
| | - Wenyi Gao
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
- *Correspondence: Wenyi Gao, ; Donglu Wu,
| | - Donglu Wu
- Key Laboratory of Effective Components of Traditional Chinese Medicine, Changchun, China
- School of Clinical Medical, Changchun University of Chinese Medicine, Changchun, China
- *Correspondence: Wenyi Gao, ; Donglu Wu,
| |
Collapse
|
8
|
Acquaviva R, Malfa GA, Loizzo MR, Xiao J, Bianchi S, Tundis R. Advances on Natural Abietane, Labdane and Clerodane Diterpenes as Anti-Cancer Agents: Sources and Mechanisms of Action. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27154791. [PMID: 35897965 PMCID: PMC9330018 DOI: 10.3390/molecules27154791] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/18/2022] [Accepted: 07/25/2022] [Indexed: 01/14/2023]
Abstract
Extensive research over the past decades has identified numerous phytochemicals that could represent an important source of anti-cancer compounds. There is an immediate need for less toxic and more effective preventive and therapeutic strategies for the treatment of cancer. Natural compounds are considered suitable candidates for the development of new anti-cancer drugs due to their pleiotropic actions on target events with multiple manners. This comprehensive review highlighted the most relevant findings achieved in the screening of phytochemicals for anticancer drug development, particularly focused on a promising class of phytochemicals such as diterpenes with abietane, clerodane, and labdane skeleton. The chemical structure of these compounds, their main natural sources, and mechanisms of action were critically discussed.
Collapse
Affiliation(s)
- Rosaria Acquaviva
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria, 95125 Catania, Italy; (R.A.); (S.B.)
- CERNUT, Research Centre on Nutraceuticals and Health Products, Department of Drug and Health Sciences, University of Catania, Viale A. Doria, 95125 Catania, Italy
| | - Giuseppe A. Malfa
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria, 95125 Catania, Italy; (R.A.); (S.B.)
- CERNUT, Research Centre on Nutraceuticals and Health Products, Department of Drug and Health Sciences, University of Catania, Viale A. Doria, 95125 Catania, Italy
- Correspondence:
| | - Monica R. Loizzo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.R.L.); (R.T.)
| | - Jianbo Xiao
- Nutrition and Bromatology Group, Department of Analytical and Food Chemistry, Faculty of Sciences, Universidade de Vigo, 32004 Ourense, Spain;
| | - Simone Bianchi
- Department of Drug and Health Sciences, University of Catania, Viale A. Doria, 95125 Catania, Italy; (R.A.); (S.B.)
| | - Rosa Tundis
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy; (M.R.L.); (R.T.)
| |
Collapse
|
9
|
Salviolone from Salvia miltiorrhiza Roots Impairs Cell Cycle Progression, Colony Formation, and Metalloproteinase-2 Activity in A375 Melanoma Cells: Involvement of P21(Cip1/Waf1) Expression and STAT3 Phosphorylation. Int J Mol Sci 2022; 23:ijms23031121. [PMID: 35163058 PMCID: PMC8835475 DOI: 10.3390/ijms23031121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/15/2022] [Accepted: 01/17/2022] [Indexed: 02/04/2023] Open
Abstract
Melanoma is a highly malignant solid tumor characterized by an elevated growth and propagation rate. Since, often, melanoma treatment cannot prevent recurrences and the appearance of metastasis, new anti-melanoma agents need to be discovered. Salvia miltiorrhiza roots are a source of diterpenoid derivatives, natural compounds with several biological activities, including antiproliferative and anticancer effects. Seven diterpenoid derivatives were purified from S. miltiorrhiza roots and identified by NMR and MS analysis. Tanshinone IIA and cryptotanshinone were detected as the main components of S. miltiorrhiza root ethanol extract. Although their antitumor activity is already known, they have been confirmed to induce a reduction in A375 and MeWo melanoma cell growth. Likewise, salviolone has been shown to impair the viability of melanoma cells without affecting the growth of normal melanocytes. The underlying anticancer activity of salviolone has been investigated and compared to that of cryptotanshinone in A375 cells, showing an increased P21 protein expression in a P53-dependent manner. In that way, salviolone, even more than cryptotanshinone, displays a multitarget effect on cell-cycle-related proteins. Besides, it modulates the phosphorylation level of the signal transducer and activator of transcription (STAT)3. Unexpectedly, salviolone and cryptotanshinone induce sustained activation of the extracellular signal-regulated kinases (ERK)1/2 and the protein kinase B (Akt). However, the blockage of ERK1/2 or Akt activities suggests that kinase activation does not hinder their ability to inhibit A375 cell growth. Finally, salviolone and cryptotanshinone inhibit to a comparable extent some crucial malignancy features of A375 melanoma cells, such as colony formation in soft agar and metalloproteinase-2 activity. In conclusion, it has been shown for the first time that salviolone, harboring a different molecular structure than tanshinone IIA and cryptotanshinone, exhibits a pleiotropic effect against melanoma by hampering cell cycle progression, STAT3 signaling, and malignant phenotype of A375 melanoma cells.
Collapse
|
10
|
Oliveira WN, Alencar EN, Rocha HAO, Amaral-Machado L, Egito EST. Nanostructured systems increase the in vitro cytotoxic effect of bullfrog oil in human melanoma cells (A2058). Biomed Pharmacother 2021; 145:112438. [PMID: 34861632 DOI: 10.1016/j.biopha.2021.112438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/06/2021] [Accepted: 11/12/2021] [Indexed: 11/17/2022] Open
Abstract
The aim of this work was to investigate the in vitro cytotoxic effect of previously developed nanocapsules, nanoemulsion, and microemulsion based on bullfrog oil (BFO) against human melanoma cells (A2058). The nanosystems were produced as described in previous studies and characterized according to droplet/particle distribution and zeta potential. The biocompatibility was evaluated by the determination of the hemolytic potential against human erythrocytes. The cytotoxicity assessment was based on MTT and cell death assays, determination of Reactive Oxygen Species (ROS) levels, and cell uptake. The nanosystems were successfully reproduced and showed hemolytic potential smaller than 10% at all oil concentrations (50 and 100 µg.mL-1) (p < 0.05). The MTT assay revealed that the nanosystems decreased the mitochondrial activity up to 92 ± 2% (p < 0.05). The study showed that the free BFO induced cell apoptosis, while all the nanostructured systems caused cell death by necrosis associated with a ROS overproduction. This can be related to the increased ability of the nanostructured systems to deliver the BFO across all cellular compartments (membrane, cytoplasm, and nucleus). Finally, these results elucidate the in vitro BFO nanosystems cytotoxic effect against human melanoma cells (A2058), revealing the emulsified ones as the most cytotoxic systems. Overall, the findings suggest that the safety and antineoplastic activity of these systems can be further investigated by in vivo studies.
Collapse
Affiliation(s)
- W N Oliveira
- Graduate Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, Rio Grande do Norte, Brazil
| | - E N Alencar
- Graduate Program in Pharmaceutical Nanotechnology, Federal University of Rio Grande do Norte (UFRN), Natal, Rio Grande do Norte, Brazil
| | - H A O Rocha
- Graduate Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, Rio Grande do Norte, Brazil
| | - L Amaral-Machado
- Graduate Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, Rio Grande do Norte, Brazil
| | - E S T Egito
- Graduate Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, Rio Grande do Norte, Brazil; Graduate Program in Pharmaceutical Nanotechnology, Federal University of Rio Grande do Norte (UFRN), Natal, Rio Grande do Norte, Brazil.
| |
Collapse
|
11
|
Antiproliferative effect of cryptotanshinone against human non-small cell lung cancer cells through inactivation of lncRNA HOTAIR /p-Akt signaling pathway. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2021.103150] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
12
|
Munakarmi S, Shrestha J, Shin HB, Lee GH, Jeong YJ. 3,3'-Diindolylmethane Suppresses the Growth of Hepatocellular Carcinoma by Regulating Its Invasion, Migration, and ER Stress-Mediated Mitochondrial Apoptosis. Cells 2021; 10:cells10051178. [PMID: 34066056 PMCID: PMC8151225 DOI: 10.3390/cells10051178] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/30/2021] [Accepted: 05/05/2021] [Indexed: 12/30/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the leading cause of cancer-related death worldwide with limited treatment options. Biomarker-based active phenolic flavonoids isolated from medicinal plants might shed some light on potential therapeutics for treating HCC. 3,3′-diindolylmethane (DIM) is a unique biologically active dimer of indole-3-carbinol (I3C), a phytochemical compound derived from Brassica species of cruciferous vegetables—such as broccoli, kale, cabbage, and cauliflower. It has anti-cancer effects on various cancers such as breast cancer, prostate cancer, endometrial cancer, and colon cancer. However, the molecular mechanism of DIM involved in reducing cancer risk and/or enhancing therapy remains unknown. The aim of the present study was to evaluate anti-cancer and therapeutic effects of DIM in human hepatoma cell lines Hep3B and HuhCell proliferation was measured with MTT and trypan blue colony formation assays. Migration, invasion, and apoptosis were measured with Transwell assays and flow cytometry analyses. Reactive oxygen species (ROS) intensity and the loss in mitochondrial membrane potential of Hep3B and Huh7 cells were determined using dihydroethidium (DHE) staining and tetramethylrhodamine ethyl ester dye. Results showed that DIM significantly suppressed HCC cell growth, proliferation, migration, and invasion in a concentration-dependent manner. Furthermore, DIM treatment activated caspase-dependent apoptotic pathway and suppressed epithelial–mesenchymal transition (EMT) via ER stress and unfolded protein response (UPR). Taken together, our results suggest that DIM is a potential anticancer drug for HCC therapy by targeting ER-stress/UPR.
Collapse
Affiliation(s)
- Suvesh Munakarmi
- Laboratory of Liver Regeneration, Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Korea;
| | - Juna Shrestha
- Alka Hospital Private Limited, Jwalakhel, Kathmandu 446010, Nepal;
| | - Hyun-Beak Shin
- Department of Surgery, Jeonbuk National University Hospital, Jeonju 54907, Korea;
| | - Geum-Hwa Lee
- Department of Pharmacology and New Drug Development Research Institute, Jeonbuk National University Hospital, Jeonju 54907, Korea;
| | - Yeon-Jun Jeong
- Laboratory of Liver Regeneration, Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Korea;
- Department of Surgery, Jeonbuk National University Hospital, Jeonju 54907, Korea;
- Correspondence:
| |
Collapse
|
13
|
Hu Y, Zhang X, Zhang J, Xia X, Li H, Qiu C, Liao Y, Chen H, He Z, Song Z, Zhou W. Activated STAT3 signaling pathway by ligature-induced periodontitis could contribute to neuroinflammation and cognitive impairment in rats. J Neuroinflammation 2021; 18:80. [PMID: 33757547 PMCID: PMC7986277 DOI: 10.1186/s12974-021-02071-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/02/2021] [Indexed: 12/11/2022] Open
Abstract
Background Increasing evidence suggests a causal link between periodontitis and cognitive disorders. Systemic inflammation initiated by periodontitis may mediate the development of cognitive impairment. Our study aims to investigate the effect of ligature-induced periodontitis on cognitive function and the role of signal transducers and activators of transcription 3 (STAT3) in this process. Materials and methods Ligature-induced periodontitis was established, and the rats were treated intraperitoneally with/without the pSTAT3 inhibitor cryptotanshinone (CTS). Alveolar bone resorption and periodontal inflammation were detected by micro-computed tomography analysis and histopathological evaluation. Locomotor activity and cognitive function were evaluated by the open field test and the Morris water maze test, respectively. The activation of microglia and astrocytes in the hippocampus and cortex was assessed by immunohistochemistry (IHC). The expression of interleukins (IL-1β, IL-6, IL-8, IL-21) in both the periphery and cortex was evaluated by RT-PCR and ELISA. The expression of TLR/NF-κB and ROS cascades was evaluated by RT-PCR. The expression of pSTAT3 and the activation of the STAT3 signaling pathway (JAK2, STAT3, and pSTAT3) in the periodontal tissue and cortex were assessed by IHC and Western blot. The expression of amyloid precursor protein (APP) and its key secretases was evaluated by RT-PCR. The level of amyloid β-protein (Aβ) and the ratio of Aβ1-40/1-42 were measured via ELISA in the plasma and cortex while IHC was used to detect the level of Aβ1-42 in the brain. Results In periodontal ligature rats, significant alveolar bone resorption and local inflammatory cell infiltration were present. Apparent increases in inflammatory cytokines (IL-1β, IL-6, IL-8, and IL-21) were detected in peripherial blood and brain. Additionally, spatial learning and memory ability was impaired, while locomotor activity was not affected. Activated microglia and astrocytes were found in the cortex and hippocampus, presenting as enlarged cell bodies and irregular protrusions. Levels of TLR/NF-kB, PPAR and ROS were altered. The STAT3 signaling pathway was activated in both the periodontal tissue and cortex, and the processing of APP by β- and γ-secretases was promoted. The changes mentioned above could be relieved by the pSTAT3 inhibitor CTS. Conclusions Ligature-induced periodontitis in rats resulted in systemic inflammation and further abnormal APP processing, leading to cognitive impairments. In this progress, the activation of the STAT3 signaling pathway may play an important role by increasing inflammatory load and promoting neuroinflammation. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02071-9.
Collapse
Affiliation(s)
- Yi Hu
- Department of Periodontology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Xu Zhang
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, 115 Jinzun Road, Shanghai, 200125, China
| | - Jing Zhang
- Department of Periodontology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Xinyi Xia
- Department of Periodontology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Huxiao Li
- Department of Periodontology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Che Qiu
- Department of Periodontology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Yue Liao
- Department of Periodontology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Huiwen Chen
- Department of Periodontology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, China
| | - Zhiyan He
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, 115 Jinzun Road, Shanghai, 200125, China
| | - Zhongchen Song
- Department of Periodontology, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases; Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, 639 Zhizaoju Road, Shanghai, 200011, China.
| | - Wei Zhou
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, 115 Jinzun Road, Shanghai, 200125, China.
| |
Collapse
|
14
|
Wang H, Pang W, Xu X, You B, Zhang C, Li D. Cryptotanshinone Attenuates Ischemia/Reperfusion-induced Apoptosis in Myocardium by Upregulating MAPK3. J Cardiovasc Pharmacol 2021; 77:370-377. [PMID: 33662979 DOI: 10.1097/fjc.0000000000000971] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 11/26/2020] [Indexed: 01/17/2023]
Abstract
ABSTRACT Chinese people have used the root of Salvia miltiorrhiza Bunge (called "Danshen" in Chinese) for centuries as an anticancer agent, anti-inflammatory agent, antioxidant, and cardiovascular disease drug. In addition, Danshen is considered to be a drug that can improve ischemia/reperfusion (I/R)-induced myocardium injury in traditional Chinese medicine. However, Danshen is a mixture that includes various bioactive substances. In this study, we aimed to identify the protective component and mechanism of Danshen on myocardium through network pharmacology and molecular simulation methods. First, cryptotanshinone (CTS) was identified as a potential active compound from Danshen that was associated with apoptosis by a network pharmacology approach. Subsequently, biological experiments validated that CTS inhibited ischemia/reperfusion-induced cardiomyocyte apoptosis in vivo and in vitro. Molecular docking techniques were used to screen key target information. Based on the simulative results, MAPKs were verified as well-connected molecules of CTS. Western blotting assays also demonstrated that CTS could enhance MAPK expression. Furthermore, we demonstrated that inhibition of the MAPK pathway reversed the CTS-mediated effect on cardiomyocyte apoptosis. Altogether, our work screened out CTS from Danshen and demonstrated that it protected cardiomyocytes from apoptosis.
Collapse
Affiliation(s)
- Hefeng Wang
- Department of Cardiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China
| | - Wenhui Pang
- Department of Otolaryngology Head and Neck Surgery, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xingsheng Xu
- Department of Cardiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China
| | - Beian You
- Department of Cardiology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, China
| | - Cuijuan Zhang
- Department of Cardiology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China; and
| | - Dan Li
- Department of Cardiology, the Affiliated Hospital of Qingdao University, Qingdao, Shandong, China; and
| |
Collapse
|
15
|
Bittner ML, Lopes R, Hua J, Sima C, Datta A, Wilson-Robles H. Comprehensive live-cell imaging analysis of cryptotanshinone and synergistic drug-screening effects in various human and canine cancer cell lines. PLoS One 2021; 16:e0236074. [PMID: 33544704 PMCID: PMC7864433 DOI: 10.1371/journal.pone.0236074] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 12/01/2020] [Indexed: 12/15/2022] Open
Abstract
Background Several studies have highlighted both the extreme anticancer effects of Cryptotanshinone (CT), a Stat3 crippling component from Salvia miltiorrhiza, as well as other STAT3 inhibitors to fight cancer. Methods Data presented in this experiment incorporates 2 years of in vitro studies applying a comprehensive live-cell drug-screening analysis of human and canine cancer cells exposed to CT at 20 μM concentration, as well as to other drug combinations. As previously observed in other studies, dogs are natural cancer models, given to their similarity in cancer genetics, epidemiology and disease progression compared to humans. Results Results obtained from several types of human and canine cancer cells exposed to CT and varied drug combinations, verified CT efficacy at combating cancer by achieving an extremely high percentage of apoptosis within 24 hours of drug exposure. Conclusions CT anticancer efficacy in various human and canine cancer cell lines denotes its ability to interact across different biological processes and cancer regulatory cell networks, driving inhibition of cancer cell survival.
Collapse
Affiliation(s)
- Michael L. Bittner
- Center for Bioinformatics and Genomic Systems Engineering, Texas A&M Engineering Experiment Station, Texas A&M University, College Station, TX, United States of America
- Translational Genomics Research Institute, Phoenix, AZ, United States of America
| | - Rosana Lopes
- Center for Bioinformatics and Genomic Systems Engineering, Texas A&M Engineering Experiment Station, Texas A&M University, College Station, TX, United States of America
- * E-mail: (RL); (HWR)
| | - Jianping Hua
- Center for Bioinformatics and Genomic Systems Engineering, Texas A&M Engineering Experiment Station, Texas A&M University, College Station, TX, United States of America
| | - Chao Sima
- Center for Bioinformatics and Genomic Systems Engineering, Texas A&M Engineering Experiment Station, Texas A&M University, College Station, TX, United States of America
| | - Aniruddha Datta
- Center for Bioinformatics and Genomic Systems Engineering, Texas A&M Engineering Experiment Station, Texas A&M University, College Station, TX, United States of America
| | - Heather Wilson-Robles
- College of Veterinary Medicine, Texas A&M University, College Station, TX, United States of America
- * E-mail: (RL); (HWR)
| |
Collapse
|
16
|
Li H, Gao C, Liu C, Liu L, Zhuang J, Yang J, Zhou C, Feng F, Sun C, Wu J. A review of the biological activity and pharmacology of cryptotanshinone, an important active constituent in Danshen. Biomed Pharmacother 2021; 137:111332. [PMID: 33548911 DOI: 10.1016/j.biopha.2021.111332] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/13/2021] [Accepted: 01/25/2021] [Indexed: 02/07/2023] Open
Abstract
Cryptotanshinone (IUPAC name: (R)-1,2,6,7,8,9-hexahydro-1,6,6-trimethyl-phenanthro(1,2-b)furan-10,11-dione), a biologically active constituent extracted from the roots and rhizomes of the plant Salvia miltiorrhiza, has been studied in depth as a medicinally active compound and shown to have efficacy in the treatment of numerous diseases and disorders. In this review, we describe in detail the current status of cryptotanshinone research, including findings relating to the structure, pharmacokinetics, pharmacological activity, and derivatives of this compound. Cryptotanshinoneh as a diverse range of pharmacological effects, including anti-cancer, anti-inflammatory, immune regulatory, neuroprotective, and anti-fibrosis activities. Studies on the molecular mechanisms underlying the activities of cryptotanshinone have established that the JAK2/STAT3, PI3K/AKT, NF-κB, AMPK, and cell cycle pathways are involved in the inhibitory and pro-apoptotic effects of cryptotanshinone on different tumor cell lines, these molecular pathways interact in a coordinated manner to inhibit cell proliferation, migration and invasion,and induce transformation, autophagy, necrosis, and cellular immunity. The anti-inflammatory mechanisms of cryptotanshinone have been found to be associated with the TLR4-MyD88/PI3K/Nrf2 and TLR4-MyD88/NF-κB/MAPK pathways, whereasthe Hedgehog, NF-κB, and Nrf-2/HO-1 pathways are regulated by cryptotanshinone to reduce organ fibrosis, and its inhibitory effects on the PI3K/AKT-eNOS pathway have been linked to neuroprotective effects. Given the potential medicinal utility of cryptotanshinone, further research is needed to verify the efficacy and safety of this compound in clinical use, evaluate its pharmacological activity, and identify molecular targets.
Collapse
Affiliation(s)
- Huayao Li
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, PR China.
| | - Chundi Gao
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, PR China.
| | - Cun Liu
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, PR China.
| | - Lijuan Liu
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261041, Shandong, PR China; Department of Basic Medical Science, Qingdao University, Qingdao, 266071, PR China.
| | - Jing Zhuang
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261041, Shandong, PR China; Qingdao Academy of Chinese Medical Sciences, Shandong University of Chinese Medicine, Qingdao, 266112, Shandong, PR China.
| | - Jing Yang
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261041, Shandong, PR China.
| | - Chao Zhou
- College of First Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, PR China; Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261041, Shandong, PR China.
| | - Fubin Feng
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261041, Shandong, PR China; Department of Basic Medical Science, Qingdao University, Qingdao, 266071, PR China.
| | - Changgang Sun
- Departmen of Oncology, Weifang Traditional Chinese Hospital, Weifang, 261041, Shandong, PR China; Chinese Medicine Innovation Institute, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, PR China.
| | - Jibiao Wu
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, PR China.
| |
Collapse
|
17
|
Anticancer activities of TCM and their active components against tumor metastasis. Biomed Pharmacother 2020; 133:111044. [PMID: 33378952 DOI: 10.1016/j.biopha.2020.111044] [Citation(s) in RCA: 171] [Impact Index Per Article: 42.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 02/07/2023] Open
Abstract
Traditional Chinese Medicine (TCM) has the characteristics of multiple targets, slight side effects and good therapeutic effects. Good anti-tumor effects are shown by Traditional Chinese Medicine prescription, Chinese patent medicine, single Traditional Chinese Medicine and Traditional Chinese medicine monomer compound. Clinically, TCM prolonged the survival time of patients and improved the life quality of patients, due to less side effects. Cancer metastasis is a complex process involving numerous steps, multiple genes and their products. During the process of tumor metastasis, firstly, cancer cell increases its proliferative capacity by reducing autophagy and apoptosis, and then the cancer cell capacity is stimulated by increasing the ability of tumors to absorb nutrients from the outside through angiogenesis. Both of the two steps can increase tumor migration and invasion. Finally, the purpose of tumor metastasis is achieved. By inhibiting autophagy and apoptosis of tumor cells, angiogenesis and EMT outside the tumor can inhibit the invasion and migration of cancer, and consequently achieve the purpose of inhibiting tumor metastasis. This review explores the research achievements of Traditional Chinese Medicine on breast cancer, lung cancer, hepatic carcinoma, colorectal cancer, gastric cancer and other cancer metastasis in the past five years, summarizes the development direction of TCM on cancer metastasis research in the past five years and makes a prospect for the future.
Collapse
|
18
|
Munakarmi S, Chand L, Shin HB, Hussein UK, Yun BS, Park HR, Jeong YJ. Anticancer effects of Poncirus fructus on hepatocellular carcinoma through regulation of apoptosis, migration, and invasion. Oncol Rep 2020; 44:2537-2546. [PMID: 33125135 PMCID: PMC7640358 DOI: 10.3892/or.2020.7790] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023] Open
Abstract
Poncirus fructus (PF) is a phytochemical compound extracted from the dry, immature fruits of Poncirus trifoliate. PF is traditionally used to treat gastrointestinal disorders, allergies, and inflammatory disease. In East Asia, PF is also known for its anticancer properties. There are numerous reports on the anticancer and anti-inflammatory effects of PF in a wide range of cancers and gastrointestinal diseases, respectively. However, the role of PF in inducing apoptosis and suppressing the invasiveness of hepatocellular carcinoma (HCC) remains unclear. This study investigated the ability of PF to induce apoptosis and inhibit the invasiveness and migratory ability of HCC cell lines (Hep3B and Huh7). Wound healing, Transwell migration and invasion, and colony-formation assays, as well as flow cytometry, were used to analyze cell proliferation, migration, invasion, and apoptosis. Epithelial-mesenchymal transition (EMT)-related and apoptotic proteins were assessed by western blotting. The mitochondrial membrane potential of the Hep3B and Huh7 cells was observed with tetramethylrhodamine ethyl ester. The reactive oxygen species (ROS) level was determined by dihydroethidium (DHE) staining. PF treatment significantly decreased the proliferation of Hep3B and Huh7 cells in a dose-dependent manner, reduced the mitochondrial membrane potential, increased ROS levels, decreased the protein levels of Bcl-2, and increased the protein levels of Bax and cleaved caspase-3 and 9, suggesting that PF mediated HCC apoptosis via a mitochondrial pathway. Our findings showed that PF prevented HCC cell migration and invasion by inhibiting the EMT process and downregulating MMP-2 and MMP-9 activities. The results suggest the potential anticancer effects of PF by inhibiting proliferation, inducing apoptosis, and reducing the invasion and migration of HCC cells.
Collapse
Affiliation(s)
- Suvesh Munakarmi
- Laboratory of Liver Regeneration, Biomedical Research Institute, Chonbuk National University Medical School, Jeonju 54907, Republic of Korea
| | - Lokendra Chand
- Laboratory of Liver Regeneration, Biomedical Research Institute, Chonbuk National University Medical School, Jeonju 54907, Republic of Korea
| | - Hyun Beak Shin
- Laboratory of Liver Regeneration, Biomedical Research Institute, Chonbuk National University Medical School, Jeonju 54907, Republic of Korea
| | - Usama Khamis Hussein
- Research Institute of Clinical Medicine of Jeonbuk National University, Biomedical Research Institute of Chonbuk National University, Jeonju 54907, Republic of Korea
| | - Bong-Sik Yun
- Division of Biotechnology, College of Environmental and BioSource Sciences, Chonbuk National University, Jeonju 54907, Republic of Korea
| | - Hae Ryong Park
- Department of Food Science and Biotechnology, Kyungnam University, Masanhappo‑gu, Changwon‑si 631‑701, Republic of Korea
| | - Yeon Jun Jeong
- Laboratory of Liver Regeneration, Biomedical Research Institute, Chonbuk National University Medical School, Jeonju 54907, Republic of Korea
| |
Collapse
|
19
|
Wu YH, Wu YR, Li B, Yan ZY. Cryptotanshinone: A review of its pharmacology activities and molecular mechanisms. Fitoterapia 2020; 145:104633. [DOI: 10.1016/j.fitote.2020.104633] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/03/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023]
|
20
|
Zhou J, Su CM, Chen HA, Du S, Li CW, Wu H, Tsai SH, Yeh YT. Cryptanshinone Inhibits the Glycolysis and Inhibits Cell Migration Through PKM2/β-Catenin Axis in Breast Cancer. Onco Targets Ther 2020; 13:8629-8639. [PMID: 32922039 PMCID: PMC7457727 DOI: 10.2147/ott.s239134] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 08/03/2020] [Indexed: 12/16/2022] Open
Abstract
Background Breast cancer is one of the most prevalent gynecologic malignancies worldwide. Despite the high sensitivity in response to chemotherapy, drug resistance occurred frequently in clinical treatment. Cryptotanshinone (CTS) is a herbal medicine and has been identified as an anti-inflammatory and anti-oxidative drug. Methods In vitro assays, including the cell proliferation assay, colony formation assay, Western blot analysis, transwell migration/invasion assays, and cell scratch assay were used to explore the biological activities and working mechanism of CTS. Breast cancer cells were also transfected with PKM2 expressing vectors to define the molecular mechanisms involved in CTS-mediated anti-tumor activity. Results We found that CTS shows anti-proliferative effects and decreases the clonogenic ability of breast cancer cells. We also found that CTS inhibited the migration and invasion activity of MCF-7 and MDA-MB-231 cells by different analyzed methods. CTS also downregulated the levels of glycolysis-related proteins, such as PKM2, LDHA, and HK2. In addition, overexpression of PKM2 recovered CTS-mediated suppression of cell proliferation, colony formation, and cell mobility of breast cancer cells. We also found PKM2 was significantly overexpressed in tumor tissues and invasive ductal breast carcinoma compared to normal tissues and patients with high PKM2 expression had worse overall survival and metastasis-free survival outcomes. Conclusion CTS inhibited the proliferation, migration, and invasion of breast cancer cells. The involved mechanism may refer to the downregulation of the PKM2/β-catenin axis.
Collapse
Affiliation(s)
- Jiefeng Zhou
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei City, Taiwan.,Ningbo AJcore Biosciences Inc, High-tech Zone, Ningbo City, People's Republic of China
| | - Chih-Ming Su
- Division of General Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan.,Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Hsin-An Chen
- Division of General Surgery, Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei City, Taiwan.,Division of General Surgery, Department of Surgery, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Shicong Du
- Ningbo AJcore Biosciences Inc, High-tech Zone, Ningbo City, People's Republic of China
| | - Chang-Wei Li
- Ningbo AllBiolife Biotech Inc, High-tech Zone, Ningbo City, People's Republic of China
| | - Haoran Wu
- Ningbo AJcore Biosciences Inc, High-tech Zone, Ningbo City, People's Republic of China
| | - Shin-Han Tsai
- Department of Emergency Medicine, Shuang Ho Hospital, Taipei Medical University, Institute of Injury Prevention and Control, Taipei Medical University, Taipei City, Taiwan
| | - Yu-Ting Yeh
- Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University, Taipei City, Taiwan.,Information Technology Office, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| |
Collapse
|
21
|
Ashrafizadeh M, Zarrabi A, Orouei S, Saberifar S, Salami S, Hushmandi K, Najafi M. Recent advances and future directions in anti-tumor activity of cryptotanshinone: A mechanistic review. Phytother Res 2020; 35:155-179. [PMID: 33507609 DOI: 10.1002/ptr.6815] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 05/29/2020] [Accepted: 07/02/2020] [Indexed: 12/13/2022]
Abstract
In respect to the enhanced incidence rate of cancer worldwide, studies have focused on cancer therapy using novel strategies. Chemotherapy is a common strategy in cancer therapy, but its adverse effects and chemoresistance have limited its efficacy. So, attempts have been directed towards minimally invasive cancer therapy using plant derived-natural compounds. Cryptotanshinone (CT) is a component of salvia miltiorrihiza Bunge, well-known as Danshen and has a variety of therapeutic and biological activities such as antioxidant, anti-inflammatory, anti-diabetic and neuroprotective. Recently, studies have focused on anti-tumor activity of CT against different cancers. Notably, this herbal compound is efficient in cancer therapy by targeting various molecular signaling pathways. In the present review, we mechanistically describe the anti-tumor activity of CT with an emphasis on molecular signaling pathways. Then, we evaluate the potential of CT in cancer immunotherapy and enhancing the efficacy of chemotherapy by sensitizing cancer cells into anti-tumor activity of chemotherapeutic agents, and elevating accumulation of anti-tumor drugs in cancer cells. Finally, we mention strategies to enhance the anti-tumor activity of CT, for instance, using nanoparticles to provide targeted drug delivery.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla, 34956, Istanbul, Turkey.,Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul, Turkey
| | - Sima Orouei
- MSc. Student, Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sedigheh Saberifar
- Department of Basic Sciences, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Saeed Salami
- DVM. Graduated, Kazerun Branch, Islamic Azad University, Kazeroon, Iran
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology & Zoonoses, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
22
|
Li L, Wu B, Zhao Q, Li J, Han Y, Fan X, Dong J, Li P. Attenuation of doxorubicin-induced cardiotoxicity by cryptotanshinone detected through association analysis of transcriptomic profiling and KEGG pathway. Aging (Albany NY) 2020; 12:9585-9603. [PMID: 32457254 PMCID: PMC7288906 DOI: 10.18632/aging.103228] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 04/16/2020] [Indexed: 05/09/2023]
Abstract
OBJECTIVE The cardiotoxicity of doxorubicin (DOX) reduces the quality of life and prognosis of cancer patients, and therefore its clinical application has been largely restricted. This study aimed to assess the effects of cryptotanshione (CPT) on DOX-induced rat cardiac insufficiency. RESULTS CPT treatment significantly suppressed apoptosis in vitro. The oral administration of CPT significantly improved cardiac function in the rat model, reduced collagen production and suppressed apoptosis and the production of reactive oxygen species in the heart tissue. Transcriptomic profiling and its relevant bioinformatics analysis showed that CPT suppressed doxorubicin-induced cardiotoxicity by inhibiting p53 signaling pathway. CONCLUSION Transcriptomic profiling and bioinformatics analysis can be used to evaluate the cardio-protective effect of CPT through inactivating p53 signaling pathway in the doxorubicin-mediated myocardial damage model. METHODS F-actin staining and flow cytometry were used to assess the effects of CPT on cardiomyocytes. In vivo, echocardiography and hemodynamic evaluation were used to assess the effects of CPT on the cardiac dysfunction in rats. Furthermore, transcriptomic profiling and bioinformatics analysis, as well as western blot analysis, were used to determine that CPT induced changes in the signaling pathways in the model.
Collapse
Affiliation(s)
- Le Li
- Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Bin Wu
- Laboratory of Platelet and Endothelium Biology, Department of Transfusion Medicine, Wuhan Hospital of Traditional Chinese and Western Medicine (Wuhan No.1 Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Qiangqiang Zhao
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Hematology, Qinghai Provincial People’s Hospital, Xi’ning, China
| | - Jian Li
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yunfeng Han
- Department of Nuclear Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohang Fan
- Department of Pathophysiology, Scholl of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junli Dong
- Laboratory of Clinical Pharmacogenetics, Department of Pharmacy, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Pengcheng Li
- Laboratory of Platelet and Endothelium Biology, Department of Transfusion Medicine, Wuhan Hospital of Traditional Chinese and Western Medicine (Wuhan No.1 Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Thaichinda S, Tancharoen S, Kanekura T, Higashi Y, Dararat P, Kikuchi K, Nararatwanchai T. Pinus maritima Extract Induces Apoptosis in Human Malignant Melanoma Cells via ROS/Caspase-3 Signaling. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20926889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Melanoma is the most aggressive type of skin cancer due to its rapid metastasis with a high recurrence rate following conventional therapy. Pine bark extract (PBE) from Pinus maritima contains numerous phenolic compounds and functions as a potent antioxidant. The present study aimed to analyze the potential anticancer properties of PBE on human malignant melanoma A375 cells. The chemical composition of PBE was determined by high-performance liquid chromatography/photodiode array detector. The effects of PBE on cell death, migration, and invasion were determined using xCELLigence Technology real-time cell analysis. Annexin/propidium iodide flow cytometry and Hoechst 33342 staining were conducted to detect cell apoptosis. PBE induced apoptosis and inhibited cell migration and invasion. Cleaved caspase-3 expression and activity were significantly increased ( P < 0.01) in cells treated with PBE compared with control cells. PBE ameliorated hydrogen peroxide (H2O2)-induced reactive oxygen species (ROS) formation. Treatment of the cells with PBE in the presence of H2O2 led to significant ( P < 0.001) reduction of matrix metallopeptidase-9, which is a mediator responsible for advanced melanoma. PBE induces A375 programmed cell death and suppresses cellular invasion by attenuating the ROS-dependent pathway associated with MMP-9 reduction.
Collapse
Affiliation(s)
- Sunisa Thaichinda
- Department of Anti-aging Medicine, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Muang, Chiang Rai, Thailand
| | - Salunya Tancharoen
- Department of Pharmacology, Faculty of Dentistry, Mahidol University, Rajthevee, Bangkok, Thailand
| | - Takuro Kanekura
- Department of Dermatology, Kagoshima University Medical and Dental Science, Sakuragaoka, Kagoshima, Japan
| | - Yuko Higashi
- Department of Dermatology, Kagoshima University Medical and Dental Science, Sakuragaoka, Kagoshima, Japan
| | - Pornpen Dararat
- Department of Pharmacology, Faculty of Dentistry, Mahidol University, Rajthevee, Bangkok, Thailand
| | - Kiyoshi Kikuchi
- Division of Brain Science, Department of Physiology, Kurume University School of Medicine, Kurume, Fukuoka, Japan
| | - Thamthiwat Nararatwanchai
- Department of Anti-aging Medicine, School of Anti-Aging and Regenerative Medicine, Mae Fah Luang University, Muang, Chiang Rai, Thailand
| |
Collapse
|
24
|
Nie X, Liu Y, Li M, Yu X, Yuan W, Huang S, Ren D, Wang Y, Wang Y. SP94 Peptide-Functionalized PEG-PLGA Nanoparticle Loading with Cryptotanshinone for Targeting Therapy of Hepatocellular Carcinoma. AAPS PharmSciTech 2020; 21:124. [PMID: 32342227 DOI: 10.1208/s12249-020-01655-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 03/07/2020] [Indexed: 12/28/2022] Open
Abstract
To achieve improved drug delivery efficiency to hepatocellular carcinoma (HCC), biodegradable poly (ethylene glycol)-poly (lactic-co-glycolic acid) (PEG-PLGA) nanoparticles (NP), surface-modified with SP94 peptide, were designed for the efficient delivery of cryptotanshinone to the tumor for the treatment of HCC. Cryptotanshinone NP and SP94-NP were prepared by using nanoprecipitation. The physicochemical and pharmaceutical properties of the NP and SP94-NP were characterized, and the release kinetics suggested that both NP and SP94-NP provided continuous, slow release of cryptotanshinone for 48 h. The in vitro cellular experiment demonstrated that SP94-NP significantly enhanced the cellular uptake of cryptotanshinone and induced high cytotoxicity and cellular apoptosis of hepatocellular carcinoma (HepG2) cells. The in vivo detecting results of targeting effect using the Cy5.5 probe evidenced that SP94-NP showed an accumulation in tumor more efficiently than that of unconjugated ones. Meanwhile, SP94-NP exhibited the smallest tumor size than other groups and showed no toxicity to body. The results of this study provide a promising nanoplatform for the targeting of HCC.
Collapse
|
25
|
Si L, Yan X, Wang Y, Ren B, Ren H, Ding Y, Zheng Q, Li D, Liu Y. Chamaejasmin B Decreases Malignant Characteristics of Mouse Melanoma B16F0 and B16F10 Cells. Front Oncol 2020; 10:415. [PMID: 32300554 PMCID: PMC7145408 DOI: 10.3389/fonc.2020.00415] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Accepted: 03/10/2020] [Indexed: 12/16/2022] Open
Abstract
Chamaejasmin B (CHB), a natural biflavone isolated from Stellera chamaejasme L., has been reported to exhibit anti-cancer properties; however, its effect in melanoma cells is not clear. Here, we aimed to investigate the anticancer effect of CHB in mouse melanoma B16F0 and B16F10 cells. We found that CHB significantly suppressed cell proliferation and promoted cell cycle arrest at G0/G1 phase in B16F0 cells; it also induced cell differentiation and increased melanin content by increasing tyrosinase (TYR) activity and mRNA levels of melanogenesis-related genes in B16F0 cells. Meanwhile, wound closure, invasion, and migration of B16F0 and B16F10 cells were dramatically inhibited. Moreover, CHB significantly increased ROS levels and decreased ΔΨm, resulting in B16F0 and B16F10 cell apoptosis. Finally, in vivo studies showed that CHB inhibited tumor growth and induced tumor apoptosis in a mouse xenograft model of murine melanoma B16F0 and B16F10 cells. Overall, CHB decreases malignant characteristics and may be a promising therapeutic agent for malignant melanoma cells via multiple signaling pathways.
Collapse
Affiliation(s)
- Lingling Si
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China.,Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, China
| | - Xinyan Yan
- People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| | - Yan Wang
- Key Laboratory of Xinjiang Endemic Phytomedicine Resources, Pharmacy School, Shihezi University, Ministry of Education, Shihezi, China
| | - Boxue Ren
- Key Laboratory of Xinjiang Endemic Phytomedicine Resources, Pharmacy School, Shihezi University, Ministry of Education, Shihezi, China
| | - Huanhuan Ren
- Key Laboratory of Xinjiang Endemic Phytomedicine Resources, Pharmacy School, Shihezi University, Ministry of Education, Shihezi, China
| | - Yangfang Ding
- Key Laboratory of Xinjiang Endemic Phytomedicine Resources, Pharmacy School, Shihezi University, Ministry of Education, Shihezi, China
| | - Qiusheng Zheng
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China.,Key Laboratory of Xinjiang Endemic Phytomedicine Resources, Pharmacy School, Shihezi University, Ministry of Education, Shihezi, China
| | - Defang Li
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| | - Ying Liu
- School of Integrated Traditional Chinese and Western Medicine, Binzhou Medical University, Yantai, China
| |
Collapse
|
26
|
Zhang Q, Wang L, Gan C, Yu Y, Li Y, Deng Y, Liu H, You J, Yin W. Cryptotanshinone Induces Apoptosis and Inhibits Migration and Invasion in Human Hepatocellular Carcinoma Cells In Vitro. Nat Prod Commun 2020. [DOI: 10.1177/1934578x19899570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Cryptotanshinone (CPT), an active quinoid diterpene isolated from Salvia miltiorrhiza Bunge, was previously reported to have potential anticancer effects. However, the mechanisms of CPT on hepatocellular carcinoma (HCC) cells are not well understood. In this study, we investigated the anticancer effects of CPT on HCC cells. Thiazolyl blue tetrazolium bromide assay showed dose-dependent and time-dependent cytotoxicity of CPT on human HCC cells, especially in HCCLM3 and Huh-7 cells. Hoechst 33258 stain, flow cytometry assay, and Western blot assay all indicated that CPT could distinctly induce the apoptosis of human HCC cells and break intracellular homeostasis by triggering the imbalance of mitochondrial transmembrane potential ( Δψm) and reactive oxygen species. In addition, CPT could significantly inhibit HCCLM3 and Huh-7 cells’ migration and invasion via the signal transducers and activators of transcription 3/matrix metalloproteinases mediated signaling pathway. Our findings demonstrated that the antitumor effects of CPT on human HCC cells were by suppressing cell proliferation, inducing cell apoptosis and impairing cell migration and invasion.
Collapse
Affiliation(s)
- Qianyu Zhang
- West China School of Public Health and Heathy Food Evaluation Research Center and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Liqun Wang
- West China School of Public Health and Heathy Food Evaluation Research Center and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Cailing Gan
- Laboratory of Liver Surgery and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Yu
- Laboratory of Liver Surgery and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Yali Li
- West China School of Public Health and Heathy Food Evaluation Research Center and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Yuanle Deng
- West China School of Public Health and Heathy Food Evaluation Research Center and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Hongyao Liu
- Laboratory of Liver Surgery and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jia You
- West China School of Public Health and Heathy Food Evaluation Research Center and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Wenya Yin
- West China School of Public Health and Heathy Food Evaluation Research Center and West China Fourth Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
27
|
Amaral-Machado L, Oliveira WN, Alencar ÉN, Cruz AKM, Rocha HAO, Ebeid K, Salem AK, Egito EST. Bullfrog oil (Rana catesbeiana Shaw) induces apoptosis, in A2058 human melanoma cells by mitochondrial dysfunction triggered by oxidative stress. Biomed Pharmacother 2019; 117:109103. [PMID: 31203130 DOI: 10.1016/j.biopha.2019.109103] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 06/03/2019] [Accepted: 06/06/2019] [Indexed: 12/18/2022] Open
Abstract
Bullfrog oil, an animal oil extracted from the adipose tissue of Rana catesbeiana Shaw, showed promising cytotoxic activity against melanoma cells and, therefore, has the potential to become a pharmaceutical active compound. However, there is a lack of information regarding the pathways involved in its pharmacological activity. Thus, the aim of this study was to investigate and elucidate the cytotoxic effect of this oil against A2058 human melanoma cells. The cytotoxic potential was evaluated by the MTT assay, the cell cycle analysis and the cell death assay. In addition, the apoptotic potential was investigated by (i) the DNA fragmentation using propidium iodide staining analysis, (ii) the evaluation of mitochondrial membrane potential and (iii) the determination of intracellular Reactive Oxygen Species (ROS) level. The results showed that the bullfrog oil was able to promote a time-dependent cytotoxic effect, decreasing cell viability to 38% after 72 h of treatment without affecting the cell cycle. Additionally, the bullfrog oil induced the apoptosis in A2058 cells, increasing up to 50 ± 13% of the intracellular ROS level, maintaining the DNA integrity and promoting an approximate decrease of 35 ± 5% in the mitochondrial membrane potential. It can be concluded that the in vitro cytotoxic effect of the bullfrog oil in A2058 human melanoma cells is mediated by oxidative stress that induces mitochondrial dysfunction, triggering the apoptosis. These unprecedented results highlight the pharmacological potential of bullfrog oil and provide important information to support studies on the development of new pharmaceutical products for complementary and alternative treatments for melanoma.
Collapse
Affiliation(s)
- Lucas Amaral-Machado
- Graduation Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil
| | | | - Éverton N Alencar
- Graduation Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil
| | | | | | - Kareem Ebeid
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, 52242, USA; Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Aliasger K Salem
- Division of Pharmaceutics and Translational Therapeutics, College of Pharmacy, University of Iowa, Iowa City, IA, 52242, USA
| | - Eryvaldo Sócrates T Egito
- Graduation Program in Health Sciences, Federal University of Rio Grande do Norte (UFRN), Natal, Brazil; Graduation Program in Pharmaceutical Sciences, UFRN, Natal, Brazil.
| |
Collapse
|
28
|
Han Z, Liu S, Lin H, Trivett AL, Hannifin S, Yang D, Oppenheim JJ. Inhibition of murine hepatoma tumor growth by cryptotanshinone involves TLR7-dependent activation of macrophages and induction of adaptive antitumor immune defenses. Cancer Immunol Immunother 2019; 68:1073-1085. [PMID: 31161238 PMCID: PMC6584221 DOI: 10.1007/s00262-019-02338-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 04/01/2019] [Indexed: 01/05/2023]
Abstract
Cryptotanshinone (CT), a purified compound initially isolated from the dried roots of Salvia militorrhiza. Bunge, exhibits cytotoxic antitumor effects on many tumors. We have shown that CT possesses the dual capacities to concomitantly inhibit the proliferation of lung cancer cells and promote the generation of antitumor immunity. In this study, we investigated whether CT could be used to treat hepatocellular carcinoma (HCC) using a mouse Hepa1-6 model. CT inhibited the proliferation of mouse hepatoma (Hepa1-6) cells in vitro by inducing Hepa1-6 cells apoptosis through the JAK2/STAT3 signaling pathway. In addition, CT activated macrophages and polarized mouse bone marrow-derived macrophages (BMM) toward an M1 phenotype in vitro, which depended on the TLR7/MyD88/NF-κB signaling pathway. Furthermore, CT significantly inhibited the growth of syngeneic Hepa1-6 hepatoma tumors, and, in combination with anti-PD-L1 cured Hepa1-6-bearing mice with the induction of long-term anti-Hepa1-6 specific immunity. Immunoprofiling of treated Hepa1-6-bearing mice revealed that CT-promoted activation of tumor-infiltrating macrophages and dendritic cells, induction of antitumor T cell response, and infiltration of effector/memory CD8 T cells in the tumor tissue. Importantly, the immunotherapeutic effects of CT and anti-PD-L1 depended on the presence of CD8 T cells. Thus, CT and anti-PD-L1 may provide an effective immunotherapeutic regimen for human HCC based on a combination of cytotoxic effects and induction of tumor-specific immunity.
Collapse
Affiliation(s)
- Zhen Han
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick National Laboratory for Cancer Research (FNLCR), Rm 21-89/31-19, Bldg 560, 1050 Boyles Street, Frederick, MD, 21702-1201, USA
| | - Shuo Liu
- Guang' Anmen Hospital, China Academy of Chinese Medical Sciences, #5 Beixian Ge, Xi Cheng District, Beijing, 100053, China
| | - Hongsheng Lin
- Guang' Anmen Hospital, China Academy of Chinese Medical Sciences, #5 Beixian Ge, Xi Cheng District, Beijing, 100053, China.
| | - Anna L Trivett
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick National Laboratory for Cancer Research (FNLCR), Rm 21-89/31-19, Bldg 560, 1050 Boyles Street, Frederick, MD, 21702-1201, USA
| | - Sean Hannifin
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick National Laboratory for Cancer Research (FNLCR), Rm 21-89/31-19, Bldg 560, 1050 Boyles Street, Frederick, MD, 21702-1201, USA
| | - De Yang
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick National Laboratory for Cancer Research (FNLCR), Rm 21-89/31-19, Bldg 560, 1050 Boyles Street, Frederick, MD, 21702-1201, USA.
| | - Joost J Oppenheim
- Cancer and Inflammation Program, Center for Cancer Research, National Cancer Institute, Frederick National Laboratory for Cancer Research (FNLCR), Rm 21-89/31-19, Bldg 560, 1050 Boyles Street, Frederick, MD, 21702-1201, USA.
| |
Collapse
|
29
|
Chengxi W, Rishuo G, QuLiang G, Laiyou W, Lianbing H, Linghao Q. The prevention effects of cryptotanshinone nanoemulsion on postoperative peritoneal adhesions. Drug Dev Ind Pharm 2019; 45:695-702. [DOI: 10.1080/03639045.2018.1529788] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Affiliation(s)
- Wang Chengxi
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Guo Rishuo
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Gu QuLiang
- School of Basic Science, Guangdong Pharmaceutical University, Guangzhou, China
| | - Wang Laiyou
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hou Lianbing
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qin Linghao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
30
|
Wang T, Wang G, Zhang Y, Zhang J, Cao W, Chen X. Effect of lentivirus-mediated overexpression or silencing of MnSOD on apoptosis of resveratrol-treated fibroblast-like synoviocytes in rheumatoid arthritis. Eur J Pharmacol 2019; 844:65-72. [DOI: 10.1016/j.ejphar.2018.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 11/28/2018] [Accepted: 12/03/2018] [Indexed: 12/17/2022]
|
31
|
Wang W, Wang Y, Liu M, Zhang Y, Yang T, Li D, Huang Y, Li Q, Bai G, Shi L. Betulinic acid induces apoptosis and suppresses metastasis in hepatocellular carcinoma cell lines in vitro and in vivo. J Cell Mol Med 2019; 23:586-595. [PMID: 30417527 PMCID: PMC6307792 DOI: 10.1111/jcmm.13964] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 08/06/2018] [Accepted: 09/19/2018] [Indexed: 01/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a high incidence and mortality malignant tumour globally. Betulinic acid (BA) is a pentacyclic triterpenoid with potential pro-apoptotic activities which widely found in many plants. In this study, we determined the effects of BA on proliferation, apoptosis, invasion, and metastasis in HCC cell lines and on tumour growth and pulmonary metastasis in mice. The results suggested that BA could inhibit cell viability and proliferation of HCC cell lines including HepG2, LM3, and MHCC97H. In addition, BA induced apoptosis of HepG2 cells characterised condensed nuclei and nuclear fragmentation. Moreover, western blot analysis showed that BA-induced apoptosis associated with increasing of pro-apoptotic protein Bax and cleaved caspase-3 and decreasing of anti-apoptotic protein Bcl-2. Meanwhile, BA also reduced the reactive oxygen species (ROS) level. Furthermore, BA also significantly inhibited HCC growth in vivo and blocked pulmonary metastasis of HCC by regulating the metastasis-related proteins including MMP-2, MMP-9, and TIMP2 without obvious toxicity. In all, the present study suggested that BA might be a promising anti-HCC drug candidate by inhibiting proliferation, inducing apoptosis, and blocking metastasis.
Collapse
Affiliation(s)
- Wei Wang
- Department of General surgeryFirst Affiliated Hospital of Jinzhou Medical UniversityJinzhouChina
| | - Yang Wang
- Graduate SchoolJinzhou Medical UniversityJinzhouChina
| | - Mingxing Liu
- Graduate SchoolJinzhou Medical UniversityJinzhouChina
| | - Yifan Zhang
- Graduate SchoolJinzhou Medical UniversityJinzhouChina
| | - Tao Yang
- Department of General surgeryFirst Affiliated Hospital of Jinzhou Medical UniversityJinzhouChina
| | - Dongsheng Li
- Department of General surgeryFirst Affiliated Hospital of Jinzhou Medical UniversityJinzhouChina
| | - Yinpeng Huang
- Department of General surgeryFirst Affiliated Hospital of Jinzhou Medical UniversityJinzhouChina
| | - Qing Li
- Department of Internal MedicineThird Affiliated Hospital of Jinzhou Medical UniversityJinzhouChina
| | - Guang Bai
- Department of General surgeryFirst Affiliated Hospital of Jinzhou Medical UniversityJinzhouChina
| | | |
Collapse
|
32
|
Zangeneh F, Vazirizadeh A, Mirshamsi MR, Fakhri A, Faizi M, Pourahmad J. Induction of Apoptosis by an Extract of Persian Gulf Marine Mollusc, Turbo Coronatus through the Production of Reactive
Oxygen Species in Mouse Melanoma Cells. Asian Pac J Cancer Prev 2018; 19:3479-3488. [PMID: 30583673 PMCID: PMC6428523 DOI: 10.31557/apjcp.2018.19.12.3479] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Objective: A variety of approaches such as surgery, chemotherapy, radiotherapy, hormonal therapy and
immunotherapy are used to treat melanomas, but unfortunately in most case, the response is very weak and often side
effects are serious. This study concerns selective toxicity of an extract of Turbo coronatus on cells and mitochondria from
a syngeneic mouse model of melanoma. Methods: Cells and mitochondria isolated from extra tumoral and melanoma
tissues were exposed toa T. coronatus crude extract and fractions obtained by gel-filtration chromatography and assayed
for mitochondrial and cellular parameters. Result: Crude extract (375, 750 and 1,500 μg/ml) and fraction 1; F1; (275,
550 and 1100 μg/ml) of T. coronatus extract induced a significant (p<0.05) increase of the reactive oxygen species
(ROS) level, swelling of mitochondria, collapse of mitochondrial membrane potential (MMP), release of cytochrome
c and caspase-3 activation only in the mitochondria and cells obtained from melanoma but not extra tumoral tissues. In
addition, the F1 fraction decreased the percentage of viable cells and induced apoptosis in melanoma cells. Conclusion:
For the first time we could demonstrate that the F1 fraction of a T. coronatus extract, selectively induces ROS mediated
cytotoxicity by directly targeting mitochondria in melanoma tissues and it may be a suitable candidate for novel drug
treatment of malignant melanomas.
Collapse
Affiliation(s)
- Fatemeh Zangeneh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy and Pharmaceutical Sciences, Research Center Shahid Beheshti University of Medical Sciences, Tehran, Iran. ,
| | | | | | | | | | | |
Collapse
|
33
|
Sun HN, Luo YH, Meng LQ, Piao XJ, Wang Y, Wang JR, Wang H, Zhang Y, Li JQ, Xu WT, Liu Y, Zhang Y, Zhang T, Han YH, Jin MH, Shen GN, Zang YQ, Cao LK, Zhang DJ, Jin CH. Cryptotanshinone induces reactive oxygen species‑mediated apoptosis in human rheumatoid arthritis fibroblast‑like synoviocytes. Int J Mol Med 2018; 43:1067-1075. [PMID: 30535477 DOI: 10.3892/ijmm.2018.4012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 11/16/2018] [Indexed: 11/06/2022] Open
Abstract
The present study investigated the mechanisms of apoptosis induced by cryptotanshinone (CT) in human rheumatoid arthritis fibroblast‑like synoviocytes (RA‑FLSs). Cell Counting kit‑8 assay was performed to determine the cytotoxic effects of CT in human RA‑FLSs, including primary RA‑FLS, HFLS‑RA and MH7A cells, and in HFLS cells derived from normal synovial tissue. Annexin V‑FITC/PI staining was used to detect the apoptotic effects of CT in HFLS‑RA and MH7A cells. Flow cytometry was performed to detect the apoptotic and reactive oxygen species (ROS) levels induced by CT in HFLS‑RA cells. Western blotting was used to assess the expression levels of proteins associated with apoptosis and with the mitogen‑activated protein kinase (MAPK), protein kinase B (Akt), and signal transducer and activator of transcription‑3 (STAT3) signaling pathways. The results demonstrated that CT treatment significantly suppressed HFLS‑RA and MH7A cell growth, whereas no clear inhibitory effect was observed in normal HFLS cells. CT exposure downregulated the expression levels of B‑cell lymphoma 2 (Bcl‑2), p‑Akt, p‑extracellular signal‑related kinase and p‑STAT3, while it upregulated the expression levels of Bcl‑2‑associated death promoter (Bad), caspase‑3, poly (ADP‑ribose) polymerase (PARP), p‑p38 and p‑c‑Jun N‑terminal kinase. Following ROS scavenging, the CT‑induced apoptosis and altered expression levels of Bcl‑2, Bad, cleaved caspase‑3 and cleaved PARP were restored. Furthermore, the Akt, MAPK and STAT3 signaling pathways were regulated by intracellular ROS. These results suggest that ROS‑mediated Akt, MAPK and STAT3 signaling pathways serve important roles in the CT‑induced apoptosis of RA‑FLSs.
Collapse
Affiliation(s)
- Hu-Nan Sun
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Ying-Hua Luo
- Department of Grass Science, College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Ling-Qi Meng
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Xian-Ji Piao
- Department of Gynaecology and Obstetrics, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, Heilongjiang 163316, P.R. China
| | - Yue Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Jia-Ru Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Hao Wang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yi Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Jin-Qian Li
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Wan-Ting Xu
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yang Liu
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yu Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Tong Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Ying-Hao Han
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Mei-Hua Jin
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Gui-Nan Shen
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Yan-Qing Zang
- Department of Food Science and Engineering, College of Food Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Long-Kui Cao
- Department of Food Science and Engineering, College of Food Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Dong-Jie Zhang
- Department of Food Science and Engineering, College of Food Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| | - Cheng-Hao Jin
- Department of Biochemistry and Molecular Biology, College of Life Science and Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang 163319, P.R. China
| |
Collapse
|
34
|
Saraf RS, Datta A, Sima C, Hua J, Lopes R, Bittner M. An in-silico study examining the induction of apoptosis by Cryptotanshinone in metastatic melanoma cell lines. BMC Cancer 2018; 18:855. [PMID: 30157799 PMCID: PMC6116360 DOI: 10.1186/s12885-018-4756-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Accepted: 08/16/2018] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Metastatic melanoma is an aggressive form of skin cancer that evades various anti-cancer treatments including surgery, radio-,immuno- and chemo-therapy. TRAIL-induced apoptosis is a desirable method to treat melanoma since, unlike other treatments, it does not harm non-cancerous cells. The pro-inflammatory response to melanoma by nF κB and STAT3 pathways makes the cancer cells resist TRAIL-induced apoptosis. We show that due to to its dual action on DR5, a death receptor for TRAIL and on STAT3, Cryptotanshinone can be used to increase sensitivity to TRAIL. METHODS The development of chemoresistance and invasive properties in melanoma cells involves several biological pathways. The key components of these pathways are represented as a Boolean network with multiple inputs and multiple outputs. RESULTS The possible mutations in genes that can lead to cancer are captured by faults in the combinatorial circuit and the model is used to theoretically predict the effectiveness of Cryptotanshinone for inducing apoptosis in melanoma cell lines. This prediction is experimentally validated by showing that Cryptotanshinone can cause enhanced cell death in A375 melanoma cells. CONCLUSION The results presented in this paper facilitate a better understanding of melanoma drug resistance. Furthermore, this framework can be used to detect additional drug intervention points in the pathway that could amplify the action of Cryptotanshinone.
Collapse
Affiliation(s)
- Radhika S. Saraf
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, US
| | - Aniruddha Datta
- Department of Electrical and Computer Engineering, Texas A&M University, College Station, US
| | - Chao Sima
- TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering (CBGSE), College Station, US
| | - Jianping Hua
- TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering (CBGSE), College Station, US
| | - Rosana Lopes
- TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering (CBGSE), College Station, US
| | - Michael Bittner
- TEES-AgriLife Center for Bioinformatics and Genomic Systems Engineering (CBGSE), College Station, US
- Translational Genomics Research Institute (TGen), Phoenix, US
| |
Collapse
|
35
|
Yang Y, Liu W, Wang J, Zhang Y, Xu W, Tao L. The different effects of natural pyrethrins and beta-cypermethrin on human hepatocyte QSG7701 cells by ROS-mediated oxidative damage. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:24230-24240. [PMID: 29948706 DOI: 10.1007/s11356-018-2503-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 06/04/2018] [Indexed: 06/08/2023]
Abstract
With the widespread use of natural pyrethrins and pyrethroids to defend pest insects, people had the sustained interest in the potential risk to environment and human health. However, the research about natural pyrethrins and beta-cypermethrin induction of cytotoxicity is still seldom. This study is about the cytotoxic effects of these on human non-target cells in vitro. The cytotoxic effect of natural pyrethrins and beta-cypermethrin on QSG7701 cells were researched by using various bioassays in vitro. The results suggested that with the natural pyrethrin concentration increased, the viability of QSG7701 cells were inhibited increasingly, and the IC50 value as calculated was approximately 42.54 and 18.68 μg/mL after the cells were treated 24 and 48 h. The proliferative potential of QSG7701 cells treated with 40 μg/mL natural pyrethrins 6 and 12 h was decreased by 67.44 and 94.74%, dramatic enhancement ROS, collapse of mitochondrial membrane potential, DNA exhibit severity of impairment, and chromatin DNA condensation. However, beta-cypermethrin has lower toxicity than natural pyrethrins. The IC50 values of beta-cypermethrin were all > 80 μg/mL, and the colony formation expression was decreased by 15.26 and 19.09%, which implied that natural pyrethrins are more significantly cytotoxic and potentially genotoxic to human hepatocyte cells.
Collapse
Affiliation(s)
- Yun Yang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Wenjing Liu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Jian Wang
- Medicine Hospital Imaging Center, Wei fang traditional Chinese hospital, Shandong, 261000, China
| | - Yang Zhang
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Wenping Xu
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Liming Tao
- Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
36
|
Cryptotanshinone inhibits proliferation yet induces apoptosis by suppressing STAT3 signals in renal cell carcinoma. Oncotarget 2018; 8:50023-50033. [PMID: 28654902 PMCID: PMC5564825 DOI: 10.18632/oncotarget.18483] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2017] [Accepted: 05/04/2017] [Indexed: 11/30/2022] Open
Abstract
It has been established that signal transducer and activator of transcription 3 serves as an oncoprotein in various human cancers; targeting it is therefore a reasonable approach for emerging cancer therapies. Cryptotanshinone, a natural compound extracted from the root of Salvia miltiorrhiza Bunge, has been identified as a potential STAT3 inhibitor. However, its functional role in renal cell carcinomas remains largely unknown. Therefore, we investigated the mode of action for cryptotanshinone. We found that cryptotanshinone substantially suppressed cancer cell growth while it promoted cell apoptosis by inhibiting the phosphorylation of STAT3 at Tyr705 and its blocking nuclear translocation. Coordinately, P-AKT, CyclinD1, C-MYC, MEKK2, and HGF were down-regulated and cell cycle progression was arrested at the G0/G1 phase, thereby attenuating cell proliferation. Moreover, the level of Cleaved-Caspase-3 was elevated while Bcl-2 and Survivin were down-regulated, accounting for the increased apoptosis. Furthermore, in vivo results revealed that cryptotanshinone effectively inhibits tumorigenesis in an A498-xenografted mouse model. Taken together, our data gives a more comprehensive understanding of how cryptotanshinone functions in renal cell carcinomas and demonstrates its potential as a powerful therapeutic approach to treat renal cell carcinomas.
Collapse
|
37
|
Yang S, Zhang Y, Luo Y, Xu B, Yao Y, Deng Y, Yang F, Ye T, Wang G, Cheng Z, Zheng Y, Xie Y. Hinokiflavone induces apoptosis in melanoma cells through the ROS-mitochondrial apoptotic pathway and impairs cell migration and invasion. Biomed Pharmacother 2018; 103:101-110. [PMID: 29635122 DOI: 10.1016/j.biopha.2018.02.076] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 02/05/2018] [Accepted: 02/19/2018] [Indexed: 02/05/2023] Open
Abstract
Melanoma, the highest degree of malignancy, is one of the most common skin tumors. However, there is no effective strategy to treat melanoma in current clinical practice. Therefore, it is urgent to find an efficient drug to overcome melanoma. Here, the in vitro anticancer effects of a natural product named hinokiflavone on three melanoma carcinoma cell lines (human melanoma A375 and CHL-1 cells, murine melanoma B16-F10 cells) and mechanisms of action were explored. The results of MTT assay revealed that hinokiflavone inhibited cell proliferation of these cell lines in a dose- and time-dependent manner. Interestingly, hinokiflavone showed low toxicity to normal liver cells. Flow cytometry assay and EdU incorporation assay indicated that hinokiflavone affected A375 and B16 cells survival by inducing apoptosis and blocking cell cycle progression at S phase in a concentration-dependent manner. Moreover, hinokiflavone enhanced the reactive oxygen species (ROS) and decreased the mitochondrial membrane potential obviously. Furthermore, hinokiflavone effectively impaired A375 cells migration and invasion, and down-regulated the expression of matrix metalloproteinase (MMP) MMP2 and MMP9. The above-mentioned results demonstrated that hinokiflavone could be a novel chemotherapeutic agent in melanoma treatment by inhibiting cell proliferation, inducing apoptosis and cell cycle arresting and blocking cell migration and invasion.
Collapse
Affiliation(s)
- Shuping Yang
- Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province 610064, PR China
| | - Yange Zhang
- Cosmetic Plastic and Burn Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province 610041, PR China
| | - Yi Luo
- Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province 610064, PR China
| | - Bocheng Xu
- Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province 610064, PR China
| | - Yuqin Yao
- Research Center for Occupational Respiratory Diseases, West China School of Public Health/No.4 West China Teaching Hospital, Sichuan University, Chengdu, Sichuan Province 610041, PR China
| | - Yuanle Deng
- Research Center for Occupational Respiratory Diseases, West China School of Public Health/No.4 West China Teaching Hospital, Sichuan University, Chengdu, Sichuan Province 610041, PR China
| | - Fangfang Yang
- Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province 610064, PR China
| | - Tinghong Ye
- Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province 610064, PR China
| | - Gang Wang
- School of Pharmacy, Zunyi Medical College, Zunyi, Guizhou Province 563003, PR China
| | - Zhiqiang Cheng
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yu Zheng
- Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province 610064, PR China.
| | - Yongmei Xie
- Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan Province 610064, PR China.
| |
Collapse
|
38
|
Liu C, Sun HN, Luo YH, Piao XJ, Wu DD, Meng LQ, Wang Y, Zhang Y, Wang JR, Wang H, Xu WT, Li JQ, Liu Y, Wu YQ, Han YH, Shen GN, Jin MH, Zang YQ, Li JC, Fang NZ, Cui YD, Jin CH. Cryptotanshinone induces ROS-mediated apoptosis in human gastric cancer cells. Oncotarget 2017; 8:115398-115412. [PMID: 29383168 PMCID: PMC5777780 DOI: 10.18632/oncotarget.23267] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 12/03/2017] [Indexed: 02/07/2023] Open
Abstract
Cryptotanshinone (CT), isolated from the plant Salvia miltiorrhiza Bunge, has been reported to have potential anticancer effects on human prostate and breast cancer cells. However, the mechanisms of action of CT on gastric cancer (GC) cells are not well understood. Here we investigated the antitumor effects of CT on GC cells and its possible molecular mechanism. We found CT suppressed viability of twelve GC cell lines in a dose-dependent manner. CT induced cell cycle arrest at the G2/M phase and mitochondrial apoptosis accompanying the accumulation of reactive oxygen species (ROS). Pretreatment with ROS inhibitor N-acetyl-L-cysteine (NAC) blocked CT-induced apoptosis. CT increased p-JNK and p-p38, and decreased p-ERK and p-STAT3 protein expression, these effects were prevented by NAC. Furthermore, a xenograft assay showed that CT significantly inhibited MKN-45 cell-induced tumor growth in vivo by increasing expression of pro-apoptotic proteins (p-JNK, p-38 and cleaved-caspase-3) and reducing expression of anti-apoptotic proteins (p-ERK and p-STAT3) without adverse effects on nude mice weight. In conclusion, CT induced apoptosis and cell cycle arrest in GC cells via ROS-mediated MAPK and AKT signaling pathways, and this CT may be a useful compound for the developing anticancer agents for GC.
Collapse
Affiliation(s)
- Chang Liu
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Hu-Nan Sun
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Ying-Hua Luo
- College of Animal Science & Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Xian-Ji Piao
- Department of Gynaecology and Obstetrics, The Fifth Affiliated Hospital of Harbin Medical University, Daqing 163316, China
| | - Dan-Dan Wu
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Ling-Qi Meng
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yue Wang
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yi Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Jia-Ru Wang
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Hao Wang
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Wan-Ting Xu
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Jin-Qian Li
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yang Liu
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yi-Qin Wu
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Ying-Hao Han
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Gui-Nan Shen
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Mei-Hua Jin
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yan-Qing Zang
- College of Food Science & Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Jing-Chun Li
- College of Animal Science & Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Nan-Zhu Fang
- Department of Animal Science, College of Agriculture, Yanbian University, Gongyuan-jie, Yanji 133002, China
| | - Yu-Dong Cui
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Cheng-Hao Jin
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| |
Collapse
|
39
|
Xu Z, Jiang H, Zhu Y, Wang H, Jiang J, Chen L, Xu W, Hu T, Cho CH. Cryptotanshinone induces ROS-dependent autophagy in multidrug-resistant colon cancer cells. Chem Biol Interact 2017; 273:48-55. [DOI: 10.1016/j.cbi.2017.06.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 05/18/2017] [Accepted: 06/05/2017] [Indexed: 02/07/2023]
|
40
|
Aloe-emodin (AE) nanoparticles suppresses proliferation and induces apoptosis in human lung squamous carcinoma via ROS generation in vitro and in vivo. Biochem Biophys Res Commun 2017. [PMID: 28629998 DOI: 10.1016/j.bbrc.2017.06.084] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Human lung squamous cell carcinoma is a deadly cancer for which present therapeutic strategies are inadequate. And traditional chemotherapy results in severe systemic toxicity. Compounds from living organisms often exert a biological activity, triggering several targets, which may be useful for the improvement of novel pharmaceuticals. Aloe-emodin (AE), a well-known natural compound, is a primary component of anthraquinones in Aloe vera and exhibits anti-proliferative and apoptotic effects on various tumor cells. However, the translational and clinical use of AE has been limited owing to its rapid degradation and poor bioavailability. To improve its efficacy, a poly (lactic-co-glycolic acid) based AE nanoparticle formulation (NanoAE) was prepared. Our study indicated that compared to the free AE, nanoAE significantly suppressed cancer cell proliferation, induced cell cycle arrest and apoptosis, evidenced by high cleavage of Caspase-3, poly (ADP-ribose) polymerase (PARP), Caspase-8 and Caspase-9. NanoAE enhanced reactive oxygen species (ROS) production, along with Mitogen-activated protein kinases (MAPKs) activation and PI3K/AKT inactivation. Cell proliferation, apoptosis and MAPKs and PI3K/AKT were dependent on ROS production in nanoAE-treated groups. In vivo, nanoAE exhibited inhibitory effects on the tumor growth with little toxicity. Together, our results indicated that nanoAE might be an effective treatment for human lung squamous cell carcinoma.
Collapse
|
41
|
Jiang G, Liu J, Ren B, Zhang L, Owusu L, Liu L, Zhang J, Tang Y, Li W. Anti-tumor and chemosensitization effects of Cryptotanshinone extracted from Salvia miltiorrhiza Bge. on ovarian cancer cells in vitro. JOURNAL OF ETHNOPHARMACOLOGY 2017; 205:33-40. [PMID: 28456578 DOI: 10.1016/j.jep.2017.04.026] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 04/25/2017] [Accepted: 04/26/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Cryptotanshinone, a natural compound isolated from the roots of Salvia miltiorrhiza Bge. (Danshen), is a commonly used traditional Chinese medicine to treat high blood pressure in some countries. It has been shown that Cryptotanshinone induces cancer cells apoptosis and impairs cell migration and invasion. However, the antiproliferation and chemosensitization effects of Cryptotanshinone on ovarian cancer and the underlying mechanism are not fully elucidated. AIM OF STUDY In this study, we evaluated the inhibitory effect of Cryptotanshinone on ovarian cancer cells and explored the underlying molecular mechanism. Additionally, the chemosensitization potential of Cryptotanshinone was evaluated in combination with cisplatin. MATERIALS AND METHODS MTT assay was used for cell viability assessment of ovarian cancer A2780 cells treated with Cryptotanshinone and/ or cisplatin. Flow cytometry was used for apoptosis analysis. Wound healing and transwell assays were used for migratory and invasive potential assessment of Cryptotanshinone-treated ovarian cancer cells. Western blot was used to investigate proteins involved in the mechanisms for metastasis and apoptosis. γH2AX immunocytochemistry was used to detect DNA damage in A2780 cells exposed to Cryptotanshinone and/or cisplatin. RESULTS Cryptotanshinone significantly induced ovarian cancer A2780 cells apoptosis by activating caspase cascade. Additionally, wound healing and transwell assays revealed that Cryptotanshinone could suppress migration and invasion of ovarian cancer cells and dramatically inhibited MMP-2 and MMP-9 expression. Furthermore, Cryptotanshinone could sensitize A2780 cells to cisplatin treatment in a dose-dependent manner. CONCLUSION Our data confirmed the anti-tumor effect of Cryptotanshinone on ovarian cancer cells and provided new findings that Cryptotanshinone could sensitize ovarian cancer cells to chemotherapy.
Collapse
Affiliation(s)
- Guoqiang Jiang
- Department of Biotechnology, Dalian Medical University, Dalian 116044, Liaoning, China
| | - Jia Liu
- Department of Biotechnology, Dalian Medical University, Dalian 116044, Liaoning, China
| | - Baoyin Ren
- Department of Biotechnology, Dalian Medical University, Dalian 116044, Liaoning, China
| | - Lin Zhang
- Department of Biotechnology, Dalian Medical University, Dalian 116044, Liaoning, China
| | - Lawrence Owusu
- Department of Integrative Medicine, Dalian Medical University, Dalian 116044, Liaoning, China; Department of Biochemistry and Biotechnology, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Likun Liu
- Department of Biotechnology, Dalian Medical University, Dalian 116044, Liaoning, China
| | - Jing Zhang
- Department of Biotechnology, Dalian Medical University, Dalian 116044, Liaoning, China
| | - Yawei Tang
- Department of Immunology, College of Basic Medical Science, Dalian Medical University, Lvshun South Road, Dalian 116044, Liaoning, China
| | - Weiling Li
- Department of Biotechnology, Dalian Medical University, Dalian 116044, Liaoning, China.
| |
Collapse
|
42
|
Yang B, Zhao S. Polydatin regulates proliferation, apoptosis and autophagy in multiple myeloma cells through mTOR/p70s6k pathway. Onco Targets Ther 2017; 10:935-944. [PMID: 28243129 PMCID: PMC5317338 DOI: 10.2147/ott.s123398] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Polydatin (PD) plays an important role in suppressing platelet aggregation, reducing blood lipid, restoring microcirculation and protecting from myocardial ischemia/reperfusion injury and shock. In addition, PD possesses anticancer activity. However, the effect and the mechanism of PD in regulating multiple myeloma (MM) cell survival and death are still unknown. METHODS Cell proliferation and apoptosis of RPMI 8226 cells, respectively, were analyzed by cell counting kit8 (CCK-8) assay and flow cytometry. The levels of caspase-3, cleaved caspase-3, caspase-9, cleaved caspase-9, Bcl-2 and Bax were analyzed by Western blot. Autophagy induced by PD was investigated by detecting the levels of Beclin 1, Atg5, LC3I, LC3II, HSP70 and HSP27. The autophagy inhibitor 3-methyladenine (3-MA), mTOR/p70s6k inhibitor rapamycin, and mTOR activator MHY1485 were used to analyze the mechanism of cell proliferation, apoptosis and autophagy influenced by PD. The phosphorylations of mTOR and p70s6k were detected by Western blot. RESULTS A gradual decrease in cell proliferation of RPMI 8226 cells was observed with an increase in PD concentrations (P<0.05). PD also induced cell apoptosis and autophagy in a concentration-dependent manner. Both 3-MA and MHY1485 reversed the inhibitory effect of PD on cell proliferation and attenuated the positive effect of PD on cell apoptosis and autophagy. The phosphorylation of mTOR and p70s6k was significantly suppressed by PD (P<0.05). Furthermore, inhibition of the mTOR/p70s6k signaling pathway by rapamycin significantly induced autophagy and apoptosis and inhibited cell viability (P<0.05). CONCLUSION PD effectively suppressed cell proliferation and induced apoptosis and autophagy of MM cells via the mTOR/p70s6k signaling pathway in a concentration-dependent manner in vitro, indicating that PD could be a potential anticancer drug for MM therapy.
Collapse
Affiliation(s)
- Baojun Yang
- Department of Pathology, General Hospital of PINGMEISHENMA Medical Group, Pingdingshan
| | - Shunxin Zhao
- Department of Respiratory Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, People's Republic of China
| |
Collapse
|
43
|
Induction of G2/M phase arrest and apoptosis by a new tetrahydroingenol diterpenoid from Euphorbia erythradenia Bioss. in melanoma cancer cells. Biomed Pharmacother 2017; 86:334-342. [DOI: 10.1016/j.biopha.2016.12.029] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 11/27/2016] [Accepted: 12/04/2016] [Indexed: 11/21/2022] Open
|