1
|
Hasan‐Abad A, Atapour A, Sobhani‐Nasab A, Motedayyen H, ArefNezhad R. Plant-Based Anticancer Compounds With a Focus on Breast Cancer. Cancer Rep (Hoboken) 2024; 7:e70012. [PMID: 39453820 PMCID: PMC11506041 DOI: 10.1002/cnr2.70012] [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: 10/18/2023] [Revised: 07/11/2024] [Accepted: 08/11/2024] [Indexed: 10/27/2024] Open
Abstract
Breast cancer is a common form of cancer among women characterized by the growth of malignant cells in the breast tissue. The most common treatments for this condition include chemotherapy, surgical intervention, radiation therapy, hormone therapy, and biological therapy. The primary issues associated with chemotherapy and radiation therapy are their adverse events and significant financial burden among patients in underdeveloped countries. This highlights the need to explore and develop superior therapeutic options that are less detrimental and more economically efficient. Plants provide an abundant supply of innovative compounds and present a promising new avenue for investigating cancer. Plants and their derivations are undergoing a revolution due to their reduced toxicity, expediency, cost-effectiveness, safety, and simplicity in comparison to conventional treatment methods. Natural products are considered promising candidates for the development of anticancer drugs, due perhaps to the diverse pleiotropic effects on target events. The effects of plant-derived products are limited to cancer cells while leaving healthy cells unaffected. Identification of compounds with strong anticancer properties and development of plant-based medications for cancer treatment might be crucial steps in breast cancer therapy. Although bioactive compounds have potent anticancer properties, they also have drawbacks that need to be resolved before their application in clinical trials and improved for the approved drugs. This study aims to give comprehensive information on known anticancer compounds, including their sources and molecular mechanisms of actions, along with opportunities and challenges in plant-based anticancer therapies.
Collapse
Affiliation(s)
| | - Amir Atapour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and TechnologiesShiraz University of Medical SciencesShirazIran
| | - Ali Sobhani‐Nasab
- Autoimmune Diseases Research CenterKashan University of Medical SciencesKashanIran
| | - Hossein Motedayyen
- Autoimmune Diseases Research CenterKashan University of Medical SciencesKashanIran
| | - Reza ArefNezhad
- Department of Anatomy, School of MedicineShiraz University of Medical SciencesShirazIran
| |
Collapse
|
2
|
Huang P, Wang X, Cao Y, Yang J, Yao R, Liang L, Cheng G, Yang L. Research progress on the use of Salvia miltiorrhiza Bunge extracts in the treatment of pulmonary diseases. Biomed Pharmacother 2024; 179:117282. [PMID: 39146764 DOI: 10.1016/j.biopha.2024.117282] [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: 06/12/2024] [Revised: 08/01/2024] [Accepted: 08/08/2024] [Indexed: 08/17/2024] Open
Abstract
Salvia miltiorrhiza Bunge extracts, known for their diverse biological activities, often have remarkable efficacy in treating pulmonary diseases overlooked due to their specific cardiovascular actions. With the recent outbreak of COVID-19, research into pulmonary-related diseases has garnered significant attention. Salvia miltiorrhiza Bunge extracts can be broadly categorized into lipophilic and hydrophilic components; however, a comprehensive summary of their mechanisms in treating pulmonary diseases is lacking. Therefore, this review aims to systematically summarize the therapeutic mechanisms of 10 major Salvia miltiorrhiza Bunge extracts in treating pulmonary fibrosis, lung cancer, acute lung injury, and chronic obstructive pulmonary disease, with the goal of identifying promising options for efficacious therapies.
Collapse
Affiliation(s)
- Peifeng Huang
- School of Integrative medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Xuezhen Wang
- School of Integrative medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yingyi Cao
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Jiaming Yang
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Rongmei Yao
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Leiqin Liang
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| | - Gong Cheng
- New Cornerstone Science Laboratory, Tsinghua University-Peking University Joint Center for Life Sciences, School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Institute of Infectious Diseases, Shenzhen Bay Laboratory, Shenzhen, Guangdong 518000, China.
| | - Long Yang
- School of Integrative medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China.
| |
Collapse
|
3
|
Zhang Q, Xia Y, Wang F, Yang D, Liang Z. Induction of ferroptosis by natural products in non-small cell lung cancer: a comprehensive systematic review. Front Pharmacol 2024; 15:1385565. [PMID: 38751790 PMCID: PMC11094314 DOI: 10.3389/fphar.2024.1385565] [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: 02/13/2024] [Accepted: 04/12/2024] [Indexed: 05/18/2024] Open
Abstract
Lung cancer is one of the leading causes of cancer-related deaths worldwide that presents a substantial peril to human health. Non-Small Cell Lung Cancer (NSCLC) is a main subtype of lung cancer with heightened metastasis and invasion ability. The predominant treatment approaches currently comprise surgical interventions, chemotherapy regimens, and radiotherapeutic procedures. However, it poses significant clinical challenges due to its tumor heterogeneity and drug resistance, resulting in diminished patient survival rates. Therefore, the development of novel treatment strategies for NSCLC is necessary. Ferroptosis was characterized by iron-dependent lipid peroxidation and the accumulation of lipid reactive oxygen species (ROS), leading to oxidative damage of cells and eventually cell death. An increasing number of studies have found that exploiting the induction of ferroptosis may be a potential therapeutic approach in NSCLC. Recent investigations have underscored the remarkable potential of natural products in the cancer treatment, owing to their potent activity and high safety profiles. Notably, accumulating evidences have shown that targeting ferroptosis through natural compounds as a novel strategy for combating NSCLC holds considerable promise. Nevertheless, the existing literature on comprehensive reviews elucidating the role of natural products inducing the ferroptosis for NSCLC therapy remains relatively sparse. In order to furnish a valuable reference and support for the identification of natural products inducing ferroptosis in anti-NSCLC therapeutics, this article provided a comprehensive review explaining the mechanisms by which natural products selectively target ferroptosis and modulate the pathogenesis of NSCLC.
Collapse
Affiliation(s)
| | | | | | | | - Zongsuo Liang
- Key Laboratory of Plant Secondary Metabolism and Regulation of Zhejiang Province, College of Life Sciences, Zhejiang Sci-Tech University, Hangzhou, China
| |
Collapse
|
4
|
Ahamed A, Hasan M, Samanta A, Alam SSM, Jamil Z, Ali S, Hoque M. Prospective pharmacological potential of cryptotanshinone in cancer therapy. PHARMACOLOGICAL RESEARCH - MODERN CHINESE MEDICINE 2023; 9:100308. [DOI: 10.1016/j.prmcm.2023.100308] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
|
5
|
Hosokawa M, Mikawa R, Hagiwara A, Okuno Y, Awaya T, Yamamoto Y, Takahashi S, Yamaki H, Osawa M, Setoguchi Y, Saito MK, Abe S, Hirai T, Gotoh S, Hagiwara M. Cryptotanshinone is a candidate therapeutic agent for interstitial lung disease associated with a BRICHOS-domain mutation of SFTPC. iScience 2023; 26:107731. [PMID: 37701577 PMCID: PMC10494175 DOI: 10.1016/j.isci.2023.107731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 08/05/2023] [Accepted: 08/23/2023] [Indexed: 09/14/2023] Open
Abstract
Interstitial lung disease (ILD) represents a large group of diseases characterized by chronic inflammation and fibrosis of the lungs, for which therapeutic options are limited. Among several causative genes of familial ILD with autosomal dominant inheritance, the mutations in the BRICHOS domain of SFTPC cause protein accumulation and endoplasmic reticulum stress by misfolding its proprotein. Through a screening system using these two phenotypes in HEK293 cells and evaluation using alveolar epithelial type 2 (AT2) cells differentiated from patient-derived induced pluripotent stem cells (iPSCs), we identified Cryptotanshinone (CPT) as a potential therapeutic agent for ILD. CPT decreased cell death induced by mutant SFTPC overexpression in A549 and HEK293 cells and ameliorated the bleomycin-induced contraction of the matrix in fibroblast-dependent alveolar organoids derived from iPSCs with SFTPC mutation. CPT and this screening strategy can apply to abnormal protein-folding-associated ILD and other protein-misfolding diseases.
Collapse
Affiliation(s)
- Motoyasu Hosokawa
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Department of Developmental Biology and Functional Genomics, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Ryuta Mikawa
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
- Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Atsuko Hagiwara
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Yukiko Okuno
- Medical Research Support Center, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Tomonari Awaya
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| | - Yuki Yamamoto
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
- Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Senye Takahashi
- Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Haruka Yamaki
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Mitsujiro Osawa
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Yasuhiro Setoguchi
- Department of Respiratory Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo 113-8519, Japan
| | - Megumu K Saito
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Shinji Abe
- Department of Respiratory Medicine Tokyo, Medical University Hospital, Shinjuku-ku, Tokyo 160-0023, Japan
| | - Toyohiro Hirai
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Shimpei Gotoh
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
- Department of Drug Discovery for Lung Diseases, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Masatoshi Hagiwara
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
6
|
An Q, Wu M, Yang C, Feng Y, Xu X, Su H, Zhang G. Salviae miltiorrhiza against human lung cancer: A review of its mechanism (Review). Exp Ther Med 2023; 25:139. [PMID: 36845955 PMCID: PMC9947574 DOI: 10.3892/etm.2023.11838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 01/10/2023] [Indexed: 02/15/2023] Open
Abstract
Lung cancer is one of the commonest malignant tumors in the world today, causing millions of mortalities every year. New methods to treat lung cancer are urgently needed. Salviae miltiorrhiza Bunge is a common Chinese medicine, often used for promoting blood circulation. In the past 20 years, Salviae miltiorrhiza has made significant progress in the treatment of lung cancer and is considered to be one of the most promising methods to fight against the disease. A great amount of research has shown that the mechanism of Salviae miltiorrhiza against human lung cancer mainly includes inhibiting the proliferation of lung cancer cells, promoting lung cancer cell apoptosis, inducing cell autophagy, regulating immunity and resisting angiogenesis. Research has shown that Salviae miltiorrhiza has certain effects on the resistance to chemotherapy drugs. The present review discussed the status and prospects of Salviae miltiorrhiza against human lung cancer.
Collapse
Affiliation(s)
- Qingwen An
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China,Key Laboratory of Blood-Stasis-Toxin Syndrome of Zhejiang Province, Hangzhou, Zhejiang 310053, P.R. China
| | - Mengting Wu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China,Key Laboratory of Blood-Stasis-Toxin Syndrome of Zhejiang Province, Hangzhou, Zhejiang 310053, P.R. China
| | - Chuqi Yang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China,Key Laboratory of Blood-Stasis-Toxin Syndrome of Zhejiang Province, Hangzhou, Zhejiang 310053, P.R. China
| | - Yewen Feng
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China,Key Laboratory of Blood-Stasis-Toxin Syndrome of Zhejiang Province, Hangzhou, Zhejiang 310053, P.R. China
| | - Xuefei Xu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China,Key Laboratory of Blood-Stasis-Toxin Syndrome of Zhejiang Province, Hangzhou, Zhejiang 310053, P.R. China
| | - Hang Su
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China,Key Laboratory of Blood-Stasis-Toxin Syndrome of Zhejiang Province, Hangzhou, Zhejiang 310053, P.R. China
| | - Guangji Zhang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China,Key Laboratory of Blood-Stasis-Toxin Syndrome of Zhejiang Province, Hangzhou, Zhejiang 310053, P.R. China,Traditional Chinese Medicine ‘Preventing Disease’ Wisdom Health Project Research Center of Zhejiang, Hangzhou, Zhejiang 310053, P.R. China,Correspondence to: Professor Guangji Zhang, School of Basic Medical Sciences, Zhejiang Chinese Medical University, 526 Binwen Road, Hangzhou, Zhejiang 310053, P.R. China
| |
Collapse
|
7
|
Noori S, Nourbakhsh M, Imani H, Deravi N, Salehi N, Abdolvahabi Z. Naringenin and cryptotanshinone shift the immune response towards Th1 and modulate T regulatory cells via JAK2/STAT3 pathway in breast cancer. BMC Complement Med Ther 2022; 22:145. [PMID: 35606804 PMCID: PMC9125892 DOI: 10.1186/s12906-022-03625-x] [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: 06/08/2021] [Accepted: 05/16/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Use of natural products has been proposed as an efficient method in modulation of immune system and treatment of cancers. The aim of this study was to investigate the potential of cryptotanshinone (CPT), naringenin, and their combination in modulating the immune response towards Th1 cells and the involvement of JAK2/STAT3 signaling pathway in these effects. METHODS Mouse models of delayed type hypersensitivity (DTH) were produced and treated with naringenin and CPT. The proliferation of spleen cells were assessed by Bromodeoxyuridine (BrdU) assay. Flowcytometry and enzyme-linked immunosorbent assay (ELISA) tests were employed to evaluate subpopulation of T-lymphocytes and the levels of cytokines, respectively. The JAK/STAT signaling pathway was analyzed by Western blotting. RESULTS We showed higher DTH, increased lymphocyte proliferation, decreased tumor growth and reduced JAK2/STAT3 phosphorylation in mice treated with naringenin and CPT. Moreover, a significant decline in the production of IL-4 and an upsurge in the production of IFN-γ by splenocytes were observed. Additionally, the population of intra-tumor CD4+CD25+Foxp3+ T cells was significantly lower in naringenin + CPT treated animals than that in controls. CONCLUSION Naringenin-CPT combination could exert immunomodulatory effects, suggesting this combination as a novel complementary therapeutic regimen for breast cancer.
Collapse
Affiliation(s)
- Shokoofe Noori
- Department of Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mitra Nourbakhsh
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Department of Biochemistry, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Hossein Imani
- Nutrition Department, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloofar Deravi
- Department of Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Niloufar Salehi
- Department of Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zohreh Abdolvahabi
- Metabolic Diseases Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| |
Collapse
|
8
|
Yang W, Han J, Gong S, Zhao J, Yu T, Ma J. Cryptotanshinone Suppressed Postmenopausal Osteoporosis by Preventing RANKL-Mediated Osteoclastogenesis against Kidney Injury. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:2821984. [PMID: 35132325 PMCID: PMC8817845 DOI: 10.1155/2022/2821984] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 01/03/2022] [Accepted: 01/07/2022] [Indexed: 11/21/2022]
Abstract
BACKGROUND Cryptotanshinone (CPT), an active component extracted from the root of Salvia miltiorrhiza Bunge, exhibits extensive favorable bioactive properties including anti-inflammatory, antioxidative, antibacterial, and antitumor effects. This study aims to investigate the effects of CPT on osteogenesis and explore related mechanisms both in vivo and in vitro. METHODS In the in vivo experiment, ovariectomized (OVX) female rats were intragastrically administered with CPT at doses of 10 mg/kg and 20 mg/kg for 13 consecutive weeks. Dual-energy X-ray absorptiometry was employed to detect bone mineral density (BMD). ELISA assay was leveraged to detect the biochemical parameters such as BUN and creatinine in the kidney samples. Bone and kidney sections were dyed by H&E and Masson staining kits. In the in vitro experiment, the RAW 264.7 cells were stimulated through the receptor activation of the nuclear factor kappa B ligand (RANKL) to establish an osteoclast differentiation model, and CPT's protective effect against bone loss was evaluated. Differentiated osteoclasts were determined by TRAP staining. While, osteoclast-marker proteins such as NFATc1, c-Fos, and cathepsin K were identified by Western blot. RESULTS The results from in vivo experiments revealed that CPT could elevate bone mass and increase bone formation markers in OVX rats. Intriguingly, CPT administration noticeably ameliorated the kidney injury in OVX rats by suppressing BUN and restoring creatinine levels. Furthermore, the results from in vitro experiments suggested that CPT downregulated the protein expression of osteoclast-associated genes such as cathepsin K, c-Fos, and NFATc1 which hinted the related potential mechanisms. CONCLUSION The evidence from in vivo and in vitro experiments suggested that CPT exerted antiosteoclastogenic effects by inhibiting the activation of osteoclast differentiation followed by suppressing the protein expressions of cathepsin K, c-Fos, and NFATc1 in osteoclast precursors, and it exhibited protective effects against kidney damage, which highlighted its advantage in clinical application.
Collapse
Affiliation(s)
- Wenjiu Yang
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong, China
| | - Jing Han
- Department of Ophthalmology, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong, China
| | - Shuo Gong
- Department of Spine Surgery, First Affiliated Hospital of Shandong First Medical University (Shandong Province Qianfoshan Hospital), Jinan, Shandong, China
| | - Jun Zhao
- Department of Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Tengbo Yu
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Jinfeng Ma
- Department of Spinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao 266003, Shandong, China
| |
Collapse
|
9
|
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
|
10
|
Garcia-Oliveira P, Otero P, Pereira AG, Chamorro F, Carpena M, Echave J, Fraga-Corral M, Simal-Gandara J, Prieto MA. Status and Challenges of Plant-Anticancer Compounds in Cancer Treatment. Pharmaceuticals (Basel) 2021; 14:ph14020157. [PMID: 33673021 PMCID: PMC7918405 DOI: 10.3390/ph14020157] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/03/2021] [Accepted: 02/06/2021] [Indexed: 12/12/2022] Open
Abstract
Nowadays, cancer is one of the deadliest diseases in the world, which has been estimated to cause 9.9 million deaths in 2020. Conventional treatments for cancer commonly involve mono-chemotherapy or a combination of radiotherapy and mono-chemotherapy. However, the negative side effects of these approaches have been extensively reported and have prompted the search of new therapeutic drugs. In this context, scientific community started to look for innovative sources of anticancer compounds in natural sources, including traditional plants. Currently, numerous studies have evaluated the anticancer properties of natural compounds derived from plants, both in vitro and in vivo. In pre-clinical stages, some promising compounds could be mentioned, such as the sulforaphane or different phenolic compounds. On the other hand, some phytochemicals obtained positive results in clinical stages and were further approved for cancer treatment, such as vinca alkaloids or the paclitaxel. Nevertheless, these compounds are not exempt of limitations, such as low solubility, restricted effect on their own, negative side-effects, etc. This review aims to compile the information about the current phytochemicals used for cancer treatment and also promising candidates, main action mechanisms and also reported limitations. In this sense, some strategies to face the limitations have been considered, such as nano-based formulations to improve solubility or chemical modification to reduce toxicity. In conclusion, although more research is still necessary to develop more efficient and safe phytochemical drugs, more of these compounds might be used in future cancer therapies.
Collapse
Affiliation(s)
- Paula Garcia-Oliveira
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
- Centro de Investigação de Montanha (CIMO), Campus de Santa Apolonia, Instituto Politécnico de Bragança, 5300-253 Bragança, Portugal
| | - Paz Otero
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
| | - Antia Gonzalez Pereira
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
- Centro de Investigação de Montanha (CIMO), Campus de Santa Apolonia, Instituto Politécnico de Bragança, 5300-253 Bragança, Portugal
| | - Franklin Chamorro
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
| | - Maria Carpena
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
| | - Javier Echave
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
| | - Maria Fraga-Corral
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
- Centro de Investigação de Montanha (CIMO), Campus de Santa Apolonia, Instituto Politécnico de Bragança, 5300-253 Bragança, Portugal
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
- Correspondence: (J.S.-G.); (M.A.P.)
| | - Miguel Angel Prieto
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
- Correspondence: (J.S.-G.); (M.A.P.)
| |
Collapse
|
11
|
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
|
12
|
Clinical application and mechanism of traditional Chinese medicine in treatment of lung cancer. Chin Med J (Engl) 2020; 133:2987-2997. [PMID: 33065603 PMCID: PMC7752681 DOI: 10.1097/cm9.0000000000001141] [Citation(s) in RCA: 83] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Lung cancer is a malignant tumor characterized by a rapid proliferation rate, less survivability, high mortality, and metastatic potential. This review focuses on updated research about the clinical application of traditional Chinese medicine (TCM) as an adjuvant therapy to lung cancer treatment and the mechanisms of TCM effect on lung cancer in vitro and in vivo. We summarized the recent 5 years of different research progress on clinical applications and antitumor mechanisms of TCM in the treatment of lung cancer. As a potent adjuvant therapy, TCM could enhance conventional treatments (chemotherapy, radiation therapy, and epidermal growth factor receptors [EGFRs] tyrosine kinase inhibitors [TKIs]) effects as well as provide synergistic effects, enhance chemotherapy drugs chemosensitivity, reverse drug resistance, reduce adverse reactions and toxicity, relieve patients’ pain and improve quality of life (QOL). After treating with TCM, lung cancer cells will induce apoptosis and/or autophagy, suppress metastasis, impact immune reaction, and therapeutic effect of EGFR-TKIs. Therefore, TCM is a promisingly potent adjuvant therapy in the treatment of lung cancer and its multiple mechanisms are worthy of an in-depth study.
Collapse
|
13
|
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: 3.4] [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
|
14
|
Liu Y, Xie X, Hou X, Shen J, Shi J, Chen H, He Y, Wang Z, Feng N. Functional oral nanoparticles for delivering silibinin and cryptotanshinone against breast cancer lung metastasis. J Nanobiotechnology 2020; 18:83. [PMID: 32473632 PMCID: PMC7260741 DOI: 10.1186/s12951-020-00638-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 05/16/2020] [Indexed: 02/07/2023] Open
Abstract
Background Breast cancer lung metastasis occurs in more than 60% of all patients with breast cancer, and most of those afflicted by it eventually die of recurrence. The tumor microenvironment plays vital roles in metastasis. Modulating the tumor microenvironment via multiple pathways could efficiently prevent or inhibit lung metastasis. Silibinin and cryptotanshinone are natural plant products that demonstrate anti-metastasis effects and modulate the tumor microenvironment via different pathways. However, they have poor aqueous solubility, membrane permeability, and oral bioavailability. Oral drug administration may help improve the quality of life and compliance of patients with breast cancer, primarily under long-term and/or follow-up therapy. Herein, we developed poly-N-(2-hydroxypropyl) methacrylamide (pHPMA)-coated wheat germ agglutinin-modified lipid-polymer hybrid nanoparticles, co-loaded with silibinin and cryptotanshinone (S/C-pW-LPNs). We assessed their oral bioavailability, and evaluated their anti-metastasis efficacy in a 4T1 breast cancer tumor-bearing nude mouse model. Results An in vitro mucus diffusion study revealed that pHPMA enhanced W-LPN mucus penetration. After oral administration, pHPMA enhanced nanoparticle distribution in rat jejunum and substantially augmented oral bioavailability. S/C-W-LPNs markedly increased 4T1 cell toxicity and inhibited cell invasion and migration. Compared to LPNs loaded with either silibinin or cryptotanshinone alone, S/C-pW-LPNs dramatically slowed tumor progression in 4T1 tumor-bearing nude mice. S/C-pW-LPNs presented with the most robust anti-metastasis activity on smooth lung surfaces and mitigated lung metastasis foci. They also downregulated tumor microenvironment biomarkers such as CD31, TGF-β1, and MMP-9 that promote metastasis. Conclusions Silibinin- and cryptotanshinone-co-loaded pW-LPNs efficiently penetrate intestinal barriers, thereby enhancing the oral bioavailability of the drug loads. These nanoparticles exhibit favorable anti-metastasis effects in breast cancer-bearing nude mice. Hence, S/C-pW-LPNs are promising oral drug nanocarriers that inhibit breast cancer lung metastasis.
Collapse
Affiliation(s)
- Ying Liu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, People's Republic of China
| | - Xingmei Xie
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, People's Republic of China
| | - Xuefeng Hou
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, People's Republic of China
| | - Junyi Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, People's Republic of China
| | - Jiangpei Shi
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, People's Republic of China
| | - Haizhen Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, People's Republic of China
| | - Yuanzhi He
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, People's Republic of China
| | - Zhi Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, People's Republic of China
| | - Nianping Feng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Zhangjiang Hi-Tech Park, Pudong New District, Shanghai, 201203, People's Republic of China.
| |
Collapse
|
15
|
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: 1.8] [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
|
16
|
Vundavilli H, Datta A, Sima C, Hua J, Lopes R, Bittner M. Cryptotanshinone Induces Cell Death in Lung Cancer by Targeting Aberrant Feedback Loops. IEEE J Biomed Health Inform 2019; 24:2430-2438. [PMID: 31825884 DOI: 10.1109/jbhi.2019.2958042] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Signaling pathways oversee highly efficient cellular mechanisms such as growth, division, and death. These processes are controlled by robust negative feedback loops that inhibit receptor-mediated growth factor pathways. Specifically, the ERK, the AKT, and the S6K feedback loops attenuate signaling via growth factor receptors and other kinase receptors to regulate cell growth. Irregularity in any of these supervised processes can lead to uncontrolled cell proliferation and possibly Cancer. These irregularities primarily occur as mutated genes, and an exhaustive search of the perfect drug combination by performing experiments can be both costly and complex. Hence, in this paper, we model the Lung Cancer pathway as a Modified Boolean Network that incorporates feedback. By simulating this network, we theoretically predict the drug combinations that achieve the desired goal for the majority of mutations. Our theoretical analysis identifies Cryptotanshinone, a traditional Chinese herb derivative, as a potent drug component in the fight against cancer. We validated these theoretical results using multiple wet lab experiments carried out on H2073 and SW900 lung cancer cell lines.
Collapse
|
17
|
Wang H, Zhang Y, Zhang Y, Liu W, Wang J. Cryptotanshinone inhibits lung cancer invasion via microRNA-133a/matrix metalloproteinase 14 regulation. Oncol Lett 2019; 18:2554-2559. [PMID: 31452742 DOI: 10.3892/ol.2019.10580] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 04/04/2019] [Indexed: 12/24/2022] Open
Abstract
Cryptotanshinone inhibits the proliferative and colony formation abilities of human non-small cell lung cancer cells (NSCLCs). The present study reported that signal transducer and activator of transcription 3 is not the only target of cryptotanshinone during the inhibition of human NSCLCs. It was identified that cryptotanshinone upregulates the expression levels of microRNA (miR)-30d-5p, miR-126-3p, miR-133a, miR-338-3p and miR-451a, and downregulates miR-21-5p, miR-96-5p, miR-182-5p and miR-205-5p. Among these, miR-133a was the most significantly upregulated. miR-133a targets and downregulates the expression of matrix metalloproteinase (MMP)14; however, MMP15, MMP16 and MMP24 were determined to be unaffected. This process was identified to be independent of tissue inhibitor of metalloproteinases 2. Cryptotanshinone also suppresses the invasion of human NSCLCs, which may be due to the inhibited expression of MMP14. In conclusion, cryptotanshinone may serve as a potential therapeutic agent in the treatment of lung cancer.
Collapse
Affiliation(s)
- Huijuan Wang
- Department of Tumor Chemotherapy, Tumor Hospital of Wuwei, Wuwei, Gansu 733000, P.R. China
| | - Yanshan Zhang
- Department of Thoracic Surgery, Tumor Hospital of Wuwei, Wuwei, Gansu 733000, P.R. China
| | - Yingguo Zhang
- Department of Thoracic Surgery, Tumor Hospital of Wuwei, Wuwei, Gansu 733000, P.R. China
| | - Wenling Liu
- Department of Thoracic Surgery, Tumor Hospital of Wuwei, Wuwei, Gansu 733000, P.R. China
| | - Jihong Wang
- Department of Thoracic Surgery, Tumor Hospital of Wuwei, Wuwei, Gansu 733000, P.R. China
| |
Collapse
|
18
|
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: 48] [Impact Index Per Article: 8.0] [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
|
19
|
Liu S, Han Z, Trivett AL, Lin H, Hannifin S, Yang D, Oppenheim JJ. Cryptotanshinone has curative dual anti-proliferative and immunotherapeutic effects on mouse Lewis lung carcinoma. Cancer Immunol Immunother 2019; 68:1059-1071. [PMID: 30972427 PMCID: PMC6584267 DOI: 10.1007/s00262-019-02326-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Accepted: 03/19/2019] [Indexed: 12/21/2022]
Abstract
Lung cancer is currently the leading cause of cancer-related mortality with very limited effective therapy. Screening of a variety of traditional Chinese medicines (TCMs) for their capacity to inhibit the proliferation of human lung cancer A549 cells and to induce the in vitro maturation of human DCs led to the identification of cryptotanshinone (CT), a compound purified from the TCM Salvia miltiorrhiza Bunge. Here, CT was shown to inhibit the proliferation of mouse Lewis lung carcinoma (LLC) cells by upregulating p53, downregulating cyclin B1 and Cdc2, and, consequently, inducing G2/M cell-cycle arrest of LLC cells. In addition, CT promoted maturation of mouse and human DCs with upregulation of costimulatory and MHC molecules and stimulated DCs to produce TNFα, IL-1β, and IL-12p70, but not IL-10 in vitro. CT-induced maturation of DCs depended on MyD88 and also involved the activation of NF-κB, p38, and JNK. CT was effective in the treatment of LLC tumors and, when used in combination with low doses of anti-PD-L1, cured LLC-bearing mice with the induction of subsequent anti-LLC long-term specific immunity. CT treatment promoted T-cell infiltration and elevated the expression of genes typical of Th1 polarization in LLC tumor tissue. The therapeutic effect of CT and low doses of anti-PD-L1 was reduced by depletion of CD4 and CD8 T cells. This paper provides the first report that CT induces immunological antitumor activities and may provide a new promising antitumor immunotherapeutic.
Collapse
Affiliation(s)
- Shuo Liu
- 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.,Guang An Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - 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
| | - 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
| | - Hongsheng Lin
- Guang An Men Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - 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
|
20
|
Ma Z, Fan Y, Wu Y, Kebebe D, Zhang B, Lu P, Pi J, Liu Z. Traditional Chinese medicine-combination therapies utilizing nanotechnology-based targeted delivery systems: a new strategy for antitumor treatment. Int J Nanomedicine 2019; 14:2029-2053. [PMID: 30962686 PMCID: PMC6435121 DOI: 10.2147/ijn.s197889] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cancer is a major public health problem, and is now the world’s leading cause of death. Traditional Chinese medicine (TCM)-combination therapy is a new treatment approach and a vital therapeutic strategy for cancer, as it exhibits promising antitumor potential. Nano-targeted drug-delivery systems have remarkable advantages and allow the development of TCM-combination therapies by systematically controlling drug release and delivering drugs to solid tumors. In this review, the anticancer activity of TCM compounds is introduced. The combined use of TCM for antitumor treatment is analyzed and summarized. These combination therapies, using a single nanocarrier system, namely codelivery, are analyzed, issues that require attention are determined, and future perspectives are identified. We carried out a systematic review of >280 studies published in PubMed since 1985 (no patents involved), in order to provide a few basic considerations in terms of the design principles and management of targeted nanotechnology-based TCM-combination therapies.
Collapse
Affiliation(s)
- Zhe Ma
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ; .,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ;
| | - Yuqi Fan
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ; .,School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yumei Wu
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ; .,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ;
| | - Dereje Kebebe
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ; .,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ; .,School of Pharmacy, Institute of Health Sciences, Jimma University, Jimma, Ethiopia
| | - Bing Zhang
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ; .,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ;
| | - Peng Lu
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ; .,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ;
| | - Jiaxin Pi
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ; .,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ;
| | - Zhidong Liu
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ; .,Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China, ;
| |
Collapse
|
21
|
Ji Y, Liu Y, Xue N, Du T, Wang L, Huang R, Li L, Yan C, Chen X. Cryptotanshinone inhibits esophageal squamous-cell carcinoma in vitro and in vivo through the suppression of STAT3 activation. Onco Targets Ther 2019; 12:883-896. [PMID: 30774375 PMCID: PMC6357882 DOI: 10.2147/ott.s187777] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Purpose Esophageal squamous-cell carcinoma (ESCC) is the most common subtype of esophageal cancer, with a poor clinical outcome. Cryptotanshinone (CTS) is the main bioactive compound from the root of Salvia miltiorrhiza Bunge. Our study aimed to investigate the anti-cancer effects and molecular mechanisms of CTS on ESCC. Materials and methods We investigated the anti-tumor activity of CTS on ESCC in vitro and in vivo. Activation of the STAT3 signaling pathway was evaluated in ESCC and HEK-Blue™ IL-6 cells. Cell viability was assessed by the MTT assay. Apoptosis and cell cycle arrest were assessed using flow cytometry. Cell migration was detected by a scratch wound assay. Results CTS inhibited STAT3 expression and IL-6-mediated STAT3 activation in esophageal cancer cells. Subsequently, CTS dose-dependently inhibited the proliferation of esophageal cancer cells via induction of cell apoptosis. Furthermore, CTS suppressed the migration of esophageal cancer cells. In vivo, CTS inhibited tumor growth of EC109 cell in xenograft mice without any obvious effect on body weight. Conclusion Our results indicated that STAT3 inhibition may be a therapeutic target for esophageal cancer. CTS could provide a potential approach for esophageal cancer therapy by influencing the janus kinase-2/STAT3 signaling pathway.
Collapse
Affiliation(s)
- Yubin Ji
- Research Center on Life Sciences and Environmental Sciences, Harbin University of Commerce, Harbin 150076, People's Republic of China
| | - Yichen Liu
- Research Center on Life Sciences and Environmental Sciences, Harbin University of Commerce, Harbin 150076, People's Republic of China.,State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China,
| | - Nina Xue
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China,
| | - Tingting Du
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China,
| | - Liyuan Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China,
| | - Rui Huang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China,
| | - Ling Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China,
| | - Chunhong Yan
- Georgia Cancer Center, Augusta University, Augusta, GA, USA,
| | - Xiaoguang Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, People's Republic of China,
| |
Collapse
|
22
|
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: 9] [Impact Index Per Article: 1.3] [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
|
23
|
Wu JP, Kang NX, Zhang MY, Gao HW, Li XR, Liu YL, Xu QM, Yang SL. Oleiferoside W from the roots of Camellia oleifera C. Abel, inducing cell cycle arrest and apoptosis in A549 cells. JOURNAL OF ASIAN NATURAL PRODUCTS RESEARCH 2018; 20:793-806. [PMID: 28679317 DOI: 10.1080/10286020.2017.1347640] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 06/23/2017] [Indexed: 06/07/2023]
Abstract
Camellia oleifera C. Abel has been widely cultivated in China, and a group of bioactive constituents such as triterpeniod saponin have been isolated from C. oleifera C. Abel. In the current study, a new triterpeniod saponin was isolated from the EtOH extract of the roots of C. oleifera C. Abel, named as oleiferoside W, and the cytotoxic properties of oleiferoside W were evaluated in non-small cell lung cancer A549 cells. At the same time the inducing apoptosis, the depolarization of mitochondrial membrane potential (Δψ), the up-regulation of related pro-apoptotic proteins, such as cleaved-PARP, cleaved-caspase-3, and the down-regulation of anti-apoptotic marker Bcl-2/Bax were measured on oleiferoside W. Furthermore, the function, inducing the generation of reactive oxygen species (ROS) and apoptosis, of oleiferoside W could be reversed by N-acetylcysteine (NAC). In conclusion, our findings showed that oleiferoside W induced apoptosis involving mitochondrial pathway and increasing intracellular ROS production in the A549 cells, suggesting that oleiferoside W may have the possibility to be a useful anticancer agent for therapy in lung cancer.
Collapse
Affiliation(s)
- Jiang-Ping Wu
- a College of Pharmaceutical Science, Bozhou Vocational and Technical College , Bozhou 236800 , China
- b College of Pharmaceutical Science, Soochow University , Suzhou 215123 , China
| | - Nai-Xin Kang
- b College of Pharmaceutical Science, Soochow University , Suzhou 215123 , China
| | - Mi-Ya Zhang
- b College of Pharmaceutical Science, Soochow University , Suzhou 215123 , China
| | - Hong-Wei Gao
- b College of Pharmaceutical Science, Soochow University , Suzhou 215123 , China
| | - Xiao-Ran Li
- b College of Pharmaceutical Science, Soochow University , Suzhou 215123 , China
| | - Yan-Li Liu
- b College of Pharmaceutical Science, Soochow University , Suzhou 215123 , China
| | - Qiong-Ming Xu
- b College of Pharmaceutical Science, Soochow University , Suzhou 215123 , China
| | - Shi-Lin Yang
- b College of Pharmaceutical Science, Soochow University , Suzhou 215123 , China
- c College of Pharmaceutical Science, Jiangxi University of Traditional Chinese Medicine , Nanchang 330006 , China
| |
Collapse
|
24
|
Chen ZM, Huang L, Li MM, Meng L, Ying SC, Xu AM. Inhibitory effects of isocryptotanshinone on gastric cancer. Sci Rep 2018; 8:9307. [PMID: 29915371 PMCID: PMC6006307 DOI: 10.1038/s41598-018-27638-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 06/07/2018] [Indexed: 02/07/2023] Open
Abstract
Gastric cancer (GC) is one of the most common digestive malignancies globally, and the prognosis of patients with advanced tumors remains poor. Isocryptotanshinone (ICTS), isolated from Salvia miltiorrhiza, was found to inhibit the proliferation of lung and breast cancer cells. However, whether ICTS has anticancer activities against GC is unknown. In the present study, we reported that the proliferation of GC cells was inhibited by ICTS in a dose- and time-dependent manner. After treatment with ICTS, GC cells were arrested in the G1/G0 phase of cell cycle and the apoptotic cells were induced in a dose-dependent manner. Additionally, ICTS suppressed the expression of cell cycle- and apoptosis-associated proteins (e.g., Cyclin D1, phosphorylated Rb, E2F1, Mcl-1, Bcl-2, and Survivin). ICTS inhibited the phosphorylation of STAT3 in a dose-dependent manner. Down-regulated STAT3 attenuated the expression of Cyclin D1, p-Rb, and Survivin, which remarkably increased the sensitivity of ICTS in GC cells; overexpression of STAT3 restored the cell growth and proliferation and the protein expression suppressed by ICTS. ICTS also suppressed the xenograft tumor growth in BALB/c nude mice. Together, these data indicate that ICTS inhibits GC proliferation by inducing G1/G0 cell cycle arrest and apoptosis via inhibiting the STAT3 signaling pathway.
Collapse
Affiliation(s)
- Zhang-Ming Chen
- Department of Gastrointestinal Surgery, Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Lei Huang
- Department of Gastrointestinal Surgery, Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| | - Miao-Miao Li
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Lei Meng
- Department of Gastrointestinal Surgery, Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Song-Cheng Ying
- Department of Immunology, School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
| | - A-Man Xu
- Department of Gastrointestinal Surgery, Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China.
| |
Collapse
|
25
|
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: 5.1] [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
|
26
|
Shen L, Lou Z, Zhang G, Xu G, Zhang G. Diterpenoid Tanshinones, the extract from Danshen (Radix Salviae
Miltiorrhizae) induced apoptosis in nine human cancer cell lines. J TRADIT CHIN MED 2017; 36:514-21. [PMID: 28459519 DOI: 10.1016/s0254-6272(16)30069-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
OBJECTIVE To identify the active anti-tumor constituents
in the extract from Danshen (Radix Salviae
Miltiorrhizae) and investigate the mechanisms underlying
the actions. METHODS First, we introduced a two-step counter-
current chromatography to extract the therapeutically
active diterpenoid, tanshinone from
Danshen (Radix Salviae Miltiorrhizae). The cholecystokinin
(CCK-8) method was used to evaluate
the inhibitory effect of diterpenoid tanshinone in
liver cancer QGY-7703, lung cancer PC9, lung cancer
A549, gastric cancer MKN-45, gastric cancer
HGC-27, colon cancer HCT116, myeloma cellU266/
RPMI8226, and human breast cancer MCF-7 in vitro.
Fluorescence staining was used to observe
the cytotoxicity ofditerpenoid tanshinone on PC9
cells. The Western blot was used to detect apoptosis-
related protein poly ADP-ribose polymerase
(PARP), cysteinyl aspartate specific proteinase3/9
(caspase3/9), and cleaved-cysteinyl aspartate specific
proteinase3/9 (cleaved-caspase3/9). The endoplasmic
reticulum stress-related activating transcription factor 4 (ATF4), phosphorylated eukaryotic
initiation factor 2α (p-eIF2α), and phosphorylated
jun amino-terminal kinase (p-JNK), and caspase-
12 were also analyzed using the Western
blot. RESULTS Diterpenoid tanshinone inhibited the
nine human tumor cell lines, with an IC50 of
4.37-29 μg/mL, with the PC9 and MCF-7 displaying
the lowest values. Fluorescence staining showed a
lethal effect of diterpenoid tanshinone on PC9
cells. The Western blot showed that the expression
of caspase3/9 protein and ATF-4 protein decreased
gradually. However, the PARP, cleaved-caspase 3/9
and the expression of p-eIF2 α, P-JNK, and caspase-
12 increased gradually, in a dose-dependent fashion. CONCLUSION We successfully introduced a
two-step counter-current chromatography method
to extract diterpenoid tanshinone, and demonstrated
its antitumor activity. Diterpenoid tanshinone
can induce apoptosis in nine human cancer cell lines.
Collapse
|
27
|
Ke F, Wang Z, Song X, Ma Q, Hu Y, Jiang L, Zhang Y, Liu Y, Zhang Y, Gong W. Cryptotanshinone induces cell cycle arrest and apoptosis through the JAK2/STAT3 and PI3K/Akt/NFκB pathways in cholangiocarcinoma cells. Drug Des Devel Ther 2017; 11:1753-1766. [PMID: 28670110 PMCID: PMC5479302 DOI: 10.2147/dddt.s132488] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Cholangiocarcinoma (CCA) is the most common biliary tract malignancy in the world with high resistance to current chemotherapies and extremely poor prognosis. The main objective of this study was to investigate the inhibitory effects of cryptotanshinone (CTS), a natural compound isolated from Salvia miltiorrhiza Bunge, on CCA both in vitro and in vivo and to explore the underlying mechanisms of CTS-induced apoptosis and cell cycle arrest. METHODS The anti-tumor activity of CTS on HCCC-9810 and RBE cells was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-tetrazolium bromide (MTT) assay and colony forming assays. Cell cycle changes were detected by flow cytometric analysis. Apoptosis was detected by annexin V/propidium iodide double staining and Hoechst 33342 staining assays. The efficacy of CTS in vivo was evaluated using a HCCC-9810 xenograft model in athymic nude mice. The expression of key proteins involved in cell apoptosis and signaling pathway in vitro was analyzed by Western blot analysis. RESULTS CTS induced potent growth inhibition, S-phase arrest, apoptosis, and colony-forming inhibition in HCCC-9810 and RBE cells in a dose-dependent manner. Intraperitoneal injection of CTS (0, 10, or 25 mg/kg) for 4 weeks significantly inhibited the growth of HCCC-9810 xenografts in athymic nude mice. CTS treatment induced S-phase arrest with a decrease of cyclin A1 and an increase of cyclin D1 protein level. Bcl-2 expression was downregulated remarkably, while Bax expression was increased after apoptosis occurred. Additionally, the activation of JAK2/STAT3 and PI3K/Akt/NFκB was significantly inhibited in CTS-treated CCA cells. CONCLUSION CTS induced CCA cell apoptosis by suppressing both the JAK2/STAT3 and PI3K/Akt/NFκB signaling pathways and altering the expression of Bcl-2/Bax family, which was regulated by these two signaling pathways. CTS may serve as a potential therapeutic agent for CCA.
Collapse
Affiliation(s)
- Fayong Ke
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
- Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Zheng Wang
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
- Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Xiaoling Song
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
- Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Qiang Ma
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
- Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Yunping Hu
- Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Lin Jiang
- Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Yijian Zhang
- Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Yingbin Liu
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
- Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Yong Zhang
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| | - Wei Gong
- Department of General Surgery, Xinhua Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
- Institute of Biliary Disease Research, School of Medicine, Shanghai Jiao Tong University, Shanghai, People’s Republic of China
| |
Collapse
|
28
|
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: 6.8] [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
|
29
|
Cryptotanshinone inhibits human glioma cell proliferation in vitro and in vivo through SHP-2-dependent inhibition of STAT3 activation. Cell Death Dis 2017; 8:e2767. [PMID: 28492557 PMCID: PMC5520699 DOI: 10.1038/cddis.2017.174] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/16/2017] [Accepted: 03/20/2017] [Indexed: 12/22/2022]
Abstract
Malignant gliomas (MGs) are one of the most common primary brain cancers in adults with a high mortality rate and relapse rate. Thus, finding better effective approaches to treat MGs has become very urgent. Here, we studied the effects of cryptotanshinone (CTS) on MGs in vitro and in vivo, and explored the underlying mechanisms. Effects of CTS in vitro on cell proliferation, cycle, migration and invasion were evaluated. The activation of JAK/STATs signaling was detected by western blot and immunofluorescenc staining. SHP-2 inhibitor or SiRNA were used to determine the involvement of SHP-2. The in vivo anti-MGs activity of CTS was studied with nude mice bearing intracerebral U87 xenografts. Our results revealed that CTS significantly inhibited the proliferation of MGs in vitro via inhibiting STAT3 signal pathway. The cell cycle was arrested at G0/G1 phase. Although CTS did not change the expression of total SHP-2 protein, the tyrosine phosphatase activity of SHP-2 protein was increased by CTS treatment in a dose-dependent manner in vivo and in vitro. SHP-2 inhibitor or SiRNA could reverse the inhibitory effect of CTS on phosphorylation of STAT3 Tyr705. In vivo study also showed that CTS inhibited the intracranial tumor growth and extended survival of nude mice bearing intracerebral U87 xenografts, confirming an inhibitory effect of CTS on MGs. Our results indicated CTS may be a potential therapeutic agent for MGs. The inhibitory action of CTS is largely attributed to the inhibition of STAT3 Tyr705 phosphorylation with a novel mechanism of upregulating the tyrosine phosphatase activity of SHP-2 protein.
Collapse
|
30
|
Wang J, Zhang G, Dai C, Gao X, Wu J, Shen L, Chen Z, Liu P. Cryptotanshinone potentiates the antitumor effects of doxorubicin on gastric cancer cells via inhibition of STAT3 activity. J Int Med Res 2017; 45:220-230. [PMID: 28222632 PMCID: PMC5536615 DOI: 10.1177/0300060516685513] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Objective To investigate the synergistic effects of cryptotanshinone (CPT) and doxorubicin (DOXO) on induction of apoptosis in human gastric cancer cells and the mechanisms. Methods Cell proliferation and apoptosis were detected using the CCK8 assay and AnnexinV/PI staining, respectively. Western blotting was used to determine the levels and phosphorylation of proteins encoded by STAT3-regulated genes and the cleaved forms of caspases and PARP. Results CPT significantly potentiated the antiproliferative effect of DOXO in gastric cancer cell lines. CPT combined with DOXO induced apoptosis and cleavage of caspases-3,-7,-9 as well as PARP. CPT or a STAT3 siRNA significantly suppressed constitutive and IL-6-induced phosphorylation of STAT3 Tyr705, decreasing the levels of proteins encoded by STAT3-target genes (Bcl-xL, Mcl-1, survivin, and XIAP). Conclusions CPT enhanced the anticancer activity of DOXO in gastric cancer cells via STAT3 inactivation and suppression STAT3-regulated antiapoptotic gene expression, indicating that DOXO combined with CPT may serve as effective therapy for gastric cancer.
Collapse
Affiliation(s)
- Jiye Wang
- 1 The Criminal Science and Technology Department, Zhejiang Police College, Hangzhou, PR China
| | - Guangji Zhang
- 3 College of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Chunyan Dai
- 2 Zhejiang Key Laboratory of Gastro-Intestinal Pathophysiology, Zhejiang Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Xiufei Gao
- 2 Zhejiang Key Laboratory of Gastro-Intestinal Pathophysiology, Zhejiang Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Jianbin Wu
- 1 The Criminal Science and Technology Department, Zhejiang Police College, Hangzhou, PR China
| | - Li Shen
- 4 Center of Post-doctoral Studies, China Academy of Chinese Medicine Science, Beijing, China
| | - Zhe Chen
- 2 Zhejiang Key Laboratory of Gastro-Intestinal Pathophysiology, Zhejiang Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Pei Liu
- 2 Zhejiang Key Laboratory of Gastro-Intestinal Pathophysiology, Zhejiang Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, PR China
| |
Collapse
|
31
|
Wu J, Xu H, Zhang L, Zhang X. Radix Astragali and Tanshinone Help Carboplatin Inhibit B16 Tumor Cell Growth. Technol Cancer Res Treat 2016; 15:583-8. [PMID: 26041399 DOI: 10.1177/1533034615588682] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Accepted: 04/30/2015] [Indexed: 11/15/2022] Open
Abstract
Background: Excessive UV radiation causes increased melanoma incidence. Postoperation chemotherapy will destroy lymphocytes and compromise immune response. Immunodepression is also detected in patients with cancers. Previous studies suggested that polysaccharide–protein complexes manifested immunomodulatory and antitumor activities. Radix Astragali (RA) extract is a product of polysaccharide–protein complexes, which has been used in the treatment of a variety of diseases because of its low toxicity to the host. Tanshinone (TA) is a derivative of phenanthrenequinone isolated from Danshen, which is suggested to inhibit tumor growth by inducing apoptosis in tumor cells. Carboplatin (CA) is a commonly used chemotherapeutic drug in melanoma treatment. Therefore, we hypothesized that the combination of RA and TA will help CA better inhibit the B16 cell growth. Purpose: The study will test that the efficacy of growth inhibition of tumor cell produced by CA + RA + TA is better than CA + RA or CA + TA. Methods: The B16 tumor cells were injected to Swiss-Hauschka (ICR) mice subcutaneously. Twenty-four hours later, mice received CA intraperitoneally, CA + RA (RA were administered gastrically at the dosage of 10 g/kg body weight), CA + TA (TA were administered gastrically at the dosage of 0.5 g/kg body weight), or no treatment (model group). Tumor weight, volume, latency, incidence, the percentage of CD4+ and CD8+ in spleen, and natural killer (NK), and cytotoxic lymphocyte (CTL) activities were measured and compared among different groups. Results: Compared with mice treated with CA + RA, CA + TA, or CA alone, the mice treated with CA + RA + TA showed (1) significantly smaller tumor weight and tumor volume; (2) significantly longer tumor latency; (3) significantly lower tumor incidence; and (4) significantly increased percentage of CD4+ and CD8+ in spleen and increased activities of NK and CTL. Conclusion: Combination of RA and TA can help CA produce more effective inhibition on B16 cell growth.
Collapse
Affiliation(s)
- Jinyi Wu
- Department of Cardiology, China-Japan Union Hospital, Jilin University, China
| | - Haiming Xu
- Department of Cardiology, China-Japan Union Hospital, Jilin University, China
| | - Lei Zhang
- Department of Cardiology, China-Japan Union Hospital, Jilin University, China
| | - Xiuying Zhang
- Department of Histology and Embryology, School of Basic Medical Sciences, Capital Medical University, China
| |
Collapse
|
32
|
Guo S, Luo W, Liu L, Pang X, Zhu H, Liu A, Lu J, Ma DL, Leung CH, Wang Y, Chen X. Isocryptotanshinone, a STAT3 inhibitor, induces apoptosis and pro-death autophagy in A549 lung cancer cells. J Drug Target 2016; 24:934-942. [PMID: 26904961 DOI: 10.3109/1061186x.2016.1157882] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is a potential drug target for chemotherapy. Cryptotanshinone (CTS) was identified as a potent STAT3 inhibitor, while the effect of other tanshinones remains unknown. In this study, the influence of eight tanshinones on STAT3 activity was initially screened and isocryptotanshinone (ICTS) significantly inhibited STAT3 activity in a dual luciferase assay. ICTS inhibited the constitutive and inducible phosphorylation of STAT3 at Y705 without affecting the phosphorylation of STAT3 at S727 in A549 lung cancer cells. Furthermore, ICTS inhibited the nuclear translocation of STAT3. Compared with CTS, ICTS exhibited a stronger inhibitory effect on STAT3 phosphorylation and on A549 cytotoxicity. ICTS induced autophagy as evidenced by the accumulation of autophagic vacuoles and the increased expression of LC3 protein and autophagosomes. ICTS-induced cell death was partially reversed by the autophagy inhibitor chloroquine. The docking assay predicted that both ICTS and CTS bind the SH2 domain of STAT3. ICTS formed hydrogen bonds and pi-pi interaction with the nearby amino acid residues of Lys591, Arg609, and Ser636. These findings suggested that ICTS, a natural compound, is a potent STAT3 inhibitor. ICTS induced apoptosis and pro-death autophagy in A549 cells.
Collapse
Affiliation(s)
- Shuhui Guo
- a State Key Laboratory of Quality Research in Chinese Medicine , Institute of Chinese Medical Sciences, University of Macau , Macau , China
| | - Weiwei Luo
- a State Key Laboratory of Quality Research in Chinese Medicine , Institute of Chinese Medical Sciences, University of Macau , Macau , China
| | - Lijuan Liu
- a State Key Laboratory of Quality Research in Chinese Medicine , Institute of Chinese Medical Sciences, University of Macau , Macau , China
| | - Xiaocong Pang
- b Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Hong Zhu
- c Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences , Zhejiang University , Hangzhou , Zhejiang , China
| | - Ailin Liu
- b Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College , Beijing , China
| | - Jinjian Lu
- a State Key Laboratory of Quality Research in Chinese Medicine , Institute of Chinese Medical Sciences, University of Macau , Macau , China
| | - Dik-Lung Ma
- d Department of Chemistry , Hong Kong Baptist University , Kowloon Tong , Hong Kong
| | - Chung-Hang Leung
- a State Key Laboratory of Quality Research in Chinese Medicine , Institute of Chinese Medical Sciences, University of Macau , Macau , China
| | - Yitao Wang
- a State Key Laboratory of Quality Research in Chinese Medicine , Institute of Chinese Medical Sciences, University of Macau , Macau , China
| | - Xiuping Chen
- a State Key Laboratory of Quality Research in Chinese Medicine , Institute of Chinese Medical Sciences, University of Macau , Macau , China
| |
Collapse
|
33
|
Anticancer activity of cryptotanshinone on acute lymphoblastic leukemia cells. Arch Toxicol 2015; 90:2275-2286. [DOI: 10.1007/s00204-015-1616-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 10/19/2015] [Indexed: 12/21/2022]
|
34
|
Gao H, Sun W, Zhao W, Hao W, Leung CH, Lu J, Chen X. Total Tanshinones-Induced Apoptosis and Autophagy Via Reactive Oxygen Species in Lung Cancer 95D Cells. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2015; 43:1265-79. [DOI: 10.1142/s0192415x1550072x] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tanshinones are a group of bioactive constituents isolated from Salvia miltiorrhiza Bunge, a widely prescribed traditional Chinese herb. In the current study, the anticancer properties of total tanshinones (TDT) were evaluated using 95D lung cancer cells. Tanshinone IIA was identified as the main component of TDT. Compared with tanshinone IIA, TDT showed more cytotoxic effects on the 95D cells. Annexin V/7-AAD double staining, the depolarization of mitochondrial membrane potential (MMP) (Δψ), the up-regulation of pro-apoptotic proteins, such as cleaved-PARP, cleaved-caspase-3, Bax, and Bad, and the down-regulation of anti-apoptotic protein Bcl-2 were evidence of TDT-induced apoptosis. Furthermore, TDT-induced autophagy as demonstrated by monodansylcadaverine (MDC) staining and the up-regulation of autophagy-associated proteins, such as LC3-II, Beclin-1, Atg3, Atg5, Atg7, and Atg12. Autophagy inhibitors, 3-methyladenine (3-MA) and bafilomycin A1, enhanced TDT-induced cell death. 3-MA pretreatment enhanced the TDT-induced up-regulation of Bax and cleaved-PARP. In addition, TDT induced the generation of reactive oxygen species (ROS), which was reversed by N-acetylcysteine (NAC). NAC also reversed TDT-induced depolarization of Δψ, MDC staining, up-regulation of Bax, cleaved-PARP, Beclin-1, LC3-II, and cell viability. In conclusion, our findings showed that TDT-induced apoptosis and protective autophagy in 95D cells mediated by increasing intracellular ROS production.
Collapse
Affiliation(s)
- Hongwei Gao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Wen Sun
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Wenwen Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Wenhui Hao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Chung-Hang Leung
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Jinjian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| |
Collapse
|
35
|
Ge Y, Yang B, Chen Z, Cheng R. Cryptotanshinone suppresses the proliferation and induces the apoptosis of pancreatic cancer cells via the STAT3 signaling pathway. Mol Med Rep 2015; 12:7782-8. [PMID: 26459366 DOI: 10.3892/mmr.2015.4379] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 08/27/2015] [Indexed: 11/06/2022] Open
Abstract
Pancreatic cancer remains a challenging disease worldwide. Cryptotanshinone (CPT) is one of the active constituents of Salvia miltiorrhiza Bunge and exhibits significant antitumor activities in several human cancer cells. However, the efficacy and molecular mechanism of CPT in pancreatic cancer remains to be elucidated. In the present study, the effect of CPT on the proliferation, apoptosis and cell cycle of human pancreatic cancer cell BxPC‑3 cells was evaluated. The results demonstrated that CPT inhibited proliferation of the BxPC‑3 cells in a concentration‑dependent manner, and significantly induced cell apoptosis and cell cycle arrest. The protein levels of cleaved caspase‑3, caspase‑9 and poly ADP ribose polymerase were upregulated, while the levels of c‑myc, survivin and cyclin D1 were downregulated following treatment with CPT. In addition, CPT decreased the activities of signal transducer and activator of transcription 3 (STAT3) and several upstream regulatory signaling pathways after 24 h. However, CPT only inhibited the phosphorylation of STAT3 Tyr705 within 30 min, without marked effects on the phosphorylation of the other proteins. These results suggested that the inhibition of STAT3 activity by CPT was directly and independent of the upstream regulators in human pancreatic cancer. The present study demonstrated that CPT exerts anticancer effects by inducing apoptosis and cell cycle arrest via inhibition of the STAT3 signaling pathway in human BxPC-3 cells.
Collapse
Affiliation(s)
- Yuqing Ge
- National Clinical Research Base of Traditional Chinese Medicine, The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Bo Yang
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Zhe Chen
- National Clinical Research Base of Traditional Chinese Medicine, The First Affiliated Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| | - Rubin Cheng
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310053, P.R. China
| |
Collapse
|
36
|
Zhang X, Luo W, Zhao W, Lu J, Chen X. Isocryptotanshinone Induced Apoptosis and Activated MAPK Signaling in Human Breast Cancer MCF-7 Cells. J Breast Cancer 2015; 18:112-8. [PMID: 26155286 PMCID: PMC4490259 DOI: 10.4048/jbc.2015.18.2.112] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Accepted: 06/05/2015] [Indexed: 02/06/2023] Open
Abstract
Purpose Isocryptotanshinone (ICTS) is a natural bioactive product that is isolated from the roots of the widely used medical herb Salvia miltiorrhiza. However, few reports exist on the mechanisms underlying the therapeutic effects of ICTS. Here, we report that ICTS has anticancer activity and describe the mechanism underlying this effect. Methods The antiproliferative effect of ICTS was determined using 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) and clonogenic assays. The effect of ICTS on the cell cycle was measured using flow cytometry. Apoptosis was determined by Hoechst 33342 staining, DNA fragmentation assays, and Western blotting for apoptotic proteins. Finally, the effect of ICTS on mitogen-activated protein kinases (MAPKs) was determined by Western blotting. Results ICTS significantly inhibited proliferation of MCF-7 and MDA-MB-231 human breast cancer cells, HepG2 human liver cancer cells, and A549 human lung cancer cells in vitro. Among the tested cell lines, MCF-7 cells showed the highest sensitivity to ICTS. ICTS significantly inhibited colony formation by MCF-7 cells. Furthermore, exposure of MCF-7 cells to ICTS induced cell cycle arrest at the G1 phase and decreased mitochondrial membrane potential. Hoechst 33342 staining and Western blot analysis for apoptotic proteins suggested that ICTS induced apoptosis in MCF-7 cells. In addition, ICTS activated MAPK signaling in MCF-7 cells by inducing time- and concentration-dependent phosphorylation of JNK, ERK, and p38 MAPK. Conclusion Our results suggest that ICTS inhibited MCF-7 cell proliferation by inducing apoptosis and activating MAPK signaling pathways.
Collapse
Affiliation(s)
- Xuenong Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Weiwei Luo
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Wenwen Zhao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Jinjian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China
| |
Collapse
|
37
|
Cryptotanshinone, a Stat3 inhibitor, suppresses colorectal cancer proliferation and growth in vitro. Mol Cell Biochem 2015; 406:63-73. [PMID: 25912550 DOI: 10.1007/s11010-015-2424-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/22/2015] [Indexed: 01/10/2023]
Abstract
Cryptotanshinone (CPT) is a natural compound extracted from herbal medicine that has been previously shown to possess antitumor properties in various types of human cancer cells. In the present study, we examined the potential role of CPT in the treatment of colorectal cancer. Using SW480, HCT116, and LOVO colorectal cancer cell lines, the effects of CPT on cell viability, apoptosis, and tumorigenicity were evaluated. The results showed that CPT significantly inhibited the growth and viability of SW480, HCT116, and LOVO cell lines by inducing apoptosis and prevented anchorage dependent growth on agar. In addition, CPT inhibited the activation of Signal transducer and activator of transcription 3 (Stat3) pathways in colorectal cancer cells. Stat3 is a transcription factor that mediates the expression of various genes associated with many cellular processes, such as inflammation and cell growth, and has been shown to promote several cancer types, including colorectal cancer. These findings indicate that CPT may be a potential candidate for the treatment and prevention of colorectal cancer in part by inhibiting the activation of Stat3.
Collapse
|
38
|
Zhang Y, Chen L, Li F, Wang H, Yao Y, Shu J, Ying MZ. Cryptotanshinone protects against adriamycin-induced mitochondrial dysfunction in cardiomyocytes. PHARMACEUTICAL BIOLOGY 2015; 54:237-42. [PMID: 25858002 DOI: 10.3109/13880209.2015.1029052] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
CONTEXT The serious side effect of Adriamycin (ADR) is cardiomyopathy. Cryptotanshinone (CRY) is widely and safely used as antioxidant with MTD more than 5 mg/g in rats (p.o). OBJECTIVE The objective of this study is to study the protection effects of CRY against ADR-induced mitochondrial dysfunction in cardiomyocytes. MATERIALS AND METHODS The chemical administration lasted for 20 days with an effective dose of CRY (p.o.) at 50 mg/kg in rats. Mitochondrial respiratory chain complex activities, ATP generation, mitochondrial membrane potential (MMP), superoxide anion free radical, oxidative stress-relative enzymes, and mitochondrial biogenesis-relative factors in normal control, ADR (i.p., 1.25 mg/kg), and ADR (i.p., 1.25 mg/kg) + CYP (p.o., 50 mg/kg) groups were detected. RESULTS 50 mg/kg CRY significantly promoted the energy production of ATP (16.99 ± 2.38 nmol/g Pro) (Pro: Protein) by increasing the complexes activities except II (p > 0.05). After the treatment of CRY, the suppressed MMP was increased while superoxide anion free radical (0.57 ± 0.07/mg Pro) was inhibited markedly. Mitochondrial biogenesis-relative factors PGC-1α, NRF-1, and TFAM were also promoted. Remarkable augmentations of NO, inducible nitric oxide synthase (iNOS), and increased activity of GSH-PX (p < 0.05) were also detected after the treatment of CRY, while no obvious changes on the activity of nitric oxide synthase (cNOS; p > 0.05) were observed. DISCUSSION AND CONCLUSION These results suggest that CRY protects against ADR-induced mitochondrial dysfunction in cardiomyocytes. It could be an ideal potential drug of cardioprotection.
Collapse
MESH Headings
- Adenosine Triphosphate/metabolism
- Animals
- Cardiomyopathies/chemically induced
- Cardiomyopathies/metabolism
- Cardiomyopathies/prevention & control
- Cardiotoxicity/prevention & control
- Disease Models, Animal
- Doxorubicin/toxicity
- Drugs, Chinese Herbal/isolation & purification
- Drugs, Chinese Herbal/pharmacology
- Drugs, Chinese Herbal/therapeutic use
- Male
- Membrane Potential, Mitochondrial/drug effects
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Oxidative Stress/drug effects
- Phenanthrenes/isolation & purification
- Phenanthrenes/pharmacology
- Phenanthrenes/therapeutic use
- Rats, Wistar
- Salvia miltiorrhiza/chemistry
Collapse
Affiliation(s)
- Yanshan Zhang
- a Department of Tumor Surgery , Wuwei Tumor Hospital , Wuwei, Gansu PR China
| | - Liang Chen
- b Department of Paediatrics , Changhai Hospital, Second Military Medical University , Shanghai , PR China
| | - Fan Li
- c International Medical Center, Chinese PLA General Hospital , Beijing PR China
| | - Huijuan Wang
- d Department of Tumor Chemotherapy , Wuwei Tumor Hospital , Wuwei, Gansu , PR China
| | - Yunyi Yao
- e Research Center for Biochemistry and Molecular Biology, Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College , Xuzhou, Jiangsu , PR China , and
| | - Jiamei Shu
- f Department of Cardiology , The Second Affiliated Hospital of Soochow University , Suzhou, Jiangsu PR China
| | - Ming-Zhong Ying
- c International Medical Center, Chinese PLA General Hospital , Beijing PR China
| |
Collapse
|