1
|
Kar E, Övenler Z, Hacıoğlu C, Kar F. Boric Acid Induces Oxidative Damage and Apoptosis Through SEMA3A/PLXNA1/NRP1 Signalling Pathway in U251 Glioblastoma Cell. J Cell Mol Med 2025; 29:e70578. [PMID: 40318008 PMCID: PMC12049150 DOI: 10.1111/jcmm.70578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/17/2025] [Accepted: 04/21/2025] [Indexed: 05/07/2025] Open
Abstract
Glioblastoma is one of the deadliest cancers with a very low chance of survival. Glioblastomas have a poor prognosis because of their infiltrative nature, which makes them difficult to totally isolate with rigorous surgery, radiation, and chemotherapy. Our aim in this study was to investigate the efficacy of boric acid, which has anti-cancer properties, on glioblastoma, which has very limited treatment options. U251 human glioblastoma cell lines were treated with IC25 (15.62 μg/mL), IC50 (31.25 μg/mL) and IC75 (62.5 μg/mL) doses of boric acid. Cell viability and proliferation levels were tested. At the same time, the activity of boric acid on cells was tested through oxidative stress, apoptosis, and semaphorin signalling pathway parameters. Our findings indicate that boric acid induced dose-dependent oxidative stress, cellular growth inhibition, apoptosis and morphological changes in U251 cells. Additionally, treatments with increasing amounts of boric acid resulted in a rise in the production of biomarkers of the semaphorin pathway, which may limit cell growth and proliferation. We found that boric acid activates apoptosis by triggering ROS formation at high doses and at the same time inhibits cell proliferation by increasing semaphorin signalling pathway expressions. Boric acid may act as an anti-cancer agent by activating different mechanisms in a dose-dependent manner.
Collapse
Affiliation(s)
- Ezgi Kar
- Department of Nutrition and Dietetics, Faculty of Health SciencesKutahya Health Sciences UniversityKutahyaTurkey
| | - Zeynep Övenler
- Faculty of MedicineKutahya Health Sciences UniversityKutahyaTurkey
| | - Ceyhan Hacıoğlu
- Department of Medical Biochemistry, Faculty of MedicineDuzce UniversityDuzceTurkey
| | - Fatih Kar
- Department of Medical Biochemistry, Faculty of MedicineKutahya Health Sciences UniversityKutahyaTurkey
| |
Collapse
|
2
|
Benedict A, Suresh V, Selvamani M, Jayaraman S, Hussein MA. Merremia emarginata Extract Potentiates the Inhibition of Human Colon Cancer Cells (HT-29) via the Modulation of Caspase-3/Bcl-2-Mediated Pathways. Cureus 2024; 16:e56300. [PMID: 38629020 PMCID: PMC11019472 DOI: 10.7759/cureus.56300] [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: 02/14/2024] [Accepted: 03/16/2024] [Indexed: 04/19/2024] Open
Abstract
Background This study investigates Merremia emarginata's curative effectiveness against colon cancer cells. M. emarginata, often known as Elika jemudu, is a Convolvulaceae family plant. The inhibitory ability of anticancer herbal extracts against cancer cell growth and mediators is tested. Aim This study aims to evaluate the potent anticancer activity of M. emarginata against colon cancer cell line (HT-29). Materials and methods M. emarginata leaves were gathered and processed using solvent extraction. Anticancer activity on colon cancer cells was measured using the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) test and cysteine aspartic acid protease-3 (caspase 3), B-cell lymphoma 2 (Bcl-2), and B-cell lymphoma-extra large (Bcl-xL) mRNA expressions. The data was reported as the mean ± SD of three separate experiments done in triplicate. The statistical analysis was carried out using one-way analysis of variance (ANOVA), with a p-value less than 0.05 indicating statistical significance. Results The cell viability test showed a gradual decrease in cell growth and proliferation as the concentration increased. The ethanolic extract of M. emarginata was found to be cytotoxic against colon caller cell lines. The extract was able to induce apoptosis of cancer as revealed by Bcl-2, Bcl-xL, and caspase-3 (p<0.05 and p<0.001) signaling pathways. Conclusion M. emarginata extracts showed good anticancer activity against colon cancer cell lines. Further work is required to establish and identify the chemical constituent responsible for its anticancer activity.
Collapse
Affiliation(s)
- Andrew Benedict
- Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, IND
| | - Vasugi Suresh
- Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, IND
| | - Muthamizh Selvamani
- Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, IND
| | - Selvaraj Jayaraman
- Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, IND
| | - Mohammed Asif Hussein
- Physiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Science, Saveetha University, Chennai, IND
| |
Collapse
|
3
|
Chen JJ, Yan QL, Bai M, Liu Q, Song SJ, Yao GD. Deoxyelephantopin, a germacrane-type sesquiterpene lactone from Elephantopus scaber, induces mitochondrial apoptosis of hepatocarcinoma cells by targeting Hsp90α in vitro and in vivo. Phytother Res 2023; 37:702-716. [PMID: 36420857 DOI: 10.1002/ptr.7654] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 09/06/2022] [Accepted: 09/10/2022] [Indexed: 11/25/2022]
Abstract
Hepatocellular carcinoma has been known as the most frequent subtype of liver cancer with a high rate of spread, metastases, and recurrence, also dismal treatment effects. However, effective therapies for HCC are still required. Nowadays, natural products have been known as a valuable source for drug discovery. In this research, 44 sesquiterpene lactones isolated from the Elephantopus scaber Linn. (Asteraceae) were tested by MTT assay for the antitumor activities. Deoxyelephantopin (DET) was found to exert significant cytotoxicity on HepG2 and Hep3B cells. Moreover, we found that DET treatment markedly reduced the growth of HCC cells in a concentration-dependent manner, which was better than sorafenib. Furthermore, DET induced mitochondrial dysfunction, oxidative stress, and cellular apoptosis. Additionally, we found that DET and sorafenib synergistically induced apoptosis and mitochondrial dysfunction in HCC cells. DET combined with sorafenib was also efficacious in tumor xenograft model. Molecular docking experiments revealed that DET had a potentially high binding affinity with Hsp90α. Moreover, Drug Affinity Responsive Target Stability assay suggested that DET could directly target Hsp90α. Additionally, the expression of Hsp90α was both decreased in vitro and in vivo. Altogether, this study revealed that DET might be a promising agent for HCC therapy by targeting Hsp90α.
Collapse
Affiliation(s)
- Jing-Jie Chen
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Qiu-Lin Yan
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Ming Bai
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Qingbo Liu
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Shao-Jiang Song
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Guo-Dong Yao
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
4
|
Naeem A, Hu P, Yang M, Zhang J, Liu Y, Zhu W, Zheng Q. Natural Products as Anticancer Agents: Current Status and Future Perspectives. Molecules 2022; 27:molecules27238367. [PMID: 36500466 PMCID: PMC9737905 DOI: 10.3390/molecules27238367] [Citation(s) in RCA: 167] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/22/2022] [Accepted: 11/28/2022] [Indexed: 12/02/2022] Open
Abstract
Natural products have been an invaluable and useful source of anticancer agents over the years. Several compounds have been synthesized from natural products by modifying their structures or by using naturally occurring compounds as building blocks in the synthesis of these compounds for various purposes in different fields, such as biology, medicine, and engineering. Multiple modern and costly treatments have been applied to combat cancer and limit its lethality, but the results are not significantly refreshing. Natural products, which are a significant source of new therapeutic drugs, are currently being investigated as potential cytotoxic agents and have shown a positive trend in preclinical research and have prompted numerous innovative strategies in order to combat cancer and expedite the clinical research. Natural products are becoming increasingly important for drug discovery due to their high molecular diversity and novel biofunctionality. Furthermore, natural products can provide superior efficacy and safety due to their unique molecular properties. The objective of the current review is to provide an overview of the emergence of natural products for the treatment and prevention of cancer, such as chemosensitizers, immunotherapeutics, combinatorial therapies with other anticancer drugs, novel formulations of natural products, and the molecular mechanisms underlying their anticancer properties.
Collapse
Affiliation(s)
- Abid Naeem
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Pengyi Hu
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Ming Yang
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Jing Zhang
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Yali Liu
- Key Laboratory of Pharmacodynamics and Safety Evaluation, Health Commission of Jiangxi Province, Nanchang Medical College, Nanchang 330006, China
- Key Laboratory of Pharmacodynamics and Quality Evaluation on Anti-Inflammatory Chinese Herbs, Jiangxi Administration of Traditional Chinese Medicine, Nanchang Medical College, Nanchang 330006, China
| | - Weifeng Zhu
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
| | - Qin Zheng
- Key Laboratory of Modern Preparation of Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China
- Correspondence:
| |
Collapse
|
5
|
Migheli R, Virdis P, Galleri G, Arru C, Lostia G, Coradduzza D, Muroni MR, Pintore G, Podda L, Fozza C, De Miglio MR. Antineoplastic Properties by Proapoptotic Mechanisms Induction of Inula viscosa and Its Sesquiterpene Lactones Tomentosin and Inuviscolide. Biomedicines 2022; 10:2739. [PMID: 36359261 PMCID: PMC9687476 DOI: 10.3390/biomedicines10112739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 10/15/2022] [Accepted: 10/24/2022] [Indexed: 08/30/2023] Open
Abstract
Cancer is a complex disease including approximately 200 different entities that can potentially affect all body tissues. Among the conventional treatments, radiotherapy and chemotherapy are most often applied to different types of cancers. Despite substantial advances in the development of innovative antineoplastic drugs, cancer remains one of the most significant causes of death, worldwide. The principal pitfall of successful cancer treatment is the intrinsic or acquired resistance to therapeutic agents. The development of more effective or synergistic therapeutic approaches to improve patient outcomes and minimize toxicity has become an urgent issue. Inula viscosa is widely distributed throughout Europe, Africa, and Asia. Used as a medicinal plant in different countries, I. viscosa has been characterized for its complex chemical composition in order to identify the bioactive compounds responsible for its biological activities, including anticancer effects. Sesquiterpene lactones (SLs) are natural, biologically active products that have attracted considerable attention due to their biological activities. SLs are alkylating agents that form covalent adducts with free cysteine residues within enzymes and key proteins favoring cancer cell cytotoxicity. They are effective inducers of apoptosis in several cancer cell types through different molecular mechanisms. This review focuses on recent advances in the cytotoxic effects of I. viscosa and SLs in the treatment of neoplastic diseases, with a special emphasis on their proapoptotic molecular mechanisms.
Collapse
Affiliation(s)
- Rossana Migheli
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Patrizia Virdis
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Grazia Galleri
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Caterina Arru
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy
| | - Giada Lostia
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | | | - Maria Rosaria Muroni
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Giorgio Pintore
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Luigi Podda
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | - Claudio Fozza
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy
| | | |
Collapse
|
6
|
Zhou Y, Liu J, Zhang J, Xu Y, Li W, Gao P, Xing Y, Huang L, Qin X, Jin S. Chinese endemic medicinal plant Bolbostemma paniculatum (Maxim.) Franquet: A comprehensive review. Front Pharmacol 2022; 13:974054. [PMID: 36160391 PMCID: PMC9490187 DOI: 10.3389/fphar.2022.974054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/08/2022] [Indexed: 11/24/2022] Open
Abstract
Bolbostemma paniculatum (Maxim.) Franquet is a unique species in China with a long history of medicinal use, which has the effects of detoxifying, dissolving lumps and dispersing swellings. And it is commonly used to treat many diseases, such as carbuncle and sore, acute mastitis, mammary cancer, scrofula and subcutaneous nodule traditionally. Modern clinical studies have found that B. paniculatum and its compounds can be used for the treatment of a variety of cancers, mastitis, hyperplasia of mammary glands, chronic lymphadenitis, cervical lymph tuberculosis and surgical wart skin diseases, and the curative effect is positive. At present, a variety of Chinese patent medicines containing B. paniculatum have been exploited and marketed in China for the treatment of cancers, breast diseases and flat warts. This review article comprehensively discussed the traditional application, botany, chemical components, pharmacological activities, and quality control of B. paniculatum, put forward some noteworthy issues and suggestions in current studies, and briefly discussed the possible development potential of this plant as well as future research perspectives. 96 compounds have been isolated from B. paniculatum, including triterpenoids, sterols, alkaloids and other components, of which triterpenoid saponins are the main bioactive components. The crude extracts and monomer compounds of B. paniculatum have a wide range of pharmacological activities, such as anti-tumor, antiviral, anti-inflammatory, immunoregulatory, and so on. Moreover, its anti-tumor mechanism involves many aspects, including inhibiting cell proliferation, promoting cell apoptosis, blocking the cell cycle, interfering with cell invasion and metastasis, suppressing angiogenesis, and regulating autophagy. While there is a lack of systematic and in-depth research on its anti-tumor active components and mechanism of action at the moment; and a tight connection between the chemical composition and pharmacological activity of B. paniculatum has also not been established. Besides, a systematic quality determination standard for B. paniculatum should also be built, in order to carry out further research.
Collapse
Affiliation(s)
- Yujiao Zhou
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Junyu Liu
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jianqiong Zhang
- Pediatric Department, Ya’an City Hospital of Traditional Chinese Medicine, Ya’an, Sichuan, China
| | - Yi Xu
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wangni Li
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Pang Gao
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yanghuan Xing
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lehong Huang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xuhua Qin
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Xuhua Qin, ; Shenrui Jin,
| | - Shenrui Jin
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- *Correspondence: Xuhua Qin, ; Shenrui Jin,
| |
Collapse
|
7
|
Wang CL, Gao MZ, Gao DM, Guo YH, Gao Z, Gao XJ, Wang JQ, Qiao MQ. Tubeimoside-1: A review of its antitumor effects, pharmacokinetics, toxicity, and targeting preparations. Front Pharmacol 2022; 13:941270. [PMID: 35910383 PMCID: PMC9335946 DOI: 10.3389/fphar.2022.941270] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Tubeimoside-1 (TBMS-1), a natural triterpenoid saponin found in traditional Chinese herbal medicine Bolbostemmatis Rhizoma, is present in numerous Chinese medicine preparations. This review aims to comprehensively describe the pharmacology, pharmacokinetics, toxicity and targeting preparations of TBMS-1, as well the therapeutic potential for cancer treatement. Information concerning TBMS-1 was systematically collected from the authoritative internet database of PubMed, Web of Science, and China National Knowledge Infrastructure applying a combination of keywords involving “tumor,” “pharmacokinetics,” “toxicology,” and targeting preparations. New evidence shows that TBMS-1 possesses a remarkable inhibitory effect on the tumors of the respiratory system, digestive system, nervous system, genital system as well as other systems in vivo and in vitro. Pharmacokinetic studies reveal that TBMS-1 is extensively distributed in various tissues and prone to degradation by the gastrointestinal tract after oral administration, causing a decrease in bioavailability. Meanwhile, several lines of evidence have shown that TBMS-1 may cause adverse and toxic effects at high doses. The development of liver-targeting and lung-targeting preparations can reduce the toxic effect of TBMS-1 and increase its efficacy. In summary, TBMS-1 can effectively control tumor treatment. However, additional research is necessary to investigate in vivo antitumor effects and the pharmacokinetics of TBMS-1. In addition, to reduce the toxicity of TBMS-1, future research should aim to modify its structure, formulate targeting preparations or combinations with other drugs.
Collapse
Affiliation(s)
- Chang-Lin Wang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Research and Innovation Team of Emotional Diseases and Syndromes in Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ming-Zhou Gao
- Research and Innovation Team of Emotional Diseases and Syndromes in Shandong University of Traditional Chinese Medicine, Jinan, China
- Institute of Traditional Chinese Medicine Innovation, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dong-Mei Gao
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Research and Innovation Team of Emotional Diseases and Syndromes in Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ying-Hui Guo
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Research and Innovation Team of Emotional Diseases and Syndromes in Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhan Gao
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Research and Innovation Team of Emotional Diseases and Syndromes in Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiang-Ju Gao
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Research and Innovation Team of Emotional Diseases and Syndromes in Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jie-Qiong Wang
- Research and Innovation Team of Emotional Diseases and Syndromes in Shandong University of Traditional Chinese Medicine, Jinan, China
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Jie-Qiong Wang, ; Ming-Qi Qiao,
| | - Ming-Qi Qiao
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Research and Innovation Team of Emotional Diseases and Syndromes in Shandong University of Traditional Chinese Medicine, Jinan, China
- *Correspondence: Jie-Qiong Wang, ; Ming-Qi Qiao,
| |
Collapse
|
8
|
MicroRNA-34a Promotes Ischemia-Induced Cardiomyocytes Apoptosis through Targeting Notch1. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1388415. [PMID: 35265142 PMCID: PMC8901351 DOI: 10.1155/2022/1388415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 01/23/2022] [Accepted: 02/03/2022] [Indexed: 11/22/2022]
Abstract
Myocardial apoptosis occurs during myocardial ischemia. This study aimed to determine the effect of microRNA-34a (miR-34a) in ischemia-induced myocardial apoptosis. Mainly, SD rats were subjected to myocardial ischemia by ligaturing the left anterior descending branch of coronary artery. After rats had myocardial infarction, HE staining and TUNEL staining confirmed a significant increase in apoptosis. The expression of miR-34a was noticeably upregulated, while the expression of Notch1 was downregulated. An increase in caspase-3 and a decrease in Bcl-2/Bax ratio were observed in myocardium. Similar results were observed in the in vitro model of cardiomyocyte ischemia and anoxia of this study. When rat cardiomyocytes were administered with serum starvation and microaerophilic system, apoptosis-related proteins were significantly increased. However, transfecting the miR-34a inhibitor into the cardiomyocyte before the serum starvation and hypoxia treatment could increase the ratio of Bcl-2/Bax and downregulate the expression of caspase-3, as well as prevent cardiomyocytes from apoptosis. As opposed to the abovementioned points, the upregulation of miR-34a expression by transfecting miR-34a mimics induced Notch1 reduce and apoptosis-related proteins increase apparently, while upregulation of Notch1 could stimulate apoptosis attributed to miR-34a. Mechanistically, we demonstrated that Notch1 is a direct target of miR-34a. In conclusion, our current results suggested that miR-34a significantly stimulates ischemia-induced cardiomyocytes apoptosis by targeting Notch1.
Collapse
|
9
|
Guo L, Yang Y, Sheng Y, Wang J, Ruan S, Han C. Mechanism of piperine in affecting apoptosis and proliferation of gastric cancer cells via ROS-mitochondria-associated signalling pathway. J Cell Mol Med 2021; 25:9513-9522. [PMID: 34464498 PMCID: PMC8505830 DOI: 10.1111/jcmm.16891] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 12/20/2022] Open
Abstract
Piperine (PIP), the main active ingredient in pepper, belongs to the cinnamamide alkaloid. PIP has been found to have functions, including anti-oxidation, immune regulation, anti-tumour and promotion of drug metabolism. The present study was mainly designed to reveal the anti-tumour effect of PIP against gastric cancer and the relevant mechanism. In brief, the undifferentiated human gastric cancer cell HGC-27 was used, which was treated with different concentrations of PIP. As a result, PIP could inhibit proliferation and induce apoptosis of HGC-27 cells in a dose-dependent manner. The mechanism of PIP was associated with ROS increase and mitochondrial damage, simultaneously, the expression of key proteins of apoptosis was affected, including Bcl-2, Bax, Cyt-c, Caspase-9 and Caspase-3. Pre-treatment of ROS scavenger NAC HGC-27 cells could significantly reduce PIP-induced apoptosis and inhibit the activation of apoptotic signals. Consistently, PIP could induce ROS to increase and activate apoptotic signals in the animal model. Therefore, the present study showed that PIP can induce the generation of ROS, thereby promoting the activation of mitochondrial apoptotic pathway and exerting anti-tumour effects.
Collapse
Affiliation(s)
- Li Guo
- Department of Center LaboratoryThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Yi Yang
- Department of PharmacyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - YongJia Sheng
- Department of PharmacyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Jin Wang
- Department of PharmacyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Shuiliang Ruan
- Department of GastroenterologyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| | - Chenyang Han
- Department of PharmacyThe Second Affiliated Hospital of Jiaxing UniversityJiaxingChina
| |
Collapse
|
10
|
Sun Y, Jing Y, Zhang Y. Serum lncRNA-ANRIL and SOX9 expression levels in glioma patients and their relationship with poor prognosis. World J Surg Oncol 2021; 19:287. [PMID: 34556140 PMCID: PMC8461887 DOI: 10.1186/s12957-021-02392-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 09/03/2021] [Indexed: 01/16/2023] Open
Abstract
Background lncRNA-CDKN2B antisense RNA 1 (ANRIL) and SRY-box transcription factor 9 (SOX9) has abnormal expression in many tumors including glioma, but the underlying molecular mechanism is unclear. This study set out to investigate the serum lncRNA-ANRIL and SOX9 levels in glioma patients and their effects on prognosis. Methods We enrolled 142 glioma patients admitted to our hospital from May 2014 to May 2016 into the research group (RG) and 120 healthy subjects receiving concurrent physical examinations into the control group (CG). Fasting peripheral blood (4 mL each) was sampled from subjects from the two groups. Using the quantitative real-time polymerase chain reaction (qRT-PCR), lncRNA-ANRIL and SOX9 were measured to explore their values in the early diagnosis of glioma. Patients from RG were followed up for 3 years to analyze the influence of lncRNA-ANRIL and SOX9 on patient prognosis. We purchased glioma cell lines U251 and U87 and grouped them according to the transfection of different plasmids. We conducted CCK8 assay to test cell proliferation, Transwell assay to test cell invasion, the flow cytometry to test cell apoptosis, and Western Blot assay to measure bcl-2 and bax protein levels. Results ANRIL and SOX9 were evidently higher in RG than in CG (P<0.01). The receiver operating characteristic (ROC) curve revealed that the diagnostic sensitivity of ANRIL combined with SOX9 for glioma was 81.62%, and the specificity was 90.83% (P<0.01). ANRIL and SOX9 were closely related to tumor grade, tumor diameter, distant metastasis, and family history of glioma (P<0.01). In total, 135 patients were successfully followed up (95.07%). Patients with high levels of ANRIL and SOX9 had a markedly poorer prognosis than those with low levels (P<0.05). ANRIL and SOX9 were markedly higher in glioma cell lines (U251 and U87) than in normal brain cells (P<0.01). The proliferation and invasion of U251 cells were notably reduced after the transfection of ANRIL and SOX9 inhibitory sequences (P<0.01), but the apoptosis was notably increased (P<0.01). Bcl-2 expression was markedly increased in lncRNA-ANRIL-inhibitor and SOX9-inhibitor (P<0.01), while bax expression was markedly reduced in lncRNA-ANRIL-inhibitor and SOX9-inhibitor (P<0.01). Conclusion lncRNA-ANRIL and SOX9 levels were higher in glioma patients than in healthy people. High-lncRNA-ANRIL and SOX9 levels were strongly associated with unfavorable prognosis of patients. The testing of biological behaviors revealed that lncRNA-ANRIL and SOX9 worked as tumor-promoting genes in glioma.
Collapse
Affiliation(s)
- Youlu Sun
- Department of Neurosurgery, Guangrao County People's Hospital, No. 180 Huayuan Road, Dongying, Guangrao County, 257300, P.R. China
| | - Yuesong Jing
- Department of Neurosurgery, The Second People's Hospital Of Dongying, Dongying, 257335, P.R. China
| | - Yuxin Zhang
- Department of Neurosurgery, Guangrao County People's Hospital, No. 180 Huayuan Road, Dongying, Guangrao County, 257300, P.R. China.
| |
Collapse
|
11
|
Xu D, Ran C, Yin L, Lin J, Fu H, Peng X, Zhao X, Shu G. Acute and Subchronic Toxicity Studies of Aristolochic Acid A in Tianfu Broilers. Animals (Basel) 2021; 11:ani11061556. [PMID: 34071750 PMCID: PMC8228413 DOI: 10.3390/ani11061556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/19/2021] [Accepted: 05/25/2021] [Indexed: 11/21/2022] Open
Abstract
Simple Summary Aristolochic acid (AA) is a chemical compound present in traditional Chinese medicine, which is widely used for anti-infection, anti-viral and anti-bacterial treatment, as antibiotics were banned in the poultry industry. However, long-term use of this drug in high doses can cause harmful damage to the kidneys of animals. Therefore, in this study, the acute toxicity test and subchronic toxicity test of AA were carried out in order to explore the nephrotoxicity mechanism of AA on Tianfu broilers. In this study, the LD50 of AA to male Tianfu broilers was 14.52 mg/kg. Subchronic exposure to high doses of AA in broilers can cause nephrotoxicity by breaking the redox balance to form oxidative stress, along with promoting apoptosis of renal cells. In conclusion, AA has been found to damage broilers’ kidneys in high doses. This study provides suggestions for the clinical application of traditional Chinese medicine containing AA in the poultry industry. Abstract Aristolochic acid (AA) is one of the components of some traditional Chinese medicines, which has high toxic potential in animals, leading to huge economic losses in the breeding industry. The purpose of this study is to evaluate the toxicology of AA on Tianfu broilers through acute and subchronic toxicity tests. The results showed that the median lethal dose of AA to Tianfu broilers was 14.52 mg/kg. After continuous intraperitoneal injection of AA solution (1.452 mg/kg) for 28 days, the swollen and necrotic renal tubular epithelial cells were histologically observed; in addition, blood urea nitrogen (BUN) and creatinine (Cre) were significantly increased, indicating AA could induce serious kidney lesions in broilers. Moreover, the ROS, the apoptosis rate and the depolarization rate of the mitochondrial membrane potential of broilers’ renal cells increased. The results of QRT-PCR showed that AA reduced the mRNA expressions of HO-1, NQO1, Raf-1 and Bcl-2, while the expressions of Bax and Caspase-3 increased, which show that AA aroused oxidative stress and promoted the apoptosis of renal cells. In conclusion, AA has been found to damage broilers’ kidneys by breaking the redox balance to form oxidative stress, along with promoting apoptosis of renal cells.
Collapse
Affiliation(s)
- Dan Xu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 610000, China; (D.X.); (X.Z.)
| | - Chonglin Ran
- Department of Veterinary Medicine, Sichuan Agricultural University, Chengdu 610000, China; (C.R.); (L.Y.); (J.L.); (H.F.); (X.P.)
| | - Lizi Yin
- Department of Veterinary Medicine, Sichuan Agricultural University, Chengdu 610000, China; (C.R.); (L.Y.); (J.L.); (H.F.); (X.P.)
| | - Juchun Lin
- Department of Veterinary Medicine, Sichuan Agricultural University, Chengdu 610000, China; (C.R.); (L.Y.); (J.L.); (H.F.); (X.P.)
| | - Hualin Fu
- Department of Veterinary Medicine, Sichuan Agricultural University, Chengdu 610000, China; (C.R.); (L.Y.); (J.L.); (H.F.); (X.P.)
| | - Xi Peng
- Department of Veterinary Medicine, Sichuan Agricultural University, Chengdu 610000, China; (C.R.); (L.Y.); (J.L.); (H.F.); (X.P.)
| | - Xiaoling Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu 610000, China; (D.X.); (X.Z.)
| | - Gang Shu
- Department of Veterinary Medicine, Sichuan Agricultural University, Chengdu 610000, China; (C.R.); (L.Y.); (J.L.); (H.F.); (X.P.)
- Correspondence:
| |
Collapse
|
12
|
Li C, Wu G, Zhao H, Dong N, Wu B, Chen Y, Lu Q. Natural-Derived Polysaccharides From Plants, Mushrooms, and Seaweeds for the Treatment of Inflammatory Bowel Disease. Front Pharmacol 2021; 12:651813. [PMID: 33981232 PMCID: PMC8108135 DOI: 10.3389/fphar.2021.651813] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 03/22/2021] [Indexed: 12/13/2022] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease impairing the gastrointestinal tract, and its incidence and prevalence have been increasing over time worldwide. IBD greatly reduces peoples' quality of life and results in several life-threatening complications, including polyp, toxic colonic dilatation, intestinal perforation, gastrointestinal bleeding, and cancerization. The current therapies for IBD mainly include drugs for noncritical patients and operation for critical patients. However, continuous use of these drugs causes serious side effects and increased drug resistance, and the demand of effective and affordable drugs with minimal side effects for IBD sufferers is urgent. Natural-derived polysaccharides are becoming a research hotspot for their therapeutic effects on IBD. This study focuses on the research progress of various natural polysaccharides from plants, seaweeds, and mushrooms for the treatment of IBD during recent 20 years. Regulation of oxidative stress, inflammatory status, gut microbiota, and immune system and protection of the intestinal epithelial barrier function are the underlying mechanisms for the natural-derived polysaccharides to treat IBD. The excellent efficacy and safety of polysaccharides make them promising candidates for IBD therapy.
Collapse
Affiliation(s)
- Cailan Li
- Department of Pharmacology, Zunyi Medical University, Zhuhai Campus, Zhuhai, China
| | - Guosong Wu
- Pharmacy Department, Baiyun Branch of Nanfang Hospital of Southern Medical University, Guangzhou, China
| | - Hualang Zhao
- Department of Pharmaceutical Sciences, Zunyi Medical University, Zhuhai Campus, Zhuhai, China
| | - Na Dong
- Department of Pharmaceutical Sciences, Zunyi Medical University, Zhuhai Campus, Zhuhai, China
| | - Bowen Wu
- Department of Pharmaceutical Sciences, Zunyi Medical University, Zhuhai Campus, Zhuhai, China
| | - Yujia Chen
- Department of Pharmaceutical Sciences, Zunyi Medical University, Zhuhai Campus, Zhuhai, China
| | - Qiang Lu
- Department of Pharmaceutical Sciences, Zunyi Medical University, Zhuhai Campus, Zhuhai, China
| |
Collapse
|
13
|
Guo Q, Li X, Cui MN, Sun JL, Ji HY, Ni BB, Yan MX. CD13: A Key Player in Multidrug Resistance in Cancer Chemotherapy. Oncol Res 2020; 28:533-540. [PMID: 32532363 PMCID: PMC7751223 DOI: 10.3727/096504020x15919605976853] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cancer is one of the most serious diseases that are harmful to human health. Systemic chemotherapy is an optimal therapeutic strategy for the treatment of cancer, but great difficulty has been encountered in its administration in the form of multidrug resistance (MDR). As an enzyme on the outer cell surface, CD13 is documented to be involved in the MDR development of tumor cells. In this review, we will focus on the role of CD13 in MDR generation based on the current evidence.
Collapse
Affiliation(s)
- Qie Guo
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao UniversityQingdao, ShandongP.R. China
| | - Xiao Li
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao UniversityQingdao, ShandongP.R. China
| | - Meng-Na Cui
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao UniversityQingdao, ShandongP.R. China
| | - Jia-Lin Sun
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao UniversityQingdao, ShandongP.R. China
| | - Hong-Yan Ji
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao UniversityQingdao, ShandongP.R. China
| | - Bei-Bei Ni
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao UniversityQingdao, ShandongP.R. China
| | - Mei-Xing Yan
- Department of Pharmacy, Qingdao Women and Childrens HospitalQingdao, ShandongP.R. China
| |
Collapse
|
14
|
Neuroprotection of Intermedin Against Cerebral Ischemia/Reperfusion Injury Through Cerebral Microcirculation Improvement and Apoptosis Inhibition. J Mol Neurosci 2020; 71:767-777. [PMID: 32910355 DOI: 10.1007/s12031-020-01697-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 09/01/2020] [Indexed: 01/06/2023]
Abstract
Ischemic stroke is the primary cause of disability and mortality worldwide. Ischemia/reperfusion (I/R)-induced microcirculatory dysfunction and organ injury generally occur after ischemic stroke. Several studies have shown that intermedin (IMD) has a regulating function on cerebral microcirculation and blood-brain barrier via relaxing cerebral vessels and improving the local blood supply after cerebral ischemia. However, a unified conclusion has not been reached, and the underlying mechanism remains unclear. To observe and analyze the changes of cerebral microcirculation perfusion of cerebral IRI by IMD post-treatment in the rats and further explore the mechanism underlying the beneficial effect of IMD on cerebral IRI. Thirty-nine rats were divided into three groups: sham, I/R, and I/R + IMD groups. After IMD ischemia post-treatment, the rat cerebral infarction rate and the degree of neurological deficit were evaluated by TTC staining and neurological function score; the changes in the amount of cerebral microcirculation implantation on the injured side of the rats were observed by laser Doppler; the pathological changes and cell ultrastructure of rat cortex and hippocampus were observed by HE staining and transmission electron microscopy; the neuron apoptosis in the rat cortex and hippocampus was detected by TUNEL staining and immunohistochemical staining. Impaired neurological function, abnormal cortical/hippocampal neuron morphology, and the proportion of cerebral infarction were significantly improved in the IMD group compared with the I/R group, which suggested a possible neuroprotective role of IMD. IMD treatment also increased the average perfusion of cerebral surface microcirculation in rats by astonished 42.7 times. Finally, IMD administration decreased the caspase-3- and Bax-positive cell numbers and apoptotic cell ratio. IMD has a significant protective effect on neuronal damage caused by cerebral I/R in rats by improving cerebral microcirculation and inhibiting apoptosis.
Collapse
|
15
|
Jia G, Yang H, Diao Z, Liu Y, Sun C. Neural stem cell conditioned medium alleviates Aβ 25-35 damage to SH-SY5Y cells through the PCMT1/MST1 pathway. Eur J Histochem 2020; 64:3135. [PMID: 32705859 PMCID: PMC7388643 DOI: 10.4081/ejh.2020.3135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 05/29/2020] [Indexed: 11/27/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive, neurodegenerative disease. Accumulating evidence suggests that protein isoaspartate methyltransferase 1 (PCMT1) is highly expressed in brain tissue (substantia nigra, blue plaque, paraventricular nucleus). In this study, we investigated the effect of neural stem cell conditioned medium alleviates Aβ25-35 damage to SH-SY5Y cells by PCMT1/MST1 pathway. Results demonstrated that Aβ25-35 significantly decreased the cell viability in time and dose dependent manner. However, Neural stem cell-complete medium (NSC-CPM) or NSC-CDM had inhibitory effect on toxicity when fibrillation of Aβ25-35 occurred in their presence and NSC-CDM had a better inhibitor result. An increase of the PCMT1 expression levels was found in Aβ25-35 + NSC-CDM group. sh-PCMT1 significantly reduced the PCMT1, the cell viability and inhibited the protective effect; induced apoptosis and increased the expression of p-MST1. Overexpression of PCMT1 group reversed the effect of Aβ25-35 inhibited the cell viability and Aβ25-35 induced the apoptosis. In conclusion, NSC-CDM corrects the damage of Aβ25-35 to cells by increasing PCMT1, reducing MST phosphorylation.
Collapse
Affiliation(s)
- Guoyong Jia
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan.
| | - Hongna Yang
- Department of Critical-care Medicine, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan.
| | - Zengyan Diao
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan.
| | - Ying Liu
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan.
| | - Congcong Sun
- Department of Neurology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan.
| |
Collapse
|
16
|
Cao LJ, Xie HT, Chu ZX, Ma Y, Wang MM, Shi Z. Tubeimoside‑1 induces apoptosis in human glioma U251 cells by suppressing PI3K/Akt‑mediated signaling pathways. Mol Med Rep 2020; 22:1527-1535. [PMID: 32627020 PMCID: PMC7339596 DOI: 10.3892/mmr.2020.11224] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 05/12/2020] [Indexed: 12/15/2022] Open
Abstract
Tubeimoside-1 (TBMS1), a traditional Chinese herb extracted from Bolbostemma paniculatum (Maxim.), induces apoptosis in a number of human cancer cell lines. TBMS1 has been reported to induce apoptosis in human glioma cells, however the mechanism remains to be elucidated. The present study explored TBMS1-induced PI3K/Akt-related pathways in human glioma cells. The human glioma U251 and the human astrocyte (HA) cell lines were treated with various concentrations of TBMS1. MTT assays were conducted to analyze cell viability. Cell cycle distribution and the rate of apoptosis were assessed using flow cytometry. BrdU incorporation and Hoechst 33342 staining were performed to analyze the cell cycle and apoptosis, respectively. Western blotting was performed to investigate protein expression levels. The results demonstrated that TBMS1 reduced cell viability in human glioma cells U251 by suppressing Akt phosphorylation. Subsequently, TBMS1 inhibited DNA synthesis and induced G2/M phase arrest by targeting the PI3K/Akt/p21 and the cyclin-dependent kinase 1/cyclin B1 signaling cascades. In addition, TBMS1 triggered apoptosis via the PI3K/Akt-mediated Bcl-2 signaling pathway. These results demonstrated that TBMS1 prevented the progression of gliomas via the PI3K/Akt-dependent pathway, which provided a theoretical basis for in vivo studies to use TBMS1 as potential therapy for the prevention of cancer.
Collapse
Affiliation(s)
- Li-Juan Cao
- Department of Pediatrics, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia Autonomous Region 028007, P.R. China
| | - Hai-Tang Xie
- Department of Pediatrics, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia Autonomous Region 028007, P.R. China
| | - Zhong-Xia Chu
- Department of Pediatrics, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia Autonomous Region 028007, P.R. China
| | - Yue Ma
- Department of Pediatrics, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia Autonomous Region 028007, P.R. China
| | - Ming-Ming Wang
- Department of Pediatrics, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia Autonomous Region 028007, P.R. China
| | - Zhuang Shi
- Department of Mongolian Medicine Hand Foot Surgery, Affiliated Hospital of Inner Mongolia University for Nationalities, Tongliao, Inner Mongolia Autonomous Region 028007, P.R. China
| |
Collapse
|
17
|
Liu S, Pan X, Cheng W, Deng B, He Y, Zhang L, Ning Y, Li J. Tubeimoside I Antagonizes Yoda1-Evoked Piezo1 Channel Activation. Front Pharmacol 2020; 11:768. [PMID: 32523536 PMCID: PMC7261832 DOI: 10.3389/fphar.2020.00768] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Accepted: 05/11/2020] [Indexed: 12/23/2022] Open
Abstract
Piezo1, a mechanosensitive Ca2+-permeable non-selective cationic ion channel protein, is involved in a wide range of biological processes and plays crucial roles in vascular development. However, the pharmacology of this protein is in its infancy. Yoda1, the first specific chemical activator of Piezo1 channels, can activate Piezo1 in absence of mechanical stimulation. Hence, we sought to identify inhibitors of Yoda1 from Traditional Chinese Medicine (TCM). Intracellular Ca2+ measurements were conducted in human umbilical vein endothelial cells (HUVECs), HEK 293T cells overexpressing TRPC5 and TRPM2 channels, as well as in CHO K1 cells overexpressing TRPV4 channels. We identified tubeimoside I (TBMS1) as a strong inhibitor of the Yoda1 response and demonstrated its selectivity for the Piezo1 channels. Similarly, Yoda1-induced inhibitory results were obtained in Piezo1 wild-type overexpressed cells, murine liver endothelial cells (MLECs), and macrophages. The physiological responses of TBMS1 were identified by isometric tension, which can inhibit Yoda1 relaxation of aortic rings. Our results demonstrated that TBMS1 can effectively antagonize Yoda1 induced Piezo1 channel activation. This study sheds light on the existence of Yoda1 inhibitors and improves the understanding of vascular pharmacology through Piezo1 channels.
Collapse
Affiliation(s)
- Silin Liu
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xianmei Pan
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wenbin Cheng
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Bo Deng
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yu He
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Lei Zhang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yile Ning
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jing Li
- Lingnan Medical Research Center, Guangzhou University of Chinese Medicine, Guangzhou, China.,The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, China.,School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
18
|
Ruan C, You L, Qiu Y, Cui X, Wu D. Tubeimoside I induces autophagy in HepG2 cells by activating the AMP-activated protein kinase signaling pathway. Oncol Lett 2020; 20:623-630. [PMID: 32537027 PMCID: PMC7291703 DOI: 10.3892/ol.2020.11604] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 04/01/2020] [Indexed: 12/13/2022] Open
Abstract
Tubeimoside I (TBMS) is a natural compound with antitumor properties. However, the detailed mechanism underlying the function of TBMS in liver cancer has not been fully elucidated. In the present study, TBMS was shown to suppress cell proliferation and induce S phase cell cycle arrest in HepG2 cells. Furthermore, TBMS treatment induced autophagy, evidenced by autophagosome accumulation, and increased the mRNA expression of Beclin 1 and microtubule-associated protein 1 light chain 3 (LC3)-I. However, flow cytometry analysis demonstrated that TBMS exerted no effect on cell apoptosis. Moreover, TBMS increased the phosphorylation of AMP-activated protein kinase (AMPK) in a concentration-dependent manner, thereby activating the AMPK signaling pathway. A specific AMPK inhibitor, compound C (CC), caused markedly suppressed TBMS-induced accumulation of LC3-II. In addition, the mRNA expression of LC3-I and Beclin 1 was also suppressed in cells treated with TBMS and CC in combination. The results of the present study provide new insights into the role of TBMS in inducing autophagy and support the potential application of TBMS for liver cancer treatment in the clinical setting.
Collapse
Affiliation(s)
- Chengxu Ruan
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350116, P.R. China
| | - Lijuan You
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350116, P.R. China
| | - Yingdong Qiu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350116, P.R. China
| | - Xiao Cui
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian 350116, P.R. China
| | - Defeng Wu
- College of Bee Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, P.R. China
| |
Collapse
|
19
|
Song S, Jiang Z, Spezia-Lindner DE, Liang T, Xu C, Wang H, Tian Y, Bai Y. BHRF1 Enhances EBV Mediated Nasopharyngeal Carcinoma Tumorigenesis through Modulating Mitophagy Associated with Mitochondrial Membrane Permeabilization Transition. Cells 2020; 9:cells9051158. [PMID: 32392902 PMCID: PMC7290790 DOI: 10.3390/cells9051158] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/28/2020] [Accepted: 04/28/2020] [Indexed: 12/15/2022] Open
Abstract
Epstein-Barr virus (EBV) is a major contributor to nasopharyngeal carcinoma (NPC) tumorigenesis. Mitochondria have been shown to be a target for tumor viral invasion, and to mediate viral tumorigenesis. In this study, we detected that mitochondrial morphological changes in tumor tissues of NPC patients infected with EBV were accompanied by an elevated expression of BHRF1, an EBV encoded protein homologue to Bcl-2. High expression of BHRF1 in human NPC cell lines enhanced tumorigenesis and metastasis features. With BHRF1 localized to mitochondria, its expression induced cyclophlin D dependent mitochondrial membrane permeabilization transition (MMPT). The MMPT further modulated mitochondrial function, increased ROS production and activated mitophagy, leading to enhanced tumorigenesis. Altogether, our results indicated that EBV-encoded BHRF1 plays an important role in NPC tumorigenesis through regulating cyclophlin D dependent MMPT.
Collapse
Affiliation(s)
- Shujie Song
- School of Public Health, Xi’an Jiaotong University, Xi’an 710061, Shaanxi, China;
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China; (Z.J.); (T.L.)
- No. 3 Hospital, the Affiliated Hospital of Northwest University School of Medicine, Xi’an 710018, Shaanxi, China;
| | - Zhiying Jiang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China; (Z.J.); (T.L.)
| | - David Ethan Spezia-Lindner
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX 78258, USA;
| | - Ting Liang
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China; (Z.J.); (T.L.)
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX 78258, USA;
| | - Chang Xu
- First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, Zhejiang, China;
| | - Haifeng Wang
- No. 3 Hospital, the Affiliated Hospital of Northwest University School of Medicine, Xi’an 710018, Shaanxi, China;
| | - Ye Tian
- No. 3 Hospital, the Affiliated Hospital of Northwest University School of Medicine, Xi’an 710018, Shaanxi, China;
- Correspondence: (Y.T.); (Y.B.)
| | - Yidong Bai
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, China; (Z.J.); (T.L.)
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX 78258, USA;
- Correspondence: (Y.T.); (Y.B.)
| |
Collapse
|
20
|
Parsa FG, Feizi MAH, Safaralizadeh R, Hosseini-Yazdi SA, Mahdavi M. Molecular mechanisms of apoptosis induction in K562 and KG1a leukemia cells by a water-soluble copper(II) thiosemicarbazone complex. J Biol Inorg Chem 2020; 25:383-394. [PMID: 32274578 DOI: 10.1007/s00775-020-01769-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 02/25/2020] [Indexed: 11/26/2022]
Abstract
Thiosemicarbazones (TSCs) and their metal complexes exhibit pronounced and selective cytotoxic potential against a broad span of cancers. Here, we assessed the anti-cancer activity of a water-soluble copper(II) complex of thiosemicarbazone (Cu-TSC) against two cancer cell lines of human leukemia. Our analysis revealed that Cu-TSC treatment results in a time and dose-dependent growth inhibition in K562 and KG1a cells while sparing normal human fibroblast (HFF2) cells. The IC50 values for the Cu-TSC treatment were measured to be 21.7 ± 1.5 µM and 50.25 ± 2.5 µM for K562 and KG1a cells, respectively. Cell cycle analysis indicated that Cu-TSC induces the accumulation of cells in the sub-G1 fraction as well as the reversible arrest in G0/G1 and G2/M phases in K562 and KG1a cells, respectively. Furthermore, the occurrence of apoptosis as the prime mode of cell death was verified through apoptotic body formation, phosphatidylserine externalization, and caspase-3 activation. Additionally, the real-time quantitative PCR analysis revealed that Cu-TSC triggers apoptosis in both cell lines via the upregulation of caspases-8, -9, and the changing of Bax/Bcl2 ratio. Finally, flow cytometric analysis confirmed that Cu-TSC treatment causes the enhancement of reactive oxygen species formation in both K562 and KG1a cells. Altogether, these findings suggest that Cu-TSC is a promising inducer of apoptosis in leukemia cells and carries potential as an anti-cancer compound.
Collapse
Affiliation(s)
| | | | - Reza Safaralizadeh
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | | - Majid Mahdavi
- Department of Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| |
Collapse
|
21
|
Sheng W, Yang H, Niu Z, Yin H. Anti-apoptosis effect of heme oxygenase-1 on lung injury after cardiopulmonary bypass. J Thorac Dis 2020; 12:1393-1403. [PMID: 32395277 PMCID: PMC7212168 DOI: 10.21037/jtd.2020.03.48] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Background This study aimed to investigate the anti-apoptosis effects of heme oxygenase-1 (HO-1) on lung injury (LI) after cardiopulmonary bypass (CPB) and its probable mechanisms. Methods One hundred and forty-four male Wistar rats were divided into 3 groups randomly: group A (control group), group B (cobalt protoporphyrin, CoPP), and group C [CoPP plus zinc protoporphyrin (ZnPP)]. Lung tissues were harvested at different time: before CPB (T0), 0 min after CPB (T1), 2 h after CPB (T2), 6 h (T3), 12 h (T4), and 24 h (T5). Results The HO-1 protein expressions in lung tissue in group B were higher than those in group A and group C in any given time, and the same as HO-1 activity (P<0.05). The expressions of Bcl-2 protein in group B at all time point after bypass were higher than those in group A and group C, and the difference was statistically significant (P<0.05). Apoptosis index (AI) in group B at any time point after bypass were lower than those in group A and group C (P<0.05). Conclusions CoPP can significantly increase the expression of HO-1 protein in lung tissue. HO-1 is still highly expressed after CPB, so as to play an important part in anti-apoptosis, and reduce LI.
Collapse
Affiliation(s)
- Wei Sheng
- Department of Cardiovascular Surgery, Qingdao Municipal Hospital, Medical College of Qingdao University, Qingdao 266071, China
| | - Haiqin Yang
- Department of Mental Intervention, Qingdao Preferential Hospital, Qingdao 266071, China
| | - Zhaozhuo Niu
- Department of Cardiovascular Surgery, Qingdao Municipal Hospital, Medical College of Qingdao University, Qingdao 266071, China
| | - Hong Yin
- Department of Cardiovascular Surgery, Qingdao Municipal Hospital, Medical College of Qingdao University, Qingdao 266071, China
| |
Collapse
|
22
|
Bi D, Li X, Li T, Li X, Lin Z, Yao L, Li H, Xu H, Hu Z, Zhang Z, Liu Q, Xu X. Characterization and Neuroprotection Potential of Seleno-Polymannuronate. Front Pharmacol 2020; 11:21. [PMID: 32153394 PMCID: PMC7044149 DOI: 10.3389/fphar.2020.00021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 01/08/2020] [Indexed: 12/22/2022] Open
Abstract
Seleno-polymannuronate (Se-PM) was prepared from alginate-derived polymannuronate (PM) through a sulfation followed by a selenylation replacement reaction. The organic selenium content of Se-PM was 437.25 μg/g and its average molecular weight was 2.36 kDa. The neuroprotection effect of Se-PM and corresponding molecular mechanisms were investigated. Our results showed that, comparing to both sulfated PM (S-PM) and PM, Se-PM remarkably inhibited the aggregation of Aβ1-42 oligomer in vitro and significantly reduced the APP and BACE1 protein expression in N2a-sw cells, highlighting the critical function of the selenium presented in Se-PM. Moreover, Se-PM decreased the expression of cytochrome c and the ratio of Bax to Bcl-2, and enhanced the mitochondrial membrane potential in N2a-sw cells. These results suggested that Se-PM treatment can markedly inhibit N2a-sw cell apoptosis and promote N2a-sw cell survival and that Se-PM might be a potential therapeutic agent for the prevention of neurodegeneration owing to its remarkable neuroprotection effect.
Collapse
Affiliation(s)
- Decheng Bi
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Xiaofan Li
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Tong Li
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Xiuting Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing, China
| | - Zhijian Lin
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Lijun Yao
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Hui Li
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Hong Xu
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Zhangli Hu
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Zhenqing Zhang
- College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Qiong Liu
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Xu Xu
- Shenzhen Key Laboratory of Marine Bioresource and Eco-environmental Science, Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| |
Collapse
|
23
|
Pioglitazone Protects Compression-Mediated Apoptosis in Nucleus Pulposus Mesenchymal Stem Cells by Suppressing Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4764071. [PMID: 31885796 PMCID: PMC6893265 DOI: 10.1155/2019/4764071] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 07/19/2019] [Accepted: 08/10/2019] [Indexed: 01/28/2023]
Abstract
Excessive compression, the main cause of intervertebral disc (IVD) degeneration, affected endogenous repair of the intervertebral disc. Pioglitazone (PGZ) is the agonist of peroxisome proliferator-activated receptor γ, which has been widely used in the treatment of diabetes mellitus. The present study aim at investigating whether pioglitazone has protective effects on compression-mediated cell apoptosis in nucleus pulposus mesenchymal stem cells (NP-MSCs) and further exploring the possible underlying mechanism. Our results indicated that the isolated cells satisfied the criteria of MSC stated by the International Society for Cellular Therapy. Besides, our research revealed that pioglitazone could protect cell viability, cell proliferation of NP-MSCs and alleviated the toxic effects caused by compression. The actin stress fibers was suppressed obviously under compression, and pioglitazone alleviated the adverse outcomes. Pioglitazone exerted protective effects on compression-induced NP-MSCs apoptosis according to annexin V/PI double-staining and TUNEL assays. Pioglitazone suppressed compression-induced NP-MSCs oxidative stress, including decreasing compression-induced overproduction of reactive oxygen species (ROS) and malondialdehyde (MDA), and alleviated compression-induced mitochondrial membrane potential (MMP) decrease. Ultrastructure collapse of the mitochondria exhibited a notable improvement by pioglitazone in compression-induced NP-MSCs according to transmission electron microscopy (TEM). Furthermore, the molecular results showed that pioglitazone significantly decreased the expression of apoptosis-associated proteins, including cyto.cytochrome c, Bax, cleaved caspase-9, and cleaved caspase-3, and promoted Bcl-2 expression. These results indicated that pioglitazone alleviated compression-induced NP-MSCs apoptosis by suppressing oxidative stress and the mitochondrial apoptosis pathway, which may be a valuable candidate for the treatment of IVD degeneration.
Collapse
|
24
|
Chen Z, Mou L, Pan Y, Feng C, Zhang J, Li J. CXCL8 Promotes Glioma Progression By Activating The JAK/STAT1/HIF-1α/Snail Signaling Axis. Onco Targets Ther 2019; 12:8125-8138. [PMID: 31686858 PMCID: PMC6783399 DOI: 10.2147/ott.s224721] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 09/12/2019] [Indexed: 12/20/2022] Open
Abstract
Background Upregulation of CXCL8 (C-X-C motif ligand 8) in tumor cells has been reported in several types of cancer, and it correlates with a poor prognosis. However, the role of CXCL8 in glioma progression remains unknown. Materials and methods In this study, we examined CXCL8 expression levels in human glioma cell lines and in sixteen human gliomas with different grades. The molecular role of CXCL8 in glioma cells was investigated using quantitative polymerase chain reaction (qRT-PCR) assays, Western blotting, CCK-8 assays, EdU assays, colony formation assays, Transwell migration and invasion assays. Results We found that high expression levels of CXCL8 were positively associated with progression and poor prognosis in human glioma. Mechanistically, CXCL8 promoted the epithelial-mesenchymal transition (EMT) in glioma cells by activating the JAK/STAT1/HIF-1α/Snail signaling pathway. Conclusion Taken together, our data provide a plausible mechanism for CXCL8-modulated glioma progression, which suggests that CXCL8 may represent a potential therapeutic target in the prevention and treatment of gliomas.
Collapse
Affiliation(s)
- Zhiming Chen
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, People's Republic of China
| | - Lei Mou
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, People's Republic of China
| | - Yiheng Pan
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, People's Republic of China
| | - Chi Feng
- Department of Neurosurgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, People's Republic of China
| | - Jingjing Zhang
- Department of Obstetrics, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, People's Republic of China
| | - Junjun Li
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, People's Republic of China
| |
Collapse
|
25
|
Ngadiono E, Hardiany NS. Advancing towards Effective Glioma Therapy: MicroRNA Derived from Umbilical Cord Mesenchymal Stem Cells' Extracellular Vesicles. Malays J Med Sci 2019; 26:5-16. [PMID: 31496889 PMCID: PMC6719885 DOI: 10.21315/mjms2019.26.4.2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 12/10/2018] [Indexed: 01/20/2023] Open
Abstract
A glioma, especially a grade IV glioblastoma, is a malignant tumour with a poor prognosis despite growing medical advancements. Researchers have been looking for better and more effective treatments targeting the molecular pathways of gliomas due to glioblastomas’ ability to develop resistance to chemotherapies. Moreover, glioma stem cells (GSC) contribute to maintaining the glioma population, which benefits from its ability to self-renew and differentiate. Recent research has reported that through the introduction of umbilical cord mesenchymal stem cells (UCMSC) into glioma cells, the growth and development of the glioma cells can be downregulated. It has more currently been found out that UCMSC release extracellular vesicles (EVs) containing miRNA that are responsible for this phenomenon. Therefore, this review analyses literature to discuss all possible miRNAs contained within the UCMSC’s EVs and to elaborate on their molecular mechanisms in halting gliomas and GSC growth. This review will also include the challenges and limitations, to account for which more in vivo research is suggested. In conclusion, this review highlights how miRNAs contained within UCMSC’s EVs are able to downregulate multiple prominent pathways in the survival of gliomas.
Collapse
Affiliation(s)
- Eko Ngadiono
- International Class Program, Faculty of Medicines Universitas Indonesia, Jakarta, Indonesia
| | - Novi Silvia Hardiany
- Department of Biochemistry & Molecular Biology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| |
Collapse
|
26
|
Yan J, Dou X, Zhou J, Xiong Y, Mo L, Li L, Lei Y. Tubeimoside-I sensitizes colorectal cancer cells to chemotherapy by inducing ROS-mediated impaired autophagolysosomes accumulation. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:353. [PMID: 31412953 PMCID: PMC6694658 DOI: 10.1186/s13046-019-1355-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 08/01/2019] [Indexed: 12/24/2022]
Abstract
Background Tubeimoside-I (TBM), a plant-derived bioactive compound, shows antitumor activity in different tumors and can enhance the efficacy of chemotherapeutic agents. However, the detail mechanism underlying remains to be elucidated. Methods The cytotoxic potential of TBM towards CRC cells was examined by CCK8 assay, colony formation, LDH release assay, flow cytometry method and Western blots. The ROS levels, autophagy, apoptosis, chemosensitivity to 5-FU or DOX, etc. were determined between control and TBM-treated CRC cells. Results In this study, we found that TBM could inhibit proliferation and induce apoptosis in colorectal cancer (CRC) cells. Intriguingly, TBM treatment could either promote autophagy initiation by ROS-induced AMPK activation, or block autophagy flux through inhibiting lysosomal hydrolytic enzymes, which leaded to massive impaired autophagylysosomes accumulation. Administration of autophagy initiation inhibitor (3-MA or selective ablation of autophagy related proteins) relieves TBM-induced CRC suppression, while combination use of autophagy flux inhibitor chloroquine (CQ) slightly augments TBM-induced cell death, suggesting that impaired autophagylysosomes accumulation contributes to TBM-induced growth inhibition in CRC cells. Notably, as an autophagy flux inhibitor, TBM works synergistically with 5-fluorouracil (5-FU) or doxorubicin (DOX) in CRC suppression. Conclusion Together, our study provides new insights regarding the anti-tumor activity of TBM against CRC, and established potential applications of TBM for CRC combination therapies in clinic. Electronic supplementary material The online version of this article (10.1186/s13046-019-1355-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jianghong Yan
- Institute of Life Science,Chongqing Medical University, Chongqing, 400016, China.,Department of Medical Laboratory Technology, Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoyun Dou
- Institute of Life Science,Chongqing Medical University, Chongqing, 400016, China
| | - Jing Zhou
- Department of Biochemistry and Molecular Biology and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yuanfeng Xiong
- Department of Medical Laboratory Technology, Chongqing Medical University, Chongqing, 400016, China
| | - Ling Mo
- Department of Medical Laboratory Technology, Chongqing Medical University, Chongqing, 400016, China
| | - Longhao Li
- Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China.
| | - Yunlong Lei
- Department of Biochemistry and Molecular Biology and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| |
Collapse
|
27
|
Cao J, Zhao E, Zhu Q, Ji J, Wei Z, Xu B, Cui H. Tubeimoside-1 Inhibits Glioblastoma Growth, Migration, and Invasion via Inducing Ubiquitylation of MET. Cells 2019; 8:E774. [PMID: 31349699 PMCID: PMC6721480 DOI: 10.3390/cells8080774] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 07/14/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022] Open
Abstract
Tubeimoside-1 (TBMS1) is one of the extracts of rhizoma bolbostemmae, which has remarkable anti-cancer function in the treatment of esophagus and gastric cancer in traditional Chinese medicine. However the mechanisms of its anti-cancer function is remain unclear. In this study, we demonstrate that TBMS1 could inhibit cell growth and metastasis in glioblastoma. MET is a member of the receptor tyrosine kinase family, which amplifies frequently in various human cancers. As an important proto-oncogene, multiple inhibitors have been developed for the therapy of cancers. Here, we found TBMS1 could reduce/decrease the protein level of MET via increasing its Ubiquitination degradation. Therefore, TBMS1 is a promising compound for the treatment of glioblastoma and an inhibitor of MET.
Collapse
Affiliation(s)
- Jiangjun Cao
- Chongqing Engineering Research Center of Antitumor Natural Drugs, Chongqing Three Gorges Medical College, Chongqing 404120, China
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
- Institute of Medicine of Southwest University, Southwest University, Chongqing 400715, China
| | - Erhu Zhao
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
| | - Qingzong Zhu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
| | - Juanli Ji
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
| | - Zekun Wei
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China
| | - Bo Xu
- Institute of Medicine of Southwest University, Southwest University, Chongqing 400715, China.
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China.
| |
Collapse
|
28
|
Ma X, Deng J, Cui X, Chen Q, Wang W. Berberine exhibits antioxidative effects and reduces apoptosis of the vaginal epithelium in bacterial vaginosis. Exp Ther Med 2019; 18:2122-2130. [PMID: 31410167 PMCID: PMC6676195 DOI: 10.3892/etm.2019.7772] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 05/31/2019] [Indexed: 12/16/2022] Open
Abstract
Bacterial vaginosis (BV) is a common type of vaginitis. Berberine is a natural alkaline product that reduces oxidative stress and apoptosis in cells. The aim of the present study was to investigate the effects of berberine on oxidative stress and apoptotic rates of BV. Vaginal epithelial and discharge samples were obtained from 60 healthy individuals and 180 patients with BV before and after one month of berberine treatment. Clinical observation was documented for all patients before and after treatment for comparison. Additionally, an in vitro study was performed; the samples were divided into groups the following groups: Control, model (H2O2-treated), LT (low-dose berberine), MT (medium-dose berberine) and HT (high-dose berberine). Expression levels of the oxidative stress related proteins were detected by western blotting. Clinical symptoms of patients with BV significantly improved following berberine treatment. Oxidative stress in vaginal discharge was significantly lower following treatment, indicated by the increased activity of superoxide dismutase (SOD) and catalase, as well as the reduced levels of malondialdehyde and H2O2. Apoptosis of the vaginal epithelial cells was also reduced, which was indicated by the reduced expression of apoptosis proteins caspase-3, cytochrome C, capase-12 and Bax, and increased expression of Bcl-2. The results of the in vitro experiments demonstrated a dose-dependent decrease in apoptosis with berberine treatment compared with levels before treatment. Oxidative stress relief was demonstrated by the reduced reactive oxygen species level and increased SOD and endothelial nitric oxide synthase levels, whereas suppression of apoptosis was further supported by the reduction in apoptotic proteins, as well as a decreased Bax/Bcl-2 ratio. Berberine exhibited effects on lowering oxidative stress in vaginal discharge and reducing oxidative damage, as well as apoptosis of the vaginal epithelium, which are beneficial to patients with bacterial vaginosis.
Collapse
Affiliation(s)
- Xiuzhen Ma
- Department of Obstetrics and Gynecology, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong 264000, P.R. China
| | - Junfeng Deng
- Department of Obstetrics and Gynecology, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong 264000, P.R. China
| | - Xinmu Cui
- Department of Obstetrics and Gynecology, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong 264000, P.R. China
| | - Qi Chen
- Department of Obstetrics and Gynecology, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong 264000, P.R. China
| | - Weihua Wang
- Department of Obstetrics and Gynecology, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong 264000, P.R. China
| |
Collapse
|
29
|
Lee CM, Lee J, Nam MJ, Choi YS, Park SH. Tomentosin Displays Anti-Carcinogenic Effect in Human Osteosarcoma MG-63 Cells via the Induction of Intracellular Reactive Oxygen Species. Int J Mol Sci 2019; 20:ijms20061508. [PMID: 30917517 PMCID: PMC6471964 DOI: 10.3390/ijms20061508] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/19/2019] [Accepted: 03/22/2019] [Indexed: 12/11/2022] Open
Abstract
Tomentosin is a natural sesquiterpene lactone extracted from various plants and is widely used as a medicine because it exhibits essential therapeutic properties. In this study, we investigated the anti-carcinogenic effects of tomentosin in human osteosarcoma MG-63 cells by performing cell migration/viability/proliferation, apoptosis, and reactive oxygen species (ROS) analysis assays. MG-63 cells were treated with various doses of tomentosin. After treatment with tomentosin, MG-63 cells were analyzed using the MTT assay, colony formation assay, cell counting assay, wound healing assay, Boyden chamber assay, zymography assay, cell cycle analysis, FITC Annexin V apoptosis assay, terminal deoxynucleotidyl transferase dUTP nick end labeling assay, western blot analysis, and ROS detection analysis. Our results indicated that tomentosin decreased cell viability and migration ability in MG-63 cells. Moreover, tomentosin induced apoptosis, cell cycle arrest, DNA damage, and ROS production in MG-63 cells. Furthermore, tomentosin-induced intracellular ROS decreased cell viability and induced apoptosis, cell cycle arrest, and DNA damage in MG-63 cells. Taken together, our results suggested that tomentosin exerted anti-carcinogenic effects in MG-63 cells by induction of intracellular ROS.
Collapse
Affiliation(s)
- Chang Min Lee
- Department of Biological Science, Gachon University, Seongnam 13120, Korea.
| | - Jongsung Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon 16419, Korea.
| | - Myeong Jin Nam
- Department of Biological Science, Gachon University, Seongnam 13120, Korea.
| | - Youn Soo Choi
- Department of Biomedical Sciences, Seoul National University Graduate School, Department of Medicine, College of Medicine, Seoul National University, Seoul 03080, Korea.
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong 30016, Korea.
| |
Collapse
|
30
|
Xie L, Chen Y, Chen J, Zhang H, Liao Y, Zhou Y, Zhou L, Qing C. Anti-tumor effects and mechanism of GA-13315, a novel gibberellin derivative, in human lung adenocarcinoma: an in vitro and in vivo study. Cell Mol Biol Lett 2019; 24:6. [PMID: 30651744 PMCID: PMC6327519 DOI: 10.1186/s11658-018-0126-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 12/03/2018] [Indexed: 12/11/2022] Open
Abstract
Objective To investigate the anti-tumor effects and the mechanism of the compound 13-chlorine-3, 15-dioxy-gibberellic acid methyl ester (GA-13315) in lung adenocarcinoma in vitro and in vivo. Methods The antiproliferative effect of GA-13313 on the A549 cell line was determined by MTT (3-[4, 5-dimethylthiazol-2-yl]-2, 5 diphenyl tetrazolium bromide) assay. A xenograft model of A549 was established to evaluate the anti-tumor effect and histopathological examination was performed to assess the toxicity of GA-13315. Apoptosis was detected by TUNEL staining in tissues and flow cytometry in cells; activation of caspase-3, caspase-8 and caspase-9 was evaluated by immunohistochemical analysis; protein levels of Bcl-2-associated X protein (Bax), B-cell lymphoma-2 (Bcl-2), caspase-4, activating transcription factor 4 (ATF4), glucose-regulated protein 78 (GRP78) and growth arrest and DNA damage-inducible gene 153 (GADD153) were determined by western blotting. Mitochondrial membrane potential (MMP) was measured by the JC-1 fluorescence probe. Results Our results showed that GA-13315 exhibited potent, dose- and time-dependent anti-proliferative activity, and the IC50 values were 37.43 ± 2.73, 28.08 ± 7.76 and 19.29 ± 7.61 μM at 24, 48, and 72 h, respectively. The xenograft experiment revealed that tumor weight and volume were significantly decreased after GA-13315 3 mg/kg and 9 mg/kg (P < 0.05) treatment, and GA-13315 had low toxicity in bone marrow, kidney and colon tissues. GA-13315 triggered remarkable apoptosis in A549 cells at the concentration of 25.6 μM and 32 μM (P < 0.05) and activated caspase-3, − 8 and − 9. Moreover, GA-13315 induced apoptosis through the mitochondrial apoptosis pathway by elevating the Bax/Bcl-2 ratio, releasing cytochrome c and activating caspase-9 in A549 cells. In the endoplasmic reticulum apoptosis pathway, the levels of caspase-4, ATF4, GRP78 and GADD153 were markedly upregulated. Conclusions This study suggests that GA-13315 can be considered as a promising chemotherapeutic agent with anticancer activity in treatment of lung cancer in future.
Collapse
Affiliation(s)
- Lin Xie
- 1Department of Medical Oncology, Third Affiliated Hospital of Kunming Medical University/ Cancer Hospital of Yunnan Province, Kunming, China
| | - Yajuan Chen
- 2School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Cancer Hospital of Yunnan Province, Kunming Medical University, NO.1168, West Chunrong Road, Chenggong Developing Area, Kunming, 650031 China
| | - Jingbo Chen
- 3Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education School of Chemical Science and Technology, Yunnan University, Kunming, China
| | - Hongbin Zhang
- 3Key Laboratory of Medicinal Chemistry for Natural Resource, Ministry of Education School of Chemical Science and Technology, Yunnan University, Kunming, China
| | - Yedan Liao
- 1Department of Medical Oncology, Third Affiliated Hospital of Kunming Medical University/ Cancer Hospital of Yunnan Province, Kunming, China
| | - Yonghong Zhou
- 1Department of Medical Oncology, Third Affiliated Hospital of Kunming Medical University/ Cancer Hospital of Yunnan Province, Kunming, China
| | - Ling Zhou
- 1Department of Medical Oncology, Third Affiliated Hospital of Kunming Medical University/ Cancer Hospital of Yunnan Province, Kunming, China
| | - Chen Qing
- 2School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology for Natural Products, Cancer Hospital of Yunnan Province, Kunming Medical University, NO.1168, West Chunrong Road, Chenggong Developing Area, Kunming, 650031 China
| |
Collapse
|
31
|
Feng X, Zhou J, Li J, Hou X, Li L, Chen Y, Fu S, Zhou L, Li C, Lei Y. Tubeimoside I induces accumulation of impaired autophagolysosome against cervical cancer cells by both initiating autophagy and inhibiting lysosomal function. Cell Death Dis 2018; 9:1117. [PMID: 30389907 PMCID: PMC6214972 DOI: 10.1038/s41419-018-1151-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 09/28/2018] [Accepted: 10/09/2018] [Indexed: 02/05/2023]
Abstract
Cervical cancer is one of the most aggressive human cancers with poor prognosis due to constant chemoresistance and repeated relapse. Tubeimoside I (TBM) has been identified as a potent antitumor agent that inhibits cancer cell proliferation by triggering apoptosis and inducing cell cycle arrest. Nevertheless, the detailed mechanism remains unclear and needs to be further elucidated, especially in cervical cancer. In this study, we found that TBM could induce proliferation inhibition and cell death in cervical cancer cells both in vitro and in vivo. Further results demonstrated that treatment with TBM could induce autophagosome accumulation, which was important to TBM against cervical cancer cells. Mechanism studies showed that TBM increased autophagosome by two pathways: First, TBM could initiate autophagy by activating AMPK that would lead to stabilization of the Beclin1-Vps34 complex via dissociating Bcl-2 from Beclin1; Second, TBM could impair lysosomal cathepsin activity and block autophagic flux, leading to accumulation of impaired autophagolysosomes. In line with this, inhibition of autophagy initiation attenuated TBM-induced cell death, whereas autophagic flux inhibition could exacerbated the cytotoxic activity of TBM in cervical cancer cells. Strikingly, as a novel lethal impaired autophagolysosome inducer, TBM might enhance the therapeutic effects of chemotherapeutic drugs towards cervical cancer, such as cisplatin and paclitaxel. Together, our study provides new insights into the molecular mechanisms of TBM in the antitumor therapy, and establishes potential applications of TBM for cervical cancer treatment in clinic.
Collapse
Affiliation(s)
- Xuping Feng
- Department of Biochemistry and Molecular Biology, and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, P.R. China.,State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| | - Jing Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| | - Jingyi Li
- School of Biological Sciences and Technology, Chengdu Medical College, Chengdu, 610083, P.R. China
| | - Xueyan Hou
- School of pharmacy, Xinxiang Medical University, Xinxiang, 453003, P. R. China
| | - Longhao Li
- Department of Biochemistry and Molecular Biology, and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, P.R. China.,Department of Oncology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Yongmin Chen
- Department of Neurology, The Affiliated Hospital of Hainan Medical College, Hainan, 570102, P.R. China
| | - Shuyue Fu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| | - Li Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, P.R. China
| | - Changlong Li
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, P.R. China
| | - Yunlong Lei
- Department of Biochemistry and Molecular Biology, and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing, 400016, P.R. China.
| |
Collapse
|
32
|
Lv J, Xing Y. Effects of UV on apoptotic factors in lens epithelial cells of an animal model. Exp Ther Med 2018; 16:2309-2312. [PMID: 30186472 PMCID: PMC6122387 DOI: 10.3892/etm.2018.6435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Accepted: 06/19/2018] [Indexed: 01/05/2023] Open
Abstract
Effect of ultraviolet (UV) on the apoptotic factors p53, Bax and Bcl-2 in lens epithelial cells (LECs) was explored. Sixty SD rats were randomly divided into control group (n=12) and model group (n=48). Rats in model group were used to construct UV radiation eyeball models. Twelve rats in model group and 3 rats in control group were sacrificed at day 1, 3, 5 and 7, and the expression of p53, Bax and Bcl-2 in LECs was detected by RT-qPCR. There was no significant difference in p53 expression between model and control group on day 1 (P>0.05), while expression level of p53 was significantly higher in model than in control group at day 3, 5 and 7 (P<0.05), and the highest expression level was found on day 7 (P<0.05). Relative expression level of Bax in model group was significantly higher than that in control group (P<0.05), and the highest expression level was found on day 7 (P<0.05). On day 1, expression level of Bcl-2 in model group was significantly lower than that in control group (P<0.05), but it began to rise on day 3 but did not differ significantly from the control group (P>0.05), and was significantly higher in model than in control group on day 5 and 7 (P<0.05), and the highest expression level was observed on day 7 (P<0.05). Linear correlation analysis showed that there was a positive correlation between p53, Bax and Bcl-2 expression and UV exposure time in LEC (r=0.82, 0.90, 0.95, P<0.05). Expression levels of p53, Bax, and Bcl-2 in LEC of rats are positively correlated with the time after UV irradiation, suggesting that UV could induce apoptosis of LEC by promoting p53, Bax and Bcl-2 expression.
Collapse
Affiliation(s)
- Juling Lv
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Yiqiao Xing
- Eye Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
33
|
Liu X, Jiang S, Tian X, Jiang Y. Expression of cleaved caspase-3 predicts good chemotherapy response but poor survival for patients with advanced primary triple-negative breast cancer. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2018; 11:4363-4373. [PMID: 31949833 PMCID: PMC6962992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 08/14/2018] [Indexed: 06/10/2023]
Abstract
OBJECTIVE To assess cleaved caspase-3 (CC3), Ki-67, and E-cadherin (E-cad) expression in relation to chemotherapy response and prognosis of patients with advanced primary triple-negative breast cancer (TNBC). METHODS CC3 expression was detected immunohistochemically in 67 pre-chemotherapy biopsy samples. Ki67 and E-cad levels were obtained from patients' medical records. RESULTS CC3-positivity (N = 32; 47.8%) was associated with a higher first-line chemotherapy overall response rate (ORR; P = 0.028) and second-line chemotherapy clinical benefit rate (CBR; P = 0.033). The Ki-67 high-risk group (N = 51; 76.1%) exhibited a reduced second-line chemotherapy CBR (P = 0.024). The E-cad negative group (N = 25; 37.3%) exhibited a lower first-line chemotherapy ORR (P = 0.044) and CBR (P<0.001), and a lower second-line chemotherapy CBR (P = 0.020). CC3, Ki-67, and E-cad were significant predictors of third-line chemotherapy ORR or CBR. Similar numbers of chemotherapy cycles were completed by the CC3-positive and -negative groups. The Ki-67 high-risk and E-cad negative groups completed fewer second-line chemotherapy cycles (P = 0.038) and fewer first-line chemotherapy cycles, respectively (P = 0.001). Kaplan-Meier analyses identified worse outcomes for the CC3-positive, Ki-67 high-risk, and E-cad negative groups than for their corresponding comparison groups (P<0.05). Multivariate Cox regression analysis identified CC3 expression and an absence of E-cad expression as independent survival factors (P<0.05). CONCLUSIONS Our CC3-positive group exhibited a better chemotherapy response, but a worse prognosis. The Ki-67 high-risk and E-cad negative groups exhibited both a worse chemotherapy response and worse prognosis.
Collapse
Affiliation(s)
- Xiaodan Liu
- Molecular Oncology Laboratory of Cancer Research Institute, The First Affiliated Hospital of China Medical UniversityShenyang, Liaoning Province, China
- Department of General Surgery, Shenyang Military General HospitalShenyang, China
| | - Shenyi Jiang
- Department of General Medical Practice, The First Affiliated Hospital of China Medical UniversityShenyang, Liaoning Province, China
| | - Xin Tian
- Molecular Oncology Laboratory of Cancer Research Institute, The First Affiliated Hospital of China Medical UniversityShenyang, Liaoning Province, China
| | - Youhong Jiang
- Molecular Oncology Laboratory of Cancer Research Institute, The First Affiliated Hospital of China Medical UniversityShenyang, Liaoning Province, China
| |
Collapse
|
34
|
Wu T, Cui H, Xu Y, Du Q, Zhao E, Cao J, Nie L, Fu G, Ren A. The effect of tubeimoside-1 on the proliferation, metastasis and apoptosis of oral squamous cell carcinoma in vitro. Onco Targets Ther 2018; 11:3989-4000. [PMID: 30022842 PMCID: PMC6044352 DOI: 10.2147/ott.s164503] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Background Tubeimoside-1 (TBMS1), a triterpenoid saponin extracted from traditional Chinese medicine tubeimoside, exerts a cytotoxic effect on several human cancer cell lines. However, no study has focused on whether TBMS1 works on oral squamous cell carcinoma (OSCC). Materials and methods We treated OSCC cells with TBMS1 to detect the effect and relevant molecular basis of TBMS1 for the first time. We chose two oral cancer cell lines, CAL27 and SCC15, for this study. First, the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenylte-trazolium bromide assay and cell proliferation 5′-bromo-2′-deoxyuridine assay were carried out to detect cell growth. Second, colony formation assay was performed to assess clonogenesis capacity. Next apoptosis was analyzed by flow cytometry. Subsequently, wound healing and transwell assays were applied to explore cell migration. Finally, Western blot was further performed to examine corresponding proteins’ expression change. Results Our data showed that TBMS1 significantly suppressed proliferation of OSCC cells in a dose- and time-dependent manner and it inhibited migration of OSCC cells as well. After treatment with TBMS1, OSCC cells underwent cell apoptosis. Furthermore, Western blot demonstrated that TBMS1 downregulated apoptosis-associated proteins such as PARP, p-ERK1/2, Bcl-2, caspase-3, caspase-7 and caspase-8 and upregulated cleaved PARP, cleaved caspase-3 and cleaved caspase-9. It could also reduce expression of c-Myc and MMP-7. Meanwhile, TBMS1 did not change the total ERK1/2 expression. Conclusion These results revealed that TBMS1 might be a potential chemotherapeutic drug for the management of OSCC.
Collapse
Affiliation(s)
- Tingting Wu
- College of Stomatology, Chongqing Medical University, Chongqing, People's Republic of China, .,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, People's Republic of China, .,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, People's Republic of China, .,Department of Oral Implantology, Stomatological Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, People's Republic of China
| | - Yamei Xu
- College of Stomatology, Chongqing Medical University, Chongqing, People's Republic of China, .,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, People's Republic of China, .,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, People's Republic of China, .,Department of Oral Implantology, Stomatological Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Quangao Du
- College of Stomatology, Chongqing Medical University, Chongqing, People's Republic of China, .,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, People's Republic of China, .,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, People's Republic of China, .,Department of Oral Implantology, Stomatological Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Erhu Zhao
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, People's Republic of China
| | - Jiangjun Cao
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing, People's Republic of China
| | - Ling Nie
- College of Stomatology, Chongqing Medical University, Chongqing, People's Republic of China, .,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, People's Republic of China, .,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, People's Republic of China, .,Department of Oral Implantology, Stomatological Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Gang Fu
- College of Stomatology, Chongqing Medical University, Chongqing, People's Republic of China, .,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, People's Republic of China, .,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, People's Republic of China, .,Department of Oral Implantology, Stomatological Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Aishu Ren
- College of Stomatology, Chongqing Medical University, Chongqing, People's Republic of China, .,Chongqing Key Laboratory of Oral Diseases and Biomedical Sciences, Stomatological Hospital of Chongqing Medical University, Chongqing, People's Republic of China, .,Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital of Chongqing Medical University, Chongqing, People's Republic of China, .,Department of Orthodontics, Stomatological Hospital of Chongqing Medical University, Chongqing, People's Republic of China,
| |
Collapse
|
35
|
Chen L, Weng Q, Li F, Liu J, Zhang X, Zhou Y. Pharmacokinetics and Bioavailability Study of Tubeimoside I in ICR Mice by UPLC-MS/MS. JOURNAL OF ANALYTICAL METHODS IN CHEMISTRY 2018; 2018:9074893. [PMID: 30116651 PMCID: PMC6079592 DOI: 10.1155/2018/9074893] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 06/19/2018] [Indexed: 06/08/2023]
Abstract
The aim of this study is to establish and validate a rapid, selective, and sensitive ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS) method to determine tubeimoside I (TBMS-I) in ICR (Institute of Cancer Research) mouse whole blood and its application in the pharmacokinetics and bioavailability study. The blood samples were precipitated by acetonitrile to extract the analytes. Chromatographic separation was performed on a UPLC BEH C18 column (2.1 mm × 50 mm, 1.7 μm). The mobile phase consisted of water with 0.1% formic acid and methanol (1 : 1, v/v) at a flow rate of 0.4 mL/min. The total eluting time was 4 min. The TBMS-I and ardisiacrispin A (internal standard (IS)) were quantitatively detected by a tandem mass spectrometry equipped with an electrospray ionization (ESI) in a positive mode by multiple reaction monitoring (MRM). A validation of this method was in accordance with the US Food and Drug Administration (FDA) guidelines. The lower limit of quantification (LLOQ) of TBMS-I was 2 ng/mL, and the calibration curve was linearly ranged from 2 to 2000 ng/mL (r2 ≥ 0.995). The relative standard deviation (RSD) of interday precision and intraday precision was both lower than 15%, and the accuracy was between 91.7% and 108.0%. The average recovery was >66.9%, and the matrix effects were from 104.8% to 111.0%. In this assay, a fast, highly sensitive, and reproducible quantitative method was developed and validated in mouse blood for the first time. The absolute availability of TBMS-I in the mouse was only 1%, exhibiting a poor oral absorption.
Collapse
Affiliation(s)
- Lianguo Chen
- Wenzhou People's Hospital, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou 325000, China
| | - Qinghua Weng
- Wenzhou People's Hospital, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou 325000, China
| | - Feifei Li
- Wenzhou People's Hospital, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou 325000, China
| | - Jinlai Liu
- Wenzhou People's Hospital, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou 325000, China
| | - Xueliang Zhang
- Wenzhou People's Hospital, The Third Clinical Institute Affiliated to Wenzhou Medical University, Wenzhou 325000, China
| | - Yunfang Zhou
- Laboratory of Clinical Pharmacy, The People's Hospital of Lishui, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China
| |
Collapse
|
36
|
Shi H, Bi H, Sun X, Dong H, Jiang Y, Mu H, Li W, Liu G, Gao R, Su J. Tubeimoside-1 inhibits the proliferation and metastasis by promoting miR-126-5p expression in non-small cell lung cancer cells. Oncol Lett 2018; 16:3126-3134. [PMID: 30127904 PMCID: PMC6096222 DOI: 10.3892/ol.2018.9051] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 06/12/2018] [Indexed: 12/27/2022] Open
Abstract
Tubeimoside-1 (TBMS1) possesses broad anticancer activities, including the cytostatic and anti-angiogenesis effects in lung cancer. However, the effect of TBMS1 on the metastasis of non-small cell lung cancer (NSCLC) cells and the potential underlying mechanism remain unclear. In the present study, a cell counting kit-8 assay revealed that TBMS1 suppressed the proliferation of NCI-H1299 cells significantly, particularly following 48 h of treatment. Further studies showed that TBMS1 notably enhanced the apoptosis, and inhibited the migration and invasion of NCI-H1299 cells upon treatment for 48 h. A total of 14 NSCLC tissues and 14 normal adjacent tissues were collected, reverse transcription-quantitative polymerase chain reaction revealed decreased expression of microRNA (miR)-126-5p in NSCLC tissues compared with adjacent NSCLC tissues, which was reversed following TBMS1 administration in NCI-H1299 cells. The overexpression of miR-126-5p induced by TBMS1 was demonstrated to target and downregulate vascular endothelial growth factor (VEGF)-A. Simultaneously, the expression of VEGF-R2 was reduced notably, along with a significant declined in the phosphorylation levels of dual specificity mitogen-activated protein kinase kinase 1 and extracellular signal-regulated kinase (ERK)1/2. Overall, the aforementioned results indicated that TBMS1 inhibited the proliferation and metastasis, and promoted the apoptosis of NCI-H1299 cells, which may be mediated by overexpressing miR-126-5p, which inactivates the VEGF-A/VEGFR2/ERK signaling pathway. Therefore, TBMS1 may be a promising drug for prevention and treatment of NSCLC.
Collapse
Affiliation(s)
- Hanbing Shi
- Department of Respiration II, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Hongxia Bi
- Department of Respiratory Medicine, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Xingyuan Sun
- Department of Neurology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Haiying Dong
- Laboratory Center of Ultrastructural Pathology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Yunfei Jiang
- Department of Respiration II, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Haijun Mu
- Department of Respiration II, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Wei Li
- Department of Respiration II, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Guohua Liu
- Department of Respiration II, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Ruizhi Gao
- Department of Respiratory Medicine, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Jiang Su
- Department of Respiratory Medicine, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| |
Collapse
|
37
|
Ceragenin CSA-13 as free molecules and attached to magnetic nanoparticle surfaces induce caspase-dependent apoptosis in human breast cancer cells via disruption of cell oxidative balance. Oncotarget 2018; 9:21904-21920. [PMID: 29774111 PMCID: PMC5955147 DOI: 10.18632/oncotarget.25105] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 03/22/2018] [Indexed: 01/31/2023] Open
Abstract
Natural antimicrobial peptides and ceragenins, as non-peptide amphipathic mimics, have been proposed as anti-cancer agents. To date, it has been confirmed that cathelicidin LL-37 and ceragenin CSA-13, both in free form and immobilized on the surface of magnetic nanoparticles (MNP@LL-37, MNP@CSA-13) induce apoptosis in colon cancer cells. Nevertheless, the question remains whether ceragenins, as synthetic analogs of LL-37 peptide and mimicking a number of its properties, act as antineoplastic agents in breast cancer cells, where LL-37 peptide stimulates oncogenesis. Considering potential anticancer activity, we determined whether CSA-13 and MNP@CSA-13 might be effective against breast cancer cells. Our study provides evidence that both CSA-13 and MNP@CSA-13 decreased viability and inhibit proliferation of MCF-7 and MDA-MB-231 cells despite the protumorigenic properties of LL-37 peptide. Flow cytometry-based analyses revealed that ceragenin treatment results in increases in dead and PI-negative/low-viability cells, which was associated with glutathione (GSH) depletion and increased reactive oxygen species (ROS) generation followed by mitochondrial membrane depolarization, caspase activation, and DNA fragmentation. These findings demonstrate that both CSA-13 and MNP@CSA-13 cause disruption of the oxidative balance of cancer cells. This novel mechanism of ceragenin-mediated eradication of cancer cells suggest that these agents may be developed as a possible treatment of breast cancer.
Collapse
|
38
|
Chen C, Wang K, Wang Q, Wang X. LncRNA HULC mediates radioresistance via autophagy in prostate cancer cells. ACTA ACUST UNITED AC 2018; 51:e7080. [PMID: 29694502 PMCID: PMC5937721 DOI: 10.1590/1414-431x20187080] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 01/02/2018] [Indexed: 12/22/2022]
Abstract
Prostate cancer (PCa) is the second leading cause of cancer death in men. Irradiation is one of the available options for treatment of PCa, however, approximately 10-45% of PCa are resistant to irradiation. We aimed to explore the role of long non-coding RNA highly upregulated in liver cancer (HULC) in the sensitivity of PCa cells to irradiation. Survival rate, cell apoptosis, cycle, expressions of related proteins, and caspase-3 activity were assessed to explore the effects of HULC on sensitivity of PCa cells to irradiation. Expression of HULC in DU-145, PC3, LNCaP, and RWPE-1 cells was determined and the influence of HULC on DU-145 cells was explored. Then, PC3 cells aberrantly expressing HULC were implanted into NOD-SCID mice for tumor xenograft study. Changes of autophagy after aberrant expression of HULC in vivo and in vitro were tested. Furthermore, the interacted protein of HULC and involved signaling pathway were investigated. In PC3 and LNCaP cells under irradiation, survival rate and cell cycle were decreased and apoptosis was increased by HULC knockdown. HULC knockdown arrested PC3 cells at G0/G1 phase. DU-145 was sensitive to irradiation, and resistance to irradiation of DU-145 cells was enhanced by HULC overexpression. Moreover, HULC knockdown enhanced the sensitivity of PC3 xenografts to irradiation. HULC knockdown promoted autophagy through interaction with Beclin-1 and inhibition of mTOR, resulting in increased apoptosis. HULC knockdown improved sensitivity of PCa cells to irradiation both in vivo and in vitro. HULC suppressed Beclin-1 phosphorylation, thereby reduced autophagy, involving the mTOR pathway.
Collapse
Affiliation(s)
- Changxuan Chen
- Tengzhou Central People's Hospital, Department of Urology, Jining Medical College, Tengzhou, China
| | - Kaizhen Wang
- Tengzhou Central People's Hospital, Department of Urology, Jining Medical College, Tengzhou, China
| | - Qian Wang
- Tengzhou Central People's Hospital, Department of Traumatology, Jining Medical College, Tengzhou, China
| | - Xin Wang
- Tengzhou Central People's Hospital, Department of Urology, Jining Medical College, Tengzhou, China
| |
Collapse
|
39
|
Zhuang J, Liu Y, Yuan Q, Liu J, Liu Y, Li H, Wang D. Blue light-induced apoptosis of human promyelocytic leukemia cells via the mitochondrial-mediated signaling pathway. Oncol Lett 2018; 15:6291-6296. [PMID: 29731847 PMCID: PMC5921239 DOI: 10.3892/ol.2018.8162] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 02/28/2018] [Indexed: 02/06/2023] Open
Abstract
Acute promyelocytic leukemia is frequently associated with dizziness, fever, nausea, hematochezia and anemia. Blue light, or light with wavelengths of 400–480 nm, transmits high levels of energy. The aim of the present study was to determine the pro-apoptotic effects of blue light (wavelength, 456 nm; radiation power, 0.25 mW/cm2) and the underlying mechanisms in a human promyelocytic leukemia cell line (HL60). Blue light reduced the viability and enhanced the mortality of HL60 cells in a time-dependent manner. Exposure to blue light for 24 h caused depolarization of the mitochondrial membrane potential and the overproduction of reactive oxygen species in HL60 cells. In a nude mouse model, 9-day exposure to blue light markedly suppressed the growth of HL60-xenografted tumors; however, it had no effect on hepatic and renal tissues. In addition, blue light abrogated the expression of B-cell lymphoma (Bcl)-2 and Bcl extra-long, while enhancing the levels of Bcl-2-associated X protein, cytochrome c, and cleaved caspases-3 and −9 in tumor tissues. The results suggested that the pro-apoptotic effects of blue light in human promyelocytic leukemia cells may be associated with the mitochondrial apoptosis signaling pathway.
Collapse
Affiliation(s)
- Jianjian Zhuang
- State Key Laboratory of Superhard Materials, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yange Liu
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Qingxia Yuan
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Junsong Liu
- State Key Laboratory of Superhard Materials, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yan Liu
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Hongdong Li
- State Key Laboratory of Superhard Materials, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun, Jilin 130012, P.R. China
| |
Collapse
|
40
|
Ye SF, Yang Y, Wu L, Ma WW, Zeng HH. Ethaselen: a novel organoselenium anticancer agent targeting thioredoxin reductase 1 reverses cisplatin resistance in drug-resistant K562 cells by inducing apoptosis. J Zhejiang Univ Sci B 2018; 18:373-382. [PMID: 28471109 DOI: 10.1631/jzus.b1600073] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
It has been reported that Ethaselen shows inhibitory effects on thioredoxin reductase (TrxR) activity and human tumor cell growth. In order to find an efficient way to reverse cisplatin resistance, we investigated the reversal effects of Ethaselen on cisplatin resistance in K562/cisplatin (CDDP) cells that were established by pulse-inducing human erythrocyte leukemic cell line K562, which are fivefold more resistant to cisplatin compared to K562 cells. The morphology and growth showed that the adhesion of K562/CDDP further decreased while the cell volume increased. The proliferation of K562/CDDP is strengthened. The antitumor activities in vitro were assessed by MTT (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) assay and combination index (CI), showing the significant synergic effects of cisplatin and Ethaselen. Focusing on apoptosis, a series of comparisons was made between K562 and K562/CDDP. Cisplatin induced higher reactive oxygen species (ROS) generation in K562 and subsequently induced the formation of mitochondrial permeability transition pores (PTPs). In addition, cisplatin increased the ratio of Bax to Bcl-2 in K562, which can influence the mitochondrial membrane permeability. PTP formation and mitochondrial membrane permeabilization eventually resulted in the release of cytochrome c and activation of the Caspase pathway. However, these effects were not clearly seen in K562/CDDP, which may be the reason for the acquired CDDP resistance. However, Ethaselen can induce a high level of ROS in K562/CDDP by TrxR activity inhibition and increased ratio of Bax to Bcl-2 in K562/CDDP by nuclear factor κB (NF-κB) suppression, which subsequently induces the release of cytochrome c in K562/CDDP. This response is partly responsible for the reversal of the cisplatin resistance in K562/CDDP cells.
Collapse
Affiliation(s)
- Suo-Fu Ye
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China.,School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yong Yang
- Keaise Clinical Examination Lab, Wuhan 430000, China
| | - Lin Wu
- Keaise Clinical Examination Lab, Wuhan 430000, China
| | - Wei-Wei Ma
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China.,School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Hui-Hui Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, China.,School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
41
|
Zhou X, Liu J, Zhang J, Wei Y, Li H. Flubendazole inhibits glioma proliferation by G2/M cell cycle arrest and pro-apoptosis. Cell Death Discov 2018. [PMID: 29531815 PMCID: PMC5841417 DOI: 10.1038/s41420-017-0017-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Flubendazole, FDA-approved anthelmintic, has been widely used in treating testinal parasites. In the recent years, Flubendazole has been reported to exert anticancer activities. On the other hand, little was known about the effects of Flubendazole on gliomas. Here we demonstrated a novel effect of flubendazole on glioma cells. We found that Flubendazole inhibited cell proliferation and promoted cell apoptosis of glioma cell lines in vitro, and suppressed tumor growth in xenograft models by intraperitoneal injection. However, Flubendazole might have no influence on cell migration. Mechanism study reaveled that Flubendazole caused cell cycle arrest in G2/M phase, which partly account for the suppressed proliferation. Consistently, Flubendazole induced P53 expression and reduced Cyclin B1 and p-cdc2 expression in glioma cells. In addition, Flubendazole promoted cell apoptosis by regulating the classical apoptosis protein BCL-2 expression. These observations suggest that Flubendazole exerts anti-proliferation and pro-apoptosis effects in Glioma through affecting the cell cycle and intrinsic apoptotic signaling, and indicate a novel utilization of Flubendazole in the treatment of Glioma.
Collapse
Affiliation(s)
- Xumin Zhou
- 1Department of Pathogen Biology and Experimental teaching center of Preventive Medicine, Guangdong Provincial Key Laboratory of Tropical Disease, School of Public Health, Southern Medical University, Guangzhou, 510515 China
| | - Jumei Liu
- 1Department of Pathogen Biology and Experimental teaching center of Preventive Medicine, Guangdong Provincial Key Laboratory of Tropical Disease, School of Public Health, Southern Medical University, Guangzhou, 510515 China
| | - Jinming Zhang
- 2Department of Respiration, Nanfang Hospital, Southern Medical University, Guangzhou, 510515 China
| | - Yong Wei
- 1Department of Pathogen Biology and Experimental teaching center of Preventive Medicine, Guangdong Provincial Key Laboratory of Tropical Disease, School of Public Health, Southern Medical University, Guangzhou, 510515 China
| | - Hua Li
- 1Department of Pathogen Biology and Experimental teaching center of Preventive Medicine, Guangdong Provincial Key Laboratory of Tropical Disease, School of Public Health, Southern Medical University, Guangzhou, 510515 China
| |
Collapse
|
42
|
Shi H, Bi H, Sun X, Dong H, Jiang Y, Mu H, Liu G, Kong W, Gao R, Su J. Antitumor effects of Tubeimoside-1 in NCI-H1299 cells are mediated by microRNA-126-5p-induced inactivation of VEGF-A/VEGFR-2/ERK signaling pathway. Mol Med Rep 2018; 17:4327-4336. [PMID: 29363720 PMCID: PMC5802206 DOI: 10.3892/mmr.2018.8459] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 01/02/2018] [Indexed: 12/15/2022] Open
Abstract
Tubeimoside-1 (TBMS1), a triterpenoid saponin isolated from the tuber of Bolbostemma paniculatum (Maxim) Franquet, serves an universal suppressive role in multiple cancer types, including lung cancer. However, the mechanism involved in non-small cell lung cancer (NSCLC) cells by which TBMS1 elicits its antitumor effects is not yet completely understood. The present study indicated that 10 µmol/l TBMS1 significantly enhanced apoptosis and notably blocked the migration and invasion of NCI-H1299 cells. These effects were reversed following transfection with miR-126-5p inhibitor into TBMS1-treated NCI-H1299 cells. Vascular endothelial growth factor-A (VEGF-A) is a target gene for miR-126-5p. Notably, results suggested that the downregulated VEGF-A and VEGFR-2 in TBMS1-treated NCI-H1299 cells were upregulated after inhibiting miR-126-5p, and overexpression of VEGF-A or VEGFR-2 could significantly reduce apoptosis and promote the migration and invasion of TBMS1-treated NCI-H1299 cells. Furthermore, TBMS1 combined with TBHQ (an ERK activator) dramatically suppressed TBMS1-induced apoptosis and stimulated TBMS1-reduced migration and invasion in NCI-H1299 cells, suggesting that TBMS1 inhibits the ERK signaling pathway and represses the growth and metastasis of NCI-H1299 cells. Further study demonstrated that either inhibiting miR-126-5p or overexpressing VEGF-A and VEGFR-2 in TBMS1-treated NCI-H1299 cells elevated the mRNA expression levels and phosphorylation levels of MEK1, as well as ERK. To conclude, TBMS1 increases miR-126-5p expression, whereas overexpressing miR-126-5p inactivates VEGF-A/VEGFR-2/ERK signaling pathway, which ultimately actuates the pro-apoptotic and anti-metastatic effects in NCI-H1299 cells. Therefore, the present findings provide a theoretical foundation for TBMS1 as a potential candidate in NSCLC treatment.
Collapse
Affiliation(s)
- Hanbing Shi
- Department of Respiration II, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Hongxia Bi
- Department of Respiratory Medicine, The Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Xingyuan Sun
- Department of Neurology, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Haiying Dong
- Laboratory Center of Ultrastructural Pathology, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Yunfei Jiang
- Department of Respiration II, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Haijun Mu
- Department of Respiration II, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Guohua Liu
- Department of Respiration II, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Weili Kong
- Department of Respiration II, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Ruizhi Gao
- Department of Respiration II, The Third Affiliated Hospital of Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| | - Jiang Su
- Department of Cardiovascular Medicine, Qiqihar Medical University, Qiqihar, Heilongjiang 161006, P.R. China
| |
Collapse
|
43
|
Yang X, Zhong DN, Qin H, Wu PR, Wei KL, Chen G, He RQ, Zhong JC. Caspase-3 over-expression is associated with poor overall survival and clinicopathological parameters in breast cancer: a meta-analysis of 3091 cases. Oncotarget 2017; 9:8629-8641. [PMID: 29492222 PMCID: PMC5823563 DOI: 10.18632/oncotarget.23667] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 10/28/2017] [Indexed: 01/11/2023] Open
Abstract
Caspase-3 is a vital executioner molecule during the apoptotic process. Numerous studies have revealed the close association of caspase-3 expression and breast cancer. Nevertheless, the prognostic value of caspase-3 expression for patients with breast cancer remains uncertain. To thoroughly analyze the prognostic effect of caspase-3 expression on the clinicopathological features and survival of breast cancer, we conducted this meta-analysis. With various search strategies, electronic databases were comprehensively searched. A total of 3091 patients from 21 studies were ultimately obtained. The analysis results indicated that increased expression of caspase-3 had a negative influence on the overall survival (OS) of breast cancer (HR = 1.73, 95%CI 1.12–2.67, P = 0.014). Subgroup analyses based on race revealed that the value of caspase-3 for evaluating patients’ OS was more useful in Asian patients (HR = 3.16, 95%CI 1.20–8.15, P = 0.020), and subgroup analyses based on study analytical methods revealed that caspase-3 was a risk factor for breast cancer patients in multivariate overall survival analyses (HR = 1.67, 95%CI 1.02–2.75, P = 0.044). As for the relationship between caspase-3 expression and breast cancer subtype as well as progression, caspase-3 might serve as a risk factor for the progestogen receptor (PR) and human epidermal growth factor receptor-2 (HER-2) subtypes (OR = 1.44, 95%CI 1.09–1.89, P = 0.010; OR = 1.76, 95%CI 1.18–2.62, P = 0.050, respectively) of breast cancer. However, no evidence showed that increased expression of caspase-3 was statistically correlated with tumor differentiation state (low/moderate or high), tumor TNM stage (I-II/III-IV) or lymph node metastasis (–/+). In conclusion, this meta-analysis revealed that increased caspase-3 expression was significantly associated with worse prognosis and two subtypes of breast cancer. More prospective studies are urgently needed to define the prognostic value of caspase-3 expression in patients with breast cancer.
Collapse
Affiliation(s)
- Xia Yang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Da-Ni Zhong
- Department of Chemotherapy, Tumor Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Hui Qin
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Pei-Rong Wu
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Kang-Lai Wei
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Rong-Quan He
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| | - Jin-Cai Zhong
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, China
| |
Collapse
|
44
|
Nucleoside reverse transcriptase inhibitor-induced rat oocyte dysfunction and low fertility mediated by autophagy. Oncotarget 2017; 9:3895-3907. [PMID: 29423092 PMCID: PMC5790509 DOI: 10.18632/oncotarget.23243] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 12/01/2017] [Indexed: 12/16/2022] Open
Abstract
Low fertility is one of the most common side effects caused by nucleoside reverse transcriptase inhibitors (NRTIs), whereas the molecular mechanism underlying this process were largely unclear. This study was conducted to investigate whether autophagy plays a role in NRTIs-induced oocyte dysfunction and low fertility in female rat. Both in vivo and in vitro experiments were conducted. For the in vivo experiment, female adult Sprague-Dawley rats were subjected to zidovudine (AZT) and lamivudine (3TC) intragastric treatment for 3, 6, 9, and 12 weeks; a control was also set. Oocytes were collected for maturation evaluation, in vitro fertilization and mitochondrial function assays, and apoptosis and autophagy analysis. For the in vitro experiment, oocytes were collected and assigned to the control, 3-methyladenine (3-MA, an effective autophagy inhibitor), AZT, AZT+3-MA, 3TC, and 3TC+3-MA groups. The oocytes were cultured with the abovementioned drugs for 24, 48, and 72 h and then, subjected to the same assays as in the in vivo study. The results showed a significant time-dependent decrease in oocyte maturation-related maker levels, oocyte cleavage rate, blastocyst formation rate, mitochondrial DNA copy number and adenosine triphosphate level, and apoptosis, and a significant increase in the reactive oxygen species levels (all P-values < 0.05), in both the in vivo and the in vitro experiments. These changes, except for the changes in the oocyte maturation-related markers, were partially attenuated by 3-MA. In conclusion, we demonstrated that NRTIs can cause rat oocyte dysfunction and low fertility, and this damage was, at least partially, mediated by autophagy.
Collapse
|
45
|
Zhang JB, Zhang L, Li SQ, Hou AH, Liu WC, Dai LL. Tubeimoside I attenuates inflammation and oxidative damage in a mice model of PM 2.5-induced pulmonary injury. Exp Ther Med 2017; 15:1602-1607. [PMID: 29434745 DOI: 10.3892/etm.2017.5597] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 10/20/2017] [Indexed: 01/20/2023] Open
Abstract
In the present study, the effects of tubeimoside I (TBMS1) on particulate matter <2.5 µm in diameter (PM2.5)-induced pulmonary injury and its mechanisms of action were investigated. Male BALB/c mice were randomly assigned into five groups (n=10/group): Control, PM2.5, PM2.5 + TBMS1 45 mg/kg, PM2.5 + TBMS1 90 mg/kg and PM2.5 + TBMS1 180 mg/kg. The dose of the PM2.5 suspension administered to the mice was 40 mg/kg via nasal instillation. The PM2.5 + TBMS1 groups received TBMS1 daily orally for 21 consecutive days, while the mice in the control and PM2.5 groups received equivalent volumes of PBS. Subsequently, lactic dehydrogenase, acid phosphatase, alkaline phosphatase, albumin, tumor necrosis factor-α and interleukin-6 protein levels in bronchoalveolar lavage fluid were determined. Oxidative stress was evaluated by detecting the protein levels of malondialdehyde, superoxide dismutase and inducible nitric oxide synthase, and the level of nitric oxide in lung tissue. Lastly, histopathological images of lung sections were obtained to observe changes in the lung tissue with treatment. The results indicated that exposure to PM2.5 induced pathological pulmonary changes, and biofilm and parenchymal cell damage, and promoted inflammation and oxidative stress. Treatment with TBMS1 attenuated the development of PM2.5-induced pulmonary injury. Its mechanisms of action were associated with reducing cytotoxic effects, levels of inflammatory mediators and oxidative damage. In conclusion, the results of the present study indicate that TBMS1 is a potential therapeutic drug for treating PM2.5-induced pulmonary injury.
Collapse
Affiliation(s)
- Jin-Bo Zhang
- Center of Preventive Treatment of Disease, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong 264000, P.R. China
| | - Lei Zhang
- Department of Heart Disease, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong 264000, P.R. China
| | - Shi-Qing Li
- Department of Encephalopathy, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong 264000, P.R. China
| | - Ai-Hua Hou
- Department of Oncology, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong 264000, P.R. China
| | - Wei-Chao Liu
- Center of Preventive Treatment of Disease, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong 264000, P.R. China
| | - Ling-Ling Dai
- Department of Oncology, Yantai Hospital of Traditional Chinese Medicine, Yantai, Shandong 264000, P.R. China
| |
Collapse
|
46
|
Li H, Liu J, Wang Y, Fu Z, Hüttemann M, Monks TJ, Chen AF, Wang JM. MiR-27b augments bone marrow progenitor cell survival via suppressing the mitochondrial apoptotic pathway in Type 2 diabetes. Am J Physiol Endocrinol Metab 2017; 313:E391-E401. [PMID: 28698281 PMCID: PMC5668595 DOI: 10.1152/ajpendo.00073.2017] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 06/15/2017] [Accepted: 07/03/2017] [Indexed: 12/24/2022]
Abstract
Bone marrow-derived progenitor cells (BMPCs) are potential candidates for autologous cell therapy in tissue repair and regeneration because of their high angiogenic potential. However, increased progenitor cell apoptosis in diabetes directly limits their success in the clinic. MicroRNAs are endogenous noncoding RNAs that regulate gene expression at the posttranscriptional level, but their roles in BMPC-mediated angiogenesis are incompletely understood. In the present study, we tested the hypothesis that the proangiogenic miR-27b inhibits BMPC apoptosis in Type 2 diabetes. Bone marrow-derived EPCs from adult male Type 2 diabetic db/db mice and their normal littermates db/+ mice were used. MiR-27b expression (real-time PCR) in EPCs was decreased after 24 h of exposure to methylglyoxal (MGO) or oxidized low-density lipoprotein but not high glucose, advanced glycation end products, the reactive oxygen species generator LY83583, or H2O2 The increase in BMPC apoptosis in the diabetic mice was rescued following transfection with a miR-27b mimic, and the increased apoptosis induced by MGO was also rescued by the miR-27b mimic. p53 protein expression and the Bax/Bcl-2 ratio in EPCs (Western blot analyses) were significantly higher in db/db mice, both of which were suppressed by miR-27b. Furthermore, mitochondrial respiration, as measured by oxygen consumption rate, was enhanced by miR-27b in diabetic BMPCs, with concomitant decrease of mitochondrial Bax/Bcl-2 ratio. The 3' UTR binding assays revealed that both Bax, and its activator RUNX1, were direct targets of miR-27b, suggesting that miR-27b inhibits Bax expression in both direct and indirect manners. miR-27b prevents EPC apoptosis in Type 2 diabetic mice, at least in part, by suppressing p53 and the Bax/Bcl-2 ratio. These findings may provide a mechanistic basis for rescuing BMPC dysfunction in diabetes for successful autologous cell therapy.
Collapse
Affiliation(s)
- Hainan Li
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan
| | - Jenny Liu
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan
| | - Yihan Wang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan
| | - Zhiyao Fu
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan
| | - Maik Hüttemann
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan
- Departments of Biochemistry and Molecular Biology, School of Medicine, Wayne State University, Detroit, Michigan
| | - Terrence J Monks
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan
- Integrated Biosciences, Wayne State University, Detroit, Michigan
| | - Alex F Chen
- Clinical Research Institute, Department of Cardiology and Endocrinology, The Sixth People's Hospital, Shanghai Jiaotong University, Shanghai, China
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; and
- Vascular Surgery Research, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Jie-Mei Wang
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, Michigan;
| |
Collapse
|
47
|
Zhang C, Chen W, Zhang X, Zheng Y, Yu F, Liu Y, Wang Y. Grape seed proanthocyanidins induce mitochondrial pathway-mediated apoptosis in human colorectal carcinoma cells. Oncol Lett 2017; 14:5853-5860. [PMID: 29113217 PMCID: PMC5661607 DOI: 10.3892/ol.2017.6992] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 08/17/2017] [Indexed: 01/22/2023] Open
Abstract
Grape seed proanthocyanidins (GSPs) have been reported to possess a wide array of pharmacological and biochemical properties. Recently, GSPs have been reported to inhibit various types of colorectal cancer; however, the mechanism(s) involved remain unclear. The present study investigated the effects of GSPs on HCT-116 human colorectal carcinoma cell line. Exposure of these cells to GSPs for 48 h resulted in a significant concentration-dependent inhibition of cell viability. Further investigation indicated that GSPs induced apoptosis of these cells. Analyses of mRNA expression levels using reverse transcription-quantitative polymerase chain reaction and protein expression levels by western blotting revealed that this was associated with increased expression levels of p53 tumor suppressor protein, cytochrome c, and pro-apoptotic proteins, apoptosis regulator Bax (Bax) and Bcl-2 homologous antagonist/killer. Furthermore, decreased expression levels of the anti-apoptotic protein, B cell lymphoma-2 and activation of caspase-2, caspase-3 and caspase-9 were demonstrated. GSP-induced loss of mitochondrial membrane potential was also detected by JC-1 assay. These findings suggested that GSPs induced colon cancer cell apoptosis via the mitochondrial signaling pathway. This provided evidence indicating that GSPs may provide potential chemotherapeutic agents for colorectal cancer.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China.,Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen, Fujian 361021, P.R. China
| | - Weili Chen
- Department of Chemistry and Physics, Heihe University, Heihe, Heilongjiang 164300, P.R. China
| | - Xuhao Zhang
- Institute of Translational Medicine, The First Hospital, Jilin University, Changchun, Jilin 130031, P.R. China
| | - Yanbing Zheng
- Department of Chemistry and Physics, Heihe University, Heihe, Heilongjiang 164300, P.R. China
| | - Fengli Yu
- Department of Chemistry and Physics, Heihe University, Heihe, Heilongjiang 164300, P.R. China
| | - Yulong Liu
- State Key Laboratory of Supramolecular Structure and Materials, Jilin University, Changchun, Jilin 130012, P.R. China
| | - Yi Wang
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
48
|
Tubeimoside-1 suppresses tumor angiogenesis by stimulation of proteasomal VEGFR2 and Tie2 degradation in a non-small cell lung cancer xenograft model. Oncotarget 2017; 7:5258-72. [PMID: 26701724 PMCID: PMC4868684 DOI: 10.18632/oncotarget.6676] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Accepted: 12/07/2015] [Indexed: 12/29/2022] Open
Abstract
Tubeimoside-1 (TBMS1) is a potent anti-tumor phytochemical. Its functional and molecular mode of action, however, remains elusive so far. Since angiogenesis is essential for tumor progression and metastasis, we herein investigated the anti-angiogenic effects of the compound. In a non-small cell lung cancer (NSCLC) xenograft model we found that treatment of CD1 nu/nu mice with TBMS1 (5 mg/kg) significantly suppressed the growth and vascularization of NCI-H460 flank tumors. Moreover, TBMS1 dose-dependently reduced vascular sprouting in a rat aortic ring assay. In vitro, TBMS1 induced endothelial cell apoptosis without decreasing the viability of NSCLC tumor cells and inhibited the migration of endothelial cells by disturbing their actin filament organization. TBMS1 further stimulated the proteasomal degradation of vascular endothelial growth factor receptor-2 (VEGFR2) and Tie2 in endothelial cells, which down-regulated AKT/mTOR signaling. These findings indicate that TBMS1 represents a novel phytochemical for anti-angiogenic treatment of cancer and other angiogenesis-related diseases.
Collapse
|
49
|
Peng Y, Zhong Y, Li G. Tubeimoside-1 suppresses breast cancer metastasis through downregulation of CXCR4 chemokine receptor expression. BMB Rep 2017; 49:502-7. [PMID: 27157541 PMCID: PMC5227143 DOI: 10.5483/bmbrep.2016.49.9.030] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Indexed: 11/20/2022] Open
Abstract
To examine the effect of TBMS1on breast cancer metastasis, and investigate the potential mechanism by which Tubeimoside-1 (TBMS1) inhibits the CXCR4 expression in breast cancer cells. The expression of CXCR4 in breast cancer cell lines was determined by immunoblotting and real-time PCR. The effect of TBMS1 on NF-κB binding activity was evaluated by EMSA assay and ChIP analysis. Cell proliferation and invasion were analyzed by MTT assay and transwell invasion assay, respectively. The effect of TBMS1 on breast cancer metastasis was further evaluated in a metastasis model of nude mice. TBMS1 suppressed the expression of CXCR4 through inhibition of NF-κB binding activity. TBMS1 inhibited CXCL12-induced invasion in breast cancer cells, while ectopic expression of CXCR4 abolished the inhibitive activity of TBMS1. TBMS1 suppressed breast cancer metastasis in the metastatic model of nude mice. TBMS1 suppressed the CXCR4-mediated metastasis of breast cancer by inhibiting NF-κB binding activity. [BMB Reports 2016; 49(9): 502-507]
Collapse
Affiliation(s)
- Yaojin Peng
- Department of Human Anatomy, Medical College of Hunan Normal University, Changsha, Hunan 410006, China
| | - Yan Zhong
- Department of Gynaecology and Obstetrics, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Gao Li
- Department of Thoracic Surgery, Hainan General Hospital, Haikou, Hannan 570311, China
| |
Collapse
|
50
|
Liu M, Zhao G, Cao S, Zhang Y, Li X, Lin X. Development of Certain Protein Kinase Inhibitors with the Components from Traditional Chinese Medicine. Front Pharmacol 2017; 7:523. [PMID: 28119606 PMCID: PMC5220067 DOI: 10.3389/fphar.2016.00523] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/15/2016] [Indexed: 12/27/2022] Open
Abstract
Traditional Chinese medicines (TCMs) have been used in China for more than two thousand years, and some of them have been confirmed to be effective in cancer treatment. Protein kinases play critical roles in control of cell growth, proliferation, migration, survival, and angiogenesis and mediate their biological effects through their catalytic activity. In recent years, numerous protein kinase inhibitors have been developed and are being used clinically. Anticancer TCMs represent a large class of bioactive substances, and some of them display anticancer activity via inhibiting protein kinases to affect the phosphoinositide 3-kinase, serine/threonine-specific protein kinases, pechanistic target of rapamycin (PI3K/AKT/mTOR), P38, mitogen-activated protein kinase (MAPK) and extracellular signal-regulated kinases (ERK) pathways. In the present article, we comprehensively reviewed several components isolated from anticancer TCMs that exhibited significantly inhibitory activity toward a range of protein kinases. These components, which belong to diverse structural classes, are reviewed herein, based upon the kinases that they inhibit. The prospects and problems in development of the anticancer TCMs are also discussed.
Collapse
Affiliation(s)
- Minghua Liu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University Luzhou, China
| | - Ge Zhao
- Department of Pharmacology, School of Pharmacy, Southwest Medical University Luzhou, China
| | - Shousong Cao
- Department of Pharmacology, School of Pharmacy, Southwest Medical University Luzhou, China
| | - Yangyang Zhang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University Luzhou, China
| | - Xiaofang Li
- Department of Pharmacology, School of Pharmacy, Southwest Medical University Luzhou, China
| | - Xiukun Lin
- Department of Pharmacology, School of Pharmacy, Southwest Medical University Luzhou, China
| |
Collapse
|