1
|
Li Y, Li G, Zuo C, Wang X, Han F, Jia Y, Shang H, Tian Y. Discovery of ganoderic acid A (GAA) PROTACs as MDM2 protein degraders for the treatment of breast cancer. Eur J Med Chem 2024; 270:116367. [PMID: 38581732 DOI: 10.1016/j.ejmech.2024.116367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 03/19/2024] [Accepted: 03/27/2024] [Indexed: 04/08/2024]
Abstract
Breast cancer is one of the most common female malignant tumors, with triple-negative breast cancer (TNBC) being the most specific, highly invasive, metastatic and associated with a poor prognosis. Our previous study showed that the natural product ganoderic acid A (GAA) has a certain affinity for MDM2. In this study, two series of novel GAA PROTACs C1-C10 and V1-V10 were designed and synthesized for the treatment of breast cancer. The antitumor activity of these compounds was evaluated against four human tumor cell lines (MCF-7, MDA-MB-231, SJSA-1, and HepG2). Among them, V9 and V10 showed stronger anti-proliferative effects against breast cancer cells, and V10 showed the best selectivity in MDA-MB-231 cells (TNBC), which was 5-fold higher than that of the lead compound GAA. Preliminary structure-activity analysis revealed that V-series GAA PROTACs had better effects than C-series, and the introduction of 2O-4O PEG linkers could significantly improve the antitumor activity. Molecular docking, surface plasmon resonance (SPR), cellular thermal shift assay (CETSA), and Western blot researches showed that both V9 and V10 could bind with MDM2, and degrade the protein through the ubiquitin-proteasome system. Molecular dynamics simulation (MD) revealed that V10 is a bifunctional molecule that can bind to von Hippel-Lindau (VHL) at one end and target MDM2 at the other. In addition, V10 promoted the upregulation of p21 in p53-mutant MDA-MB-231 cells, and induced apoptosis via down-regulation of the bcl-2/bax ratio and the expression of cyclin B1. Finally, in vivo experiments showed that, V10 also exhibited good tumor inhibitory activity in xenografted TNBC zebrafish models, with an inhibition rate of 27.2% at 50 μg/mL. In conclusion, our results suggested that V10 has anti-tumor effects on p53-mutant breast cancer in vitro and in vivo, and may be used as a novel lead compound for the future development of TNBC.
Collapse
Affiliation(s)
- Yan Li
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Guangyu Li
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Chenwei Zuo
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Xiaolin Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Fang Han
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Yi Jia
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China
| | - Hai Shang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China.
| | - Yu Tian
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100193, China.
| |
Collapse
|
2
|
Khorashad JS, Rizzo S, Tonks A. Reactive oxygen species and its role in pathogenesis and resistance to therapy in acute myeloid leukemia. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2024; 7:5. [PMID: 38434766 PMCID: PMC10905166 DOI: 10.20517/cdr.2023.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/24/2024] [Accepted: 02/19/2024] [Indexed: 03/05/2024]
Abstract
Relapse following a short clinical response to therapy is the major challenge for the management of acute myeloid leukemia (AML) patients. Leukemic stem cells (LSC), as the source of relapse, have been investigated for their metabolic preferences and their alterations at the time of relapse. As LSC rely on oxidative phosphorylation (OXPHOS) for energy requirement, reactive oxygen species (ROS), as by-products of OXPHOS, have been investigated for their role in the effectiveness of the standard AML therapy. Increased levels of non-mitochondrial ROS, generated by nicotinamide adenine dinucleotide phosphate oxidase, in a subgroup of AML patients add to the complexity of studying ROS. Although there are various studies presenting the contribution of ROS to AML pathogenesis, resistance, and its inhibition or activation as a target, a model that can clearly explain its role in AML has not been conceptualized. This is due to the heterogeneity of AML, the dynamics of ROS production, which is influenced by factors such as the type of treatment, cell differentiation state, mitochondrial activity, and also the heterogeneous generation of non-mitochondrial ROS and limited available data on their interaction with the microenvironment. This review summarizes these challenges and the recent progress in this field.
Collapse
Affiliation(s)
- Jamshid Sorouri Khorashad
- Department of Immunology and inflammation, Imperial College London, London, W12 0NN, UK
- Department of Molecular Pathology, Institute of Cancer Research, Sutton, SM2 5PT, UK
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Sian Rizzo
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| | - Alex Tonks
- Department of Haematology, Division of Cancer & Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| |
Collapse
|
3
|
Totini CH, Umehara E, Reis IMA, Lago JHG, Branco A. Chemistry and Bioactivity of the Genus Persea - A Review. Chem Biodivers 2023; 20:e202300947. [PMID: 37539983 DOI: 10.1002/cbdv.202300947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/05/2023]
Abstract
This review provides the first comprehensive appraisal of bioactive compounds and their biological activities in Persea species from 1950 to 2023. Relevant articles from reputable databases, including PubMed, Web of Science, Science Direct and Google Scholar were collected, leading to the isolation of about 141 metabolite compounds, mainly flavonoids, terpenoids, fatty alcohols, lignoids, and γ-lactone derivatives. These compounds exhibit diverse biological activities, including insecticidal, antifeedant, nematicidal, antibacterial, antifungal, antiviral, cytotoxic, anti-inflammatory, and antioxidant properties. The review emphasizes the significant chemical and pharmacological potential of different Persea species, encouraging further research in various fields and medicine. Valuable insights into potential applications of Persea plants are provided.
Collapse
Affiliation(s)
- Carlos H Totini
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, 09210-580, Santo Andre, SP, Brazil
| | - Eric Umehara
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, 09210-580, Santo Andre, SP, Brazil
| | - Isabella M A Reis
- Departamento de Saúde, Universidade Estadual de Feira de Santana, 44036-900, Feira de Santana, BA, Brazil
| | - João Henrique G Lago
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, 09210-580, Santo Andre, SP, Brazil
| | - Alexsandro Branco
- Departamento de Saúde, Universidade Estadual de Feira de Santana, 44036-900, Feira de Santana, BA, Brazil
| |
Collapse
|
4
|
Asaro RJ, Profumo E, Buttari B, Cabrales P. The Double-Edged Sword of Erythrocytes in Health and Disease via Their Adhesiveness. Int J Mol Sci 2023; 24:10382. [PMID: 37373527 DOI: 10.3390/ijms241210382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Their widespread presence throughout the vasculature, coupled with their reactivity, and thereby to their potential to release reactive oxidative species, or to utilize their anti-oxidative capacities, has promoted much discussion of the role(s) of red blood cells (RBCs) in the progression of health or, alternatively, a wide range of disease states. Moreover, these role(s) have been linked to the development of adhesiveness and, in fact, thereby to the essential pathway to their eventual clearance, e.g., by macrophages in the spleen. These disparate roles coupled with the mechanisms involved are reviewed and given. Following an analysis, novel perspectives are provided; these perspectives can lead to novel assays for identifying the potential for RBC adhesiveness as suggested herein. We describe this paradigm, that involves RBC adhesiveness, hemolysis, and ghost formation, with examples including, inter alia, the progression of atherosclerosis and the suppression of tumor growth along with other disease states.
Collapse
Affiliation(s)
- Robert J Asaro
- Department of Structural Engineering, University of California, La Jolla, CA 92093-0085, USA
| | - Elisabetta Profumo
- Department of Cardiovascular and Endocrine-Metabolic Diseases, and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Brigitta Buttari
- Department of Cardiovascular and Endocrine-Metabolic Diseases, and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Pedro Cabrales
- Department of Bioengineering, University of California, La Jolla, CA 92093-0085, USA
| |
Collapse
|
5
|
Deng Y, Peng D, Yang C, Zhao L, Li J, Lu L, Zhu X, Li S, Aschner M, Jiang Y. Preventive treatment with sodium para-aminosalicylic acid inhibits manganese-induced apoptosis and inflammation via the MAPK pathway in rat thalamus. Drug Chem Toxicol 2023; 46:59-68. [PMID: 34875954 DOI: 10.1080/01480545.2021.2008127] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Excessive exposure to manganese (Mn) may lead to neurotoxicity, referred to as manganism. In several studies, sodium para-aminosalicylic acid (PAS-Na) has shown efficacy against Mn-induced neurodegeneration by attenuating the neuroinflammatory response. The present study investigated the effect of Mn on inflammation and apoptosis in the rat thalamus, as well as the underlying mechanism of the PAS-Na protective effect. The study consisted of sub-acute (Mn treatment for 4 weeks) and sub-chronic (Mn and PAS-Na treatment for 8 weeks) experiments. In the sub-chronic experiments, pro-inflammatory cytokines, namely tumor necrosis factor α (TNF-α), interleukin 1β (IL-1β), and cyclooxygenase 2 (COX-2) were significantly increased in the Mn-exposed group compared to the control II. PAS-Na treatment led to a significant reduction in the Mn-induced neuroinflammation by inhibiting IL-1β and COX-2 mRNA expression and reducing IL-1β secretion and JNK/p38 MAPK pathway activity. Furthermore, immunohistochemical analysis showed that the expression of caspase-3 was significantly increased in both the sub-acute and sub-chronic experimental paradigms concomitant with a significant decrease in B-cell lymphoma 2 (Bcl-2) in the thalamus of Mn-treated rats. PAS-Na also decreased the expression levels of several apoptotic markers downstream of the MAPK pathway, including Bcl-2/Bax and caspase-3, while up-regulating anti-apoptotic Bcl-2 proteins. In conclusion, Mn exposure led to inflammation in the rat thalamus concomitant with apoptosis, which was mediated via the MAPK signaling pathway. PAS-Na treatment antagonized effectively Mn-induced neurotoxicity by inhibiting the MAPK activity in the same brain region.
Collapse
Affiliation(s)
- Yue Deng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Dongjie Peng
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Chun Yang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Lin Zhao
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Junyan Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Lili Lu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Xiaojuan Zhu
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | - Shaojun Li
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| | | | - Yueming Jiang
- Department of Toxicology, School of Public Health, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Guangxi Medical University, Nanning, China
| |
Collapse
|
6
|
Chen CY, Kao CL, Yeh HC, Li HT, Cheng MJ, Li WJ. A Novel Biaryl Ether from Cinnamomum osmophloeum. Chem Nat Compd 2022. [DOI: 10.1007/s10600-022-03799-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
7
|
Cirsilineol inhibits cell growth and induces apoptosis in glioma C6 cells via inhibiting MAPK and PI3K/Akt/mTOR signaling pathways. APPLIED NANOSCIENCE 2022. [DOI: 10.1007/s13204-021-02229-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
8
|
Orientin reverses acetaminophen-induced acute liver failure by inhibiting oxidative stress and mitochondrial dysfunction. J Pharmacol Sci 2022; 149:11-19. [DOI: 10.1016/j.jphs.2022.01.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 01/07/2022] [Accepted: 01/27/2022] [Indexed: 11/18/2022] Open
|
9
|
Bailly C, Vergoten G. Interaction of obtusilactone B and related butanolide lactones with the barrier-to-autointegration factor 1 (BAF1). A computational study. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2021; 2:100059. [PMID: 34909681 PMCID: PMC8663951 DOI: 10.1016/j.crphar.2021.100059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/08/2021] [Accepted: 09/20/2021] [Indexed: 11/06/2022] Open
Abstract
The barrier-to-autointegration factor 1 (BAF1) protein is a DNA-binding protein implicated in nuclear envelop repair and reformation after mitosis. This nuclear protein is frequently overexpressed in cancer cells and plays a role in the occurrence and development of different tumors. It is a potential therapeutic target for gastric cancer, breast cancer and other malignancies. For this reason, BAF1 inhibitors are searched. The butanolide lactone obtusilactone B (Ob-B) has been found to inhibit VRK1-dependent phosphorylation of BAF1, upon direct binding to the nuclear protein. Taking advantage of the known crystallographic structure of BAF1, we have elaborated molecular models of Ob-B bound to BAF1 to delimit the binding site and binding configuration. The long endoolefinic alkyl side chain of Ob-B extends into a small groove on the protein surface, and the adjacent exomethylene-γ-lactone moiety occupies a pocket comprising to the Ser-4 phosphorylation site of BAF1. Twenty butanolide lactones structurally close to ObB were screened for BAF1 binding. Several natural products with BAF1-binding capacity potentially superior to Ob-B were identified, including mahubanolide, kotomolide B, epilitsenolide D2, and a few other known anticancer plant natural products. Our study provides new ideas to guide the discovery and design of BAF1 inhibitors. Obtusilactone B (Ob-B) is an anticancer inhibitor of VRK1-mediated BAF1 phosphorylation. Molecular models of Ob-B bound to BAF1 have been constructed and the binding site determined. Screening of 20 butanolide lactones led to the identification of new potential BAF1 binders. Mahubanolide, kotomolide B and epilitsenolide D2 emerge as potential BAF1 inhibitors.
Collapse
Affiliation(s)
| | - Gérard Vergoten
- University of Lille, Inserm, INFINITE - U1286, Institut de Chimie Pharmaceutique Albert Lespagnol (ICPAL), Faculté de Pharmacie, 3 rue du Professeur Laguesse, BP-83, F-59006, Lille, France
| |
Collapse
|
10
|
Zunica ERM, Axelrod CL, Cho E, Spielmann G, Davuluri G, Alexopoulos SJ, Beretta M, Hoehn KL, Dantas WS, Stadler K, King WT, Pergola K, Irving BA, Langohr IM, Yang S, Hoppel CL, Gilmore LA, Kirwan JP. Breast cancer growth and proliferation is suppressed by the mitochondrial targeted furazano[3,4-b]pyrazine BAM15. Cancer Metab 2021; 9:36. [PMID: 34627389 PMCID: PMC8502397 DOI: 10.1186/s40170-021-00274-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 09/22/2021] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Enhanced metabolic plasticity and diversification of energy production is a hallmark of highly proliferative breast cancers. This contributes to poor pharmacotherapy efficacy, recurrence, and metastases. We have previously identified a mitochondrial-targeted furazano[3,4-b]pyrazine named BAM15 that selectively reduces bioenergetic coupling efficiency and is orally available. Here, we evaluated the antineoplastic properties of uncoupling oxidative phosphorylation from ATP production in breast cancer using BAM15. METHODS The anticancer effects of BAM15 were evaluated in human triple-negative MDA-MB-231 and murine luminal B, ERα-negative EO771 cells as well as in an orthotopic allograft model of highly proliferative mammary cancer in mice fed a standard or high fat diet (HFD). Untargeted transcriptomic profiling of MDA-MB-231 cells was conducted after 16-h exposure to BAM15. Additionally, oxidative phosphorylation and electron transfer capacity was determined in permeabilized cells and excised tumor homogenates after treatment with BAM15. RESULTS BAM15 increased proton leak and over time, diminished cell proliferation, migration, and ATP production in both MDA-MB-231 and EO771 cells. Additionally, BAM15 decreased mitochondrial membrane potential, while inducing apoptosis and reactive oxygen species accumulation in MDA-MB-231 and EO771 cells. Untargeted transcriptomic profiling of MDA-MB-231 cells further revealed inhibition of signatures associated with cell survival and energy production by BAM15. In lean mice, BAM15 lowered body weight independent of food intake and slowed tumor progression compared to vehicle-treated controls. In HFD mice, BAM15 reduced tumor growth relative to vehicle and calorie-restricted weight-matched controls mediated in part by impaired cell proliferation, mitochondrial respiratory function, and ATP production. LC-MS/MS profiling of plasma and tissues from BAM15-treated animals revealed distribution of BAM15 in adipose, liver, and tumor tissue with low abundance in skeletal muscle. CONCLUSIONS Collectively, these data indicate that mitochondrial uncoupling may be an effective strategy to limit proliferation of aggressive forms of breast cancer. More broadly, these findings highlight the metabolic vulnerabilities of highly proliferative breast cancers which may be leveraged in overcoming poor responsiveness to existing therapies.
Collapse
Affiliation(s)
- Elizabeth R M Zunica
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA.,Department of Nutrition, Case Western Reserve University, Cleveland, OH, 44109, USA.,Clinical Oncology and Metabolism, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Christopher L Axelrod
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA.,Department of Translational Services, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Eunhan Cho
- School of Kinesiology, Louisiana State University, Baton Rouge, LA, USA
| | | | - Gangarao Davuluri
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA.,Sarcopenia and Malnutrition Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Stephanie J Alexopoulos
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Martina Beretta
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Kyle L Hoehn
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, New South Wales, 2052, Australia
| | - Wagner S Dantas
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Krisztian Stadler
- Department of Oxidative Stress and Disease, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - William T King
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA.,Department of Translational Services, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Kathryn Pergola
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA.,Department of Translational Services, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Brian A Irving
- School of Kinesiology, Louisiana State University, Baton Rouge, LA, USA
| | - Ingeborg M Langohr
- Department of Pathobiological Sciences, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Shengping Yang
- Department of Biostatistics, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Charles L Hoppel
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA.,Department of Pharmacology, Case Western Reserve University, Cleveland, OH, 44109, USA
| | - L Anne Gilmore
- Clinical Oncology and Metabolism, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA.,Department of Clinical Nutrition, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - John P Kirwan
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA. .,Department of Nutrition, Case Western Reserve University, Cleveland, OH, 44109, USA. .,Department of Translational Services, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA.
| |
Collapse
|
11
|
Maleki Dana P, Jahanshahi M, Badehnoosh B, Shafabakhsh R, Asemi Z, Hallajzadeh J. Inhibitory effects of berberine on ovarian cancer: Beyond apoptosis. Med Chem Res 2021. [DOI: 10.1007/s00044-021-02763-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
12
|
Cucurbitacin D Induces G2/M Phase Arrest and Apoptosis via the ROS/p38 Pathway in Capan-1 Pancreatic Cancer Cell Line. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:6571674. [PMID: 33029168 PMCID: PMC7527894 DOI: 10.1155/2020/6571674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 08/18/2020] [Accepted: 09/12/2020] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer has a poor prognosis with a five-year survival rate of less than 10%. Moreover, chemotherapy is mostly rendered ineffective owing to chemotherapy resistance and cytotoxicity. Therefore, the development of effective therapeutic strategies and novel drugs against pancreatic cancer is an urgent need. Cucurbitacin D (CuD), a plant steroid derived from Trichosanthes kirilowii, is an anticancer agent effective against various cancer cell lines. However, the anticancer activity and molecular mechanism of CuD in pancreatic cancer remain unknown. Therefore, we aimed to investigate the anticancer activity and molecular mechanism of CuD in the human pancreatic cancer cell line, Capan-1. CuD induced cell cycle arrest at the G2/M phase, apoptosis, and reactive oxygen species generation in Capan-1 cell line. In addition, CuD induced the activation of the p38 MAPK signaling pathway that regulates apoptosis, which was also inhibited by N-acetyl-L-cysteine and the p38 inhibitor SB203580. These data suggest that CuD induces cell cycle arrest and apoptosis via the ROS/p38 pathway in Capan-1 pancreatic cancer cell line; hence, CuD is a promising candidate that should be explored further for its effectiveness as an anticancer agent against pancreatic cancer.
Collapse
|
13
|
Coulidiati TH, Dantas BB, Faheina-Martins GV, de Morais Gomes ER, Gonçalves JCR, de Araújo DAM. Proapoptotic Effects of triazol-1,4-Naphthoquinones Involve Intracellular ROS Production and MAPK/ERK Pathway in Human Leukemia Cells. Anticancer Agents Med Chem 2020; 20:2089-2098. [PMID: 32698747 DOI: 10.2174/1871520620666200721124221] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 06/01/2020] [Accepted: 06/17/2020] [Indexed: 01/01/2023]
Abstract
BACKGROUND The natural products constitute an important source of antitumor and cytotoxic agents. Naphthoquinones are effectively quinones present in different plants, with demonstrated anticancer activities. A recent study conducted by our group demonstrated the antileukemic potential of two novel triazol-1,4- naphthoquinones derivatives, PTN (2-(4-Phenyl-1H-1,2,3-triazol-1-yl)-1,4-naphthoquinone) and MPTN (2-[4- (4-Methoxyphenyl)-1H-1,2,3-triazol-1-yl]-1,4-naphthoquinone). Although, the mechanisms underlying the proapoptotic effects of PTN and MPTN have not been fully elucidated so far. OBJECTIVE The aim of this study was to evaluate the proapoptotic mechanism of PTN and MPTN in human acute leukemia cells. METHODS We used fluorescence microscopy to observe acridine orange and annexin V staining cells. Flow cytometry assay has also been used for ROS quantification, BAX and cytochrome c proteins expression and apoptosis analysis. MTT assay and western blotting technique have been performed as well for MAPK pathway analysis. RESULTS By using the acridine orange and annexin V staining with fluorescence microscopy, we have characterized the proapoptotic effects of PTN and MPTN in HL-60 cells involving the intrinsic mitochondrial pathway, since these compounds promoted an increase in the intracellular BAX and cytochrome c protein levels (p<0.05). We further demonstrated that apoptosis induction in HL-60 cells was mediated by increasing intracellular ROS levels via ERK but not p38 MAPKs pathway. CONCLUSION Taken together, these results have demonstrated that PTN and MPTN are promising tools for the development of new anti-leukemic drugs.
Collapse
Affiliation(s)
- Tangbadioa H Coulidiati
- Department of Life and Earth Sciences, Research and Training Unit in Science and Technology, University Norbert Zongo of Koudougou, BP 376, Koudougou, Burkina Faso,Laboratory of Cellular and Molecular Biotechnology, Department of Biotechnology, Federal University of Paraíba, 58051-900, João Pessoa-PB, Brazil
| | - Bruna B Dantas
- Laboratory of Cellular and Molecular Biotechnology, Department of Biotechnology, Federal University of Paraíba, 58051-900, João Pessoa-PB, Brazil
| | - Glaucia V Faheina-Martins
- Laboratory of Cellular and Molecular Biotechnology, Department of Biotechnology, Federal University of Paraíba, 58051-900, João Pessoa-PB, Brazil
| | - Enéas Ricardo de Morais Gomes
- Laboratory of Cellular and Molecular Biotechnology, Department of Biotechnology, Federal University of Paraíba, 58051-900, João Pessoa-PB, Brazil
| | - Juan C R Gonçalves
- Laboratory of Cellular and Molecular Biotechnology, Department of Biotechnology, Federal University of Paraíba, 58051-900, João Pessoa-PB, Brazil,Department of Pharmaceutical Sciences, Federal University of Paraíba, 58051-900, João Pessoa-PB, Brazil
| | - Demetrius A Machado de Araújo
- Laboratory of Cellular and Molecular Biotechnology, Department of Biotechnology, Federal University of Paraíba, 58051-900, João Pessoa-PB, Brazil
| |
Collapse
|
14
|
Nunes FO, de Almeida JM, Ferreira AMT, da Cruz LA, Jacob CMB, Garcez WS, Garcez FR. Antitrypanosomal butanolides from Aiouea trinervis. EXCLI JOURNAL 2020; 19:323-333. [PMID: 32327956 PMCID: PMC7174576 DOI: 10.17179/excli2020-1088] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 02/26/2020] [Indexed: 11/21/2022]
Abstract
In a search for new antitrypanosomal agents in the Brazilian flora, the ethanol extract of the xylopodium from Aiouea trinervis (Lauraceae) exhibited in vitro activity against the epimastigote forms of Trypanosoma cruzi, the etiological agent of Chagas disease. Bioassay-guided chromatographic fractionation of the ethanol extract afforded three butanolides, isoobtusilactone A (1), epilitsenolide C2 (2), and epilitsenolide C1 (3). Butanolides 1 and 3 were more active against T. cruzi epimastigotes than the reference drug benznidazole (by 8.9-fold and 3.2-fold, respectively), while 2 proved inactive. Compounds 1 and 3 showed low cytotoxicity in mammalian Vero cells (CC50> 156 μmol L-1) and high selectivity index (SI) values for epimastigotes (SI = 56.8 and 28.6, respectively), and 1 was more selective than benznidazole (SI = 46.5). Butanolide 1 at 24 μmol L-1 also led to cell cycle alterations in epimastigote forms, and inhibited the growth of amastigote cells in more than 70 %. In silico ADMET properties of 1 were also analyzed and predicted favorable drug-like characteristics. This butanolide also complied with Lipinski's rule of five and was not predicted as interference compound (PAINS). This is the first report on the isolation of these bioactive butanolides under the guidance of in vitro trypanocidal activity against T. cruzi.
Collapse
Affiliation(s)
- Felipe Oliveira Nunes
- Instituto de Química, Universidade Federal de Mato Grosso do Sul, Av. Senador Filinto Muller 1555, 79074-460 Campo Grande-MS, Brazil
| | - Júlio Menta de Almeida
- Instituto de Biociências, Universidade Federal de Mato Grosso do Sul, Av. Costa e Silva s/n, 79070-900 Campo Grande-MS, Brazil
| | - Alda Maria Teixeira Ferreira
- Instituto de Biociências, Universidade Federal de Mato Grosso do Sul, Av. Costa e Silva s/n, 79070-900 Campo Grande-MS, Brazil
| | - Letícia Alves da Cruz
- Instituto de Biociências, Universidade Federal de Mato Grosso do Sul, Av. Costa e Silva s/n, 79070-900 Campo Grande-MS, Brazil
| | - Camila Mareti Bonin Jacob
- Instituto de Biociências, Universidade Federal de Mato Grosso do Sul, Av. Costa e Silva s/n, 79070-900 Campo Grande-MS, Brazil
| | - Walmir Silva Garcez
- Instituto de Química, Universidade Federal de Mato Grosso do Sul, Av. Senador Filinto Muller 1555, 79074-460 Campo Grande-MS, Brazil
| | - Fernanda Rodrigues Garcez
- Instituto de Química, Universidade Federal de Mato Grosso do Sul, Av. Senador Filinto Muller 1555, 79074-460 Campo Grande-MS, Brazil
| |
Collapse
|
15
|
|
16
|
Pristimerin induces apoptosis and autophagy via activation of ROS/ASK1/JNK pathway in human breast cancer in vitro and in vivo. Cell Death Discov 2019; 5:125. [PMID: 31396402 PMCID: PMC6680048 DOI: 10.1038/s41420-019-0208-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 07/10/2019] [Accepted: 07/15/2019] [Indexed: 12/11/2022] Open
Abstract
Breast cancer is the most common malignant tumor in women, and progress toward long-term survival has stagnated. Pristimerin, a natural quinonemethide triterpenoid, exhibits potential anti-tumor effects on various cancers. However, the underlying mechanism remains poorly understood. In this study, we found that pristimerin reduced the viability of breast cancer cells in vitro and the growth of xenografts in vivo, and these reductions were accompanied by thioredoxin-1 (Trx-1) inhibition and ASK1 and JNK activation. The results showed that pristimerin inhibited cell cycle progression and triggered cell apoptosis and autophagy. Furthermore, we found that the generation of reactive oxygen species (ROS) was a critical mediator in pristimerin-induced cell death. Enhanced ROS generation by pristimerin activated the ASK1/JNK signaling pathway. Inhibition of ROS with N-acetyl cysteine (NAC) significantly decreased pristimerin-induced cell death by inhibiting the phosphorylation of ASK1 and JNK. Taken together, these results suggest a critical role for the ROS/ASK1/JNK pathway in the anticancer activity of pristimerin.
Collapse
|
17
|
Ghosh S, Pal A, Ray M. Methylglyoxal in combination with 5-Fluorouracil elicits improved chemosensitivity in breast cancer through apoptosis and cell cycle inhibition. Biomed Pharmacother 2019; 114:108855. [DOI: 10.1016/j.biopha.2019.108855] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 03/27/2019] [Accepted: 04/02/2019] [Indexed: 10/27/2022] Open
|
18
|
Sharma S, Wu SY, Jimenez H, Xing F, Zhu D, Liu Y, Wu K, Tyagi A, Zhao D, Lo HW, Metheny-Barlow L, Sun P, Bourland JD, Chan MD, Thomas A, Barbault A, D'Agostino RB, Whitlow CT, Kirchner V, Blackman C, Pasche B, Watabe K. Ca 2+ and CACNA1H mediate targeted suppression of breast cancer brain metastasis by AM RF EMF. EBioMedicine 2019; 44:194-208. [PMID: 31129098 PMCID: PMC6604768 DOI: 10.1016/j.ebiom.2019.05.038] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Brain metastases are a major cause of death in patients with metastatic breast cancer. While surgical resection and radiation therapy are effective treatment modalities, the majority of patients will succumb from disease progression. We have developed a novel therapy for brain metastases that delivers athermal radiofrequency electromagnetic fields that are amplitude-modulated at breast cancer specific frequencies (BCF). METHODS 27.12 MHz amplitude-modulated BCF were administered to a patient with a breast cancer brain metastasis by placing a spoon-shaped antenna on the anterior part of the tongue for three one-hour treatments every day. In preclinical models, a BCF dose, equivalent to that delivered to the patient's brain, was administered to animals implanted with either brain metastasis patient derived xenografts (PDXs) or brain-tropic cell lines. We also examined the efficacy of combining radiation therapy with BCF treatment. Additionally, the mechanistic underpinnings associated with cancer inhibition was identified using an agnostic approach. FINDINGS Animal studies demonstrated a significant decrease in growth and metastases of brain-tropic cell lines. Moreover, BCF treatment of PDXs established from patients with brain metastases showed strong suppression of their growth ability. Importantly, BCF treatment led to significant and durable regression of brain metastasis of a patient with triple negative breast cancer. The tumour inhibitory effect was mediated by Ca2+ influx in cancer cells through CACNA1H T-type voltage-gated calcium channels, which, acting as the cellular antenna for BCF, activated CAMKII/p38 MAPK signalling and inhibited cancer stem cells through suppression of β-catenin/HMGA2 signalling. Furthermore, BCF treatment downregulated exosomal miR-1246 level, which in turn decreased angiogenesis in brain environment. Therefore, targeted growth inhibition of breast cancer metastases was achieved through CACNA1H. INTERPRETATION We demonstrate that BCF, as a single agent or in combination with radiation, is a novel treatment approach to the treatment of brain metastases. This paradigm shifting modality warrants further clinical trials for this unmet medical need.
Collapse
Affiliation(s)
- Sambad Sharma
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Shih-Ying Wu
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Hugo Jimenez
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Fei Xing
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Dongqin Zhu
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Yin Liu
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Kerui Wu
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Abhishek Tyagi
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Dan Zhao
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Hui-Wen Lo
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Linda Metheny-Barlow
- Department of Radiation Oncology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Peiqing Sun
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - John D Bourland
- Department of Radiation Oncology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Michael D Chan
- Department of Radiation Oncology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Alexandra Thomas
- Department of Hematology and Oncology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | | | - Ralph B D'Agostino
- Department of Biostatistical Sciences, Division of Public Health Sciences, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Christopher T Whitlow
- Department of Radiology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | | | - Carl Blackman
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Boris Pasche
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America
| | - Kounosuke Watabe
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Winston-Salem, NC, United States of America.
| |
Collapse
|
19
|
Fu T, Houël E, Amusant N, Touboul D, Genta-Jouve G, Della-Negra S, Fisher GL, Brunelle A, Duplais C. Biosynthetic investigation of γ-lactones in Sextonia rubra wood using in situ TOF-SIMS MS/MS imaging to localize and characterize biosynthetic intermediates. Sci Rep 2019; 9:1928. [PMID: 30760744 PMCID: PMC6374367 DOI: 10.1038/s41598-018-37577-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 12/06/2018] [Indexed: 12/22/2022] Open
Abstract
Molecular analysis by parallel tandem mass spectrometry (MS/MS) imaging contributes to the in situ characterization of biosynthetic intermediates which is crucial for deciphering the metabolic pathways in living organisms. We report the first use of TOF-SIMS MS/MS imaging for the cellular localization and characterization of biosynthetic intermediates of bioactive γ-lactones rubrynolide and rubrenolide in the Amazonian tree Sextonia rubra (Lauraceae). Five γ-lactones, including previously reported rubrynolide and rubrenolide, were isolated using a conventional approach and their structural characterization and localization at a lateral resolution of ~400 nm was later achieved using TOF-SIMS MS/MS imaging analysis. 2D/3D MS imaging at subcellular level reveals that putative biosynthetic γ-lactones intermediates are localized in the same cell types (ray parenchyma cells and oil cells) as rubrynolide and rubrenolide. Consequently, a revised metabolic pathway of rubrynolide was proposed, which involves the reaction between 2-hydroxysuccinic acid and 3-oxotetradecanoic acid, contrary to previous studies suggesting a single polyketide precursor. Our results provide insights into plant metabolite production in wood tissues and, overall, demonstrate that combining high spatial resolution TOF-SIMS imaging and MS/MS structural characterization offers new opportunities for studying molecular and cellular biochemistry in plants.
Collapse
Affiliation(s)
- Tingting Fu
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Université Paris-Sud, Université Paris-Saclay, Avenue de la Terrasse, 91198, Gif-sur-Yvette, France.,Institut de Physique Nucléaire, UMR8608, IN2P3-CNRS, Université Paris-Sud, Université Paris-Saclay, 91406, Orsay, France
| | - Emeline Houël
- CNRS UMR8172 EcoFoG, AgroParisTech, CIRAD, INRA, Université des Antilles, Université de Guyane, 97300, Cayenne, France
| | - Nadine Amusant
- CIRAD UMR93 EcoFoG, AgroParisTech, CNRS, INRA, Université des Antilles, Université de Guyane, 97310, Kourou, France
| | - David Touboul
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Université Paris-Sud, Université Paris-Saclay, Avenue de la Terrasse, 91198, Gif-sur-Yvette, France
| | - Grégory Genta-Jouve
- Université Paris Descartes, UMR CNRS 8638 COMETE, 4 avenue de l'observatoire, 75006, Paris, France
| | - Serge Della-Negra
- Institut de Physique Nucléaire, UMR8608, IN2P3-CNRS, Université Paris-Sud, Université Paris-Saclay, 91406, Orsay, France
| | | | - Alain Brunelle
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Université Paris-Sud, Université Paris-Saclay, Avenue de la Terrasse, 91198, Gif-sur-Yvette, France
| | - Christophe Duplais
- CNRS UMR8172 EcoFoG, AgroParisTech, CIRAD, INRA, Université des Antilles, Université de Guyane, 97300, Cayenne, France.
| |
Collapse
|
20
|
Wang JN, Che Y, Yuan ZY, Lu ZL, Li Y, Zhang ZR, Li N, Li RD, Wan J, Sun HD, Sun N, Puno PT, He J. Acetyl-macrocalin B suppresses tumor growth in esophageal squamous cell carcinoma and exhibits synergistic anti-cancer effects with the Chk1/2 inhibitor AZD7762. Toxicol Appl Pharmacol 2019; 365:71-83. [PMID: 30633885 DOI: 10.1016/j.taap.2019.01.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2018] [Revised: 01/04/2019] [Accepted: 01/04/2019] [Indexed: 11/17/2022]
Abstract
Natural products derived from herbal medicines have become a major focus of anti-cancer drug discovery studies. Acetyl-macrocalin B (A-macB) is an ent-diterpenoid isolated from Isodon silvatica. This study aimed to examine the effect and molecular action of A-macB in esophageal squamous cell carcinoma (ESCC) and explore possible drug synergistic modalities. A-macB induced cellular reactive oxygen species (ROS) generation, initiated the p38 mitogen-activated protein kinase (MAPK) signaling pathway, and triggered the caspase-9-dependent apoptosis cascade in ESCC cells. The ROS scavenger N-acetylcysteine (NAC) and the specific p38 inhibitor SB203580 reversed the effects of A-macB on the p38 network and thus rescued ESCC cells from apoptosis. The cellular ROS increase was at least partially due to the suppression of glutathione-S-transferase P1 (GSTP1) by A-macB. A-macB also upregulated the Chk1/Chk2-Cdc25C/Cdc2/Cyclin B1 axis to induce G2/M phase arrest. The cell growth inhibition induced by A-macB was further enhanced by AZD7762, a specific Chk1/Chk2 inhibitor, with a combination index (CI) of <1. Moreover, A-macB efficiently suppressed xenograft growth without inducing significant toxicity, and AZD7762 potentiated the effects of A-macB in the suppression of tumor growth in vivo. Taken together, A-macB is a promising lead compound for ESCC and exerts synergistic anti-cancer effects with AZD7762.
Collapse
Affiliation(s)
- Jing-Nan Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yun Che
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zu-Yang Yuan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhi-Liang Lu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yuan Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Zhi-Rong Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ning Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Ren-Da Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jun Wan
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, China
| | - Han-Dong Sun
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, China
| | - Nan Sun
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| | - Pema-Tenzin Puno
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, Yunnan, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
21
|
Lin CL, Perng MH, Li WJ, Li HT, Chen CY. Chemical Constituents of the Roots of Cinnamomum randaiense. Chem Nat Compd 2018. [DOI: 10.1007/s10600-018-2432-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
22
|
Synthetic Dibenzoxanthene Derivatives Induce Apoptosis Through Mitochondrial Pathway in Human Hepatocellular Cancer Cells. Appl Biochem Biotechnol 2018. [DOI: 10.1007/s12010-018-2721-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
23
|
Heo JR, Lee GA, Kim GS, Hwang KA, Choi KC. Phytochemical-induced reactive oxygen species and endoplasmic reticulum stress-mediated apoptosis and differentiation in malignant melanoma cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2018; 39:100-110. [PMID: 29433671 DOI: 10.1016/j.phymed.2017.12.006] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 11/03/2017] [Accepted: 12/06/2017] [Indexed: 06/08/2023]
Abstract
BACKGROUND Phytochemicals are derived from plants, vegetables and daily products and exert chemopreventive effects. Malignant melanoma is highly metastatic, and melanoma patients can develop chemotherapeutic resistance against conventional melanoma therapies. METHODS In the present study, we investigated the anti-cancer effect of the phytochemicals kaempferol (Kaem), genistein (Gen), and 3'3-diindolylmethane (DIM) on melanoma cell viability. We also evaluated the altered expression of cell cycle-related genes. We verified the production of intracellular reactive oxygen species (ROS) and endoplasmic reticulum (ER) stress at both the protein and cellular level using a western blot, TUNEL assay, and Dihydrodichlorofluorescein diacetate (DCF-DA) assay. RESULTS Treatment of A375SM melanoma cells with phytochemicals resulted in inhibition of cell growth. Treatment with phytochemicals increased the gene expression of p21 and decreased the gene expression of cyclin E and/or cyclin B. The three phytochemicals activated the ROS-p38-p53 apoptotic pathway by increasing the level of phosphorylated p38 MAPK and p53, and they activated the ER stress-mediated apoptotic pathway by increasing the level of phosphorylated eIF2α and C/EBP homologous protein (CHOP). Both the ROS-p38-p53 and ER stress-mediated pathway induced the mitochondrial apoptotic pathway by attenuating Bcl-2 expression and upregulating BAX. Detection of morphological changes demonstrated that Kaem and Gen can induce differentiation in A375SM cell line. CONCLUSION These results indicate that phytochemicals are potentially useful in treatments for melanoma due to their ability to inhibit melanoma cell growth and division via the ROS and ER stress pathway.
Collapse
Affiliation(s)
- Jae-Rim Heo
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Geum-A Lee
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Gyu-Sik Kim
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Kyung-A Hwang
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea
| | - Kyung-Chul Choi
- Laboratory of Biochemistry and Immunology, College of Veterinary Medicine, Chungbuk National University, Cheongju, Chungbuk 28644, Republic of Korea.
| |
Collapse
|
24
|
Zhang Y, Xiao F, Zhong C, Zeng M, Zou L. Retracted Article: Cd induces G2/M cell cycle arrest by up-regulating miR-133b via directly targeting PPP2R2D in L02 hepatocytes. Metallomics 2018; 10:1510-1523. [DOI: 10.1039/c8mt00243f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This study could provide a novel epigenetic mechanism for Cd-induced acute hepatotoxicity and it would offer new targets for its intervention.
Collapse
Affiliation(s)
- Yujing Zhang
- Hunan Normal University School of Medicine
- Changsha 410013
- P. R. China
- Central South University Xiangya School of Public Health
- Changsha 410078
| | - Fang Xiao
- Central South University Xiangya School of Public Health
- Changsha 410078
- P. R. China
| | - Caigao Zhong
- Central South University Xiangya School of Public Health
- Changsha 410078
- P. R. China
| | - Ming Zeng
- Central South University Xiangya School of Public Health
- Changsha 410078
- P. R. China
| | - Lianhong Zou
- Hunan Normal University School of Medicine
- Changsha 410013
- P. R. China
| |
Collapse
|
25
|
Apoptosis signal-regulating kinase 1 mediates the inhibitory effect of hepatocyte nuclear factor-4α on hepatocellular carcinoma. Oncotarget 2017; 7:27408-21. [PMID: 27050273 PMCID: PMC5053659 DOI: 10.18632/oncotarget.8478] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2015] [Accepted: 03/16/2016] [Indexed: 12/24/2022] Open
Abstract
Previous studies provided substantial evidence of a striking suppressive effect of hepatocyte nuclear factor 4α (HNF4α) on hepatocellular carcinoma (HCC). Apoptosis signal-regulating kinase 1 (ASK1) is involved in death receptor-mediated apoptosis and may acts as a tumor suppressor in hepatocarcinogenesis. However, the status and function of ASK1 during HCC progression are unclear. In this study, we found that HNF4α increased ASK1 expression by directly binding to its promoter. ASK1 expression was dramatically suppressed and correlated with HNF4α levels in HCC tissues. Reduced ASK1 expression was associated with aggressive tumors and poor prognosis for human HCC. Moreover, ASK1 inhibited the malignant phenotype of HCC cells in vitro. Intratumoral ASK1 injection significantly suppressed the growth of subcutaneous HCC xenografts in nude mice. More interestingly, systemic ASK1 delivery strikingly inhibited the growth of orthotopic HCC nodules in NOD/SCID mice. In addition, inhibition of endogenous ASK1 partially reversed the suppressive effects of HNF4α on HCC. Collectively, this study highlights the suppressive effect of ASK1 on HCC and its biological significance in HCC development. These outcomes broaden the knowledge of ASK1 function in HCC progression, and provide a novel potential prognostic biomarker and therapeutic target for advanced HCC.
Collapse
|
26
|
Rusnak L, Fu H. Regulation of ASK1 signaling by scaffold and adaptor proteins. Adv Biol Regul 2017; 66:23-30. [PMID: 29102394 DOI: 10.1016/j.jbior.2017.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/12/2017] [Accepted: 10/13/2017] [Indexed: 06/07/2023]
Abstract
The mitogen-activated protein kinase (MAPK) signaling pathway is a three-tiered kinase cascade where mitogen-activated protein kinase kinase kinases (MAP3Ks) lead to the activation of mitogen-activated protein kinase kinases (MAP2K), and ultimately MAPK proteins. MAPK signaling can promote a diverse set of biological outcomes, ranging from cell death to proliferation. There are multiple mechanisms which govern MAPK output, such as the duration and strength of the signal, cellular localization to upstream and downstream binding partners, pathway crosstalk and the binding to scaffold and adaptor molecules. This review will focus on scaffold and adaptor proteins that bind to and regulate apoptosis signal-regulating kinase 1 (ASK1), a MAP3K protein with a critical role in mediating stress response pathways.
Collapse
Affiliation(s)
- Lauren Rusnak
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University, Atlanta, GA 30322, USA; Graduate Program in Cancer Biology, Emory University, Atlanta, GA 30322, USA.
| | - Haian Fu
- Department of Pharmacology and Emory Chemical Biology Discovery Center, Emory University, Atlanta, GA 30322, USA; Graduate Program in Cancer Biology, Emory University, Atlanta, GA 30322, USA; Department of Hematology & Medical Oncology, Emory University, Atlanta, GA 30322, USA; Winship Cancer Institute, Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
27
|
He J, Chen X, Li B, Zhou W, Xiao J, He K, Zhang J, Xiang G. Chaetocin induces cell cycle arrest and apoptosis by regulating the ROS-mediated ASK-1/JNK signaling pathways. Oncol Rep 2017; 38:2489-2497. [PMID: 28849240 DOI: 10.3892/or.2017.5921] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Accepted: 08/07/2017] [Indexed: 12/11/2022] Open
Abstract
The present study demonstrated that chaetocin, a natural small-molecule product produced by Chaetomium fungal species and a potential anticancer agent, inhibited the viability and invasive ability of the human intrahepatic cholangio-carcinoma cell line CCLP-1 in vivo and in vitro as revealed by CCK-8 and Transwell invasion assays and mouse xenograft tumor experiments. As determined using flow cytometry and intracellular ROS assays, chaetocin was found to induce cell cycle arrest and oxidative stress, leading to CCLP-1 cell apoptosis. Cell apoptosis can be initiated via different apoptotic signaling pathways under oxidative stress. As determined by western blot analysis, expression levels of the apoptosis signal-regulating kinase 1 (ASK-1) signalosome and its downstream c-Jun N-terminal kinase (JNK) signaling pathway were increased under oxidative stress stimulation. These findings indicate that chaetocin arrests the cell cycle and induces apoptosis by regulating the reactive oxygen species-mediated ASK-1/JNK signaling pathways.
Collapse
Affiliation(s)
- Jingliang He
- Department of General Surgery, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, P.R. China
| | - Xiaoxun Chen
- Department of Gastrointestinal Surgery, The Guigang City People's Hospital, Guigang, Guangxi 537100, P.R. China
| | - Bowei Li
- The Third Clinical Medical College of Southern Medical University, Guangzhou, Guangdong 510317, P.R. China
| | - Wenjie Zhou
- Department of General Surgery, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, P.R. China
| | - Jinfeng Xiao
- Department of General Surgery, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, P.R. China
| | - Ke He
- Department of General Surgery, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, P.R. China
| | - Jinqian Zhang
- Department of Laboratory Medicine, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, P.R. China
| | - Guoan Xiang
- Department of General Surgery, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong 510317, P.R. China
| |
Collapse
|
28
|
|
29
|
Nakayama K, Murata S, Ito H, Iwasaki K, Villareal MO, Zheng YW, Matsui H, Isoda H, Ohkohchi N. Terpinen-4-ol inhibits colorectal cancer growth via reactive oxygen species. Oncol Lett 2017; 14:2015-2024. [PMID: 28781645 PMCID: PMC5530226 DOI: 10.3892/ol.2017.6370] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 03/19/2017] [Indexed: 12/22/2022] Open
Abstract
Terpinen-4-ol (TP4O) is the main component of the essential oil extracted from Melaleuca alternifolia, known as the tea tree, of the botanical family Myrtaceae. The anticancer effects of TP4O have been reported in several cancer cell lines. Previous reports have demonstrated that TP4O exerts anticancer effects by inducing apoptotic cell death in several cell lines; however, the underlying molecular mechanisms of these effects remain unclear. In the present study, the anticancer effects of TP4O against the colorectal cancer (CRC) cell lines HCT116 and RKO were evaluated using WST-8 and bromodeoxyuridine assays. The mechanism of cell death was investigated by the measurement of caspase-3/7, Annexin V and lactate dehydrogenase release. Reactive oxygen species (ROS) levels induced by TP4O were evaluated by electron spin resonance and quantitative measurement of dihydroethidium. Localization of the ROS derived from mitochondria was observed by confocal inverted microscopy. Protein levels of ROS scavengers were assessed by western blotting analysis. To confirm the role of ROS, cell viability was measured in the presence of antioxidant reagents. In an in vivo xenograft model of ICR-SCID mice implanted with HCT116 cells, 200 mg/kg TP4O was injected locally, and tumor growth was compared with that of the control. TP4O induced apoptotic cell death in HCT116 and RKO cells in a dose-dependent manner, and TP4O also increased the levels of ROS generated by mitochondria. TP4O-induced cell death was rescued by administration of antioxidant regents. In vivo, TP4O inhibited the proliferation of HCT116 xenografts compared with that of the control group. The results of the present study suggest that TP4O induces apoptosis in CRC cells through ROS generation. Furthermore, TP4O is potentially useful for the development of novel therapies against CRC.
Collapse
Affiliation(s)
- Ken Nakayama
- Department of Surgery, Division of Gastroenterological and Hepatobiliary Surgery, and Organ Transplantation, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Soichiro Murata
- Department of Regenerative Medicine, Yokohama University, Yokohama, Kanagawa 236-0004, Japan
| | - Hiromu Ito
- Department of Gastroenterology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Kenichi Iwasaki
- Department of Surgery, Division of Gastroenterological and Hepatobiliary Surgery, and Organ Transplantation, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Myra Orlina Villareal
- Faculty of Life and Environmental Sciences, University of Tsukuba Alliance of Research on North Africa, University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
| | - Yun-Wen Zheng
- Department of Surgery, Division of Gastroenterological and Hepatobiliary Surgery, and Organ Transplantation, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Hirofumi Matsui
- Department of Gastroenterology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| | - Hiroko Isoda
- Faculty of Life and Environmental Sciences, University of Tsukuba Alliance of Research on North Africa, University of Tsukuba, Tsukuba, Ibaraki 305-8572, Japan
| | - Nobuhiro Ohkohchi
- Department of Surgery, Division of Gastroenterological and Hepatobiliary Surgery, and Organ Transplantation, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki 305-8575, Japan
| |
Collapse
|
30
|
Guon TE, Chung HS. Moringa oleifera fruit induce apoptosis via reactive oxygen species-dependent activation of mitogen-activated protein kinases in human melanoma A2058 cells. Oncol Lett 2017; 14:1703-1710. [PMID: 28789398 DOI: 10.3892/ol.2017.6288] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 03/09/2017] [Indexed: 01/18/2023] Open
Abstract
The present study was performed to determine the effect of Moringa oleifera fruit extract on the apoptosis of human melanoma A2058 cells. A2058 cells were treated for 72 h with Moringa oleifera fruit extract at 50-100 µg/ml, and cell viability with apoptotic changes was examined. The involvement of reactive oxygen species (ROS) and mitogen-activated protein kinases (MAPKs) was examined. It was revealed that Moringa oleifera fruit extract significantly inhibited the cell viability and promoted apoptosis of A2058 cells in a concentration-dependent manner. Moringa oleifera fruit extract-treated A2058 cells exhibited increased activities of cleaved caspase-9 and caspase-3. It also caused an enhancement of MAPK phosphorylation and ROS production. The pro-apoptotic activity of Moringa oleifera fruit extract was significantly reversed by pretreatment with the c-Jun N-terminal kinase (JNK) inhibitor SP600125, extracellular-signal-regulated kinase (ERK) inhibitor PD98058 or ROS inhibitor N-acetyl-L-cysteine. Taken together, Moringa oleifera fruit extract is effective in inducing mitochondrial apoptosis of A2058 cells, which is mediated through induction of ROS formation, and JNK and ERK activation. Moringa oleifera fruit extract may thus have therapeutic benefits for human melanoma A2058 cells.
Collapse
Affiliation(s)
- Tae Eun Guon
- Department of Food and Nutrition, College of Natural Sciences, Duksung Women's University, Seoul 01369, Republic of Korea
| | - Ha Sook Chung
- Department of Food and Nutrition, College of Natural Sciences, Duksung Women's University, Seoul 01369, Republic of Korea
| |
Collapse
|
31
|
Roy A, Ahir M, Bhattacharya S, Parida PK, Adhikary A, Jana K, Ray M. Induction of mitochondrial apoptotic pathway in triple negative breast carcinoma cells by methylglyoxal via generation of reactive oxygen species. Mol Carcinog 2017; 56:2086-2103. [PMID: 28418078 DOI: 10.1002/mc.22665] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/14/2017] [Accepted: 04/13/2017] [Indexed: 12/21/2022]
Abstract
Triple negative breast cancer (TNBC) tends to form aggressive tumors associated with high mortality and morbidity which urge the need for development of new therapeutic strategies. Recently, the normal metabolite Methylglyoxal (MG) has been documented for its anti-proliferative activity against human breast cancer. However, the mode of action of MG against TNBC remains open to question. In our study, we investigated the anticancer activity of MG in MDA MB 231 and 4T1 TNBC cell lines and elucidated the underlying mechanisms. MG dose-dependently caused cell death, induced apoptosis, and generated ROS in both the TNBC cell lines. Furthermore, such effects were attenuated in presence of ROS scavenger N-Acetyl cysteine. MG triggered mitochondrial cytochrome c release in the cytosol and up-regulated Bax while down-regulated anti-apoptotic protein Bcl-2. Additionally, MG treatment down-regulated phospho-akt and inhibited the nuclear translocation of the p65 subunit of NF-κB. MG exhibited a tumor suppressive effect in BALB/c mouse 4T1 breast tumor model as well. The cytotoxic effect was studied using MTT assay. Apoptosis, ROS generation, and mitochondrial dysfunction was evaluated by flow cytometry as well as fluorescence microscopy. Western blot assay was performed to analyze proteins responsible for apoptosis. This study demonstrated MG as a potent anticancer agent against TNBC both in vitro and in vivo. The findings will furnish fresh insights into the treatment of this subgroup of breast cancer.
Collapse
Affiliation(s)
- Anirban Roy
- Department of Biophysics, Bose Institute, Kolkata, West Bengal, India
| | - Manisha Ahir
- Centre for Research in Nanoscience and Nanotechnology, University of Calcutta, Kolkata, West Bengal, India
| | - Saurav Bhattacharya
- Centre for Research in Nanoscience and Nanotechnology, University of Calcutta, Kolkata, West Bengal, India
| | | | - Arghya Adhikary
- Centre for Research in Nanoscience and Nanotechnology, University of Calcutta, Kolkata, West Bengal, India
| | - Kuladip Jana
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal, India
| | - Manju Ray
- Department of Biophysics, Bose Institute, Kolkata, West Bengal, India
| |
Collapse
|
32
|
Wang D, Jiao C, Zhu Y, Liang D, Zao M, Meng X, Gao J, He Y, Liu W, Hou J, Zhong Z, Cheng Z. Activation of CXCL12/CXCR4 renders colorectal cancer cells less sensitive to radiotherapy via up-regulating the expression of survivin. Exp Biol Med (Maywood) 2016; 242:429-435. [PMID: 27798120 DOI: 10.1177/1535370216675068] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Colorectal cancer is the most common malignancy of the gastrointestinal tract. Surgical treatment combined with radiotherapy is the main treatment course for colorectal cancer; nevertheless, radio-resistance is commonly encountered during the treatment course and seriously influences the therapeutic efficacy. We tested the hypothesis that the CXCL12/CXCR4 axis is closely related to radiotherapy sensitivity in colorectal cancer cells. Here, we found that the decrease in cell viability and the increase in cell death induced by radiotherapy were attenuated by CXCL12 treatment, and the inhibition of CXCR4 promoted colorectal cancer cells to be more sensitive to radiotherapy. We also examined the critical roles of CXCL12/CXCR4 in cell survival and found that radiotherapy induced Bax expression and facilitated the activity of caspase-3 and caspase-9, which were reversed by CXCL12 treatment. Cell apoptosis was enhanced by the inhibition of CXCR4 under radiotherapy conditions. Furthermore, treatment with CXCL12 resulted in an increased expression of survivin, and the inhibitory roles of CXCL12 in radiotherapy-induced apoptosis were mitigated by survivin knockdown. These results indicate that CXCL12/CXCR4 protects colorectal cancer cells against radiotherapy via survivin, implying an important underlying mechanism of resistance to radiotherapy during colorectal cancer therapy.
Collapse
Affiliation(s)
- Dawei Wang
- 1 Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Chengbin Jiao
- 2 Department of General Surgery, the First Affiliated Hospital of Jiamusi University, Jiamusi 154002, P.R. China
| | - Yanli Zhu
- 2 Department of General Surgery, the First Affiliated Hospital of Jiamusi University, Jiamusi 154002, P.R. China
| | - Deshen Liang
- 1 Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Ming Zao
- 1 Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Xiangyu Meng
- 1 Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Jianwei Gao
- 1 Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China
| | - Yunlong He
- 2 Department of General Surgery, the First Affiliated Hospital of Jiamusi University, Jiamusi 154002, P.R. China
| | - Weixin Liu
- 2 Department of General Surgery, the First Affiliated Hospital of Jiamusi University, Jiamusi 154002, P.R. China
| | - Jie Hou
- 2 Department of General Surgery, the First Affiliated Hospital of Jiamusi University, Jiamusi 154002, P.R. China
| | - Zhaohua Zhong
- 3 Department of Microbiology, Harbin Medical University, Nangang, Harbin 150081, P.R. China
| | - Zhuoxin Cheng
- 1 Department of General Surgery, the First Affiliated Hospital of Harbin Medical University, Harbin 150001, P.R. China.,2 Department of General Surgery, the First Affiliated Hospital of Jiamusi University, Jiamusi 154002, P.R. China
| |
Collapse
|
33
|
Shan F, Shao Z, Jiang S, Cheng Z. Erlotinib induces the human non-small-cell lung cancer cells apoptosis via activating ROS-dependent JNK pathways. Cancer Med 2016; 5:3166-3175. [PMID: 27726288 PMCID: PMC5119972 DOI: 10.1002/cam4.881] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2016] [Accepted: 08/02/2016] [Indexed: 12/23/2022] Open
Abstract
Although erlotinib (ERL) has drawn more and more attention toward its anticancer properties effect, the underlying mechanisms of ERL's anticancer properties effect remain unclear yet. So, the aim of this research was to explore the underlying anticancer mechanisms of ERL and to explore whether the reactive oxygen species (ROS)‐dependent c‐Jun N‐terminal kinase (JNK) pathway contributed to the anticancer properties provided by ERL. In our study, we used MTT assay to detect the anticell growth ability of ERL on human non–small‐cell lung cancer cell lines (A549). The extent of cell apoptosis was determined by Hoechst 33342 staining and fluorescence‐activated cell sorter (FACS) assay. Then, DCFH‐DA and JC‐1 staining were used to monitor intracellular reactive oxygen species (ROS) and mitochondrial membrane potential (MMP), respectively. Finally, the effect of ERL on phosphorylation state of JNK protein and downstream apoptosis concerned proteins were detected by western blotting assay. Results showed that ERL significantly suppressed the growth and reproduction of A549 cells with the concentration rising up in vitro. Hoechst 33342 staining and FACS assay also confirmed the proapoptosis effect of ERL on A549 cells with the concentration rising up. Furthermore, exposure of A549 cells to ERL increased the intracellular ROS production. As expected, intracellular ROS activated the proapoptotic JNK signaling pathway and inhibited the activation of EFGR signaling pathway. Our results also revealed that ERL could induce cell‐cycle arrest at G0/G1 period. Activation of JNK protein decreased MMP and downregulated content of antiapoptotic protein Bcl‐2 concomitant with the upregulated content of proapoptotic protein Bax in A549 cells. In addition, c‐Jun and cleaved caspase‐3 were also activated by the phosphorylated JNK induced by ERL. All of these proapoptosis effect of ERL was reversed by administration of N‐acetylcysteine (NAC), which performed as a ROS scavenger. Our results suggest that ERL induces A549 cells apoptosis via activating ROS‐dependent JNK pathways in human non–small lung cancer cells that provide a new experimental foundation for cancer therapy.
Collapse
Affiliation(s)
- Fenglian Shan
- Medical College, Qingdao University, Qingdao, Shandong, 266021, China.,Affilitated Hospital of Jining Medical University, Jining, Shandong, 272000, China
| | - Zewei Shao
- Jining Medical University, Jining, Shandong, 272000, China
| | - Shenghua Jiang
- Medical College, Qingdao University, Qingdao, Shandong, 266021, China.,Affilitated Hospital of Jining Medical University, Jining, Shandong, 272000, China
| | - Zhaozhong Cheng
- Medical College, Qingdao University, Qingdao, Shandong, 266021, China
| |
Collapse
|
34
|
Phenazine-1-carboxylic acid-induced programmed cell death in human prostate cancer cells is mediated by reactive oxygen species generation and mitochondrial-related apoptotic pathway. J Appl Biomed 2016. [DOI: 10.1016/j.jab.2016.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
35
|
Zhang R, Qiao H, Chen S, Chen X, Dou K, Wei L, Zhang J. Berberine reverses lapatinib resistance of HER2-positive breast cancer cells by increasing the level of ROS. Cancer Biol Ther 2016; 17:925-34. [PMID: 27416292 DOI: 10.1080/15384047.2016.1210728] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Lapatinib, a novel tyrosine kinase inhibitor of HER2/EGFR, is used to treat HER2-positive breast cancer. However, acquired drug resistance has limited the clinical therapeutic efficacy of lapatinib. Our previous study found that inhibition of autophagy can reduce the proliferation, DNA synthesis, and colony-forming capacity of lapatinib-resistant cells. Berberine has attracted extensive attention due to its wide range of biochemical and pharmacological effects in breast cancer treatment. It has been reported that berberine can induce oxidative stress and the mitochondrial-related apoptotic pathway in human breast cancer cells. In our current study, we found that a new combination therapy of berberine with lapatinib overcame lapatinib resistance. Furthermore, we found that berberine induced apoptosis of lapatinib-resistant cells through upregulating the level of ROS. Specially, lapatinib activated both the c-Myc/pro-Nrf2 pathway and GSK-3β signaling to stabilize Nrf2 and maintain a low level of ROS in resistant cells. However, berberine can upset the ROS balance by downregulating c-Myc to reverse the lapatinib resistance. Our finding provides a novel strategy of using berberine to overcome lapatinib resistance.
Collapse
Affiliation(s)
- Ruohan Zhang
- a The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology , The Fourth Military Medical University , Xi'an, Shaanxi , China.,b Department of Hepato-Biliary and Pancreto-Splenic Surgery , Xijing Hospital, The Fourth Military Medical University , Xi'an , China
| | - Hongyu Qiao
- c Department of Cardiology , Xijing Hospital, The Fourth Military Medical University , Xi'an , China
| | - Suning Chen
- d Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University , Xi'an , China
| | - Xu Chen
- e Cadet Brigade of the Fourth Military Medical University, The Fourth Military Medical University , Xi'an , China
| | - Kefeng Dou
- b Department of Hepato-Biliary and Pancreto-Splenic Surgery , Xijing Hospital, The Fourth Military Medical University , Xi'an , China
| | - Li Wei
- f Department of Obstetrics and Gynecology , Xijing Hospital, The Fourth Military Medical University , Xi'an , China
| | - Jian Zhang
- a The State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology , The Fourth Military Medical University , Xi'an, Shaanxi , China
| |
Collapse
|
36
|
Oxidative stress, polarization of macrophages and tumour angiogenesis: Efficacy of caffeic acid. Chem Biol Interact 2016; 256:111-24. [PMID: 27378625 DOI: 10.1016/j.cbi.2016.06.027] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 06/23/2016] [Accepted: 06/29/2016] [Indexed: 12/22/2022]
Abstract
Macrophage polarization is a process when macrophage expresses different functional programs in response to microenvironmental signals and two extreme forms exist; M1 and M2 macrophages. M1 macrophages are highly microbicidal and anticancer with enhanced ability to kill and phagocytose pathogens, upregulate pro-inflammatory cytokines and reactive molecular species, and present antigens; M2 macrophages and the related tumour associated macrophages (TAMs) regulate tissue remodelling and promote tissue repair and angiogenesis and can amplification of metabolic pathways that can suppress adaptive immune responses. It is demonstrated that ROS production, critical for the activation and functions of M1 macrophages, is necessary for the differentiation of M2 macrophages and TAMs, and that antioxidant therapy blocks TAMs differentiation and tumorigenesis in mouse models of cancer. In order to study how caffeic acid (CA), a natural antioxidant, affects macrophage function, polarization, angiogenesis and tumour growth we injected mice with Ehrlich ascites tumour (EAT) cells and treated them for 10 days with CA in a dose of 40 and/or 80 mg kg(-1.) Macrophage polarization was further characterized by quantifying secreted pro- and anti-inflammatory cytokines, nitric oxide and arginase 1 activity. CA may increase the cytotoxic actions of M1 macrophages and inhibit tumour growth; inhibitory activity on TAMs may be mediated through its antioxidative activity. Taken together, we conclude that the antitumour activity of CA was the result of the synergistic activities of different mechanisms by which CA acts on proliferation, angiogenesis, immunomodulation and survival. The continuous administration of CA efficiently blocked the occurrence of TAMs and markedly suppressed tumorigenesis in mouse cancer models. Targeting TAMs by antioxidants can be a potentially effective method for cancer treatment.
Collapse
|
37
|
Chang HS, Chen IS. Chemical constituents and bioactivity of Formosan lauraceous plants. J Food Drug Anal 2016; 24:247-263. [PMID: 28911577 PMCID: PMC9339549 DOI: 10.1016/j.jfda.2015.10.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2014] [Revised: 09/10/2015] [Accepted: 10/13/2015] [Indexed: 01/12/2023] Open
Abstract
Taiwan is rich in lauraceous plants. A review of 197 references based on the chemical analysis and bioactivity of indigenous lauraceous plants carried out by native scientists from 1963 to 2014 has been compiled. About 303 new compounds and thousands of known compounds comprising alkaloids and non-alkaloids with diverse structures have been isolated or identified from indigenous plants belonging to the 11 lauraceous genera. The volatile components, however, have been excluded from this review. This review provides an overview of the past efforts of Taiwan scientists working on secondary metabolites and their bioactivity in native lauraceous plants. The potential of lauraceous plants worthy of further study is also noted. The contents will be helpful for the chemotaxonomy of Lauraceae and be of value for the development of native Formosan lauraceous plants.
Collapse
Affiliation(s)
- Hsun-Shuo Chang
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung,
Taiwan, ROC
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung,
Taiwan, ROC
| | - Ih-Sheng Chen
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung,
Taiwan, ROC
- Graduate Institute of Natural Products, College of Pharmacy, Kaohsiung Medical University, Kaohsiung,
Taiwan, ROC
- Corresponding author. School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, 100 Shih-Chuan First Road, Kaohsiung City 80708, Taiwan, ROC. E-mail address: (I.-S. Chen)
| |
Collapse
|
38
|
Ding W, Hu Z, Zhang Z, Ma Q, Tang H, Ma Z. Physapubescin B Exhibits Potent Activity against Human Prostate Cancer In Vitro and In Vivo. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2015; 63:9504-9512. [PMID: 26415552 DOI: 10.1021/acs.jafc.5b03045] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
The present data showed that a natural compound isolated from the plant Physalis pubescens L. (Solanaceae), physapubescin B, exhibited antitumor activity against prostate cancer in vitro and in vivo. Treating prostate cancer cells with physapubescin B resulted in the accumulation of cells in the G2/M phase, which was associated with reduced Cdc25C levels and increased levels of CyclinB1, P21 as well as p-Cdk1 (Tyr15). Additionally, reactive oxygen species (ROS) generation was increased in physapubescin B-treated PC-3 cells. Furthermore, the physapubescin B-induced decrease of Cdc25C protein expression together with the G2/M phase cell cycle arrest were significantly abrogated by antioxidant NAC and GSH. Our data also demonstrated that physapubescin B exhibited strong in vivo antitumor efficacy in human prostate cancer PC3 xenograft. In conclusion, our results provide clear evidence that physapubescin B exhibits antitumor activity both in vitro and in vivo and deserves further development as an anticancer agent.
Collapse
Affiliation(s)
- Wanjing Ding
- Institute of Marine Biology, Ocean College of Zhejiang University , Hangzhou, Zhejiang 310058, China
| | - Zhijuan Hu
- Institute of Marine Biology, Ocean College of Zhejiang University , Hangzhou, Zhejiang 310058, China
| | - Zhewen Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University , Hangzhou, Zhejiang 310058, China
| | - Qiaoqiao Ma
- Institute of Marine Biology, Ocean College of Zhejiang University , Hangzhou, Zhejiang 310058, China
| | - Huifang Tang
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University , Hangzhou, Zhejiang 310058, China
| | - Zhongjun Ma
- Institute of Marine Biology, Ocean College of Zhejiang University , Hangzhou, Zhejiang 310058, China
| |
Collapse
|
39
|
Coulidiati TH, Dantas BB, Faheina-Martins GV, Gonçalves JCR, do Nascimento WS, de Oliveira RN, Camara CA, Oliveira EJ, Lara A, Gomes ER, Araújo DAM. Distinct effects of novel naphtoquinone-based triazoles in human leukaemic cell lines. J Pharm Pharmacol 2015; 67:1682-95. [DOI: 10.1111/jphp.12474] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2014] [Accepted: 07/06/2015] [Indexed: 12/11/2022]
Abstract
Abstract
Objectives
The aim of this study was to investigate the cytotoxic effect of new 1,4-naphthoquinone- 1,2,3-triazoles, named C2 to C8 triazole derivatives, towards human cancer cell lines.
Methods
The effect on cell viability was assessed by MTT and propidium iodide assays. The cytotoxic effect of C2 and C3 in K562 and HL-60 cells were analyzed by flow cytometry, DNA fragmentation and reactive oxygen species (ROS) production. Western blot and q-PCR procedures were also performed.
Key findings
C2 and C3 inhibited both K562 and HL-60 cells growth in a concentration-dependent manner. C2 presented the highest cytotoxic activity with an IC50 of approximately 14 μm and 41 μm for HL-60 and K562 cells, respectively, while being less toxic to normal peripheral blood monocyte cells. Both derivatives induced cellular changes in HL-60 cells, characteristic of apoptosis, such as mitochondrial membrane depolarization, phosphatidylserine externalization, increasing sub-G1 phase, DNA fragmentation, downregulating Bcl-2 protein and upregulating Bax protein. In K562 cells, C2 and C3 induced S-phase arrest of cell cycle, which was associated with upregulation of p21. The effect of these derivatives in HL-60 cells can be related to the ROS intracellular level.
Conclusion
Taken together our results showed that C2 and C3 triazole derivatives presented the best potential for drug design.
Collapse
Affiliation(s)
- Tangbadioa H. Coulidiati
- Departamento de Biotecnologia, Centro de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Bruna B. Dantas
- Departamento de Biotecnologia, Centro de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Glaucia V. Faheina-Martins
- Departamento de Biotecnologia, Centro de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Juan C. R. Gonçalves
- Departamento de Biotecnologia, Centro de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Wilson S. do Nascimento
- Laboratório de Síntese de Compostos Bioativos, Departamento de Ciências Moleculares, Universidade Federal Rural de Pernambuco, Recife, Brazil
| | - Ronaldo N. de Oliveira
- Laboratório de Síntese de Compostos Bioativos, Departamento de Ciências Moleculares, Universidade Federal Rural de Pernambuco, Recife, Brazil
| | - Celso A. Camara
- Laboratório de Síntese de Compostos Bioativos, Departamento de Ciências Moleculares, Universidade Federal Rural de Pernambuco, Recife, Brazil
| | - Eduardo J. Oliveira
- Departamento de Biotecnologia, Centro de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Aline Lara
- Departamento de Biotecnologia, Centro de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Eneas R. Gomes
- Departamento de Biotecnologia, Centro de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, Brazil
| | - Demetrius A. M. Araújo
- Departamento de Biotecnologia, Centro de Biotecnologia, Universidade Federal da Paraíba, João Pessoa, Brazil
| |
Collapse
|
40
|
Liu W, Ning R, Chen RN, Huang XF, Dai QS, Hu JH, Wang YW, Wu LL, Xiong J, Hu G, Guo QL, Yang J, Wang H. Aspafilioside B induces G2/M cell cycle arrest and apoptosis by up-regulating H-Ras and N-Ras via ERK and p38 MAPK signaling pathways in human hepatoma HepG2 cells. Mol Carcinog 2015; 55:440-57. [PMID: 25683703 DOI: 10.1002/mc.22293] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Revised: 12/22/2014] [Accepted: 12/30/2014] [Indexed: 01/28/2023]
Abstract
We recently establish that aspafilioside B, a steroidal saponin extracted from Asparagus filicinus, is an active cytotoxic component. However, its antitumor activity is till unknown. In this study, the anticancer effect of aspafilioside B against HCC cells and the underlying mechanisms were investigated. Our results showed that aspafilioside B inhibited the growth and proliferation of HCC cell lines. Further study revealed that aspafilioside B could significantly induce G2 phase cell cycle arrest and apoptosis, accompanying the accumulation of reactive oxygen species (ROS), but blocking ROS generation with N-acetyl-l-cysteine (NAC) could not prevent G2/M arrest and apoptosis. Additionally, treatment with aspafilioside B induced phosphorylation of extracellular signal-regulated kinase (ERK) and p38 MAP kinase. Moreover, both ERK inhibitor PD98059 and p38 inhibitor SB203580 almost abolished the G2/M phase arrest and apoptosis induced by aspafilioside B, and reversed the expression of cell cycle- and apoptosis-related proteins. We also found that aspafilioside B treatment increased both Ras and Raf activation, and transfection of cells with H-Ras and N-Ras shRNA almost attenuated aspafilioside B-induced G2 phase arrest and apoptosis as well as the ERK and p38 activation. Finally, in vivo, aspafilioside B suppressed tumor growth in mouse xenograft models, and the mechanism was the same as in vitro study. Collectively, these findings indicated that aspafilioside B may up-regulate H-Ras and N-Ras, causing c-Raf phosphorylation, and lead to ERK and p38 activation, which consequently induced the G2 phase arrest and apoptosis. This study provides the evidence that aspafilioside B is a promising therapeutic agent against HCC.
Collapse
Affiliation(s)
- Wei Liu
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Rui Ning
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Rui-Ni Chen
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Xue-Feng Huang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Qin-Sheng Dai
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Jin-Hua Hu
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Yu-Wen Wang
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Li-Li Wu
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Jing Xiong
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Gang Hu
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Qing-Long Guo
- Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, Nanjing, China
| | - Jian Yang
- Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Hao Wang
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
41
|
Zou Y, Xiong H, Xiong H, Lu T, Zhu F, Luo Z, Yuan X, Wang Y. A polysaccharide from Andrographis paniculata induces mitochondrial-mediated apoptosis in human hepatoma cell line (HepG2). Tumour Biol 2015; 36:5179-86. [DOI: 10.1007/s13277-015-3172-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 01/26/2015] [Indexed: 01/29/2023] Open
|
42
|
Cho YC, Park JE, Park BC, Kim JH, Jeong DG, Park SG, Cho S. Cell cycle-dependent Cdc25C phosphatase determines cell survival by regulating apoptosis signal-regulating kinase 1. Cell Death Differ 2015; 22:1605-17. [PMID: 25633196 DOI: 10.1038/cdd.2015.2] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Revised: 10/16/2014] [Accepted: 11/17/2014] [Indexed: 11/09/2022] Open
Abstract
Cdc25C (cell division cycle 25C) phosphatase triggers entry into mitosis in the cell cycle by dephosphorylating cyclin B-Cdk1. Cdc25C exhibits basal phosphatase activity during interphase and then becomes activated at the G2/M transition after hyperphosphorylation on multiple sites and dissociation from 14-3-3. Although the role of Cdc25C in mitosis has been extensively studied, its function in interphase remains elusive. Here, we show that during interphase Cdc25C suppresses apoptosis signal-regulating kinase 1 (ASK1), a member of mitogen-activated protein (MAP) kinase kinase kinase family that mediates apoptosis. Cdc25C phosphatase dephosphorylates phospho-Thr-838 in the activation loop of ASK1 in vitro and in interphase cells. In addition, knockdown of Cdc25C increases the activity of ASK1 and ASK1 downstream targets in interphase cells, and overexpression of Cdc25C inhibits ASK1-mediated apoptosis, suggesting that Cdc25C binds to and negatively regulates ASK1. Furthermore, we showed that ASK1 kinase activity correlated with Cdc25C activation during mitotic arrest and enhanced ASK1 activity in the presence of activated Cdc25C resulted from the weak association between ASK1 and Cdc25C. In cells synchronized in mitosis following nocodazole treatment, phosphorylation of Thr-838 in the activation loop of ASK1 increased. Compared with hypophosphorylated Cdc25C, which exhibited basal phosphatase activity in interphase, hyperphosphorylated Cdc25C exhibited enhanced phosphatase activity during mitotic arrest, but had significantly reduced affinity to ASK1, suggesting that enhanced ASK1 activity in mitosis was due to reduced binding of hyperphosphorylated Cdc25C to ASK1. These findings suggest that Cdc25C negatively regulates proapoptotic ASK1 in a cell cycle-dependent manner and may play a role in G2/M checkpoint-mediated apoptosis.
Collapse
Affiliation(s)
- Y-C Cho
- College of Pharmacy, Chung-Ang University, Seoul 156-756, Republic of Korea
| | - J E Park
- Medical Proteomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - B C Park
- Medical Proteomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - J-H Kim
- Targeted Gene Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-333, Republic of Korea
| | - D G Jeong
- Medical Proteomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - S G Park
- Medical Proteomics Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 305-806, Republic of Korea
| | - S Cho
- College of Pharmacy, Chung-Ang University, Seoul 156-756, Republic of Korea
| |
Collapse
|
43
|
Liang R, Shen XL, Zhang B, Li Y, Xu W, Zhao C, Luo Y, Huang K. Apoptosis signal-regulating kinase 1 promotes Ochratoxin A-induced renal cytotoxicity. Sci Rep 2015; 5:8078. [PMID: 25627963 PMCID: PMC5389036 DOI: 10.1038/srep08078] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 01/05/2015] [Indexed: 01/04/2023] Open
Abstract
Oxidative stress and apoptosis are involved in Ochratoxin A (OTA)-induced renal cytotoxicity. Apoptosis signal-regulating kinase 1 (ASK1) is a Mitogen-Activated Protein Kinase Kinase Kinase (MAPKKK, MAP3K) family member that plays an important role in oxidative stress-induced cell apoptosis. In this study, we performed RNA interference of ASK1 in HEK293 cells and employed an iTRAQ-based quantitative proteomics approach to globally investigate the regulatory mechanism of ASK1 in OTA-induced renal cytotoxicity. Our results showed that ASK1 knockdown alleviated OTA-induced ROS generation and Δψm loss and thus desensitized the cells to OTA-induced apoptosis. We identified 33 and 24 differentially expressed proteins upon OTA treatment in scrambled and ASK1 knockdown cells, respectively. Pathway classification and analysis revealed that ASK1 participated in OTA-induced inhibition of mRNA splicing, nucleotide metabolism, the cell cycle, DNA repair, and the activation of lipid metabolism. We concluded that ASK1 plays an essential role in promoting OTA-induced renal cytotoxicity.
Collapse
Affiliation(s)
- Rui Liang
- Laboratory of food safety and molecular biology, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, P.R. China
| | - Xiao Li Shen
- 1] Laboratory of food safety and molecular biology, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, P.R. China [2] School of Public Health, Zunyi Medical University, Zunyi, Guizhou 563003, P.R. China
| | - Boyang Zhang
- Laboratory of food safety and molecular biology, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, P.R. China
| | - Yuzhe Li
- Laboratory of food safety and molecular biology, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, P.R. China
| | - Wentao Xu
- Laboratory of food safety and molecular biology, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, P.R. China
| | - Changhui Zhao
- Department of Nutrition and Food Science, University of Maryland, College Park, MD 20742, USA
| | - YunBo Luo
- Laboratory of food safety and molecular biology, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, P.R. China
| | - Kunlun Huang
- Laboratory of food safety and molecular biology, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, P.R. China
| |
Collapse
|
44
|
Xie J, Xu Y, Huang X, Chen Y, Fu J, Xi M, Wang L. Berberine-induced apoptosis in human breast cancer cells is mediated by reactive oxygen species generation and mitochondrial-related apoptotic pathway. Tumour Biol 2014; 36:1279-88. [PMID: 25352028 DOI: 10.1007/s13277-014-2754-7] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 10/20/2014] [Indexed: 12/30/2022] Open
Abstract
Berberine has drawn extensive attention toward their wide range of biochemical and pharmacological effects, including antineoplastic effect in recent years, but the precise mechanisms remain unclear. Treatment of human breast cancer cells (MCF-7 and MDA-MB-231 cells) with berberine induced inhibition of cell viability in concentration- and time-dependent manner irrespective of their estrogen receptor (ER) expression. Hoechst33342 staining confirmed berberine induced breast cancer cell apoptosis in time-dependent manner. Because apoptosis induction is considered to be a crucial strategy for cancer prevention and therapy, berberine may be an effective chemotherapeutic agent against breast cancer. To explore the precise mechanism, berberine-induced oxidative stress and mitochondrial-related apoptotic pathway in human breast cancer cells were investigated in this study. In both MCF-7 and MDA-MB-231 cells, berberine increased the production of reactive oxygen species (ROS), which activated the pro-apoptotic JNK signaling. Phosphorylated JNK triggered mitochondria membrane potential (ΔΨm) depolarization and downregulation expression of anti-apoptotic protein Bcl-2 concomitant with the upregulation expression of pro-apoptotic protein Bax. Downregulation of anti-apoptotic Bcl-2 family protein in parallel with loss of ΔΨm, leading to increased the release of cytochrome c and apoptosis-inducing factor (AIF) from mitochondria, and eventually triggered the caspase-dependent and caspase-independent apoptosis. Taken together, our study reveled that berberine exerted an antitumor activity in breast cancer cells by reactive oxygen species generation and mitochondrial-related apoptotic pathway. These finding provide an insight into the potential of berberine for breast cancer therapy.
Collapse
Affiliation(s)
- Juan Xie
- State key Laboratory of Reproductive Medicine, Department of Pharmacy, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing, 210029, China,
| | | | | | | | | | | | | |
Collapse
|
45
|
Chaube SK, Shrivastav TG, Tiwari M, Prasad S, Tripathi A, Pandey AK. Neem (Azadirachta indica L.) leaf extract deteriorates oocyte quality by inducing ROS-mediated apoptosis in mammals. SPRINGERPLUS 2014; 3:464. [PMID: 25197620 PMCID: PMC4155053 DOI: 10.1186/2193-1801-3-464] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Accepted: 08/18/2014] [Indexed: 01/04/2023]
Abstract
Neem (Azadirachta indica L.) leaf has been widely used in ayurvedic system of medicine for fertility regulation for a long time. The molecular mechanism by which neem leaf regulates female fertility remains poorly understood. Animal studies suggest that aqueous neem leaf extract (NLE) induces reactive oxygen species (ROS) - mediated granulosa cell apoptosis. Granulosa cell apoptosis deprives oocytes from nutrients, survival factors and cell cycle proteins required for the achievement of meiotic competency of follicular oocytes prior to ovulation. Under this situation, follicular oocyte becomes more susceptible towards apoptosis after ovulation. The increased level of hydrogen peroxide (H2O2) inside the follicular fluid results in the transfer of H2O2 from follicular fluid to the oocyte. The increased level of H2O2 induces p53 activation and over expression of Bax protein that modulates mitochondrial membrane potential and trigger cytochrome c release. The increased cytosolic cytochrome c level induces caspase-9 and caspase-3 activities that trigger destruction of structural and specific proteins leading to DNA fragmentation and thereby oocyte apoptosis. Based on these animal studies, we propose that NLE induces generation of ROS and mitochondria-mediated apoptosis both in granulosa cells as well as in follicular oocyte. The induction of apoptosis deteriorates oocyte quality and thereby limits reproductive outcome in mammals.
Collapse
Affiliation(s)
- Shail K Chaube
- Cell Physiology Laboratory, Biochemistry Unit, Department of Zoology, Banaras Hindu University, Varanasi, 221005 India
| | - Tulsidas G Shrivastav
- Department of Reproductive Biomedicine, National Institute of Health and Family Welfare, Baba Gang Nath Marg, Munirka, New Delhi, 110067 India
| | - Meenakshi Tiwari
- Cell Physiology Laboratory, Biochemistry Unit, Department of Zoology, Banaras Hindu University, Varanasi, 221005 India
| | - Shilpa Prasad
- Cell Physiology Laboratory, Biochemistry Unit, Department of Zoology, Banaras Hindu University, Varanasi, 221005 India
| | - Anima Tripathi
- Cell Physiology Laboratory, Biochemistry Unit, Department of Zoology, Banaras Hindu University, Varanasi, 221005 India
| | - Ajai K Pandey
- Department of Kayachikitsa, Faculty of Ayurveda, Banaras Hindu University, Varanasi, 221005 India
| |
Collapse
|
46
|
Song W, Hu P, Shan Y, Du M, Liu A, Ye R. Cartilage polysaccharide induces apoptosis in K562 cells through a reactive oxygen species-mediated caspase pathway. Food Funct 2014; 5:2486-93. [PMID: 25112602 DOI: 10.1039/c4fo00476k] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
In this study, a polysaccharide (PS) was successfully extracted from porcine cartilage and its effect on chronic myeloid leukemia was examined using human K562 cells. The results of cell proliferation assays indicated that the PS inhibited cancer cell growth at different concentrations. Morphological and biochemical changes characteristic of apoptosis were observed and confirmed by PI staining and TUNEL assay. The nuclear DNA, RNA and proteins of the cancer cells subjected to PS treatment were irreversibly destroyed by reactive oxygen species (ROS), additionally, the ROS effected on the cells directly. The apoptotic signals altered the permeability of the mitochondrial outer membrane, thereby resulted in the release of apoptotic factors into the cytoplasm that induced apoptosis. As caspase-3/7, 8 and 9 were expressed, it was speculated that both intrinsic and extrinsic pathways were involved in the PS-induced apoptosis.
Collapse
Affiliation(s)
- Wei Song
- School of Food Science and Engineering, Harbin Institute of Technology, Harbin 150090, China.
| | | | | | | | | | | |
Collapse
|
47
|
Wang Y, He QY, Chiu JF. Dioscin induced activation of p38 MAPK and JNK via mitochondrial pathway in HL-60 cell line. Eur J Pharmacol 2014; 735:52-8. [PMID: 24755146 DOI: 10.1016/j.ejphar.2014.04.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2013] [Revised: 04/09/2014] [Accepted: 04/10/2014] [Indexed: 02/05/2023]
Abstract
Saponins have shown promise in cancer prevention and therapy; however, little is known about the detailed signaling pathways underlying their anticancer activities. In the present study, we examined the mechanisms of action of dioscin, a glucosides saponin isolated from Polygonatum zanlanscianense pump, in human myeloblast leukemia HL-60 cells. Dioscin suppressed HL-60 cell growth in a dose-dependent manner. This inhibition was due to the induction of apoptosis as revealed by the externalization of phosphatidylserine, and cleavages of lamin A/C and PARP-1. Treatment with dioscin induced apoptosis through activation of caspases 3, 7, 8, 9, and 10. Phosphorylation of p38 MAPK and JNK contributed to dioscin-induced apoptosis upstream of caspase activation. Using various inhibitors and antioxidant agents, we found that mitochondrial derived reactive oxygen species and depletion of mitochondrial transmembrane potential lead to the phosphorylation of p38 MAPK and JNK. Taken together, our results demonstrated that dioscin induces apoptosis by activation of p38 MAPK and JNK through the caspase-dependent mitochondrial death pathway. This work suggests that dioscin may be used as a drug lead for the treatment of myeloblast leukemia.
Collapse
Affiliation(s)
- Ying Wang
- State Key Laboratory of Quality Research in Chinese Medicine and Institute of Chinese Medical Sciences, University of Macau, Macao SAR, China.
| | - Qing-Yu He
- Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes and Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Jen-Fu Chiu
- Open Laboratory for Tumor Molecular Biology, Department of Biochemistry, Shantou University Medical College, Shantou, China; Department of Anatomy, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
48
|
Jang JH, Cho YC, Kim KH, Lee KS, Lee J, Kim DE, Park JS, Jang BC, Kim S, Kwon TK, Park JW. BAI, a novel Cdk inhibitor, enhances farnesyltransferase inhibitor LB42708-mediated apoptosis in renal carcinoma cells through the downregulation of Bcl-2 and c-FLIP (L). Int J Oncol 2014; 45:1680-90. [PMID: 24993441 DOI: 10.3892/ijo.2014.2534] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 05/26/2014] [Indexed: 11/06/2022] Open
Abstract
Previously, we reported the potential of a novel Cdk inhibitor, 2-[1,1'-biphenyl]-4-yl-N-[5-(1,1-dioxo-1λ6-isothiazolidin-2-yl)-1H-indazol-3-yl]acetamide (BAI) as a cancer chemotherapeutic agent. In this study, we investigated mechanisms by which BAI modulates FTI-mediated apoptosis in human renal carcinoma Caki cells. BAI synergizes with FTI to activate DEVDase, cleavage of poly ADP-ribose polymerase (PARP), and degradation of various anti-apoptotic proteins in Caki cells. BAI plus LB42708-induced apoptosis was inhibited by pretreatment with pan-caspase inhibitor, z-VAD-fmk, but not by overexpression of CrmA. The ROS scavenger, N-acetylcysteine (NAC) did not reduce BAI plus LB4270-induced apoptosis. Co-treatment of BAI and LB42708 reduced the mitochondrial membrane potential (MMP, ∆Ψm) in a time-dependent manner, and induced release of AIF and cytochrome c from mitochondria in Caki cells. Furthermore, BAL plus LB42708 induced downregulation of anti-apoptotic proteins [c-FLIP (L), c-FLIP (s), Bcl-2, XIAP, and Mcl-1 (L)]. Especially, we found that BAI plus LB42708-induced apoptosis was significantly attenuated by overexpression of Bcl-2 and partially blocked by overexpression of c-FLIP (L). Taken together, our results show that the activity of BAI plus LB42708 modulate multiple components in apoptotic response of human renal Caki cells, and indicate a potential as combinational therapeutic agents for preventing cancer such as renal carcinoma.
Collapse
Affiliation(s)
- Ji Hoon Jang
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Yoon Chul Cho
- Department of Urology, Dongguk University, College of Medicine, Gyeongju, Republic of Korea
| | - Ki Ho Kim
- Department of Urology, Dongguk University, College of Medicine, Gyeongju, Republic of Korea
| | - Kyung Seop Lee
- Department of Urology, Dongguk University, College of Medicine, Gyeongju, Republic of Korea
| | - Jinho Lee
- Department of Chemistry, Keimyung University, Daegu, Republic of Korea
| | - Dong Eun Kim
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Jun-Soo Park
- Chronic Disease Research Center, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Byeong-Churl Jang
- Chronic Disease Research Center, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Shin Kim
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Taeg Kyu Kwon
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| | - Jong-Wook Park
- Department of Immunology, School of Medicine, Keimyung University, Daegu, Republic of Korea
| |
Collapse
|
49
|
Liao YJ, Bai HY, Li ZH, Zou J, Chen JW, Zheng F, Zhang JX, Mai SJ, Zeng MS, Sun HD, Pu JX, Xie D. Longikaurin A, a natural ent-kaurane, induces G2/M phase arrest via downregulation of Skp2 and apoptosis induction through ROS/JNK/c-Jun pathway in hepatocellular carcinoma cells. Cell Death Dis 2014; 5:e1137. [PMID: 24651440 PMCID: PMC3973226 DOI: 10.1038/cddis.2014.66] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 01/15/2014] [Accepted: 01/27/2014] [Indexed: 12/26/2022]
Abstract
Hepatocellular carcinoma (HCC) is the most common form of primary liver cancer, and is also highly resistant to conventional chemotherapy treatments. In this study, we report that Longikaurin A (LK-A), an ent-kaurane diterpenoid isolated from the plant Isodon ternifolius, induced cell cycle arrest and apoptosis in human HCC cell lines. LK-A also suppressed tumor growth in SMMC-7721 xenograft models, without inducing any notable major organ-related toxicity. LK-A treatment led to reduced expression of the proto-oncogene S phase kinase-associated protein 2 (Skp2) in SMMC-7721 cells. Lower Skp2 levels correlated with increased expression of p21 and p-cdc2 (Try15), and a corresponding decrease in protein levels of Cyclin B1 and cdc2. Overexpression of Skp2 significantly inhibited LK-A-induced cell cycle arrest in SMMC-7721 cells, suggesting that LK-A may target Skp2 to arrest cells at the G2/M phase. LK-A also induced reactive oxygen species (ROS) production and apoptosis in SMMC-7721 cells. LK-A induced phosphorylation of c-Jun N-terminal kinase (JNK), but not extracellular signal-regulated kinase and P38 MAP kinase. Treatment with, the JNK inhibitor SP600125 prevented LK-A-induced apoptosis in SMMC-7721 cells. Moreover, the antioxidant N-acetylcysteine prevented phosphorylation of both JNK and c-Jun. Taken together, these data indicate that LK-A induces cell cycle arrest and apoptosis in cancer cells by dampening Skp2 expression, and thereby activating the ROS/JNK/c-Jun signaling pathways. LK-A is therefore a potential lead compound for development of antitumor drugs targeting HCC.
Collapse
Affiliation(s)
- Y-J Liao
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - H-Y Bai
- 1] Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China [2] Department of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Z-H Li
- Department of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - J Zou
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - J-W Chen
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - F Zheng
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Guangzhou, China
| | - J-X Zhang
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - S-J Mai
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - M-S Zeng
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - H-D Sun
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - J-X Pu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, China
| | - D Xie
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
50
|
Hsieh CJ, Kuo PL, Hsu YC, Huang YF, Tsai EM, Hsu YL. Arctigenin, a dietary phytoestrogen, induces apoptosis of estrogen receptor-negative breast cancer cells through the ROS/p38 MAPK pathway and epigenetic regulation. Free Radic Biol Med 2014; 67:159-70. [PMID: 24140706 DOI: 10.1016/j.freeradbiomed.2013.10.004] [Citation(s) in RCA: 111] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2013] [Revised: 09/13/2013] [Accepted: 10/02/2013] [Indexed: 12/31/2022]
Abstract
This study investigates the anticancer effect of arctigenin (ATG), a natural lignan product of Arctium lappa L., in human breast cancer MDA-MB-231 cells. Results indicate that ATG inhibits MDA-MB-231 cell growth by inducing apoptosis in vitro and in vivo. ATG triggers the mitochondrial caspase-independent pathways, as indicated by changes in Bax/Bcl-2 ratio, resulting in AIF and EndoG nuclear translocation. ATG increased cellular reactive oxygen species (ROS) production by increasing p22(phox)/NADPH oxidase 1 interaction and decreasing glutathione level. ATG clearly increases the activation of p38 MAPK, but not JNK and ERK1/2. Antioxidant EUK-8, a synthetic catalytic superoxide and hydrogen peroxide scavenger, significantly decreases ATG-mediated p38 activation and apoptosis. Blocking p38 with a specific inhibitor suppresses ATG-mediated Bcl-2 downregulation and apoptosis. Moreover, ATG activates ATF-2, a transcription factor activated by p38, and then upregulates histone H3K9 trimethylation in the Bcl-2 gene promoter region, resulting in Bcl-2 downregulation. Taken together, the results demonstrate that ATG induces apoptosis of MDA-MB-231 cells via the ROS/p38 MAPK pathway and epigenetic regulation of Bcl-2 by upregulation of histone H3K9 trimethylation.
Collapse
Affiliation(s)
- Chia-Jung Hsieh
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Chinese Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Po-Lin Kuo
- Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan; Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Ying-Chan Hsu
- Department of Chinese Medicine, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Ya-Fang Huang
- Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan
| | - Eing-Mei Tsai
- Center for Resources, Research, and Development, Kaohsiung Medical University, Kaohsiung 807, Taiwan; Department of Obstetrics & Gynecology, Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan.
| | - Ya-Ling Hsu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan.
| |
Collapse
|