1
|
Xu Z, Shi Y, Zhu L, Luo J, Hu Q, Jiang S, Xiao M, Jiang X, Wang H, Xu Y, Jin W, Zhou Y, Wang P, Wang K. Novel SERCA2 inhibitor Diphyllin displays anti-tumor effect in non-small cell lung cancer by promoting endoplasmic reticulum stress and mitochondrial dysfunction. Cancer Lett 2024; 598:217075. [PMID: 38909775 DOI: 10.1016/j.canlet.2024.217075] [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: 02/27/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/25/2024]
Abstract
Abnormal calcium signaling is associated with non-small cell lung cancer (NSCLC) malignant progression, poor survival and chemotherapy resistance. Targeting endoplasmic reticulum (ER) Ca2+ channels or pumps to block calcium uptake in the ER induces ER stress and concomitantly promotes mitochondrial calcium uptake, leading to mitochondrial dysfunction and ultimately inducing cell death. Here, we identified Diphyllin was a potential specific inhibitor of endoplasmic reticulum (ER) calcium-importing protein sarco/endoplasmic-reticulum Ca2+ ATPase 2 (SERCA2). In vitro and in vivo studies showed that Diphyllin increased NSCLC cell apoptosis, along with inhibition of cell proliferation and migration. Mechanistically, Diphyllin promoted ER stress by directly inhibiting SERCA2 activity and decreasing ER Ca2+ levels. At the same time, the accumulated Ca2+ in cytoplasm flowed into mitochondria to increase reactive oxygen species (ROS) and decrease mitochondrial membrane potential (MMP), leading to cytochrome C (Cyto C) release and mitochondrial dysfunction. In addition, we found that Diphyllin combined with cisplatin could have a synergistic anti-tumor effect in vitro and in vivo. Taken together, our results suggested that Diphyllin, as a potential novel inhibitor of SERCA2, exerts anti-tumor effects by blocking ER Ca2+ uptake and thereby promoting ER stress and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Zhiyong Xu
- Department of Respiratory and Critical Care Medicine, Center for Oncology Medical, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China; Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu, 322000, China
| | - Yueli Shi
- Department of Respiratory and Critical Care Medicine, Center for Oncology Medical, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China; Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu, 322000, China
| | - Liang Zhu
- Department of Rheumatology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, Zhejiang, China
| | - Jianhua Luo
- Department of Respiratory and Critical Care Medicine, Center for Oncology Medical, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China; Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu, 322000, China; Department of Respiratory Medicine, Taizhou Municipal Hospital, Taizhou, 318000, Zhejiang, China
| | - Qiongjie Hu
- Department of Respiratory and Critical Care Medicine, Center for Oncology Medical, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China; Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu, 322000, China
| | - Sujing Jiang
- Department of Respiratory and Critical Care Medicine, Center for Oncology Medical, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China; Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu, 322000, China
| | - Mingshu Xiao
- Department of Respiratory and Critical Care Medicine, Center for Oncology Medical, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China; Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu, 322000, China
| | - Xinyuan Jiang
- Department of Respiratory and Critical Care Medicine, Center for Oncology Medical, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China; Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu, 322000, China
| | - Huan Wang
- Department of Respiratory and Critical Care Medicine, Center for Oncology Medical, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China; Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu, 322000, China
| | - Yun Xu
- Department of Respiratory and Critical Care Medicine, Center for Oncology Medical, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China; Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu, 322000, China
| | - Wei Jin
- Department of Respiratory and Critical Care Medicine, Center for Oncology Medical, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Yan Zhou
- Department of Respiratory and Critical Care Medicine, Center for Oncology Medical, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China; Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu, 322000, China
| | - Pingli Wang
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, Zhejiang, China
| | - Kai Wang
- Department of Respiratory and Critical Care Medicine, Center for Oncology Medical, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China; Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancer, Yiwu, 322000, China.
| |
Collapse
|
2
|
Zhang YZ, Lai HL, Huang C, Jiang ZB, Yan HX, Wang XR, Xie C, Huang JM, Ren WK, Li JX, Zhai ZR, Yao XJ, Wu QB, Leung ELH. Tanshinone IIA induces ER stress and JNK activation to inhibit tumor growth and enhance anti-PD-1 immunotherapy in non-small cell lung cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 128:155431. [PMID: 38537440 DOI: 10.1016/j.phymed.2024.155431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/02/2024] [Accepted: 02/06/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) remains at the forefront of new cancer cases, and there is an urgent need to find new treatments or improve the efficacy of existing therapies. In addition to the application in the field of cerebrovascular diseases, recent studies have revealed that tanshinone IIA (Tan IIA) has anticancer activity in a variety of cancers. PURPOSE To investigate the potential anticancer mechanism of Tan IIA and its impact on immunotherapy in NSCLC. METHODS Cytotoxicity and colony formation assays were used to detect the Tan IIA inhibitory effect on NSCLC cells. This research clarified the mechanisms of Tan IIA in anti-tumor and programmed death-ligand 1 (PD-L1) regulation by using flow cytometry, transient transfection, western blotting and immunohistochemistry (IHC) methods. Besides, IHC was also used to analyze the nuclear factor of activated T cells 1 (NFAT2) expression in NSCLC clinical samples. Two animal models including xenograft mouse model and Lewis lung cancer model were used for evaluating tumor suppressive efficacy of Tan IIA. We also tested the efficacy of Tan IIA combined with programmed cell death protein 1 (PD-1) inhibitors in Lewis lung cancer model. RESULTS Tan IIA exhibited good NSCLC inhibitory effect which was accompanied by endoplasmic reticulum (ER) stress response and increasing Ca2+ levels. Moreover, Tan IIA could suppress the NFAT2/ Myc proto oncogene protein (c-Myc) signaling, and it also was able to control the Jun Proto-Oncogene(c-Jun)/PD-L1 axis in NSCLC cells through the c-Jun N-terminal kinase (JNK) pathway. High NFAT2 levels were potential factors for poor prognosis in NSCLC patients. Finally, animal experiments data showed a stronger immune activation phenotype, when we performed treatment of Tan IIA combined with PD-1 monoclonal antibody. CONCLUSION The findings of our research suggested a novel mechanism for Tan IIA to inhibit NSCLC, which could exert anti-cancer effects through the JNK/NFAT2/c-Myc pathway. Furthermore, Tan IIA could regulate tumor PD-L1 levels and has the potential to improve the efficacy of PD-1 inhibitors.
Collapse
Affiliation(s)
- Yi-Zhong Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics of Innovative Drug Discovery, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Huan-Ling Lai
- Guangzhou National Laboratory, No. 9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, Guangdong Province, China
| | - Chen Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics of Innovative Drug Discovery, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Ze-Bo Jiang
- Affiliated Zhuhai Hospital, Southern Medical University, Zhuhai Hospital of Integrated Traditional Chinese & Western Medicine, Zhuhai 519000, Guangdong, China
| | - Hao-Xin Yan
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics of Innovative Drug Discovery, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Xuan-Run Wang
- Cancer Center, Faculty of Health Science, University of Macau, Macau (SAR), China. MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), China; State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau (SAR), China
| | - Chun Xie
- Cancer Center, Faculty of Health Science, University of Macau, Macau (SAR), China. MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), China; State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau (SAR), China
| | - Ju-Min Huang
- Cancer Center, Faculty of Health Science, University of Macau, Macau (SAR), China. MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), China; State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau (SAR), China
| | - Wen-Kang Ren
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics of Innovative Drug Discovery, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Jia-Xin Li
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics of Innovative Drug Discovery, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Zhi-Ran Zhai
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics of Innovative Drug Discovery, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China
| | - Xiao-Jun Yao
- Faculty of Applied Sciences, Macao Polytechnic University, 999078, Macao.
| | - Qi-Biao Wu
- State Key Laboratory of Quality Research in Chinese Medicine, Dr. Neher's Biophysics of Innovative Drug Discovery, Macau Institute For Applied Research in Medicine and Health, Macau University of Science and Technology, Macau (SAR), China.
| | - Elaine Lai-Han Leung
- Cancer Center, Faculty of Health Science, University of Macau, Macau (SAR), China. MOE Frontiers Science Center for Precision Oncology, University of Macau, Macau (SAR), China; State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau (SAR), China.
| |
Collapse
|
3
|
Sanati M, Afshari AR, Kesharwani P, Sahebkar A. Recent advances in codelivery of curcumin and siRNA as anticancer therapeutics. Eur Polym J 2023; 198:112444. [DOI: 10.1016/j.eurpolymj.2023.112444] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024]
|
4
|
Pouliquen DL, Trošelj KG, Anto RJ. Curcuminoids as Anticancer Drugs: Pleiotropic Effects, Potential for Metabolic Reprogramming and Prospects for the Future. Pharmaceutics 2023; 15:1612. [PMID: 37376060 DOI: 10.3390/pharmaceutics15061612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 05/21/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
The number of published studies on curcuminoids in cancer research, including its lead molecule curcumin and synthetic analogs, has been increasing substantially during the past two decades. Insights on the diversity of inhibitory effects they have produced on a multitude of pathways involved in carcinogenesis and tumor progression have been provided. As this wealth of data was obtained in settings of various experimental and clinical data, this review first aimed at presenting a chronology of discoveries and an update on their complex in vivo effects. Secondly, there are many interesting questions linked to their pleiotropic effects. One of them, a growing research topic, relates to their ability to modulate metabolic reprogramming. This review will also cover the use of curcuminoids as chemosensitizing molecules that can be combined with several anticancer drugs to reverse the phenomenon of multidrug resistance. Finally, current investigations in these three complementary research fields raise several important questions that will be put among the prospects for the future research related to the importance of these molecules in cancer research.
Collapse
Affiliation(s)
- Daniel L Pouliquen
- Université d'Angers, Inserm, CNRS, Nantes Université, CRCI2NA, F-49000 Angers, France
| | - Koraljka Gall Trošelj
- Laboratory for Epigenomics, Division of Molecular Medicine, Ruđer Bošković Institute, 10000 Zagreb, Croatia
| | - Ruby John Anto
- Molecular Bioassay Laboratory, Institute of Advanced Virology, Thiruvananthapuram 695317, India
| |
Collapse
|
5
|
Fan M, Gao J, Zhou L, Xue W, Wang Y, Chen J, Li W, Yu Y, Liu B, Shen Y, Xu Q. Highly expressed SERCA2 triggers tumor cell autophagy and is a druggable vulnerability in triple-negative breast cancer. Acta Pharm Sin B 2022; 12:4407-4423. [PMID: 36561988 PMCID: PMC9764070 DOI: 10.1016/j.apsb.2022.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 03/13/2022] [Accepted: 03/23/2022] [Indexed: 12/25/2022] Open
Abstract
Chemoresistance remains a major obstacle to successful treatment of triple negative breast cancer (TNBC). Identification of druggable vulnerabilities is an important aim for TNBC therapy. Here, we report that SERCA2 expression correlates with TNBC progression in human patients, which promotes TNBC cell proliferation, migration and chemoresistance. Mechanistically, SERCA2 interacts with LC3B via LIR motif, facilitating WIPI2-independent autophagosome formation to induce autophagy. Autophagy-mediated SERCA2 degradation induces SERCA2 transactivation through a Ca2+/CaMKK/CREB-1 feedback. Moreover, we found that SERCA2-targeting small molecule RL71 enhances SERCA2-LC3B interaction and induces excessive autophagic cell death. The increase in SERCA2 expression predisposes TNBC cells to RL71-induced autophagic cell death in vitro and in vivo. This study elucidates a mechanism by which TNBC cells maintain their high autophagy activity to induce chemoresistance, and suggests increased SERCA2 expression as a druggable vulnerability for TNBC.
Collapse
Affiliation(s)
- Minmin Fan
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing 210023, China,The First Clinical Medical College, Nanjing University of Chinese Medicine, Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, Nanjing 210023, China
| | - Jian Gao
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Lin Zhou
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Wenwen Xue
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Yixuan Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Jingwei Chen
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Wuhao Li
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Ying Yu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing 210023, China
| | - Bo Liu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Collaborative Innovation Center of Biotherapy, Chengdu 610041, China
| | - Yan Shen
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing 210023, China,Corresponding authors. Tel./fax: +86 25 89687620.
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing Drum Tower Hospital, School of Life Sciences, Nanjing University, Nanjing 210023, China,Corresponding authors. Tel./fax: +86 25 89687620.
| |
Collapse
|
6
|
Handa T, Sasaki H, Takao M, Tano M, Uchida Y. Proteomics-based investigation of cerebrovascular molecular mechanisms in cerebral amyloid angiopathy by the FFPE-LMD-PCT-SWATH method. Fluids Barriers CNS 2022; 19:56. [PMID: 35778717 PMCID: PMC9250250 DOI: 10.1186/s12987-022-00351-x] [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: 05/08/2022] [Accepted: 06/21/2022] [Indexed: 11/25/2022] Open
Abstract
Background Cerebral amyloid angiopathy (CAA) occurs in 80% of patients with Alzheimer’s disease (AD) and is mainly caused by the abnormal deposition of Aβ in the walls of cerebral blood vessels. Cerebrovascular molecular mechanisms in CAA were investigated by using comprehensive and accurate quantitative proteomics. Methods Concerning the molecular mechanisms specific to CAA, formalin-fixed paraffin-embedded (FFPE) sections were prepared from patients having AD neuropathologic change (ADNC) with severe cortical Aβ vascular deposition (ADNC +/CAA +), and from patients having ADNC without vascular deposition of Aβ (ADNC +/CAA −; so called, AD). Cerebral cortical vessels were isolated from FFPE sections using laser microdissection (LMD), processed by pressure cycling technology (PCT), and applied to SWATH (sequential window acquisition of all theoretical fragment ion spectra) proteomics. Results The protein expression levels of 17 proteins in ADNC +/CAA +/H donors (ADNC +/CAA + donors with highly abundant Aβ in capillaries) were significantly different from those in ADNC +/CAA − and ADNC −/CAA − donors. Furthermore, we identified 56 proteins showing more than a 1.5-fold difference in average expression levels between ADNC +/CAA + and ADNC −/CAA − donors, and were significantly correlated with the levels of Aβ or Collagen alpha-2(VI) chain (COL6A2) (CAA markers) in 11 donors (6 ADNC +/CAA + and 5 ADNC −/CAA −). Over 70% of the 56 proteins showed ADNC +/CAA + specific changes in protein expression. The comparative analysis with brain parenchyma showed that more than 90% of the 56 proteins were vascular-specific pathological changes. A literature-based pathway analysis showed that 42 proteins are associated with fibrosis, oxidative stress and apoptosis. This included the increased expression of Heat shock protein HSP 90-alpha, CD44 antigen and Carbonic anhydrase 1 which are inhibited by potential drugs against CAA. Conclusions The combination of LMD-based isolation of vessels from FFPE sections, PCT-assisted sample processing and SWATH analysis (FFPE-LMD-PCT-SWATH method) revealed for the first time the changes in the expression of many proteins that are involved in fibrosis, ROS production and cell death in ADNC +/CAA + (CAA patients) vessels. The findings reported herein would be useful for developing a better understanding of the pathology of CAA and for promoting the discovery and development of drugs and biomarkers for CAA. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00351-x.
Collapse
Affiliation(s)
- Takumi Handa
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hayate Sasaki
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Masaki Takao
- Department of Neurology and Brain Bank, Mihara Memorial Hospital, Isesaki, Japan.,Department of Clinical Laboratory, National Center of Neurology and Psychiatry, National Center Hospital, Kodaira, Japan
| | - Mitsutoshi Tano
- Department of Neurology and Brain Bank, Mihara Memorial Hospital, Isesaki, Japan
| | - Yasuo Uchida
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan. .,Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, Japan.
| |
Collapse
|
7
|
Liu J, Li L, Zhang B, Xu ZP. MnO2-shelled Doxorubicin/Curcumin nanoformulation for enhanced colorectal cancer chemo-immunotherapy. J Colloid Interface Sci 2022; 617:315-325. [DOI: 10.1016/j.jcis.2022.02.132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/15/2022] [Accepted: 02/27/2022] [Indexed: 02/09/2023]
|
8
|
Upadhyay A, Kundu P, Ramu V, Kondaiah P, Chakravarty AR. BODIPY-Tagged Platinum(II) Curcumin Complexes for Endoplasmic Reticulum-Targeted Red Light PDT. Inorg Chem 2022; 61:1335-1348. [PMID: 34990135 DOI: 10.1021/acs.inorgchem.1c02745] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
[Pt(RB)(Cur)]NO3 (RBC), [Pt(IRB)(Cur)]NO3 (IRBC), and [Pt(L)(Cur)]NO3 (PBC), where HCur is curcumin, L is 1-benzyl-2-(2-pyridyl)benzimidazole, and RB and IRB are red-light-active non-iodo and diiodo-BODIPY tagged to L, respectively, were synthesized and characterized, and their anticancer activities were studied (BODIPY, boron-dipyrromethene). RBC and IRBC displayed BODIPY-centered absorption bands within 615-635 nm along with the respective curcumin bands at 445 and 492 nm in 10% dimethyl sulfoxide (DMSO)-Dulbecco's phosphate-buffered saline (DPBS). Emission bands were observed at 723 and 845 nm for RBC and IRBC, respectively, in 10% DMSO-DPBS. RBC (ΦΔ, 0.27) and IRBC (ΦΔ, 0.40) generated singlet oxygen in red light (λ = 642 nm) as evidenced from 1,3-diphenylisobenzofuran (DPBF) titrations. The formation of 1O2 from BODIPY and HO• from the curcumin was evidenced from the mechanistic pUC19 DNA photocleavage studies. The BODIPY complexes showed photocytotoxicity in A549, HeLa, and MDA-MB-231 cells while being less toxic in the dark [IC50: 1.3-6.9 μM, red light; 7.2-12.8 μM, 400-700 nm visible light]. The emissive RBC displayed localization in the endoplasmic reticulum (ER). Apoptotic cell death was evidenced from the Annexin-V/fluorescein isothiocyanate (FITC)/propidium iodide (PI) assay and green fluorescence in red light in the Fluo-4 AM assay due to ER stress, and mitochondrial dysfunction was evidenced from the 5,5,6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolylcarbocyanine iodide (JC-1) assay in A549 cells.
Collapse
|
9
|
Curcumin and Carnosic Acid Cooperate to Inhibit Proliferation and Alter Mitochondrial Function of Metastatic Prostate Cancer Cells. Antioxidants (Basel) 2021; 10:antiox10101591. [PMID: 34679726 PMCID: PMC8533243 DOI: 10.3390/antiox10101591] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2021] [Revised: 10/04/2021] [Accepted: 10/07/2021] [Indexed: 12/24/2022] Open
Abstract
Anticancer activities of plant polyphenols have been demonstrated in various models of neoplasia. However, evidence obtained in numerous in vitro studies indicates that proliferation arrest and/or killing of cancer cells require quite high micromolar concentrations of polyphenols that are difficult to reach in vivo and can also be (geno)toxic to at least some types of normal cells. The ability of certain polyphenols to synergize with one another at low concentrations can be used as a promising strategy to effectively treat human malignancies. We have recently reported that curcumin and carnosic acid applied at non-cytotoxic concentrations synergistically cooperate to induce massive apoptosis in acute myeloid leukemia cells, but not in normal hematopoietic and non-hematopoietic cells, via sustained cytosolic calcium overload. Here, we show that the two polyphenols can also synergistically suppress the growth of DU145 and PC-3 metastatic prostate cancer cell cultures. However, instead of cell killing, the combined treatment induced a marked inhibition of cell proliferation associated with G0/G1 cell cycle arrest. This was preceded by transient elevation of cytosolic calcium levels and prolonged dissipation of the mitochondrial membrane potential, without generating oxidative stress, and was associated with defective oxidative phosphorylation encompassing mitochondrial dysfunction. The above effects were concomitant with a significant downregulation of mRNA and protein expression of the oncogenic kinase SGK1, the mitochondria-hosted mTOR component. In addition, a moderate decrease in SGK1 phosphorylation at Ser422 was observed in polyphenol-treated cells. The mTOR inhibitor rapamycin produced a similar reduction in SGK1 mRNA and protein levels as well as phosphorylation. Collectively, our findings suggest that the combination of curcumin and carnosic acid at potentially bioavailable concentrations may effectively target different types of cancer cells by distinct modes of action. This and similar combinations merit further exploration as an anticancer modality.
Collapse
|
10
|
Curcumin-Loaded Nanoparticles Impair the Pro-Tumor Activity of Acid-Stressed MSC in an In Vitro Model of Osteosarcoma. Int J Mol Sci 2021; 22:ijms22115760. [PMID: 34071200 PMCID: PMC8198446 DOI: 10.3390/ijms22115760] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/14/2022] Open
Abstract
In the tumor microenvironment, mesenchymal stromal cells (MSCs) are key modulators of cancer cell behavior in response to several stimuli. Intratumoral acidosis is a metabolic trait of fast-growing tumors that can induce a pro-tumorigenic phenotype in MSCs through the activation of the NF-κB-mediated inflammatory pathway, driving tumor clonogenicity, invasion, and chemoresistance. Recent studies have indicated that curcumin, a natural ingredient extracted from Curcuma longa, acts as an NF-κB inhibitor with anti-inflammatory properties. In this work, highly proliferating osteosarcoma cells were used to study the ability of curcumin to reduce the supportive effect of MSCs when stimulated by acidosis. Due to the poor solubility of curcumin in biological fluids, we used spherical polymeric nanoparticles as carriers (SPN-curc) to optimize its uptake by MSCs. We showed that SPN-curc inhibited the release of inflammatory cytokines (IL6 and IL8) by acidity-stimulated MSCs at a higher extent than by free curcumin. SPN-curc treatment was also successful in blocking tumor stemness, migration, and invasion that were driven by the secretome of acid-stressed MSCs. Overall, these data encourage the use of lipid–polymeric nanoparticles encapsulating NF-κB inhibitors such as curcumin to treat cancers whose progression is stimulated by an activated mesenchymal stroma.
Collapse
|
11
|
de Seabra Rodrigues Dias IR, Lo HH, Zhang K, Law BYK, Nasim AA, Chung SK, Wong VKW, Liu L. Potential therapeutic compounds from traditional Chinese medicine targeting endoplasmic reticulum stress to alleviate rheumatoid arthritis. Pharmacol Res 2021; 170:105696. [PMID: 34052360 DOI: 10.1016/j.phrs.2021.105696] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/25/2021] [Accepted: 05/25/2021] [Indexed: 02/06/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory autoimmune disease which affects about 0.5-1% of people with symptoms that significantly impact a sufferer's lifestyle. The cells involved in propagating RA tend to display pro-inflammatory and cancer-like characteristics. Medical drug treatment is currently the main avenue of RA therapy. However, drug options are limited due to severe side effects, high costs, insufficient disease retardation in a majority of patients, and therapeutic effects possibly subsiding over time. Thus there is a need for new drug therapies. Endoplasmic reticulum (ER) stress, a condition due to accumulation of misfolded proteins in the ER, and subsequent cellular responses have been found to be involved in cancer and inflammatory pathologies, including RA. ER stress protein markers and their modulation have therefore been suggested as therapeutic targets, such as GRP78 and CHOP, among others. Some current RA therapeutic drugs have been found to have ER stress-modulating properties. Traditional Chinese Medicines (TCMs) frequently use natural products that affect multiple body and cellular targets, and several medicines and/or their isolated compounds have been found to also have ER stress-modulating capabilities, including TCMs used in RA treatment by Chinese Medicine practitioners. This review encourages, in light of the available information, the study of these RA-treating, ER stress-modulating TCMs as potential new pharmaceutical drugs for use in clinical RA therapy, along with providing a list of other ER stress-modulating TCMs utilized in treatment of cancers, inflammatory diseases and other diseases, that have potential use in RA treatment given similar ER stress-modulating capacity.
Collapse
Affiliation(s)
- Ivo Ricardo de Seabra Rodrigues Dias
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Hang Hong Lo
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Kaixi Zhang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Betty Yuen Kwan Law
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, China
| | - Ali Adnan Nasim
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Sookja Kim Chung
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau, China; Faculty of Medicine, Macau University of Science and Technology, Macau, China.
| | - Vincent Kam Wai Wong
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macau, China; State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, China.
| | - Liang Liu
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, China.
| |
Collapse
|
12
|
Lv X, Zhang W, Xia S, Huang Z, Shi P. Clioquinol inhibits cell growth in a SERCA2-dependent manner. J Biochem Mol Toxicol 2021; 35:e22727. [PMID: 33511738 DOI: 10.1002/jbt.22727] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 01/06/2021] [Accepted: 01/19/2021] [Indexed: 11/06/2022]
Abstract
Clioquinol has been reported to act as a potential therapy for neurodegenerative diseases and cancer. However, the underlying mechanism is unclear. We have previously reported that clioquinol induces S-phase cell cycle arrest through the elevation of calcium levels in human neurotypic SH-SY5Y cells. In this study, different types of cells were observed to detect if the effect of clioquinol on intracellular calcium levels is cell type-specific. The Cell Counting Kit-8 assay showed that clioquinol exhibited varying degrees of concentration-dependent cytotoxicity in different cell lines, and that the growth inhibition caused by it was not related to cell source or carcinogenesis. In addition, the inhibition of cell growth by clioquinol was positively associated with its effect on intracellular calcium content ([Ca2+ ]i ). Furthermore, the elevation of [Ca2+ ]i induced by clioquinol led to S-phase cell cycle arrest. Similar to our previous studies, the increase in [Ca2+ ]i was attributed to changes in the expression levels of the calcium pump SERCA2. Comparison of expression levels of SERCA2 between cell lines showed that cells with high levels of SERCA2 were more sensitive to clioquinol. In addition, analysis using UALCAN and the Human Protein Atlas also showed that the expression of SERCA2 in the corresponding human tissues was similar to that of the cells tested in this study, suggesting potential in the application of clioquinol in the future. In summary, our results expand the understanding of the molecular mechanism of clioquinol and provide an important strategy for the rational use of clioquinol.
Collapse
Affiliation(s)
- Xiaoguang Lv
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Wei Zhang
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| | - Shengli Xia
- Department of Orthopedics, Shanghai University of Medicine & Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Zhiwei Huang
- Key Lab of Science & Technology of Eco-textile, Ministry of Education, College of Chemistry, Chemical Engineering and Biotechnology, Donghua University, Shanghai, China
| | - Ping Shi
- State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, China
| |
Collapse
|
13
|
Pagliaro L, Marchesini M, Roti G. Targeting oncogenic Notch signaling with SERCA inhibitors. J Hematol Oncol 2021; 14:8. [PMID: 33407740 PMCID: PMC7789735 DOI: 10.1186/s13045-020-01015-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 12/02/2020] [Indexed: 12/26/2022] Open
Abstract
P-type ATPase inhibitors are among the most successful and widely prescribed therapeutics in modern pharmacology. Clinical transition has been safely achieved for H+/K+ ATPase inhibitors such as omeprazole and Na+/K+-ATPase inhibitors like digoxin. However, this is more challenging for Ca2+-ATPase modulators due to the physiological role of Ca2+ in cardiac dynamics. Over the past two decades, sarco-endoplasmic reticulum Ca2+-ATPase (SERCA) modulators have been studied as potential chemotherapy agents because of their Ca2+-mediated pan-cancer lethal effects. Instead, recent evidence suggests that SERCA inhibition suppresses oncogenic Notch1 signaling emerging as an alternative to γ-secretase modulators that showed limited clinical activity due to severe side effects. In this review, we focus on how SERCA inhibitors alter Notch1 signaling and show that Notch on-target-mediated antileukemia properties of these molecules can be achieved without causing overt Ca2+ cellular overload.
Collapse
Affiliation(s)
- Luca Pagliaro
- Department of Medicine and Surgery, University of Parma, 43126, Parma, Italy
| | - Matteo Marchesini
- Department of Medicine and Surgery, University of Parma, 43126, Parma, Italy
| | - Giovanni Roti
- Department of Medicine and Surgery, University of Parma, 43126, Parma, Italy.
| |
Collapse
|
14
|
Zhu G, Shen Q, Jiang H, Ji O, Zhu L, Zhang L. Curcumin inhibited the growth and invasion of human monocytic leukaemia SHI-1 cells in vivo by altering MAPK and MMP signalling. PHARMACEUTICAL BIOLOGY 2020; 58:25-34. [PMID: 31854220 PMCID: PMC6968541 DOI: 10.1080/13880209.2019.1701042] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Revised: 11/16/2019] [Accepted: 11/30/2019] [Indexed: 05/20/2023]
Abstract
Context: Curcumin, a polyphenolic compound extracted from the rhizome of the tropical plant Curcuma longa L. (Zingiberaceae), has been considered as a cancer chemopreventive drug by American National Cancer Institute.Objective: To examine the effect of curcumin on acute monocytic leukaemia SHI-1 cells in vivo.Materials and methods: The SHI-1 cells (1 × 106 cells in 0.1 mL PBS) were injected subcutaneously into the right flanks of the female SCID mice. Curcumin dissolved in olive oil (15 and 30 mg/kg) was administered (i.p.) to mice once a day for 15 days while the control group received olive oil injection. Tumour proliferation and apoptosis were examined by PCNA, TUNEL and cleaved caspase-3 staining. The expression of MAPK, NF-κB, MMP9, MMP2 and vimentin were confirmed by RT-PCR, immunohistochemistry or western blotting.Results: Administration of curcumin significantly inhibited tumour growth, as the tumour weight decreased from 0.67 g (control) to 0.47 g (15 mg/kg) and 0.35 g (30 mg/kg). Curcumin inhibited the expression of PCNA and increased the degree of TUNEL and cleaved caspase-3 staining in tumour tissue. The results of western blotting showed that curcumin treatment inhibited NF-κB and ERK signalling while activating p38 and JNK. Moreover, curcumin attenuated the mRNA transcription and protein expression of MMP2 and MMP9. Curcumin also suppressed the level of vimentin.Discussion and conclusions: Our study demonstrates that curcumin can inhibit the growth and invasion of human monocytic leukaemia in vivo, suggesting the possible use of curcumin for anti-metastasis in leukaemia and the value of determining its unique target.
Collapse
Affiliation(s)
- Guohua Zhu
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
- CONTACT Guohua Zhu First Clinical College, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qun Shen
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
- Department of Hematology, First Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Hong Jiang
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ou Ji
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Lingling Zhu
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Linyang Zhang
- First Clinical College, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
15
|
Dai Z, Tang J, Gu Z, Wang Y, Yang Y, Yang Y, Yu C. Eliciting Immunogenic Cell Death via a Unitized Nanoinducer. NANO LETTERS 2020; 20:6246-6254. [PMID: 32786942 DOI: 10.1021/acs.nanolett.0c00713] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Utilizing chemotherapeutics to induce immunogenic cell death (ICD) is a promising strategy to sensitize tumor cells and induce anticancer immunity. However, the application of traditional ICD inducers, such as chemodrugs, is largely hindered by their low tumor selectivity and severe side effects. Here, a new unitized ICD nanoinducer with high potency and cancer cell specificity is reported to achieve effective cancer immunotherapy. This nanoinducer is composed of disulfide-bond-incorporated organosilica nanoparticles, curcumin (CUR), and iron oxide nanoparticles, which can deplete intracellular glutathione, produce hydroxyl radicals, and induce cancer-cell-specific Ca2+ depletion as well as thioredoxin reductase inhibition. While the components are unable to induce ICD individually, their complementary pharmaceutical activities significantly elevate intracellular oxidative stress and endoplasmic reticulum stress in parallel. Consequently, ICD and systemic antitumor immunity can be elicited. Compared to the conventional ICD inducer doxorubicin, the unitized nanoinducer exhibits significantly improved ICD-inducing activity and cancer cell selectivity.
Collapse
Affiliation(s)
- Zan Dai
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
| | - Jie Tang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
| | - Zhengying Gu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
| | - Yue Wang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
| | - Yang Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
| | - Yannan Yang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St Lucia, Brisbane, QLD 4072, Australia
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200241, P.R. China
| |
Collapse
|
16
|
Peterková L, Kmoníčková E, Ruml T, Rimpelová S. Sarco/Endoplasmic Reticulum Calcium ATPase Inhibitors: Beyond Anticancer Perspective. J Med Chem 2020; 63:1937-1963. [PMID: 32030976 DOI: 10.1021/acs.jmedchem.9b01509] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The sarco/endoplasmic reticulum calcium ATPase (SERCA), which plays a key role in the maintenance of Ca2+ ion homeostasis, is an extensively studied enzyme, the inhibition of which has a considerable impact on cell life and death decision. To date, several SERCA inhibitors have been thoroughly studied and the most notable one, a derivative of the sesquiterpene lactone thapsigargin, is gradually approaching a clinical application. Meanwhile, new compounds with SERCA-inhibiting properties of natural, synthetic, or semisynthetic origin are being discovered and/or developed; some of these might also be suitable for the development of new drugs with improved performance. This review brings an up-to-date comprehensive overview of recently discovered compounds with the potential of SERCA inhibition, discusses their mechanism of action, and highlights their potential clinical applications, such as cancer treatment.
Collapse
Affiliation(s)
- Lucie Peterková
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 5, 166 28, Prague 6, Czech Republic
| | - Eva Kmoníčková
- Faculty of Medicine in Pilsen, Charles University, Alej Svobody 76, 323 00 Pilsen, Czech Republic
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 5, 166 28, Prague 6, Czech Republic
| | - Silvie Rimpelová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology Prague, Technická 5, 166 28, Prague 6, Czech Republic.,Faculty of Medicine in Pilsen, Charles University, Alej Svobody 76, 323 00 Pilsen, Czech Republic
| |
Collapse
|
17
|
Lv X, Zheng Q, Li M, Huang Z, Peng M, Sun J, Shi P. Clioquinol induces S-phase cell cycle arrest through the elevation of the calcium level in human neurotypic SH-SY5Y cells. Metallomics 2020; 12:173-182. [DOI: 10.1039/c9mt00260j] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Clioquinol elevates intracellular calcium levels in a non-chelating manner, leading to S-phase cell cycle arrest in human neurotypic SH-SY5Y cells.
Collapse
Affiliation(s)
- Xiaoguang Lv
- State Key Laboratory of Bioreactor Engineering
- East China University of Science and Technology
- Shanghai 200237
- China
| | - Qiaoqiao Zheng
- State Key Laboratory of Bioreactor Engineering
- East China University of Science and Technology
- Shanghai 200237
- China
| | - Ming Li
- State Key Laboratory of Bioreactor Engineering
- East China University of Science and Technology
- Shanghai 200237
- China
| | - Zhiwei Huang
- Key Lab of Science & Technology of Eco-textile
- Ministry of Education
- College of Chemistry
- Chemical Engineering and Biotechnology
- Donghua University
| | - Min Peng
- Qinghai Key Laboratory of Qinghai-Tibet Plateau Biological Resources
- Northwest Institute of Plateau Biology
- The Chinese Academy of Sciences
- Xining 810001
- China
| | - Jing Sun
- Qinghai Key Laboratory of Qinghai-Tibet Plateau Biological Resources
- Northwest Institute of Plateau Biology
- The Chinese Academy of Sciences
- Xining 810001
- China
| | - Ping Shi
- State Key Laboratory of Bioreactor Engineering
- East China University of Science and Technology
- Shanghai 200237
- China
| |
Collapse
|
18
|
Chang JC, Hu WF, Lee WS, Lin JH, Ting PC, Chang HR, Shieh KR, Chen TI, Yang KT. Intermittent Hypoxia Induces Autophagy to Protect Cardiomyocytes From Endoplasmic Reticulum Stress and Apoptosis. Front Physiol 2019; 10:995. [PMID: 31447690 PMCID: PMC6692635 DOI: 10.3389/fphys.2019.00995] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 07/18/2019] [Indexed: 12/25/2022] Open
Abstract
Intermittent hypoxia (IH), characterized as cyclic episodes of short-period hypoxia followed by normoxia, occurs in many physiological and pathophysiological conditions such as pregnancy, athlete, obstructive sleep apnea, and asthma. Hypoxia can induce autophagy, which is activated in response to protein aggregates, in the proteotoxic forms of cardiac diseases. Previous studies suggested that autophagy can protect cells by avoiding accumulation of misfolded proteins, which can be generated in response to ischemia/reperfusion (I/R) injury. The objective of the present study was to determine whether IH-induced autophagy can attenuate endoplasmic reticulum (ER) stress and cell death. In this study, H9c2 cell line, rat primary cultured cardiomyocytes, and C57BL/6 male mice underwent IH with an oscillating O2 concentration between 4 and 20% every 30 min for 1-4 days in an incubator. The levels of LC3, an autophagy indicator protein and CHOP and GRP78 (ER stress-related proteins) were measured by Western blotting analyses. Our data demonstrated that the autophagy-related proteins were upregulated in days 1-3, while the ER stress-related proteins were downregulated on the second day after IH. Treatment with H2O2 (100 μM) for 24 h caused ER stress and increased the level of ER stress-related proteins, and these effects were abolished by pre-treatment with IH condition. In response to the autophagy inhibitor, the level of ER stress-related proteins was upregulated again. Taken together, our data suggested that IH could increase myocardial autophagy as an adaptive response to prevent the ER stress and apoptosis.
Collapse
Affiliation(s)
- Jui-Chih Chang
- Department of Surgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wei-Fen Hu
- Master Program in Medical Physiology, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wen-Sen Lee
- Graduate Institute of Medical Sciences, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jian-Hong Lin
- PhD Program in Pharmacology and Toxicology, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Pei-Ching Ting
- Department of Surgery, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Huai-Ren Chang
- School of Medicine, Tzu Chi University, Hualien, Taiwan.,Division of Cardiology, Department of Internal Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Kun-Ruey Shieh
- School of Medicine, Tzu Chi University, Hualien, Taiwan.,Master Program in Medical Physiology, School of Medicine, Tzu Chi University, Hualien, Taiwan.,Department of Physiology, School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Tsung-I Chen
- Center for Physical Education, College of Education and Communication, Tzu Chi University, Hualien, Taiwan.,Institute of Education, College of Education and Communication, Tzu Chi University, Hualien, Taiwan
| | - Kun-Ta Yang
- Master Program in Medical Physiology, School of Medicine, Tzu Chi University, Hualien, Taiwan.,Department of Physiology, School of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
19
|
Shakeri A, Zirak MR, Wallace Hayes A, Reiter R, Karimi G. Curcumin and its analogues protect from endoplasmic reticulum stress: Mechanisms and pathways. Pharmacol Res 2019; 146:104335. [DOI: 10.1016/j.phrs.2019.104335] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 06/27/2019] [Accepted: 06/28/2019] [Indexed: 02/07/2023]
|
20
|
Yen CL, Liao YC, Chen RF, Huang YF, Chung WC, Lo PC, Chang CF, Wu PC, Shieh DB, Jiang ST, Shieh CC. Targeted Delivery of Curcumin Rescues Endoplasmic Reticulum–Retained Mutant NOX2 Protein and Avoids Leukocyte Apoptosis. THE JOURNAL OF IMMUNOLOGY 2019; 202:3394-3403. [DOI: 10.4049/jimmunol.1801599] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 04/05/2019] [Indexed: 12/15/2022]
|
21
|
Zhu T, Chen C, Wang S, Zhang Y, Zhu D, Li L, Luo J, Kong L. Cellular target identification of Withangulatin A using fluorescent analogues and subsequent chemical proteomics. Chem Commun (Camb) 2019; 55:8231-8234. [DOI: 10.1039/c9cc03653a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Find the target of Withangulatin A with the combination of fluorescent probes and chemical proteomics.
Collapse
Affiliation(s)
- Tianyu Zhu
- State Key Laboratory of Natural Medicines
- Department of Natural Medicinal Chemistry
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Chen Chen
- State Key Laboratory of Natural Medicines
- Department of Natural Medicinal Chemistry
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Sisi Wang
- State Key Laboratory of Natural Medicines
- Department of Natural Medicinal Chemistry
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Yi Zhang
- State Key Laboratory of Natural Medicines
- Department of Natural Medicinal Chemistry
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Dongrong Zhu
- State Key Laboratory of Natural Medicines
- Department of Natural Medicinal Chemistry
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Lingnan Li
- State Key Laboratory of Natural Medicines
- Department of Natural Medicinal Chemistry
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Jianguang Luo
- State Key Laboratory of Natural Medicines
- Department of Natural Medicinal Chemistry
- China Pharmaceutical University
- Nanjing 210009
- China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines
- Department of Natural Medicinal Chemistry
- China Pharmaceutical University
- Nanjing 210009
- China
| |
Collapse
|
22
|
Zhang Y, Lin XY, Zhang JH, Xie ZL, Deng H, Huang YF, Huang XH. Apoptosis of mouse myeloma cells induced by curcumin via the Notch3-p53 signaling axis. Oncol Lett 2019; 17:127-134. [PMID: 30655747 PMCID: PMC6313093 DOI: 10.3892/ol.2018.9591] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 08/15/2018] [Indexed: 12/19/2022] Open
Abstract
Resistance to apoptosis is a characteristic of cancer. Curcumin has become a potential anticancer drug for its pro-apoptotic effects, but the underlying mechanisms remain unclear. Furthermore, the Notch3-p53 signaling axis serves an important role in cell fate. The present study was designed to investigate the antitumor effect of curcumin by the Notch3-p53 axis in mouse myeloma P3X63Ag8 cells. The effects of curcumin on the viability of P3X63Ag8 cells were evaluated using an MTT assay. Quantitative expression of the Notch3-p53 signaling axis-associated genes was measured by reverse transcription-quantitative polymerase chain reaction, and western blot analysis was used to investigate the expression of proteins. Additionally, flow cytometry was used to measure the ratio of apoptosis. The results demonstrated that curcumin could significantly inhibit cell viability. No significant pro-apoptotic effect was observed when the concentration of curcumin was <30 µM. At 30 µM, curcumin-treated cells exhibited an apoptotic phenomenon, and the ratio of late apoptosis increased with the concentration of curcumin, and reached 28.4 and 51.8% in the medium- and high-dose groups, respectively. Curcumin inhibited the expression of Notch3, while the middle- and high-dose groups promoted p53. The expression of Notch3-responsive genes Hes family BHLH transcription factor 1 and Hes-related family transcription factor with YRPW motif 1 were notably promoted. Curcumin treatment significantly downregulated B-cell lymphoma-2 (Bcl-2) at the mRNA and protein levels, but upregulated Bcl-2-associated X. These data indicated that curcumin exhibited antitumor effects in mouse myeloma cells with induction of apoptosis by affecting the Notch3-p53 signaling axis.
Collapse
Affiliation(s)
- Ying Zhang
- Fujian Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, P.R. China
| | - Xin-Yu Lin
- Fujian Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, P.R. China
- Department of Zoology, College of Life Sciences, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, P.R. China
| | - Jiao-Hui Zhang
- Fujian Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, P.R. China
| | - Zheng-Lu Xie
- Fujian Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, P.R. China
| | - Hui Deng
- Fujian Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, P.R. China
| | - Yi-Fang Huang
- Fujian Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, P.R. China
| | - Xiao-Hong Huang
- Fujian Key Laboratory of Traditional Chinese Veterinary Medicine and Animal Health, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, P.R. China
| |
Collapse
|
23
|
The Role of the Anti-Aging Protein Klotho in IGF-1 Signaling and Reticular Calcium Leak: Impact on the Chemosensitivity of Dedifferentiated Liposarcomas. Cancers (Basel) 2018; 10:cancers10110439. [PMID: 30441794 PMCID: PMC6266342 DOI: 10.3390/cancers10110439] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 11/09/2018] [Indexed: 01/23/2023] Open
Abstract
By inhibiting Insulin-Like Growth Factor-1-Receptor (IGF-1R) signaling, Klotho (KL) acts like an aging- and tumor-suppressor. We investigated whether KL impacts the aggressiveness of liposarcomas, in which IGF-1R signaling is frequently upregulated. Indeed, we observed that a higher KL expression in liposarcomas is associated with a better outcome for patients. Moreover, KL is downregulated in dedifferentiated liposarcomas (DDLPS) compared to well-differentiated tumors and adipose tissue. Because DDLPS are high-grade tumors associated with poor prognosis, we examined the potential of KL as a tool for overcoming therapy resistance. First, we confirmed the attenuation of IGF-1-induced calcium (Ca2+)-response and Extracellular signal-Regulated Kinase 1/2 (ERK1/2) phosphorylation in KL-overexpressing human DDLPS cells. KL overexpression also reduced cell proliferation, clonogenicity, and increased apoptosis induced by gemcitabine, thapsigargin, and ABT-737, all of which are counteracted by IGF-1R-dependent signaling and activate Ca2+-dependent endoplasmic reticulum (ER) stress. Then, we monitored cell death and cytosolic Ca2+-responses and demonstrated that KL increases the reticular Ca2+-leakage by maintaining TRPC6 at the ER and opening the translocon. Only the latter is necessary for sensitizing DDLPS cells to reticular stressors. This was associated with ERK1/2 inhibition and could be mimicked with IGF-1R or MEK inhibitors. These observations provide a new therapeutic strategy in the management of DDLPS.
Collapse
|
24
|
Pesakhov S, Nachliely M, Barvish Z, Aqaqe N, Schwartzman B, Voronov E, Sharoni Y, Studzinski GP, Fishman D, Danilenko M. Cancer-selective cytotoxic Ca2+ overload in acute myeloid leukemia cells and attenuation of disease progression in mice by synergistically acting polyphenols curcumin and carnosic acid. Oncotarget 2017; 7:31847-61. [PMID: 26870993 PMCID: PMC5077981 DOI: 10.18632/oncotarget.7240] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2015] [Accepted: 01/19/2016] [Indexed: 12/20/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive hematologic malignancy characterized by extremely heterogeneous molecular and biologic abnormalities that hamper the development of effective targeted treatment modalities. While AML cells are highly sensitive to cytotoxic Ca2+ overload, the feasibility of Ca2+- targeted therapy of this disease remains unclear. Here, we show that apoptotic response of AML cells to the synergistically acting polyphenols curcumin (CUR) and carnosic acid (CA), combined at low, non-cytotoxic doses of each compound was mediated solely by disruption of cellular Ca2+ homeostasis. Specifically, activation of caspase cascade in CUR+CA-treated AML cells resulted from sustained elevation of cytosolic Ca2+ (Ca2+cyt) and was not preceded by endoplasmic reticulum stress or mitochondrial damage. The CUR+CA-induced Ca2+cyt rise did not involve excessive influx of extracellular Ca2+ but, rather, occurred due to massive Ca2+ release from intracellular stores concomitant with inhibition of Ca2+cyt extrusion through the plasma membrane. Notably, the CUR+CA combination did not alter Ca2+ homeostasis and viability in non-neoplastic hematopoietic cells, suggesting its cancer-selective action. Most importantly, co-administration of CUR and CA to AML-bearing mice markedly attenuated disease progression in two animal models. Collectively, our results provide the mechanistic and translational basis for further characterization of this combination as a prototype of novel Ca2+-targeted pharmacological tools for the treatment of AML.
Collapse
Affiliation(s)
- Stella Pesakhov
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Matan Nachliely
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Zeev Barvish
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel.,Permanent address: Blood Bank Institute, Soroka University Medical Center, Beer Sheva 85025, Israel
| | - Nasma Aqaqe
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel.,Permanent address: Department of Pathology, Sackler Faculty of Medicine Tel-Aviv University, Tel-Aviv 69978, Israel
| | - Bar Schwartzman
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Elena Voronov
- The Shraga Segal Department of Microbiology and Immunology, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Yoav Sharoni
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - George P Studzinski
- Department of Pathology and Laboratory Medicine, Rutgers-New Jersey Medical School, Newark, NJ 07103, USA
| | - Daniel Fishman
- Department of Physiology and Cell Biology, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Michael Danilenko
- Department of Clinical Biochemistry and Pharmacology, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| |
Collapse
|
25
|
Fan L, Xiao Q, Chen Y, Chen G, Duan J, Tao W. Pekinenin E Inhibits the Growth of Hepatocellular Carcinoma by Promoting Endoplasmic Reticulum Stress Mediated Cell Death. Front Pharmacol 2017; 8:424. [PMID: 28706487 PMCID: PMC5489557 DOI: 10.3389/fphar.2017.00424] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 06/14/2017] [Indexed: 01/27/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant primary liver cancer with poor prognosis. In the present study, we report that pekinenin E (PE), a casbane diterpenoid derived from the roots of Euphorbia pekinensis, has a strong antitumor activity against human HCC cells both in vitro and in vivo. PE suppressed the growth of human HCC cells Hep G2 and SMMC-7721. In addition, PE-mediated endoplasmic reticulum (ER) stress caused increasing expressions of C/EBP homologous protein (CHOP), leading to apoptosis in HCC cells both in vitro and in vivo. Inhibition of ER stress with CHOP small interfering RNA or 4-phenyl-butyric acid partially reversed PE-induced cell death. Furthermore, PE induced S cell cycle arrest, which could also be partially reversed by CHOP knockdown. In all, these findings suggest that PE causes ER stress-associated cell death and cell cycle arrest, and it may serve as a potent agent for curing human HCC.
Collapse
Affiliation(s)
- Lu Fan
- School of Medicine and Life Sciences, Nanjing University of Chinese MedicineNanjing, China
| | - Qingling Xiao
- School of Basic Biomedical Science, Nanjing University of Chinese MedicineNanjing, China
| | - Yanyan Chen
- School of Basic Biomedical Science, Nanjing University of Chinese MedicineNanjing, China
| | - Gang Chen
- School of Basic Biomedical Science, Nanjing University of Chinese MedicineNanjing, China
| | - Jinao Duan
- Jiangsu Key Laboratory for High Technology Research of TCM Formulae and Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese MedicineNanjing, China
| | - Weiwei Tao
- School of Basic Biomedical Science, Nanjing University of Chinese MedicineNanjing, China
| |
Collapse
|
26
|
Liu Q, Chen Z, Jiang G, Zhou Y, Yang X, Huang H, Liu H, Du J, Wang H. Epigenetic down regulation of G protein-coupled estrogen receptor (GPER) functions as a tumor suppressor in colorectal cancer. Mol Cancer 2017; 16:87. [PMID: 28476123 PMCID: PMC5418684 DOI: 10.1186/s12943-017-0654-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 04/24/2017] [Indexed: 02/07/2023] Open
Abstract
Background Estrogenic signals are suggested to have protection roles in the development of colorectal cancer (CRC). The G protein-coupled estrogen receptor (GPER) has been reported to mediate non-genomic effects of estrogen in hormone related cancers except CRC. Its expression and functions in CRC were investigated. Methods The expression of GPER and its associations with clinicopathological features were examined. The mechanisms were further investigated using cells, mouse xenograft models, and clinical human samples. Results GPER was significantly (p < 0.01) down regulated in CRC tissues compared with their matched adjacent normal tissues in our two cohorts and three independent investigations from Oncomine database. Patients whose tumors expressing less (n = 36) GPER showed significant (p < 0.01) poorer survival rate as compared with those with greater levels of GPER (n = 54). Promoter methylation and histone H3 deacetylation were involved in the down regulation of GPER in CRC cell lines and clinical tissues. Activation of GPER by its specific agonist G-1 inhibited proliferation, induced cell cycle arrest, mitochondrial-related apoptosis and endoplasmic reticulum (ER) stress of CRC cells. The upregulation of reactive oxygen species (ROS) induced sustained ERK1/2 activation participated in G-1 induced cell growth arrest. Further, G-1 can inhibit the phosphorylation, nuclear localization, and transcriptional activities of NF-κB via both canonical IKKα/ IκBα pathways and phosphorylation of GSK-3β. Xenograft model based on HCT-116 cells confirmed that G-1 can suppress the in vivo progression of CRC. Conclusions Epigenetic down regulation of GPER acts as a tumor suppressor in colorectal cancer and its specific activation might be a potential approach for CRC treatment. Electronic supplementary material The online version of this article (doi:10.1186/s12943-017-0654-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Qiao Liu
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Zhuojia Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Guanmin Jiang
- Hunan Cancer Hospital & The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410013, China
| | - Yan Zhou
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Xiangling Yang
- Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Institute of Human Virology, Key Laboratory of Tropical Disease Control (Ministry of Education), Sun Yat-sen University, Guangzhou, 510655, China
| | - Hongbin Huang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Huanliang Liu
- Guangdong Institute of Gastroenterology and the Sixth Affiliated Hospital, Institute of Human Virology, Key Laboratory of Tropical Disease Control (Ministry of Education), Sun Yat-sen University, Guangzhou, 510655, China
| | - Jun Du
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Hongsheng Wang
- Department of Microbial and Biochemical Pharmacy, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
27
|
Feng C, Xia Y, Zou P, Shen M, Hu J, Ying S, Pan J, Liu Z, Dai X, Zhuge W, Liang G, Ruan Y. Curcumin analog L48H37 induces apoptosis through ROS-mediated endoplasmic reticulum stress and STAT3 pathways in human lung cancer cells. Mol Carcinog 2017; 56:1765-1777. [PMID: 28218464 DOI: 10.1002/mc.22633] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Revised: 02/07/2017] [Accepted: 02/16/2017] [Indexed: 12/19/2022]
Abstract
Lung cancer is the leading cause of cancer-related deaths. Curcumin is a well-known natural product with anticancer ability, however, its poor bioavailability and pharmacokinetic profiles have limited its application in anticancer therapy. Previously, we reported that L48H37, a novel analog of curcumin with higher bioavailability, ameliorated LPS-induced inflammation, but the anticancer effect of L48H37 is still unknown. In the present study, we have investigated the effects of L48H37 in human lung cancer cells. Our results show that L48H37 decreases lung cancer cell growth and colony formation. These alterations were mediated through induction of G2/M cell cycle arrest and apoptosis in lung cancer cells. After L48H37 treatment, ER stress-related proteins were increased, and the expression of p-STAT3 was decreased in a dose-dependent manner. L48H37 also induced the accumulation of ROS in lung cancer cells, and pretreatment with NAC could fully reverse L48H37-induced reactive oxygen species (ROS) increase. Blocking ROS was able to reverse L48H37-induced endoplasmic reticulum (ER) stress, cell cycle arrest, and apoptosis. Finally, we show that L48H37 inhibits the growth of lung cancer xenografts without exhibiting toxicity. Treatment of mice bearing human lung cancer xenografts with L48H37 was also associated with indices of ER stress activation. In summary, our results provide evidence for a novel anti-tumor candidate for the treatment of lung cancer.
Collapse
Affiliation(s)
- Chen Feng
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yiqun Xia
- Department of Digestive Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Peng Zou
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Miaoshan Shen
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jie Hu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shilong Ying
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jialing Pan
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhiguo Liu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuanxuan Dai
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weishan Zhuge
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yeping Ruan
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China.,College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
28
|
Zou P, Xia Y, Chen W, Chen X, Ying S, Feng Z, Chen T, Ye Q, Wang Z, Qiu C, Yang S, Liang G. EF24 induces ROS-mediated apoptosis via targeting thioredoxin reductase 1 in gastric cancer cells. Oncotarget 2017; 7:18050-64. [PMID: 26919110 PMCID: PMC4951270 DOI: 10.18632/oncotarget.7633] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/14/2016] [Indexed: 12/22/2022] Open
Abstract
Gastric cancer (GC) is one of the leading causes of cancer mortality in the world, and finding novel agents for the treatment of advanced gastric cancer is of urgent need. Diphenyl difluoroketone (EF24), a molecule having structural similarity to curcumin, exhibits potent anti-tumor activities by arresting cell cycle and inducing apoptosis. Although EF24 demonstrates potent anticancer efficacy in numerous types of human cancer cells, the cellular targets of EF24 have not been fully defined. We report here that EF24 may interact with the thioredoxin reductase 1 (TrxR1), an important selenocysteine (Sec)-containing antioxidant enzyme, to induce reactive oxygen species (ROS)-mediated apoptosis in human gastric cancer cells. By inhibiting TrxR1 activity and increasing intracellular ROS levels, EF24 induces a lethal endoplasmic reticulum stress in human gastric cancer cells. Importantly, knockdown of TrxR1 sensitizes cells to EF24 treatment. In vivo, EF24 treatment markedly reduces the TrxR1 activity and tumor cell burden, and displays synergistic lethality with 5-FU against gastric cancer cells. Targeting TrxR1 with EF24 thus discloses a previously unrecognized mechanism underlying the biological activity of EF24, and reveals that TrxR1 is a good target for gastric cancer therapy.
Collapse
Affiliation(s)
- Peng Zou
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yiqun Xia
- Department of Digestive Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weiqian Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xi Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shilong Ying
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhiguo Feng
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tongke Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qingqing Ye
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhe Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chenyu Qiu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shulin Yang
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
29
|
Harati K, Behr B, Daigeler A, Hirsch T, Jacobsen F, Renner M, Harati A, Wallner C, Lehnhardt M, Becerikli M. Curcumin and Viscum album Extract Decrease Proliferation and Cell Viability of Soft-Tissue Sarcoma Cells: An In Vitro Analysis of Eight Cell Lines Using Real-Time Monitoring and Colorimetric Assays. Nutr Cancer 2017; 69:340-351. [PMID: 28045549 DOI: 10.1080/01635581.2017.1263349] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND The cytostatic effects of the polyphenol curcumin and Viscum album extract (VAE) were assessed in soft-tissue sarcoma (STS) cells. METHODS Eight human STS cell lines were used: fibrosarcoma (HT1080), liposarcoma (SW872, T778, MLS-402), synovial sarcoma (SW982, SYO1, 1273), and malignant fibrous histiocytoma (U2197). Primary human fibroblasts served as control cells. Cell proliferation, viability, and cell index (CI) were analyzed by BrdU assay, MTT assay, and real-time cell analysis (RTCA). RESULTS As indicated by BrdU and MTT, curcumin significantly decreased the cell proliferation of five cell lines (HT1080, SW872, SYO1, 1273, and U2197) and the viability of two cell lines (SW872 and SW982). VAE led to significant decreases of proliferation in eight cell lines (HT1080, SW872, T778, MLS-402, SW982, SYO1, 1293, and U2197) and reduced viability in seven STS lines (HT1080, SW872, T778, MLS-402, SW982, SYO1, and 1273). As indicated by RTCA for 160 h, curcumin decreased the CI of all synovial sarcoma cell lines as well as T778 and HT1080. VAE diminished the CI in most of the synovial sarcoma (SW982, SYO1) and liposarcoma (SW872, T778) cell lines as well as HT1080. Primary fibroblasts were not affected adversely by the two compounds in RTCA. CONCLUSION Curcumin and VAE can inhibit the proliferation and viability of STS cells.
Collapse
Affiliation(s)
- K Harati
- a Department of Plastic Surgery , Burn Center, Hand Center, Sarcoma Reference Center, BG-University Hospital Bergmannsheil Bochum , Bochum , Germany
| | - B Behr
- a Department of Plastic Surgery , Burn Center, Hand Center, Sarcoma Reference Center, BG-University Hospital Bergmannsheil Bochum , Bochum , Germany
| | - A Daigeler
- a Department of Plastic Surgery , Burn Center, Hand Center, Sarcoma Reference Center, BG-University Hospital Bergmannsheil Bochum , Bochum , Germany
| | - T Hirsch
- a Department of Plastic Surgery , Burn Center, Hand Center, Sarcoma Reference Center, BG-University Hospital Bergmannsheil Bochum , Bochum , Germany
| | - F Jacobsen
- a Department of Plastic Surgery , Burn Center, Hand Center, Sarcoma Reference Center, BG-University Hospital Bergmannsheil Bochum , Bochum , Germany
| | - M Renner
- b Institute of Pathology, University of Heidelberg , Heidelberg , Germany
| | - A Harati
- c Department of Neurosurgery , Klinikum Dortmund , Dortmund , Germany
| | - C Wallner
- a Department of Plastic Surgery , Burn Center, Hand Center, Sarcoma Reference Center, BG-University Hospital Bergmannsheil Bochum , Bochum , Germany
| | - M Lehnhardt
- a Department of Plastic Surgery , Burn Center, Hand Center, Sarcoma Reference Center, BG-University Hospital Bergmannsheil Bochum , Bochum , Germany
| | - M Becerikli
- a Department of Plastic Surgery , Burn Center, Hand Center, Sarcoma Reference Center, BG-University Hospital Bergmannsheil Bochum , Bochum , Germany
| |
Collapse
|
30
|
Guo H, Cao A, Chu S, Wang Y, Zang Y, Mao X, Wang H, Wang Y, Liu C, Zhang X, Peng W. Astragaloside IV Attenuates Podocyte Apoptosis Mediated by Endoplasmic Reticulum Stress through Upregulating Sarco/Endoplasmic Reticulum Ca 2+-ATPase 2 Expression in Diabetic Nephropathy. Front Pharmacol 2016; 7:500. [PMID: 28066247 PMCID: PMC5174081 DOI: 10.3389/fphar.2016.00500] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 12/05/2016] [Indexed: 12/18/2022] Open
Abstract
Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) plays a central role in the pathogenesis of diabetes. This protein has been recognized as a potential target for diabetic therapy. In this study, we identified astragaloside IV (AS-IV) as a potent modulator of SERCA inhibiting renal injury in diabetic status. Increasing doses of AS-IV (2, 6, and 18 mg kg-1 day-1) were administered intragastrically to db/db mice for 8 weeks. Biochemical and histopathological approaches were conducted to evaluate the therapeutic effects of AS-IV. Cultured mouse podocytes were used to further explore the underlying mechanism in vitro. AS-IV dose-dependently increased SERCA activity and SERCA2 expression, and suppressed ER stress-mediated and mitochondria-mediated apoptosis in db/db mouse kidney. AS-IV also normalized glucose tolerance and insulin sensitivity, improved renal function, and ameliorated glomerulosclerosis and renal inflammation in db/db mice. In palmitate stimulated podocytes, AS-IV markedly improved inhibitions of SERCA activity and SERCA2 expression, restored intracellular Ca2+ homeostasis, and attenuated podocyte apoptosis in a dose-dependent manner with a concomitant abrogation of ER stress as evidenced by the downregulation of GRP78, cleaved ATF6, phospho-IRE1α and phospho-PERK, and the inactivation of both ER stress-mediated and mitochondria-mediated apoptotic pathways. Furthermore, SERCA2b knockdown eliminated the effect of AS-IV on ER stress and ER stress-mediated apoptotic pathway, whereas its overexpression exhibited an anti-apoptotic effect. Our data obtained from in vivo and in vitro studies demonstrate that AS-IV attenuates renal injury in diabetes subsequent to inhibiting ER stress-induced podocyte apoptosis through restoring SERCA activity and SERCA2 expression.
Collapse
Affiliation(s)
- Hengjiang Guo
- Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Aili Cao
- Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Shuang Chu
- Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Yi Wang
- Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Yingjun Zang
- Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Xiaodong Mao
- Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Hao Wang
- Department of Nephrology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Yunman Wang
- Department of Nephrology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Cheng Liu
- Experimental Research Center, Putuo Hospital, Shanghai University of Traditional Chinese Medicine Shanghai, China
| | - Xuemei Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University Shanghai, China
| | - Wen Peng
- Laboratory of Renal Disease, Putuo Hospital, Shanghai University of Traditional Chinese MedicineShanghai, China; Department of Nephrology, Putuo Hospital, Shanghai University of Traditional Chinese MedicineShanghai, China
| |
Collapse
|
31
|
Ma Z, Fan C, Yang Y, Di S, Hu W, Li T, Zhu Y, Han J, Xin Z, Wu G, Zhao J, Li X, Yan X. Thapsigargin sensitizes human esophageal cancer to TRAIL-induced apoptosis via AMPK activation. Sci Rep 2016; 6:35196. [PMID: 27731378 PMCID: PMC5059685 DOI: 10.1038/srep35196] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Accepted: 09/26/2016] [Indexed: 12/22/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a promising anticancer agent for esophageal squamous cell carcinoma (ESCC). Forced expression of CHOP, one of the key downstream transcription factors during endoplasmic reticulum (ER) stress, upregulates the death receptor 5 (DR5) levels and promotes oxidative stress and cell death. In this study, we show that ER stress mediated by thapsigargin promoted CHOP and DR5 synthesis thus sensitizing TRAIL treatment, which induced ESCC cells apoptosis. These effects were reversed by DR5 siRNA in vitro and CHOP siRNA both in vitro and in vivo. Besides, chemically inhibition of AMPK by Compound C and AMPK siRNA weakened the anti-cancer effect of thapsigargin and TRAIL co-treatment. Therefore, our findings suggest ER stress effectively sensitizes human ESCC to TRAIL-mediated apoptosis via the TRAIL-DR5-AMPK signaling pathway, and that activation of ER stress may be beneficial for improving the efficacy of TRAIL-based anti-cancer therapy.
Collapse
Affiliation(s)
- Zhiqiang Ma
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Chongxi Fan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Yang Yang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Shouyin Di
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Wei Hu
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Tian Li
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Yifang Zhu
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Jing Han
- Department of Ophthalmology, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Zhenlong Xin
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Guiling Wu
- Department of Biomedical Engineering, The Fourth Military Medical University, 169 Changle West Road, Xi'an 710032, China
| | - Jing Zhao
- Department of Thoracic Surgery, Beijing Military General Hospital, 5 DongSi ShiTiao Road 100070, Beijing 100700, China
| | - Xiaofei Li
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, 1 Xinsi Road, Xi'an 710038, China
| |
Collapse
|
32
|
Talalaeva OS, Zverev YF, Bryukhanov VM. Mechanisms of Antiradical Activity of 2,3,5,6,8-Pentahydroxy-7-Ethyl-1,4-Naphthoquinone (A Review). Pharm Chem J 2016. [DOI: 10.1007/s11094-016-1450-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
33
|
Small molecule RL71 targets SERCA2 at a novel site in the treatment of human colorectal cancer. Oncotarget 2016; 6:37613-25. [PMID: 26608678 PMCID: PMC4741952 DOI: 10.18632/oncotarget.6068] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2015] [Accepted: 09/26/2015] [Indexed: 12/14/2022] Open
Abstract
While targeted agents are an important part of the treatment arsenal for colorectal cancer, there is still a lack of efficient small-molecule targeted agents based on the understanding of pathogenic molecular mechanisms. In this study, curcumin analog RL71 displayed potent cytotoxicity towards human colon cancer cells with an IC50 of 0.8 μM in SW480 cells and inhibited xenotransplanted tumor growth in a dose-dependent manner. Using affinity chromatography, we identified sarco/endoplasmic reticulum calcium-ATPase (SERCA) 2 as the binding target of RL71. Most notably, RL71 demonstrated special binding to SERCA2 at a novel site with the lowest estimated free energy −8.89 kcal mol−1, and the SERCA2 residues critical for RL71 binding were identified. RL71 suppressed the Ca2+-ATPase activity of SERCA2 both in vitro and in vivo, accompanied by the induction of endoplasmic reticulum stress leading to apoptosis and G2/M cycle arrest in SW480 cells. In addition, RL71 showed synergistic cytotoxicity with the pan-SERCA inhibitor thapsigargin. These results suggest that RL71 could be a selective small-molecule inhibitor of SERCA2, and that it may serve as a lead compound for the study of targeted colorectal cancer therapy.
Collapse
|
34
|
Ye H, Wei X, Wang Z, Zhang S, Ren J, Yao S, Shi L, Yang L, Qiu P, Wu J, Liang G. A novel double carbonyl analog of curcumin induces the apoptosis of human lung cancer H460 cells via the activation of the endoplasmic reticulum stress signaling pathway. Oncol Rep 2016; 36:1640-8. [DOI: 10.3892/or.2016.4911] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 04/29/2016] [Indexed: 11/05/2022] Open
|
35
|
Zou P, Zhang J, Xia Y, Kanchana K, Guo G, Chen W, Huang Y, Wang Z, Yang S, Liang G. ROS generation mediates the anti-cancer effects of WZ35 via activating JNK and ER stress apoptotic pathways in gastric cancer. Oncotarget 2016; 6:5860-76. [PMID: 25714022 PMCID: PMC4467407 DOI: 10.18632/oncotarget.3333] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 01/03/2015] [Indexed: 12/18/2022] Open
Abstract
Gastric cancer is one of the leading causes of cancer mortality in the world, and finding novel agents and strategies for the treatment of advanced gastric cancer is of urgent need. Curcumin is a well-known natural product with anti-cancer ability, but is limited by its poor chemical stability. In this study, an analog of curcumin with high chemical stability, WZ35, was designed and evaluated for its anti-cancer effects and underlying mechanisms against human gastric cancer. WZ35 showed much stronger anti-proliferative effects than curcumin, accompanied by dose-dependent induction of cell cycle arrest and apoptosis in gastric cancer cells. Mechanistically, our data showed that WZ35 induced reactive oxygen species (ROS) production, resulting in the activation of both JNK-mitochondrial and ER stress apoptotic pathways and eventually cell apoptosis in SGC-7901 cells. Blockage of ROS production totally reversed WZ35-induced JNK and ER stress activation as well as cancer cell apoptosis. In vivo, WZ35 showed a significant reduction in SGC-7901 xenograft tumor size in a dose-dependent manner. Taken together, this work provides a novel anticancer candidate for the treatment of gastric cancer, and importantly, reveals that increased ROS generation might be an effective strategy in human gastric cancer treatment.
Collapse
Affiliation(s)
- Peng Zou
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou Zhejiang, China.,School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
| | - Junru Zhang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou Zhejiang, China
| | - Yiqun Xia
- Department of Digestive Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Karvannan Kanchana
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou Zhejiang, China
| | - Guilong Guo
- Department of Oncological Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenbo Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou Zhejiang, China
| | - Yi Huang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou Zhejiang, China
| | - Zhe Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou Zhejiang, China
| | - Shulin Yang
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou Zhejiang, China
| |
Collapse
|
36
|
Seo JA, Kim B, Dhanasekaran DN, Tsang BK, Song YS. Curcumin induces apoptosis by inhibiting sarco/endoplasmic reticulum Ca2+ ATPase activity in ovarian cancer cells. Cancer Lett 2016; 371:30-7. [DOI: 10.1016/j.canlet.2015.11.021] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 11/13/2015] [Accepted: 11/14/2015] [Indexed: 11/25/2022]
|
37
|
Moustapha A, Pérétout PA, Rainey NE, Sureau F, Geze M, Petit JM, Dewailly E, Slomianny C, Petit PX. Curcumin induces crosstalk between autophagy and apoptosis mediated by calcium release from the endoplasmic reticulum, lysosomal destabilization and mitochondrial events. Cell Death Discov 2015; 1:15017. [PMID: 27551451 PMCID: PMC4979459 DOI: 10.1038/cddiscovery.2015.17] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 06/23/2015] [Indexed: 12/22/2022] Open
Abstract
Curcumin, a major active component of turmeric (Curcuma longa, L.), has anticancer effects. In vitro studies suggest that curcumin inhibits cancer cell growth by activating apoptosis, but the mechanism underlying these effects is still unclear. Here, we investigated the mechanisms leading to apoptosis in curcumin-treated cells. Curcumin induced endoplasmic reticulum stress causing calcium release, with a destabilization of the mitochondrial compartment resulting in apoptosis. These events were also associated with lysosomal membrane permeabilization and of caspase-8 activation, mediated by cathepsins and calpains, leading to Bid cleavage. Truncated tBid disrupts mitochondrial homeostasis and enhance apoptosis. We followed the induction of autophagy, marked by the formation of autophagosomes, by staining with acridine orange in cells exposed curcumin. At this concentration, only the early events of apoptosis (initial mitochondrial destabilization with any other manifestations) were detectable. Western blotting demonstrated the conversion of LC3-I to LC3-II (light chain 3), a marker of active autophagosome formation. We also found that the production of reactive oxygen species and formation of autophagosomes following curcumin treatment was almost completely blocked by N-acetylcystein, the mitochondrial specific antioxidants MitoQ10 and SKQ1, the calcium chelators, EGTA-AM or BAPTA-AM, and the mitochondrial calcium uniporter inhibitor, ruthenium red. Curcumin-induced autophagy failed to rescue all cells and most cells underwent type II cell death following the initial autophagic processes. All together, these data imply a fail-secure mechanism regulated by autophagy in the action of curcumin, suggesting a therapeutic potential for curcumin. Offering a novel and effective strategy for the treatment of malignant cells.
Collapse
Affiliation(s)
- A Moustapha
- INSERM U1124 'Toxicologie, Pharmacologie et Signalisation Cellulaire', Université Paris-Descartes, Centre Universitaire des Saints-Pères , Paris, France
| | - P A Pérétout
- INSERM U1124 'Toxicologie, Pharmacologie et Signalisation Cellulaire', Université Paris-Descartes, Centre Universitaire des Saints-Pères , Paris, France
| | - N E Rainey
- INSERM U1124 'Toxicologie, Pharmacologie et Signalisation Cellulaire', Université Paris-Descartes, Centre Universitaire des Saints-Pères , Paris, France
| | - F Sureau
- Université Pierre et Marie Curie-Paris 6, Laboratoire Jean Perrin , Paris, France
| | - M Geze
- Muséum National d'Histoire Naturelles, CeMIM/USM 0504, 'Biologie Fonctionnelles des Protozoaires' 57 , Paris, France
| | - J-M Petit
- Muséum National d'Histoire Naturelles, UMR 7245 CNRS/MNHN 'Molécules de Communication et Adaptation des Micro-organismes' 57 , Paris, France
| | - E Dewailly
- Laboratoire de Physiologie cellulaire, INSERM U800, Université des Sciences et Techniques de Lille 1 , Villeneuve d'Ascq, France
| | - C Slomianny
- Laboratoire de Physiologie cellulaire, INSERM U800, Université des Sciences et Techniques de Lille 1 , Villeneuve d'Ascq, France
| | - P X Petit
- INSERM U1124 'Toxicologie, Pharmacologie et Signalisation Cellulaire', Université Paris-Descartes, Centre Universitaire des Saints-Pères , Paris, France
| |
Collapse
|
38
|
Farooqi AA, Li KT, Fayyaz S, Chang YT, Ismail M, Liaw CC, Yuan SSF, Tang JY, Chang HW. Anticancer drugs for the modulation of endoplasmic reticulum stress and oxidative stress. Tumour Biol 2015; 36:5743-52. [PMID: 26188905 PMCID: PMC4546701 DOI: 10.1007/s13277-015-3797-0] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2015] [Accepted: 07/10/2015] [Indexed: 12/20/2022] Open
Abstract
Prior research has demonstrated how the endoplasmic reticulum (ER) functions as a multifunctional organelle and as a well-orchestrated protein-folding unit. It consists of sensors which detect stress-induced unfolded/misfolded proteins and it is the place where protein folding is catalyzed with chaperones. During this folding process, an immaculate disulfide bond formation requires an oxidized environment provided by the ER. Protein folding and the generation of reactive oxygen species (ROS) as a protein oxidative byproduct in ER are crosslinked. An ER stress-induced response also mediates the expression of the apoptosis-associated gene C/EBP-homologous protein (CHOP) and death receptor 5 (DR5). ER stress induces the upregulation of tumor necrosis factor-related apoptosis inducing ligand (TRAIL) receptor and opening new horizons for therapeutic research. These findings can be used to maximize TRAIL-induced apoptosis in xenografted mice. This review summarizes the current understanding of the interplay between ER stress and ROS. We also discuss how damage-associated molecular patterns (DAMPs) function as modulators of immunogenic cell death and how natural products and drugs have shown potential in regulating ER stress and ROS in different cancer cell lines. Drugs as inducers and inhibitors of ROS modulation may respectively exert inducible and inhibitory effects on ER stress and unfolded protein response (UPR). Reconceptualization of the molecular crosstalk among ROS modulating effectors, ER stress, and DAMPs will lead to advances in anticancer therapy.
Collapse
Affiliation(s)
- Ammad Ahmad Farooqi
- Institute of Biomedical and Genetic Engineering (IBGE), KRL Hospital, Islamabad, Pakistan,
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Zou P, Xia Y, Chen T, Zhang J, Wang Z, Chen W, Chen M, Kanchana K, Yang S, Liang G. Selective killing of gastric cancer cells by a small molecule targeting ROS-mediated ER stress activation. Mol Carcinog 2015; 55:1073-86. [DOI: 10.1002/mc.22351] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Revised: 05/18/2015] [Accepted: 05/28/2015] [Indexed: 12/29/2022]
Affiliation(s)
- Peng Zou
- School of Environmental and Biological Engineering; Nanjing University of Science and Technology; Nanjing Jiangsu China
- Chemical Biology Research Center; School of Pharmaceutical Sciences; Wenzhou Medical University; Wenzhou Zhejiang China
| | - Yiqun Xia
- Department of Digestive Diseases; the First Affiliated Hospital of Wenzhou Medical University; Wenzhou Zhejiang China
| | - Tongke Chen
- Laboratory Animal Center; Wenzhou Medical University; Wenzhou Zhejiang China
| | - Junru Zhang
- Chemical Biology Research Center; School of Pharmaceutical Sciences; Wenzhou Medical University; Wenzhou Zhejiang China
| | - Zhe Wang
- Chemical Biology Research Center; School of Pharmaceutical Sciences; Wenzhou Medical University; Wenzhou Zhejiang China
| | - Wenbo Chen
- Chemical Biology Research Center; School of Pharmaceutical Sciences; Wenzhou Medical University; Wenzhou Zhejiang China
| | - Minxiao Chen
- Chemical Biology Research Center; School of Pharmaceutical Sciences; Wenzhou Medical University; Wenzhou Zhejiang China
| | - Karvannan Kanchana
- Chemical Biology Research Center; School of Pharmaceutical Sciences; Wenzhou Medical University; Wenzhou Zhejiang China
| | - Shulin Yang
- School of Environmental and Biological Engineering; Nanjing University of Science and Technology; Nanjing Jiangsu China
| | - Guang Liang
- Chemical Biology Research Center; School of Pharmaceutical Sciences; Wenzhou Medical University; Wenzhou Zhejiang China
| |
Collapse
|
40
|
Hu Y, Wang L, Wang L, Wu X, Wu X, Gu Y, Shu Y, Sun Y, Shen Y, Xu Q. Preferential cytotoxicity of bortezomib toward highly malignant human liposarcoma cells via suppression of MDR1 expression and function. Toxicol Appl Pharmacol 2015; 283:1-8. [DOI: 10.1016/j.taap.2014.12.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 11/12/2014] [Accepted: 12/27/2014] [Indexed: 02/05/2023]
|
41
|
Curcumin inhibits aerobic glycolysis and induces mitochondrial-mediated apoptosis through hexokinase II in human colorectal cancer cells in vitro. Anticancer Drugs 2015; 26:15-24. [DOI: 10.1097/cad.0000000000000132] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
42
|
SBF-1 exerts strong anticervical cancer effect through inducing endoplasmic reticulum stress-associated cell death via targeting sarco/endoplasmic reticulum Ca(2+)-ATPase 2. Cell Death Dis 2014; 5:e1581. [PMID: 25522275 PMCID: PMC4649847 DOI: 10.1038/cddis.2014.538] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 10/02/2014] [Accepted: 11/03/2014] [Indexed: 12/18/2022]
Abstract
Cervical cancer is one of the most common carcinomas in the genital system. In the present study, we report that SBF-1, a synthetic steroidal glycoside, has a strong antigrowth activity against human cervical cancer cells in vitro and in vivo. SBF-1 suppressed the growth, migration and colony formation of HeLa cells. In addition, severe endoplasmic reticulum (ER) stress was triggered by SBF-1, and 4-phenyl-butyric acid, a chemical chaperone, partially reversed SBF-1-induced cell death. To uncover the target protein of SBF-1, the compound was labeled with biotin. The biotin-labeled SBF-1 bound to sarco/ER Ca2+-ATPase 2 (SERCA2) and colocalized with SERCA2 in HeLa cells. Moreover, SBF-1 inhibited SERCA activity, depleted ER Ca2+ and increased cytosolic Ca2+ levels. 1,2-Bis(o-aminophenoxy)ethane-N,N,N′,N′-tetraacetic acid, a chelator of Ca2+, partially blocked SBF-1-induced ER stress and growth inhibition. Importantly, knockdown of SERCA2 increased the sensitivity of HeLa cells to SBF-1-induced ER stress and cell death, whereas overexpression of SERCA2 decreased this sensitivity. Furthermore, SBF-1 induced growth suppression and apoptosis in HeLa xenografts, which is closely related to the induction of ER stress and inhibition of SERCA activity. Finally, SERCA2 expression was elevated in human cervical cancer tissues (n=299) and lymph node metastasis (n=8), as compared with normal cervix tissues (n=23), with a positive correlation with clinical stages. In all, these results suggest that SBF-1 disrupts Ca2+ homeostasis and causes ER stress-associated cell death through directly binding to SERCA2 and inhibiting SERCA activity. Our findings also indicate that SERCA2 is a potential therapeutic target for human cervical cancer.
Collapse
|
43
|
Garg AD, Maes H, van Vliet AR, Agostinis P. Targeting the hallmarks of cancer with therapy-induced endoplasmic reticulum (ER) stress. Mol Cell Oncol 2014; 2:e975089. [PMID: 27308392 PMCID: PMC4905250 DOI: 10.4161/23723556.2014.975089] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 09/24/2014] [Accepted: 09/25/2014] [Indexed: 12/19/2022]
Abstract
The endoplasmic reticulum (ER) is at the center of a number of vital cellular processes such as cell growth, death, and differentiation, crosstalk with immune or stromal cells, and maintenance of proteostasis or homeostasis, and ER functions have implications for various pathologies including cancer. Recently, a number of major hallmarks of cancer have been delineated that are expected to facilitate the development of anticancer therapies. However, therapeutic induction of ER stress as a strategy to broadly target multiple hallmarks of cancer has been seldom discussed despite the fact that several primary or secondary ER stress-inducing therapies have been found to exhibit positive clinical activity in cancer patients. In the present review we provide a brief historical overview of the major discoveries and milestones in the field of ER stress biology with important implications for anticancer therapy. Furthermore, we comprehensively discuss possible strategies enabling the targeting of multiple hallmarks of cancer with therapy-induced ER stress.
Collapse
Affiliation(s)
- Abhishek D Garg
- Cell Death Research & Therapy (CDRT) Laboratory; Department for Cellular and Molecular Medicine; KU Leuven University of Leuven ; Leuven, Belgium
| | - Hannelore Maes
- Cell Death Research & Therapy (CDRT) Laboratory; Department for Cellular and Molecular Medicine; KU Leuven University of Leuven ; Leuven, Belgium
| | - Alexander R van Vliet
- Cell Death Research & Therapy (CDRT) Laboratory; Department for Cellular and Molecular Medicine; KU Leuven University of Leuven ; Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) Laboratory; Department for Cellular and Molecular Medicine; KU Leuven University of Leuven ; Leuven, Belgium
| |
Collapse
|
44
|
Fan L, Li A, Li W, Cai P, Yang B, Zhang M, Gu Y, Shu Y, Sun Y, Shen Y, Wu X, Hu G, Wu X, Xu Q. Novel role of Sarco/endoplasmic reticulum calcium ATPase 2 in development of colorectal cancer and its regulation by F36, a curcumin analog. Biomed Pharmacother 2014; 68:1141-8. [DOI: 10.1016/j.biopha.2014.10.014] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Accepted: 10/16/2014] [Indexed: 11/29/2022] Open
|
45
|
Tan KL, Ali A, Du Y, Fu H, Jin HX, Chin TM, Khan M, Go ML. Synthesis and evaluation of bisbenzylidenedioxotetrahydrothiopranones as activators of endoplasmic reticulum (ER) stress signaling pathways and apoptotic cell death in acute promyelocytic leukemic cells. J Med Chem 2014; 57:5904-18. [PMID: 24960549 PMCID: PMC4216202 DOI: 10.1021/jm401352a] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Curcumin is known to trigger ER-stress induced cell death of acute promyelocytic leukemic (APL) cells by intercepting the degradation of nuclear co-repressor (N-CoR) protein which has a key role in the pathogenesis of APL. Replacing the heptadienedione moiety of curcumin with a monocarbonyl cross-conjugated dienone embedded in a tetrahydrothiopyranone dioxide ring resulted in thiopyranone dioxides that were more resilient to hydrolysis and had greater growth inhibitory activities than curcumin on APL cells. Several members intercepted the degradation of misfolded N-CoR and triggered the signaling cascade in the unfolded protein response (UPR) which led to apoptotic cell death. Microarray analysis showed that genes involved in protein processing pathways that were germane to the activation of the UPR were preferentially up-regulated in treated APL cells, supporting the notion that the UPR was a consequential mechanistic pathway affected by thiopyranone dioxides. The Michael acceptor reactivity of the scaffold may have a role in exacerbating ER stress in APL cells.
Collapse
Affiliation(s)
- Kheng-Lin Tan
- Department of Pharmacy, National University of Singapore , 18 Science Drive 4, 117543, Republic of Singapore
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Erlotinib promotes endoplasmic reticulum stress-mediated injury in the intestinal epithelium. Toxicol Appl Pharmacol 2014; 278:45-52. [PMID: 24768708 DOI: 10.1016/j.taap.2014.04.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 04/10/2014] [Accepted: 04/15/2014] [Indexed: 01/23/2023]
Abstract
Erlotinib, a popular drug for treating non-small cell lung cancer (NSCLC), causes diarrhea in approximately 55% of patients receiving this drug. In the present study, we found that erlotinib induced barrier dysfunction in rat small intestine epithelial cells (IEC-6) by increasing epithelial permeability and down-regulating E-cadherin. The mRNA levels of various pro-inflammatory cytokines (Il-6, Il-25 and Il-17f) were increased after erlotinib treatment in IEC-6 cells. Erlotinib concentration- and time-dependently induced apoptosis and endoplasmic reticulum (ER) stress in both IEC-6 and human colon epithelial cells (CCD 841 CoN). Intestinal epithelial injury was also observed in male C57BL/6J mice administrated with erlotinib. Knockdown of C/EBP homologous protein (CHOP) with small interference RNA partially reversed erlotinib-induced apoptosis, production of IL-6 and down-regulation of E-cadherin in cultured intestinal epithelial cells. In conclusion, erlotinib caused ER stress-mediated injury in the intestinal epithelium, contributing to its side effects of diarrhea in patients.
Collapse
|
47
|
In vitro efficacy of curcumin on Trichomonas vaginalis. Wien Klin Wochenschr 2014; 126 Suppl 1:S32-6. [DOI: 10.1007/s00508-014-0522-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 02/04/2014] [Indexed: 11/26/2022]
|
48
|
Wang L, Li W, Yang Y, Hu Y, Gu Y, Shu Y, Sun Y, Wu X, Shen Y, Xu Q. High expression of sarcoplasmic/endoplasmic reticulum Ca(2+)-ATPase 2b blocks cell differentiation in human liposarcoma cells. Life Sci 2014; 99:37-43. [PMID: 24508653 DOI: 10.1016/j.lfs.2014.01.067] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2013] [Revised: 12/15/2013] [Accepted: 01/17/2014] [Indexed: 11/27/2022]
Abstract
AIMS We have previously reported that elevated expression of sarcoplasmic/endoplasmic reticulum Ca(2+)-ATPase 2 (SERCA2) was related to the malignant degree of different types of human liposarcoma. Here, we investigated the effects of high SERCA2b expression on proliferation and differentiation of preadipocyte-like human liposarcoma cell line SW872 cells. MAIN METHODS SW872 cells were stably transfected with human SERCA2b expressing plasmid. Adipocyte differentiation was assayed by adipogenic gene and protein expression. Cell proliferation, formation of reactive oxygen species (ROS) and phosphorylation of peroxisome proliferator activated receptor gamma (PPAR-γ) and extracellular signal-regulated kinase (ERK) were determined by MTT assay, 2, 7-dichlorofluorescein diacetate (DCF-DA) assay and western blot analysis, respectively. KEY FINDINGS High expression of SERCA2b promoted cell proliferation and blocked the differentiation potential of SW872 cells under both in vitro and in vivo differentiation-inducing environment. Moreover, high expression of SERCA2b induced accumulation of ROS and enhanced ERK signaling, thus leading to inactivation of PPAR-γ and down-regulation of adipocyte-specific genes. SIGNIFICANCE The results revealed a novel role of SERCA2b in facilitating the blockade of human liposarcoma differentiation, which helps provide a molecular target for therapeutic interventions of human liposarcoma.
Collapse
Affiliation(s)
- Lu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Wanshuai Li
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yang Yang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yamei Hu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yanhong Gu
- Department of Clinical Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongqian Shu
- Department of Clinical Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yang Sun
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Xuefeng Wu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yan Shen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.
| | - Qiang Xu
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.
| |
Collapse
|
49
|
Abstract
Curcumin (diferuloylmethane) is the biphenolic active compound of turmeric. Curcumin has been used for hundreds of years to treat various ailments. Curcumin has been reported to exert numerous pharmacological effects by modulating multiple molecular targets including those involved in the pathogenesis of cancer. Cancer has been characterized as the dysregulation of cell signaling pathways through gradual alteration of regulatory proteins and through gene mutation. Curcumin is a highly pleiotropic molecule that modulates several intracellular signaling pathways in cancer. The pleiotropic activities of curcumin have been attributed to its novel molecular structure. Based on its β-diketone moiety, curcumin exists in keto-enol tautomers, and this tautomerism favors interaction and binding with a wide range of enzymes. Several studies have shown modulation of numerous signaling enzymes by curcumin including, LOX, COX-2, XO, proteasomes, Ca(2+)-ATPase of sarcoplasmic reticulum, MMPs, HAT, HDAC, DNMT1, DNA polymerase λ, ribonucleases, GloI, protein kinases (PKA, PKB, PKC, v-Src, GSK-3β, ErbB2), protein reductases (TrxR1, AR), GSH, ICDHs, peroxidases (Prx1, Prx2, Prx6) by treatment with curcumin. Various biophysical analyses have been reported, which shows the underlying molecular interaction of curcumin with multiple targets in terms of binding affinities. The current chapter describes how curcumin binds and modulates multiple enzymes involved cancer. Published clinical trial studies with curcumin in cancer management will also be discussed.
Collapse
Affiliation(s)
- Adeeb Shehzad
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea
| | - Raheem Shahzad
- School of Applied Biosciences, College of Agriculture and Life Sciences, Kyungpook National University, Daegu, Republic of Korea
| | - Young Sup Lee
- School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu, Republic of Korea.
| |
Collapse
|
50
|
Liu Z, Sun Y, Ren L, Huang Y, Cai Y, Weng Q, Shen X, Li X, Liang G, Wang Y. Evaluation of a curcumin analog as an anti-cancer agent inducing ER stress-mediated apoptosis in non-small cell lung cancer cells. BMC Cancer 2013; 13:494. [PMID: 24156374 PMCID: PMC4015692 DOI: 10.1186/1471-2407-13-494] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 10/17/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Recent advances have highlighted the importance of the endoplasmic reticulum (ER) in cell death processes. Pharmacological interventions that effectively enhance tumor cell death through activating ER stress have attracted a great deal of attention for anti-cancer therapy. METHODS A bio-evaluation on 113 curcumin analogs against four cancer cell lines was performed through MTT assay. Furthermore, real time cell assay and flow cytometer were used to evaluate the apoptotic induction of (1E,4E)-1,5-bis(5-bromo-2-ethoxyphenyl)penta-1,4-dien-3-one (B82). Western blot, RT-qPCR, and siRNA were then utilized to confirm whether B82-induced apoptosis is mediated through activating ER stress pathway. Finally, the in vivo anti-tumor effect of B82 was evaluated. RESULTS B82 exhibited strong anti-tumor activity in non-small cell lung cancer (NSCLC) H460 cells. Treatment with B82 significantly induced apoptosis in H460 cells in vitro and inhibited H460 tumor growth in vivo. Further studies demonstrated that the B82-induced apoptosis is mediated by activating ER stress both in vitro and in vivo. CONCLUSIONS A new monocarbonyl analog of curcumin, B82, exhibited anti-tumor effects on H460 cells via an ER stress-mediated mechanism. B82 could be further explored as a potential anticancer agent for the treatment of NSCLC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, University Town, 325035 Wenzhou, Zhejiang, China.
| | | |
Collapse
|