51
|
Dutta R, Khalil R, Green R, Mohapatra SS, Mohapatra S. Withania Somnifera (Ashwagandha) and Withaferin A: Potential in Integrative Oncology. Int J Mol Sci 2019; 20:ijms20215310. [PMID: 31731424 PMCID: PMC6862083 DOI: 10.3390/ijms20215310] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 10/18/2019] [Accepted: 10/20/2019] [Indexed: 12/16/2022] Open
Abstract
Ashwagandha (Withania Somnifera, WS), belonging to the family Solanaceae, is an Ayurvedic herb known worldwide for its numerous beneficial health activities since ancient times. This medicinal plant provides benefits against many human illnesses such as epilepsy, depression, arthritis, diabetes, and palliative effects such as analgesic, rejuvenating, regenerating, and growth-promoting effects. Several clinical trials of the different parts of the herb have demonstrated safety in patients suffering from these diseases. In the last two decades, an active component of Withaferin A (WFA) has shown tremendous cytotoxic activity suggesting its potential as an anti-carcinogenic agent in treatment of several cancers. In spite of enormous progress, a thorough elaboration of the proposed mechanism and mode of action is absent. Herein, we provide a comprehensive review of the properties of WS extracts (WSE) containing complex mixtures of diverse components including WFA, which have shown inhibitory properties against many cancers, (breast, colon, prostate, colon, ovarian, lung, brain), along with their mechanism of actions and pathways involved.
Collapse
Affiliation(s)
- Rinku Dutta
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (R.D.); (R.K.); (R.G.)
- Center for Research and Education in Nanobioengineering, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Roukiah Khalil
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (R.D.); (R.K.); (R.G.)
- Center for Research and Education in Nanobioengineering, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Ryan Green
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (R.D.); (R.K.); (R.G.)
- Center for Research and Education in Nanobioengineering, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Shyam S Mohapatra
- Center for Research and Education in Nanobioengineering, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
- Department of Internal Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
- James A Haley VA Hospital, Tampa, FL 33612, USA
| | - Subhra Mohapatra
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA; (R.D.); (R.K.); (R.G.)
- Center for Research and Education in Nanobioengineering, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
- James A Haley VA Hospital, Tampa, FL 33612, USA
- Correspondence: ; Tel.: +1-813-974-4127
| |
Collapse
|
52
|
Hsu JHM, Chang PMH, Cheng TS, Kuo YL, Wu ATH, Tran TH, Yang YH, Chen JM, Tsai YC, Chu YS, Huang TH, Huang CYF, Lai JM. Identification of Withaferin A as a Potential Candidate for Anti-Cancer Therapy in Non-Small Cell Lung Cancer. Cancers (Basel) 2019; 11:cancers11071003. [PMID: 31319622 PMCID: PMC6678286 DOI: 10.3390/cancers11071003] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 07/15/2019] [Indexed: 12/21/2022] Open
Abstract
Low response rate and recurrence are common issues in lung cancer; thus, identifying a potential compound for these patients is essential. Utilizing an in silico screening method, we identified withaferin A (WA), a cell-permeable steroidal lactone initially extracted from Withania somnifera, as a potential anti-lung cancer and anti-lung cancer stem-like cell (CSC) agent. First, we demonstrated that WA exhibited potent cytotoxicity in several lung cancer cells, as evidenced by low IC50 values. WA concurrently induced autophagy and apoptosis and the activation of reactive oxygen species (ROS), which plays an upstream role in mediating WA-elicited effects. The increase in p62 indicated that WA may modulate the autophagy flux followed by apoptosis. In vivo research also demonstrated the anti-tumor effect of WA treatment. We subsequently demonstrated that WA could inhibit the growth of lung CSCs, decrease side population cells, and inhibit lung cancer spheroid-forming capacity, at least through downregulation of mTOR/STAT3 signaling. Furthermore, the combination of WA and chemotherapeutic drugs, including cisplatin and pemetrexed, exerted synergistic effects on the inhibition of epidermal growth factor receptor (EGFR) wild-type lung cancer cell viability. In addition, WA can further enhance the cytotoxic effect of cisplatin in lung CSCs. Therefore, WA alone or in combination with standard chemotherapy is a potential treatment option for EGFR wild-type lung cancer and may decrease the occurrence of cisplatin resistance by inhibiting lung CSCs.
Collapse
Affiliation(s)
- Jade H-M Hsu
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Peter M-H Chang
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 112, Taiwan
- Faculty of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Tai-Shan Cheng
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei 112, Taiwan
| | - Yu-Lun Kuo
- Department of Computer Science and Information Engineering, National Taiwan University, Taipei 106, Taiwan
| | - Alexander T-H Wu
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Thu-Ha Tran
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei 112, Taiwan
- Taiwan International Graduate Program in Molecular Medicine, National Yang-Ming University and Academia Sinica, Taipei 112, Taiwan
| | - Yun-Hsuan Yang
- The Ph.D. Program in Pharmaceutical Biotechnology, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Jing-Ming Chen
- Graduate Institute of Applied Science and Engineering, College of Science and Engineering, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Yu-Chen Tsai
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Yeh-Shiu Chu
- Brain Research Center, National Yang-Ming University, Taipei 112, Taiwan
| | - Tse-Hung Huang
- Graduate Institute of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Chang Gung University, Taoyuan 333, Taiwan.
- School of Nursing, National Taipei University of Nursing and Health Sciences, Taipei 110, Taiwan.
- Department of Traditional Chinese Medicine, Chang Gung Memorial Hospital, Keelung 204, Taiwan.
- Graduate Institute of Health Industry Technology, Chang Gung University of Science and Technology, Taoyuan 333, Taiwan.
| | - Chi-Ying F Huang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang-Ming University, Taipei 112, Taiwan.
- Institute of Biopharmaceutical Sciences, National Yang-Ming University, Taipei 112, Taiwan.
| | - Jin-Mei Lai
- The Ph.D. Program in Pharmaceutical Biotechnology, College of Medicine, Fu Jen Catholic University, New Taipei City 242, Taiwan.
- Graduate Institute of Applied Science and Engineering, College of Science and Engineering, Fu Jen Catholic University, New Taipei City 242, Taiwan.
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei City 242, Taiwan.
| |
Collapse
|
53
|
Binoy A, Nedungadi D, Katiyar N, Bose C, Shankarappa SA, Nair BG, Mishra N. Plumbagin induces paraptosis in cancer cells by disrupting the sulfhydryl homeostasis and proteasomal function. Chem Biol Interact 2019; 310:108733. [PMID: 31276663 DOI: 10.1016/j.cbi.2019.108733] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 06/27/2019] [Accepted: 07/01/2019] [Indexed: 12/20/2022]
Abstract
Plumbagin (PLB) is an active secondary metabolite extracted from the roots of Plumbago rosea. In this study, we report that plumbagin effectively induces paraptosis by triggering extensive cytoplasmic vacuolation followed by cell death in triple negative breast cancer cells (MDA-MB-231), cervical cancer cells (HeLa) and non-small lung cancer cells (A549) but not in normal lung fibroblast cells (WI-38). The vacuoles originated from the dilation of the endoplasmic reticulum (ER) and were found to be empty. The cell death induced by plumbagin was neither apoptotic nor autophagic. Plumbagin induced ER stress mainly by inhibiting the chymotrypsin-like activity of 26S proteasome as also evident from the accumulation of polyubiquitinated proteins. The vacuolation and cell death were found to be independent of reactive oxygen species generation but was effectively inhibited by thiol antioxidant suggesting that plumbagin could modify the sulfur homeostasis in the cellular milieu. Plumbagin also resulted in a decrease in mitochondrial membrane potential eventually decreasing the ATP production. This is the first study to show that Plumbagin induces paraptosis through proteasome inhibition and disruption of sulfhydryl homeostasis and thus further opens up the lead molecule to potential therapeutic strategies for apoptosis-resistant cancers.
Collapse
Affiliation(s)
- Anupama Binoy
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O., Kollam, 690525, Kerala, India
| | - Divya Nedungadi
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O., Kollam, 690525, Kerala, India
| | - Neeraj Katiyar
- Center for Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - Chinchu Bose
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O., Kollam, 690525, Kerala, India
| | - Sahadev A Shankarappa
- Center for Nanosciences & Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, 682041, Kerala, India
| | - Bipin G Nair
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O., Kollam, 690525, Kerala, India
| | - Nandita Mishra
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Amritapuri, Clappana P.O., Kollam, 690525, Kerala, India.
| |
Collapse
|
54
|
The Role of Herbal Bioactive Components in Mitochondria Function and Cancer Therapy. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2019; 2019:3868354. [PMID: 31308852 PMCID: PMC6594309 DOI: 10.1155/2019/3868354] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/27/2019] [Accepted: 06/02/2019] [Indexed: 12/13/2022]
Abstract
Mitochondria are highly dynamic double-membrane organelles which play a well-recognized role in ATP production, calcium homeostasis, oxidation-reduction (redox) status, apoptotic cell death, and inflammation. Dysfunction of mitochondria has long been observed in a number of human diseases, including cancer. Targeting mitochondria metabolism in tumors as a cancer therapeutic strategy has attracted much attention for researchers in recent years due to the essential role of mitochondria in cancer cell growth, apoptosis, and progression. On the other hand, a series of studies have indicated that traditional medicinal herbs, including traditional Chinese medicines (TCM), exert their potential anticancer effects as an effective adjunct treatment for alleviating the systemic side effects of conventional cancer therapies, for reducing the risk of recurrence and cancer mortality and for improving the quality of patients' life. An amazing feature of these structurally diverse bioactive components is that majority of them target mitochondria to provoke cancer cell-specific death program. The aim of this review is to summarize the in vitro and in vivo studies about the role of these herbs, especially their bioactive compounds in the modulation of the disturbed mitochondrial function for cancer therapy.
Collapse
|
55
|
De S, Das S, Sengupta S. Involvement of HuR in the serum starvation induced autophagy through regulation of Beclin1 in breast cancer cell-line, MCF-7. Cell Signal 2019; 61:78-85. [PMID: 31102648 DOI: 10.1016/j.cellsig.2019.05.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 05/09/2019] [Accepted: 05/14/2019] [Indexed: 01/09/2023]
Abstract
Starvation is a cellular stress that induces autophagy, a conserved cellular self-digestion mechanism that allows cells to degrade and recycle damaged proteins and organelles. The present study illustrated that during serum deprivation, Beclin1, a crucial gene that is essential for autophagosome formation in autophagy, gets controlled post-transcriptionally in breast cancer cell-line MCF-7. RNA affinity chromatography and co-immunoprecipitation confirmed the association of HuR with 3'-UTR of beclin1 mRNA. After cytosolic translocation, HuR enhances beclin1 protein synthesis in response to serum starvation by enhancing the association of beclin1 mRNA to the polysomes. Partial silencing of HuR resulted in reduction of beclin1 expression both at mRNA and protein levels, which in turn decreased starvation-induced autophagic flux. Thus, in conclusion, fine-tuning of beclin1 gene expression at post-transcriptional level by HuR is one of the key regulatory mechanisms of starvation induced autophagy in breast cancer cell-line, MCF-7.
Collapse
Affiliation(s)
- Soumasree De
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92 A.P.C. Road, Kolkata 700009, India
| | - Sayantani Das
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92 A.P.C. Road, Kolkata 700009, India
| | - Sumita Sengupta
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92 A.P.C. Road, Kolkata 700009, India.
| |
Collapse
|
56
|
Theochares B, Vohnoutka R, Boumil E, Shea TB. Beneficial and Deleterious Impact of a Nutritional Supplementation for Inhibition of Proliferation of Neuroblastoma in Culture. Nutr Cancer 2019; 71:1345-1354. [PMID: 31058554 DOI: 10.1080/01635581.2019.1604006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Neuroblastoma, a cancer of the sympathetic nervous system, primarily affects infants and children ≤10 yr of age. High-risk neuroblastoma is associated with low survival rates and increased risks of treatment-related side-effects. Therefore, effective treatments that increase survival and reduce adverse side-effects are crucial. Cucurbitacin E (CucE), a nutritional supplement shown to have potential as an alternative to chemotherapy, was investigated for potential impact on neuroblastoma alone and in combination with the standard chemotherapeutic agent, paclitaxel, (PAC). CucE and PAC each inhibited proliferation of murine neuroblastoma cells in culture. Combined treatment with CucE and PAC also induced morphological differentiation. However, both differentiation and antiproliferative effects were reversible. Consequently, while nutritional supplementation represents a potential therapeutic approach toward treatment of cancer, certain nutritional/chemotherapeutic combinations may induce transient rather than permanent effects. Transient inhibition of proliferation by nutritional supplementation could inadvertently protect carcinogenic cells from toxicity otherwise induced by a chemotherapeutic agent. Combinatorial treatments involving nutritional supplements should therefore be utilized with caution.
Collapse
Affiliation(s)
- Brittany Theochares
- Laboratory for Neuroscience, Department of Biological Sciences, University of Massachusetts, Lowell , One University Avenue , Lowell , Massachusetts , USA
| | - Rishel Vohnoutka
- Laboratory for Neuroscience, Department of Biological Sciences, University of Massachusetts, Lowell , One University Avenue , Lowell , Massachusetts , USA
| | - Edward Boumil
- Laboratory for Neuroscience, Department of Biological Sciences, University of Massachusetts, Lowell , One University Avenue , Lowell , Massachusetts , USA
| | - Thomas B Shea
- Laboratory for Neuroscience, Department of Biological Sciences, University of Massachusetts, Lowell , One University Avenue , Lowell , Massachusetts , USA
| |
Collapse
|
57
|
RNAi Screening-based Identification of USP10 as a Novel Regulator of Paraptosis. Sci Rep 2019; 9:4909. [PMID: 30894572 PMCID: PMC6427038 DOI: 10.1038/s41598-019-40982-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 01/31/2019] [Indexed: 01/11/2023] Open
Abstract
Accumulating reports demonstrate that apoptosis does not explain all the effects of cancer therapy due to the innate and acquired apoptotic resistance of malignant cancer cells. Recently, paraptosis, a type of programmed cell death accompanied by dilation of mitochondria and/or the endoplasmic reticulum (ER), has garnered interest in cancer research as an alternative way to kill apoptosis-resistant cancers. We describe here the adaptation and validation of a high-content cell-based assay to screen and identify novel paraptotic regulators employing the malignant breast cancer cells undergoing curcumin-induced paraptosis. We used YFP-Mito cells, which express fluorescence selectively in mitochondria, to select paraptosis-related genes whose corresponding siRNAs appeared to modulate mitochondrial dilation, a morphological feature of paraptosis. From the selected 38 candidate genes, we chose ubiquitin specific peptidase 10 (USP10), a ubiquitin specific protease, as a strongly active candidate that warranted further evaluation of its involvement in paraptosis. We found that both siRNA-mediated knockdown of USP10 and treatment with the USP10 inhibitor, spautin-1, effectively attenuated curcumin-induced paraptosis. This systematic assay, in which a siRNA library is screened for the ability to ameliorate paraptotic changes in mitochondria, may enable researchers to identify potent regulators of paraptosis and new candidate genes/drugs to combat malignant breast cancer.
Collapse
|
58
|
Xu WT, Shen GN, Luo YH, Piao XJ, Wang JR, Wang H, Zhang Y, Li JQ, Feng YC, Zhang Y, Zhang T, Wang SN, Wang CY, Jin CH. New naphthalene derivatives induce human lung cancer A549 cell apoptosis via ROS-mediated MAPKs, Akt, and STAT3 signaling pathways. Chem Biol Interact 2019; 304:148-157. [PMID: 30871965 DOI: 10.1016/j.cbi.2019.03.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 02/28/2019] [Accepted: 03/07/2019] [Indexed: 11/16/2022]
Abstract
1,4-Naphthoquinone compounds are a class of organic compounds derived from naphthalene. They exert a wide variety of biological effects, but when used as anticancer drugs, have varying levels of side effects. In the present study, in order to reduce toxicity and improve the antitumor activity, we synthesized two novel 1,4-naphthoquinone derivatives, 2-(butane-1-sulfinyl)-1,4-naphthoquinone (BSQ) and 2-(octane-1-sulfinyl)-1,4-naphthoquinone (OSQ). We investigated the antitumor effects of BSQ and OSQ in human lung cancer cells and the underlying molecular mechanisms of these effects, focusing on the relationship between these compounds and reactive oxygen species (ROS) production. MTT assay and trypan blue exclusion assay results showed that BSQ and OSQ had significant cytotoxic effects in human lung cancer cells. Flow cytometry results indicated that the number of apoptotic cells and the intracellular ROS levels significantly increased after treatment with BSQ and OSQ. However, cell apoptosis was inhibited by pretreatment with the ROS scavenger N-acetyl-l-cysteine (NAC). Western blotting results showed that BSQ and OSQ increased the expression levels of p-p38 kinase and p-c-Jun N-terminal kinase (p-JNK), and decreased the expression levels of p-extracellular signal-regulated kinase (p-ERK), p-protein kinase B (p-Akt), and p-signal transducer and activator of transcription-3 (p-STAT3). These phenomena were blocked by mitogen-activated protein kinase (MAPK) inhibitors, Akt inhibitors and NAC. In conclusion, BSQ and OSQ induce human lung cancer A549 cell apoptosis by ROS-mediated MAPKs, Akt, and STAT3 signaling pathways. Therefore, BSQ and OSQ may be therapeutic potential agents for the treatment of human lung cancer.
Collapse
Affiliation(s)
- Wan-Ting Xu
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Gui-Nan Shen
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Ying-Hua Luo
- College of Animal Science & Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Xian-Ji Piao
- Department of Gynaecology and Obstetrics, The Fifth Affiliated Hospital of Harbin Medical University, Daqing, Heilongjiang, 163316, China
| | - Jia-Ru Wang
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Hao Wang
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Yi Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Jin-Qian Li
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Yu-Chao Feng
- College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Yu Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Tong Zhang
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Shi-Nong Wang
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China
| | - Chang-Yuan Wang
- College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China.
| | - Cheng-Hao Jin
- Department of Biochemistry and Molecular Biology, College of Life Science & Technology, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China; College of Food Science, Heilongjiang Bayi Agricultural University, Daqing, Heilongjiang, 163319, China.
| |
Collapse
|
59
|
Chung AH, Leisner TM, Dardis GJ, Bivins MM, Keller AL, Parise LV. CIB1 depletion with docetaxel or TRAIL enhances triple-negative breast cancer cell death. Cancer Cell Int 2019; 19:26. [PMID: 30740034 PMCID: PMC6360800 DOI: 10.1186/s12935-019-0740-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Accepted: 01/29/2019] [Indexed: 12/27/2022] Open
Abstract
Background Patients diagnosed with triple negative breast cancer (TNBC) have limited treatment options and often suffer from resistance and toxicity due to chemotherapy. We previously found that depleting calcium and integrin-binding protein 1 (CIB1) induces cell death selectively in TNBC cells, while sparing normal cells. Therefore, we asked whether CIB1 depletion further enhances tumor-specific killing when combined with either the commonly used chemotherapeutic, docetaxel, or the cell death-inducing ligand, TRAIL. Methods We targeted CIB1 by RNA interference in MDA-MB-436, MDA-MB-231, MDA-MB-468, docetaxel-resistant MDA-MB-436 TNBC cells and ME16C normal breast epithelial cells alone or combination with docetaxel or TRAIL. Cell death was quantified via trypan blue exclusion using flow cytometry and cell death mechanisms were analyzed by Western blotting. Cell surface levels of TRAIL receptors were measured by flow cytometry analysis. Results CIB1 depletion combined with docetaxel significantly enhanced tumor-specific cell death relative to each treatment alone. The enhanced cell death strongly correlated with caspase-8 activation, a hallmark of death receptor-mediated apoptosis. The death receptor TRAIL-R2 was upregulated in response to CIB1 depletion, which sensitized TNBC cells to the ligand TRAIL, resulting in a synergistic increase in cell death. In addition to death receptor-mediated apoptosis, both combination treatments activated a non-apoptotic mechanism, called paraptosis. Interestingly, these combination treatments also induced nearly complete death of docetaxel-resistant MDA-MB-436 cells, again via apoptosis and paraptosis. In contrast, neither combination treatment induced cell death in normal ME16C cells. Conclusion Novel combinations of CIB1 depletion with docetaxel or TRAIL selectively enhance naive and docetaxel-resistant TNBC cell death while sparing normal cell. Therefore, combination therapies that target CIB1 could prove to be a safe and durable strategy for treatment of TNBC and potentially other cancers. Electronic supplementary material The online version of this article (10.1186/s12935-019-0740-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Alexander H Chung
- 1Department of Pharmacology, University of North Carolina at Chapel Hill, CB #7365, Chapel Hill, NC 27599 USA
| | - Tina M Leisner
- 2Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, CB #7260, Chapel Hill, NC 27599 USA
| | - Gabrielle J Dardis
- 2Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, CB #7260, Chapel Hill, NC 27599 USA
| | - Marissa M Bivins
- 1Department of Pharmacology, University of North Carolina at Chapel Hill, CB #7365, Chapel Hill, NC 27599 USA
| | - Alana L Keller
- 2Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, CB #7260, Chapel Hill, NC 27599 USA
| | - Leslie V Parise
- 2Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, CB #7260, Chapel Hill, NC 27599 USA.,3Lineberger Comprehensive Cancer Center, Chapel Hill, NC USA
| |
Collapse
|
60
|
Bhargava P, Malik V, Liu Y, Ryu J, Kaul SC, Sundar D, Wadhwa R. Molecular Insights Into Withaferin-A-Induced Senescence: Bioinformatics and Experimental Evidence to the Role of NFκB and CARF. J Gerontol A Biol Sci Med Sci 2018; 74:183-191. [DOI: 10.1093/gerona/gly107] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Indexed: 12/27/2022] Open
Affiliation(s)
- Priyanshu Bhargava
- DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Japan
| | - Vidhi Malik
- DAILAB, Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology (IIT)-Delhi, India
| | - Ye Liu
- DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Jihoon Ryu
- DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Sunil C Kaul
- DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| | - Durai Sundar
- DAILAB, Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology (IIT)-Delhi, India
| | - Renu Wadhwa
- DAILAB, DBT-AIST International Center for Translational and Environmental Research (DAICENTER), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Japan
| |
Collapse
|
61
|
Xue J, Li R, Zhao X, Ma C, Lv X, Liu L, Liu P. Morusin induces paraptosis-like cell death through mitochondrial calcium overload and dysfunction in epithelial ovarian cancer. Chem Biol Interact 2018; 283:59-74. [PMID: 29421517 DOI: 10.1016/j.cbi.2018.02.003] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 01/12/2018] [Accepted: 02/01/2018] [Indexed: 01/02/2023]
Abstract
Epithelial ovarian cancer (EOC) is the leading cause of death among all gynecological cancers. Morusin, a prenylated flavonoid extracted from the root bark of Morus australis, has been reported to exhibit anti-tumor activity against various human cancers except EOC. In the present study, we explored the potential anti-cancer activity of morusin against EOC in vitro and in vivo and possible underlying mechanisms for the first time. We first found that morusin effectively inhibited EOC cell proliferation and survival in vitro and suppressed tumor growth in vivo. Then we observed that treatment of EOC cells with morusin resulted in paraptosis-like cell death, a novel mode of non-apoptotic programmed cell death that is characterized by extensive cytoplasmic vacuolation due to dilation of the endoplasmic reticulum (ER) and mitochondria and lack of apoptotic hallmarks. In addition, we discovered that morusin induced obvious increase in mitochondrial Ca2+ levels, accumulation of ER stress markers, generation of reactive oxygen species (ROS), and loss of mitochondrial membrane potential (Δψm) in EOC cells. Furthermore, pretreatment with 4, 4'-diisothiocyanostilbene-2, 2'-disulfonic acid (DIDS), a chemical inhibitor of voltage-dependent anion channel (VDAC) on the outer mitochondrial membrane, effectively inhibited mitochondrial Ca2+ influx, cytoplasmic vacuolation and cell death induced by morusin in EOC cells. Moreover, DIDS pretreatment also suppressed morusin-induced accumulation of ER stress markers, ROS production and depletion of Δψm. Consistently, tumor xenograft assays showed that co-treatment with DIDS partially reversed the inhibitory effects of morusin on tumor growth in vivo and inhibited the increased levels of ER stress markers induced by morusin in tumor tissues. Collectively, our results suggest that VDAC-mediated Ca2+ influx into mitochondria and subsequent mitochondrial Ca2+ overload contribute to mitochondrial swelling and dysfunction, leading to morusin-induced paraptosis-like cell death in EOC. This study may provide alternative therapeutic strategies for EOC exhibiting resistance to apoptosis.
Collapse
Affiliation(s)
- Jing Xue
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 West Wenhua Road, Jinan, 250012, Shandong Province, People's Republic of China.
| | - Rui Li
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 West Wenhua Road, Jinan, 250012, Shandong Province, People's Republic of China.
| | - Xinrui Zhao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 West Wenhua Road, Jinan, 250012, Shandong Province, People's Republic of China.
| | - Congcong Ma
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 West Wenhua Road, Jinan, 250012, Shandong Province, People's Republic of China.
| | - Xin Lv
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 West Wenhua Road, Jinan, 250012, Shandong Province, People's Republic of China.
| | - Lidong Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 West Wenhua Road, Jinan, 250012, Shandong Province, People's Republic of China.
| | - Peishu Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, 107 West Wenhua Road, Jinan, 250012, Shandong Province, People's Republic of China.
| |
Collapse
|
62
|
Kim IY, Kwon M, Choi MK, Lee D, Lee DM, Seo MJ, Choi KS. Ophiobolin A kills human glioblastoma cells by inducing endoplasmic reticulum stress via disruption of thiol proteostasis. Oncotarget 2017; 8:106740-106752. [PMID: 29290985 PMCID: PMC5739770 DOI: 10.18632/oncotarget.22537] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2017] [Accepted: 10/28/2017] [Indexed: 12/25/2022] Open
Abstract
Ophiobolin A (OP-A), a fungal sesterterpene from Bipolaris oryzae, was recently shown to have anti-glioma activity. We show here that OP-A induces paraptosis-like cell death accompanied by dilation of the endoplasmic reticulum (ER) in glioma cells, and that CHOP-mediated ER stress plays a critical role in this process. OP-A-induced ER-derived dilation and cell death were found to be independent of reactive oxygen species, but were effectively blocked by various thiol antioxidants. We observed that OP-A can react with cysteinyl thiols to form Michael adducts, suggesting that the ability of OP-A to covalently modify free sulfhydryl groups on proteins may cause protein misfolding and the accumulation of misfolded proteins, leading to paraptosis-like cell death. Taken together, these results indicate that the disruption of thiol proteostasis may critically contribute to the anti-glioma activity of OP-A.
Collapse
Affiliation(s)
- In Young Kim
- Department of Biochemistry, Ajou University School of Medicine, Suwon, Korea.,BK21 Plus Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - MiRi Kwon
- Department of Biochemistry, Ajou University School of Medicine, Suwon, Korea.,BK21 Plus Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Min-Koo Choi
- College of Pharmacy, Dankook University, Cheonan, Korea
| | - Dongjoo Lee
- College of Pharmacy, Ajou University, Suwon, Korea
| | - Dong Min Lee
- Department of Biochemistry, Ajou University School of Medicine, Suwon, Korea.,BK21 Plus Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Min Ji Seo
- Department of Biochemistry, Ajou University School of Medicine, Suwon, Korea.,BK21 Plus Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| | - Kyeong Sook Choi
- Department of Biochemistry, Ajou University School of Medicine, Suwon, Korea.,BK21 Plus Program, Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, Korea
| |
Collapse
|
63
|
Knopf KM, Murphy BL, MacMillan SN, Baskin JM, Barr MP, Boros E, Wilson JJ. In Vitro Anticancer Activity and in Vivo Biodistribution of Rhenium(I) Tricarbonyl Aqua Complexes. J Am Chem Soc 2017; 139:14302-14314. [PMID: 28948792 PMCID: PMC8091166 DOI: 10.1021/jacs.7b08640] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Seven rhenium(I) complexes of the general formula fac-[Re(CO)3(NN)(OH2)]+ where NN = 2,2'-bipyridine (8), 4,4'-dimethyl-2,2'-bipyridine (9), 4,4'-dimethoxy-2,2'-bipyridine (10), dimethyl 2,2'-bipyridine-4,4'-dicarboxylate (11), 1,10-phenanthroline (12), 2,9-dimethyl-1,10-phenanthroline (13), or 4,7-diphenyl-1,10-phenanthroline (14), were synthesized and characterized by 1H NMR spectroscopy, IR spectroscopy, mass spectrometry, and X-ray crystallography. With the exception of 11, all complexes exhibited 50% growth inhibitory concentration (IC50) values that were less than 20 μM in HeLa cells, indicating that these compounds represent a new potential class of anticancer agents. Complexes 9, 10, and 13 were as effective in cisplatin-resistant cells as wild-type cells, signifying that they circumvent cisplatin resistance. The mechanism of action of the most potent complex, 13, was explored further by leveraging its intrinsic luminescence properties to determine its intracellular localization. These studies indicated that 13 induces cytoplasmic vacuolization that is lysosomal in nature. Additional in vitro assays indicated that 13 induces cell death without causing an increase in intracellular reactive oxygen species or depolarization of the mitochondrial membrane potential. Further studies revealed that the mode of cell death does not fall into one of the canonical categories such as apoptosis, necrosis, paraptosis, and autophagy, suggesting that a novel mode of action may be operative for this class of rhenium compounds. The in vivo biodistribution and metabolism of complex 13 and its 99mTc analogue 13* were also evaluated in naı̈ve mice. Complexes 13 and 13* exhibited comparable biodistribution profiles with both hepatic and renal excretion. High-performance liquid chromatography inductively coupled plasma mass-spectrometry (HPLC-ICP-MS) analysis of mouse blood plasma and urine postadministration showed considerable metabolic stability of 13, rendering this potent complex suitable for in vivo applications. These studies have shown the biological properties of this class of compounds and demonstrated their potential as promising theranostic anticancer agents that can circumvent cisplatin resistance.
Collapse
Affiliation(s)
- Kevin M. Knopf
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Brendan L. Murphy
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Samantha N. MacMillan
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Jeremy M. Baskin
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, NY 14853, USA
- Weill Institute for Cell & Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Martin P. Barr
- Thoracic Oncology Research Group, Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, St. James’s Hospital and Trinity College Dublin, Dublin, Ireland
| | - Eszter Boros
- A. A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, 149 13 Street, Suite 2301, Charlestown, MA 02129, USA
| | - Justin J. Wilson
- Department of Chemistry & Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
64
|
De S, Das S, Mukherjee S, Das S, Sengupta Bandyopadhyay S. Establishment of twist-1 and TGFBR2 as direct targets of microRNA-20a in mesenchymal to epithelial transition of breast cancer cell-line MDA-MB-231. Exp Cell Res 2017; 361:85-92. [PMID: 28987542 DOI: 10.1016/j.yexcr.2017.10.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 10/03/2017] [Indexed: 12/16/2022]
Abstract
Messenchymal to epithelial transition (MET) is a significant physiological phenomenon involved in embryogenesis and cancer. This study aims at investigating the mechanism of microRNA-20a (miR-20a) mediated regulation of mesenchymal to epithelial transition and identification of its direct target genes in breast cancer cell-line, MDA-MB-231. Reduced migratory and invasive property, altered cellular morphology along with reduced capability for attachment to basement membrane was acquired by over-expression of miR-20a in invasive MDA-MB-231 cell-line initially expressing low level of this micro-RNA, indicating direct correlation between abundance of miR-20a and metastatic property. The switch from mesenchymal to epithelial cells mediated by miR-20a involved post-transcriptional down-regulation of twist1, which in turn controls downstream epithelial markers like E-cadherin, claudin and mesenchymal markers like N-cadherin, fibronectin, the crucial players of mesenchymal to epithelial transition (MET). Furthermore, another key component, TGF-β and one of its receptors (TGFBR2) were found to be down-regulated by miR-20a. Additionally, reporter assay established that post-transcriptional down-regulation of TGFBR2 occurred through direct binding of miR-20a to its 3'UTR, thus abrogating the TGF-β signaling pathway resulting in inhibition of MET. Delineating the underlying molecular mechanism of miR-20a-mediated MET and defining the target genes will help us to introduce a miRNA-mediated effective therapeutic strategy against breast cancer.
Collapse
Affiliation(s)
- Soumasree De
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92A.P.C. Road, Kolkata, WB 700009, India
| | - Sayantani Das
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92A.P.C. Road, Kolkata, WB 700009, India
| | - Srimoyee Mukherjee
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92A.P.C. Road, Kolkata, WB 700009, India
| | - Sainy Das
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92A.P.C. Road, Kolkata, WB 700009, India
| | - Sumita Sengupta Bandyopadhyay
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, 92A.P.C. Road, Kolkata, WB 700009, India.
| |
Collapse
|
65
|
Kessel D, Reiners JJ. Effects of Combined Lysosomal and Mitochondrial Photodamage in a Non-small-Cell Lung Cancer Cell Line: The Role of Paraptosis. Photochem Photobiol 2017; 93:1502-1508. [PMID: 28696570 DOI: 10.1111/php.12805] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 06/14/2017] [Indexed: 12/16/2022]
Abstract
We previously reported that a low level of lysosomal photodamage potentiated the phototoxic effect of subsequent mitochondrial photodamage mediated by the benzoporphyrin derivative (BPD) in murine hepatoma 1c1c7 cells. This was attributed to release of Ca2+ from damaged lysosomes and a calpain-mediated conversion of the autophagy-related protein ATG5 to a pro-apoptotic fragment. We now report a comparison of these results with those obtained with the human non-small-cell lung cancer A549 cell line. A549 cells contained lower levels of ATG5 and were less responsive than 1c1c7 cultures to the PDT combination. A rapid appearance of caspase 3/7 activation together with formation of condensed chromatin indicated initiation of apoptosis in both cell lines, but to a lesser extent in A549 cultures. Both cell lines became highly vacuolated within 16 h of combination PDT or an equivalent phototoxic dose from BPD alone. The vacuole periphery was labeled with a fluorescent probe for the endoplasmic reticulum (ER), and vacuole formation was prevented by presence of the protein synthesis inhibitor cycloheximide. These effects are characteristics of a caspase-independent death mode termed paraptosis previously associated with ER stress. These studies suggest that paraptosis may be a more frequent outcome of PDT than has hitherto been realized.
Collapse
Affiliation(s)
- David Kessel
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI
| | - John J Reiners
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI.,Institute of Environmental Health Sciences, Wayne State University, Detroit, MI
| |
Collapse
|
66
|
Saralamma VVG, Kim EH, Lee HJ, Raha S, Lee WS, Heo JD, Lee SJ, Won CK, Kim GS. Flavonoids: A new generation molecule to stimulate programmed cell deaths in cancer cells. ACTA ACUST UNITED AC 2017. [DOI: 10.12729/jbtr.2017.18.1.030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|