1
|
Gorska-Arcisz M, Popeda M, Braun M, Piasecka D, Nowak JI, Kitowska K, Stasilojc G, Okroj M, Romanska HM, Sadej R. FGFR2-triggered autophagy and activation of Nrf-2 reduce breast cancer cell response to anti-ER drugs. Cell Mol Biol Lett 2024; 29:71. [PMID: 38745155 PMCID: PMC11092031 DOI: 10.1186/s11658-024-00586-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/29/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Genetic abnormalities in the FGFR signalling occur in 40% of breast cancer (BCa) patients resistant to anti-ER therapy, which emphasizes the potential of FGFR-targeting strategies. Recent findings indicate that not only mutated FGFR is a driver of tumour progression but co-mutational landscapes and other markers should be also investigated. Autophagy has been recognized as one of the major mechanisms underlying the role of tumour microenvironment in promotion of cancer cell survival, and resistance to anti-ER drugs. The selective autophagy receptor p62/SQSTM1 promotes Nrf-2 activation by Keap1/Nrf-2 complex dissociation. Herein, we have analysed whether the negative effect of FGFR2 on BCa cell response to anti-ER treatment involves the autophagy process and/or p62/Keap1/Nrf-2 axis. METHODS The activity of autophagy in ER-positive MCF7 and T47D BCa cell lines was determined by analysis of expression level of autophagy markers (p62 and LC3B) and monitoring of autophagosomes' maturation. Western blot, qPCR and proximity ligation assay were used to determine the Keap1/Nrf-2 interaction and Nrf-2 activation. Analysis of 3D cell growth in Matrigel® was used to assess BCa cell response to applied treatments. In silico gene expression analysis was performed to determine FGFR2/Nrf-2 prognostic value. RESULTS We have found that FGFR2 signalling induced autophagy in AMPKα/ULK1-dependent manner. FGFR2 activity promoted dissociation of Keap1/Nrf-2 complex and activation of Nrf-2. Both, FGFR2-dependent autophagy and activation of Nrf-2 were found to counteract the effect of anti-ER drugs on BCa cell growth. Moreover, in silico analysis showed that high expression of NFE2L2 (gene encoding Nrf-2) combined with high FGFR2 expression was associated with poor relapse-free survival (RFS) of ER+ BCa patients. CONCLUSIONS This study revealed the unknown role of FGFR2 signalling in activation of autophagy and regulation of the p62/Keap1/Nrf-2 interdependence, which has a negative impact on the response of ER+ BCa cells to anti-ER therapies. The data from in silico analyses suggest that expression of Nrf-2 could act as a marker indicating potential benefits of implementation of anti-FGFR therapy in patients with ER+ BCa, in particular, when used in combination with anti-ER drugs.
Collapse
Affiliation(s)
- Monika Gorska-Arcisz
- Laboratory of Enzymology and Molecular Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland
| | - Marta Popeda
- Department of Pathomorphology, Medical University of Gdansk, Gdansk, Poland
| | - Marcin Braun
- Department of Pathology, Chair of Oncology, Medical University of Lodz, Pomorska 251, 92-213, Lodz, Poland
| | - Dominika Piasecka
- Laboratory of Enzymology and Molecular Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland
| | - Joanna I Nowak
- Department of Histology, Medical University of Gdansk, Gdansk, Poland
| | - Kamila Kitowska
- Laboratory of Enzymology and Molecular Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland
| | - Grzegorz Stasilojc
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Marcin Okroj
- Department of Cell Biology and Immunology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Hanna M Romanska
- Department of Pathology, Chair of Oncology, Medical University of Lodz, Pomorska 251, 92-213, Lodz, Poland.
| | - Rafal Sadej
- Laboratory of Enzymology and Molecular Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Debinki 1, 80-211, Gdansk, Poland.
| |
Collapse
|
2
|
Barzegar-Fallah A, Alimoradi H, Dunlop JL, Torbati E, Baird SK. Serotonin type-3 receptor antagonists selectively kill melanoma cells through classical apoptosis, microtubule depolymerisation, ERK activation, and NF-κB downregulation. Cell Biol Toxicol 2023; 39:1119-1135. [PMID: 34654991 DOI: 10.1007/s10565-021-09667-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 09/28/2021] [Indexed: 12/26/2022]
Abstract
Malignant melanoma is a highly metastatic tumour, resistant to treatment. Serotonin type-3 (5-HT3) receptor antagonists, such as tropisetron and ondansetron, are well-tolerated antiemetic drugs commonly used to prevent nausea caused by chemotherapy or radiotherapy. We investigated the anticancer effects of these drugs on melanoma cancer cell lines WM-266-4 and B16F10 with or without paclitaxel. We constructed IC50 curves and performed Chou-Talalay analysis, using data obtained with the MTT assay. Flow cytometry and fluorescent microscopy were used to examine characteristics of the cell cycle, cell death and cytoskeleton changes. Protein levels and activation were analysed by western blotting and molecular docking studies carried out. Data were analysed by one way ANOVA and post hoc testing. Ondansetron and tropisetron showed selective concentration-dependent cytotoxicity in melanoma cell lines WM-266-4 and B16F10. The effect in combination with paclitaxel was synergistic. The drugs did not cause cell cycle arrest but did promote characteristics of classical apoptosis, including accumulation of subG1 DNA, cleaved caspase-3, mitochondrial membrane permeability and phosphatidylserine exposure. As well, the cytosolic calcium level in the melanoma cells was enhanced, phosphorylated ERK1/2 induced and NF-κB inhibited. Finally, the formation of microtubules was shown to be impaired in melanoma cells treated with ondansetron or tropisetron. Docking studies were used to predict that these drugs could bind to the colchicine binding site on the tubulin molecule. Antiemetic drugs, already given in combination with chemotherapy, may enhance the cytotoxic effect of chemotherapy, following successful delivery to the tumour site.
Collapse
Affiliation(s)
- Anita Barzegar-Fallah
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Houman Alimoradi
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Jessica L Dunlop
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
| | - Elham Torbati
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand
- Department of Biochemistry, University of Otago, Dunedin, New Zealand
| | - Sarah K Baird
- Department of Pharmacology and Toxicology, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
3
|
Salihi A, Al-Naqshabandi MA, Khudhur ZO, Housein Z, Hama HA, Abdullah RM, Hussen BM, Alkasalias T. Gasotransmitters in the tumor microenvironment: Impacts on cancer chemotherapy (Review). Mol Med Rep 2022; 26:233. [PMID: 35616143 PMCID: PMC9178674 DOI: 10.3892/mmr.2022.12749] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/07/2022] [Indexed: 11/23/2022] Open
Abstract
Nitric oxide, carbon monoxide and hydrogen sulfide are three endogenous gasotransmitters that serve a role in regulating normal and pathological cellular activities. They can stimulate or inhibit cancer cell proliferation and invasion, as well as interfere with cancer cell responses to drug treatments. Understanding the molecular pathways governing the interactions between these gases and the tumor microenvironment can be utilized for the identification of a novel technique to disrupt cancer cell interactions and may contribute to the conception of effective and safe cancer therapy strategies. The present review discusses the effects of these gases in modulating the action of chemotherapies, as well as prospective pharmacological and therapeutic interfering approaches. A deeper knowledge of the mechanisms that underpin the cellular and pharmacological effects, as well as interactions, of each of the three gases could pave the way for therapeutic treatments and translational research.
Collapse
Affiliation(s)
- Abbas Salihi
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region 44001, Iraq
- Center of Research and Strategic Studies, Lebanese French University, Erbil, Kurdistan Region 44002, Iraq
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, SE-17165 Stockholm, Sweden
| | - Mohammed A. Al-Naqshabandi
- Department of Clinical Biochemistry, College of Health Sciences, Hawler Medical University, Erbil, Kurdistan Region 44001, Iraq
| | - Zhikal Omar Khudhur
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Kurdistan Region 44001, Iraq
| | - Zjwan Housein
- Department of Medical Laboratory Technology, Technical Health and Medical College, Erbil Polytechnique University, Erbil, Kurdistan Region 44002, Iraq
| | - Harmand A. Hama
- Department of Biology, Faculty of Education, Tishk International University, Erbil, Kurdistan Region 44002, Iraq
| | - Ramyar M. Abdullah
- College of Medicine, Hawler Medical University, Erbil, Kurdistan Region 44002, Iraq
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region 44002, Iraq
| | - Twana Alkasalias
- General Directorate of Scientific Research Center, Salahaddin University-Erbil, Erbil, Kurdistan Region 44002, Iraq
- Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, SE-17176 Stockholm, Sweden
| |
Collapse
|
4
|
Duan L, Calhoun S, Shim D, Perez RE, Blatter LA, Maki CG. Fatty acid oxidation and autophagy promote endoxifen resistance and counter the effect of AKT inhibition in ER-positive breast cancer cells. J Mol Cell Biol 2021; 13:433-444. [PMID: 33755174 PMCID: PMC8436705 DOI: 10.1093/jmcb/mjab018] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 02/22/2021] [Accepted: 02/24/2021] [Indexed: 11/24/2022] Open
Abstract
Tamoxifen (TAM) is the first-line endocrine therapy for estrogen receptor-positive (ER+) breast cancer (BC). However, acquired resistance occurs in ∼50% cases. Meanwhile, although the PI3K/AKT/mTOR pathway is a viable target for treatment of endocrine therapy-refractory patients, complex signaling feedback loops exist, which can counter the effectiveness of inhibitors of this pathway. Here, we analyzed signaling pathways and metabolism in ER+ MCF7 BC cell line and their TAM-resistant derivatives that are co-resistant to endoxifen using immunoblotting, quantitative polymerase chain reaction, and the Agilent Seahorse XF Analyzer. We found that activation of AKT and the energy-sensing kinase AMPK was increased in TAM and endoxifen-resistant cells. Furthermore, ERRα/PGC-1β and their target genes MCAD and CPT-1 were increased and regulated by AMPK, which coincided with increased fatty acid oxidation (FAO) and autophagy in TAM-resistant cells. Inhibition of AKT feedback-activates AMPK and ERRα/PGC-1β-MCAD/CPT-1 with a consequent increase in FAO and autophagy that counters the therapeutic effect of endoxifen and AKT inhibitors. Therefore, our results indicate increased activation of AKT and AMPK with metabolic reprogramming and increased autophagy in TAM-resistant cells. Simultaneous inhibition of AKT and FAO/autophagy is necessary to fully sensitize resistant cells to endoxifen.
Collapse
Affiliation(s)
- Lei Duan
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Sarah Calhoun
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Daeun Shim
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Ricardo E Perez
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| | - Lothar A Blatter
- Department of Molecular Biophysics and Physiology, Rush University Medical Center, Chicago, IL 60612, USA
| | - Carl G Maki
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
5
|
Cao W, Gao J, Zhang Y, Li A, Yu P, Cao N, Liang J, Tang X. Autophagy up-regulated by MEK/ERK promotes the repair of DNA damage caused by aflatoxin B1. Toxicol Mech Methods 2021; 32:87-96. [PMID: 34396909 DOI: 10.1080/15376516.2021.1968985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Aflatoxin B1 (AFB1), a kind of mycotoxin, exerts its cytotoxicity by increasing the oxidative damage of target organs, especially the liver. In vivo and in vitro experiments were carried out to elucidate the toxic mechanism of AFB1. The results of MTT, cloning-formation, flow cytometry, immunocytochemistry, Reverse transcription PCR (RT-PCR) and western blot showed that AFB1 activated NOX2 gp91 phox, inhibited proliferation and migration, and blocked cell cycle at G0/G1 period of HHL-5 cells. Autophagy promoted the repair of NOX2-dependent DNA damage. NOX2/gp91 phox mainly activates MEK/ERK pathway and then up-regulates autophagy. In vivo experiments have shown that AFB1 (0.75 mg/kg daily orally, 4 weeks) had no significant changes in the size and shape of the liver in mice. However, these treatments lead to structural abnormalities of hepatocytes and DNA damage. In summary, AFB1 caused intracellular oxidative stress and DNA damage, NOX2/gp91-phox activates the MEK/ERK pathway, and upregulated autophagy to promote the repair of DNA damage. We concluded that by increasing the level of autophagy, the ability of anti-AFB1 toxicity of liver can be increased.
Collapse
Affiliation(s)
- Weiya Cao
- Medical School, Anhui University of Science and Technology, Huainan, China.,Institute of Environment-friendly Materials and Occupational Health, Anhui University of Science and Technology, Wuhu, China
| | - Jiafeng Gao
- Medical School, Anhui University of Science and Technology, Huainan, China.,Institute of Environment-friendly Materials and Occupational Health, Anhui University of Science and Technology, Wuhu, China
| | - Yinci Zhang
- Medical School, Anhui University of Science and Technology, Huainan, China.,Institute of Environment-friendly Materials and Occupational Health, Anhui University of Science and Technology, Wuhu, China
| | - Amin Li
- Medical School, Anhui University of Science and Technology, Huainan, China.,Institute of Environment-friendly Materials and Occupational Health, Anhui University of Science and Technology, Wuhu, China
| | - Pan Yu
- Medical School, Anhui University of Science and Technology, Huainan, China.,Institute of Environment-friendly Materials and Occupational Health, Anhui University of Science and Technology, Wuhu, China
| | - Niandie Cao
- Medical School, Anhui University of Science and Technology, Huainan, China.,Institute of Environment-friendly Materials and Occupational Health, Anhui University of Science and Technology, Wuhu, China
| | - Jiaojiao Liang
- Medical School, Anhui University of Science and Technology, Huainan, China.,Institute of Environment-friendly Materials and Occupational Health, Anhui University of Science and Technology, Wuhu, China
| | - Xiaolong Tang
- Medical School, Anhui University of Science and Technology, Huainan, China.,Institute of Environment-friendly Materials and Occupational Health, Anhui University of Science and Technology, Wuhu, China
| |
Collapse
|
6
|
da Silva MACN, Costa JH, Pacheco-Fill T, Ruiz ALTG, Vidal FCB, Borges KRA, Guimarães SJA, de Azevedo-Santos APS, Buglio KE, Foglio MA, Barbosa MDCL, Nascimento MDDSB, de Carvalho JE. Açai ( Euterpe oleracea Mart.) Seed Extract Induces ROS Production and Cell Death in MCF-7 Breast Cancer Cell Line. Molecules 2021; 26:molecules26123546. [PMID: 34200718 PMCID: PMC8230419 DOI: 10.3390/molecules26123546] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/06/2021] [Accepted: 06/07/2021] [Indexed: 01/11/2023] Open
Abstract
Euterpe oleracea Mart. (açai) is a native palm from the Amazon region. There are various chemical constituents of açai with bioactive properties. This study aimed to evaluate the chemical composition and cytotoxic effects of açai seed extract on breast cancer cell line (MCF-7). Global Natural Products Social Molecular Networking (GNPS) was applied to identify chemical compounds present in açai seed extract. LC-MS/MS and molecular networking were employed to detect the phenolic compounds of açai. The antioxidant activity of açai seed extract was measured by DPPH assay. MCF-7 breast cancer cell line viability was evaluated by MTT assay. Cell death was evaluated by flow cytometry and time-lapse microscopy. Autophagy was evaluated by orange acridin immunofluorescence assay. Reactive oxygen species (ROS) production was evaluated by DAF assay. From the molecular networking, fifteen compounds were identified, mainly phenolic compounds. The açai seed extract showed cytotoxic effects against MCF-7, induced morphologic changes in the cell line by autophagy and increased the ROS production pathway. The present study suggests that açai seed extract has a high cytotoxic capacity and may induce autophagy by increasing ROS production in breast cancer. Apart from its antioxidant activity, flavonoids with high radical scavenging activity present in açai also generated NO (nitric oxide), contributing to its cytotoxic effect and autophagy induction.
Collapse
Affiliation(s)
- Marcos Antonio Custódio Neto da Silva
- Post-Graduate Program in Internal Medicine, Faculty of Medical Science, Universidade Estadual de Campinas, Rua Tessália Vieira de Camargo, 126, Cidade Universitária Zeferino Vaz. CEP, Campinas 13083-887, SP, Brazil;
| | - Jonas Henrique Costa
- Institute of Chemistry, Universidade Estadual de Campinas, CP 6154, Campinas 13083-970, SP, Brazil; (J.H.C.); (T.P.-F.)
| | - Taícia Pacheco-Fill
- Institute of Chemistry, Universidade Estadual de Campinas, CP 6154, Campinas 13083-970, SP, Brazil; (J.H.C.); (T.P.-F.)
| | - Ana Lúcia Tasca Gois Ruiz
- Faculty of Pharmaceutical Sciences, Universidade Estadual de Campinas, Campinas 13083-859, SP, Brazil; (A.L.T.G.R.); (K.E.B.); (M.A.F.)
| | - Flávia Castello Branco Vidal
- Post-Graduate Program in Adult Heath, Department of Patology, Federal University of Maranhão (UFMA), São Luís 65080-805, MA, Brazil; (F.C.B.V.); (K.R.A.B.)
| | - Kátia Regina Assunção Borges
- Post-Graduate Program in Adult Heath, Department of Patology, Federal University of Maranhão (UFMA), São Luís 65080-805, MA, Brazil; (F.C.B.V.); (K.R.A.B.)
| | - Sulayne Janaina Araújo Guimarães
- Post-Graduate Program in Health Sicencies, Federal University of Maranhão (UFMA), São Luís 65080-805, MA, Brazil; (S.J.A.G.); (A.P.S.d.A.-S.)
| | - Ana Paula Silva de Azevedo-Santos
- Post-Graduate Program in Health Sicencies, Federal University of Maranhão (UFMA), São Luís 65080-805, MA, Brazil; (S.J.A.G.); (A.P.S.d.A.-S.)
| | - Kaio Eduardo Buglio
- Faculty of Pharmaceutical Sciences, Universidade Estadual de Campinas, Campinas 13083-859, SP, Brazil; (A.L.T.G.R.); (K.E.B.); (M.A.F.)
| | - Mary Ann Foglio
- Faculty of Pharmaceutical Sciences, Universidade Estadual de Campinas, Campinas 13083-859, SP, Brazil; (A.L.T.G.R.); (K.E.B.); (M.A.F.)
| | - Maria do Carmo Lacerda Barbosa
- Post-Graduate Program in Family Health, Department of Medicine I, Federal University of Maranhão (UFMA), São Luís 65080-805, MA, Brazil;
| | - Maria do Desterro Soares Brandão Nascimento
- Post-Graduate Program in Adult Heath, Department of Patology, Federal University of Maranhão (UFMA), São Luís 65080-805, MA, Brazil; (F.C.B.V.); (K.R.A.B.)
- Correspondence: (M.d.D.S.B.N.); (J.E.d.C.)
| | - João Ernesto de Carvalho
- Faculty of Pharmaceutical Sciences, Universidade Estadual de Campinas, Campinas 13083-859, SP, Brazil; (A.L.T.G.R.); (K.E.B.); (M.A.F.)
- Correspondence: (M.d.D.S.B.N.); (J.E.d.C.)
| |
Collapse
|
7
|
Kuo EY, Chang HL, Lin ST, Lee TM. High Light-Induced Nitric Oxide Production Induces Autophagy and Cell Death in Chlamydomonas reinhardtii. FRONTIERS IN PLANT SCIENCE 2020; 11:772. [PMID: 32587598 PMCID: PMC7298128 DOI: 10.3389/fpls.2020.00772] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/15/2020] [Indexed: 05/05/2023]
Abstract
Autophagy plays a role in regulating important cellular functions in response to stress conditions. The role of nitric oxide (NO) in the regulation of autophagy in Chlamydomonas reinhardtii has been not studied. Illumination of C. reinhardtii cells under a high light (HL, 1,600 μmol m-2 s-1) condition induced a NO burst through NO synthase- and nitrate reductase-independent routes, and cell death. The abundance of CrATG8 protein, an autophagy marker of C. reinhardtii, increased after HL illumination along with a linear increase in the transcript abundance of autophagy-associated genes (CrVPS34, CrATG1, CrATG3, CrATG4, CrATG6, CrATG7, CrATG8, and CrATG12), which were suppressed in the presence of an NO scavenger, 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide (cPTIO). The cells were treated with NO donors, S-nitroso-N-acetyl-penicillamine, and S-nitrosoglutathione, under a normal light (50 μmol m-2 s-1) condition to elucidate the role of NO in autophagy activation and cell death. Treatment with 0.05 mM or 0.1 mM NO donors increased the abundance of ATG8 protein and CrATG transcripts, which were suppressed in the presence of cPTIO. Moreover, treatment with 0.05 mM NO donors did not affect cell viability, while 0.1 mM NO donors elicited a transient decrease in cell growth and death that recovered after 12 h. The transient effect could be prevented by the presence of cPTIO. However, treatment with 1 mM H2O2 and 0.1 mM NO donors enhanced autophagy induction and resulted in cell death after 24 h. The interaction of H2O2 and NO can be prevented by cPTIO treatment. This implies that NO is critical for the interaction of H2O2 and NO that induces cell death and autophagy. Furthermore, exposure to 0.1 mM NO donors under a non-lethal HL condition (750 μmol m-2 s-1) evoked autophagy and cell death. In conclusion, the present findings demonstrated that the NO-mediated autophagy pathway is activated in C. reinhardtii under lethal high intensity illumination and may interact with H2O2 for HL-induced cell death. The relationships between autophagy and cell death are discussed.
Collapse
Affiliation(s)
- Eva YuHua Kuo
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Hsueh-Ling Chang
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Shu-Tseng Lin
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Tse-Min Lee
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
- Doctoral Degree Program in Marine Biotechnology, National Sun Yat-sen University, Kaohsiung, Taiwan
- *Correspondence: Tse-Min Lee,
| |
Collapse
|
8
|
Tamoxifen citrate/Coenzyme Q10 as smart nanocarriers Bitherapy for Breast Cancer: Cytotoxicity, genotoxicity, and antioxidant activity. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.02.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
9
|
Duan L, Perez RE, Lai X, Chen L, Maki CG. The histone demethylase JMJD2B is critical for p53-mediated autophagy and survival in Nutlin-treated cancer cells. J Biol Chem 2019; 294:9186-9197. [PMID: 31036564 DOI: 10.1074/jbc.ra118.007122] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/24/2019] [Indexed: 01/27/2023] Open
Abstract
Autophagy promotes cancer cell survival in response to p53 activation by the anticancer agent Nutlin-3a (Nutlin). We reported previously that Nutlin kills MDM2-amplified cancer cells and that this killing is associated with an inhibition of glucose metabolism, reduced α-ketoglutarate (α-KG) levels, and reduced autophagy. In the current report, using SJSA1, U2OS, A549, and MHM cells, we found that Nutlin alters histone methylation in an MDM2 proto-oncogene-dependent manner and that this, in turn, regulates autophagy-related gene (ATG) expression and cell death. In MDM2-amplified cells, Nutlin increased histone (H) 3 lysine (K) 9 and K36 trimethylation (me3) coincident with reduced autophagy and increased apoptosis. Blocking histone methylation restored autophagy and rescued these cells from Nutlin-induced killing. In MDM2-nonamplified cells, H3K9me3 and H3K36me3 levels were either reduced or not changed by the Nutlin treatment, and this coincided with increased autophagy and cell survival. Blocking histone demethylation reduced autophagy and sensitized these cells to Nutlin-induced killing. Further experiments suggested that MDM2 amplification increases histone methylation in Nutlin-treated cells by causing depletion of the histone demethylase Jumonji domain-containing protein 2B (JMJD2B). Finally, JMJD2B knockdown or inhibition increased H3K9/K36me3 levels, decreased ATG gene expression and autophagy, and sensitized MDM2-nonamplified cells to apoptosis. Together, these results support a model in which MDM2- and JMJD2B-regulated histone methylation levels modulate ATG gene expression, autophagy, and cell fate in response to the MDM2 antagonist Nutlin-3a.
Collapse
Affiliation(s)
- Lei Duan
- From the Department of Cell & Molecular Medicine, Rush University Medical Center, Chicago, Illinois 60612
| | - Ricardo E Perez
- From the Department of Cell & Molecular Medicine, Rush University Medical Center, Chicago, Illinois 60612
| | - Xin Lai
- Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China, and
| | - Ling Chen
- Department of Laboratory Medicine, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei, China 442000
| | - Carl G Maki
- From the Department of Cell & Molecular Medicine, Rush University Medical Center, Chicago, Illinois 60612,
| |
Collapse
|
10
|
Duan L, Perez RE, Maki CG. Alpha ketoglutarate levels, regulated by p53 and OGDH, determine autophagy and cell fate/apoptosis in response to Nutlin-3a. Cancer Biol Ther 2018; 20:252-260. [PMID: 30289354 PMCID: PMC6370392 DOI: 10.1080/15384047.2018.1523858] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 08/26/2018] [Indexed: 02/06/2023] Open
Abstract
Activated p53 can promote apoptosis or cell cycle arrest. Differences in energy metabolism can influence cell fate in response to activated p53. Nutlin-3a is a preclinical drug and small molecule activator of p53. Alpha-ketoglutarate (αKG) levels were reduced in cells sensitive to Nutlin-3a-induced apoptosis and increased in cells resistant to this apoptosis. Add-back of a cell-permeable αKG analog (DMKG) rescued cells from apoptosis in response to Nutlin-3a. OGDH is a component of the αKGDH complex that converts αKG to succinate. OGDH knockdown increased endogenous αKG levels and also rescued cells from Nutlin-3a-induced apoptosis. We previously showed reduced autophagy and ATG gene expression contributes to Nutlin-3a-induced apoptosis. DMKG and OGDH knockdown restored autophagy and ATG gene expression in Nutlin-3a-treated cells. These studies indicate αKG levels, regulated by p53 and OGDH, determine autophagy and apoptosis in response to Nutlin-3a.
Collapse
Affiliation(s)
- Lei Duan
- a Department of Cell & Molecular Medicine , Rush University Medical Center , Chicago , IL , USA
| | - Ricardo E Perez
- a Department of Cell & Molecular Medicine , Rush University Medical Center , Chicago , IL , USA
| | - Carl G Maki
- a Department of Cell & Molecular Medicine , Rush University Medical Center , Chicago , IL , USA
| |
Collapse
|
11
|
Abstract
Macroautophagy/autophagy is vital for intracellular quality control and homeostasis. Therefore, careful regulation of autophagy is very important. In the past 10 years, a number of studies have reported that estrogenic effectors affect autophagy. However, some results, especially those regarding the modulatory effect of 17β-estradiol (E2) on autophagy seem inconsistent. Moreover, several clinical trials are already in place combining both autophagy inducers and autophagy inhibitors with endocrine therapies for breast cancer. Not all patients experience benefit, which further confuses and complicates our understanding of the main effects of autophagy in estrogen-related cancer. In view of the importance of the crosstalk between estrogen signaling and autophagy, this review summarizes the estrogenic effectors reported to affect autophagy, subcellular distribution and translocation of estrogen receptors, autophagy-targeted transcription factors (TFs), miRNAs, and histone modifications regulated by E2. Upon stimulation with estrogen, there will always be opposing functional actions, which might occur between different receptors, receptors on TFs, TFs on autophagy genes, or even histone modifications on transcription. The huge signaling network downstream of estrogen can promote autophagy and reduce overstimulated autophagy at the same time, which allows autophagy to be regulated by estrogen in a restricted range. To help understand how the estrogenic regulation of autophagy affects cell fate, a hypothetical model is presented here. Finally, we discuss some exciting new directions in the field. We hope this might help to better understand the multiple associations between estrogen and autophagy, the pathogenic mechanisms of many estrogen-related diseases, and to design novel and efficacious therapeutics. Abbreviations: AP-1, activator protein-1; HATs, histone acetyltransferases; HDAC, histone deacetylases; HOTAIR, HOX transcript antisense RNA.
Collapse
Affiliation(s)
- Jin Xiang
- a Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences , Wuhan University , Wuhan , PR China
| | - Xiang Liu
- a Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences , Wuhan University , Wuhan , PR China
| | - Jing Ren
- a Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences , Wuhan University , Wuhan , PR China
| | - Kun Chen
- a Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences , Wuhan University , Wuhan , PR China
| | - Hong-Lu Wang
- a Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences , Wuhan University , Wuhan , PR China
| | - Yu-Yang Miao
- a Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences , Wuhan University , Wuhan , PR China
| | - Miao-Miao Qi
- a Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Ministry of Education, School of Pharmaceutical Sciences , Wuhan University , Wuhan , PR China
| |
Collapse
|
12
|
Rodenak-Kladniew B, Castro A, Stärkel P, De Saeger C, García de Bravo M, Crespo R. Linalool induces cell cycle arrest and apoptosis in HepG2 cells through oxidative stress generation and modulation of Ras/MAPK and Akt/mTOR pathways. Life Sci 2018; 199:48-59. [PMID: 29510199 DOI: 10.1016/j.lfs.2018.03.006] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 02/20/2018] [Accepted: 03/02/2018] [Indexed: 12/11/2022]
Abstract
AIMS Linalool is a plant-derived monoterpene with anticancer activity, however its mechanisms of action remain poorly understood. The aim of this work was to elucidate the anticancer mechanisms of action of linalool in hepatocellular carcinoma (HCC) HepG2 cells. MAIN METHODS Cell viability and proliferation were determined by WST-1 assay and BrdU incorporation, respectively. Cell cycle analysis was assessed through flow cytometry (FC) and western blot (WB). Apoptosis was determined by caspase-3 activity, TUNEL assay and WB. Reactive oxygen species (ROS) and mitochondrial membrane potential (MMP) were analyzed by FC and fluorescence microscopy. Expression of Ras, MAPKs (ERK, JNK and p38) and Akt/mTOR pathways were evaluated by WB. KEY FINDINGS Linalool (0-2.5 mM) dose-dependently inhibited cell proliferation by inducing G0/G1 cell cycle arrest, through Cdk4 and cyclin A downregulation, p21 and p27 upregulation, and apoptosis, characterized by MMP loss, caspase-3 activation, PARP cleavage and DNA fragmentation. Low concentrations of linalool (1.0 mM) reduced membrane-bound Ras and Akt activity whereas higher amounts (2.0 mM) triggered mTOR inhibition and ROS generation, in correlation with MAPKs activation and Akt phosphorylation. ROS scavenger N-acetyl-L-cysteine partially rescued HepG2 cell growth and prevented MPP depolarization, ERK and JNK activation. Moreover, specific ERK and Akt phosphorylation inhibitors potentiated linalool anti-cancer activity, pointing Akt and ERK activation as pro-survival mechanisms in response to higher concentrations of linalool. SIGNIFICANCE This report reveals that linalool induces G0/G1 arrest and apoptosis in HepG2 cells involving Ras, MAPKs and Akt/mTOR pathways and suggests that linalool is a promising anticancer agent for HCC therapy.
Collapse
Affiliation(s)
- Boris Rodenak-Kladniew
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CONICET-UNLP, CCT-La Plata, Facultad de Ciencias Médicas, La Plata, Argentina
| | - Agustina Castro
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CONICET-UNLP, CCT-La Plata, Facultad de Ciencias Médicas, La Plata, Argentina
| | - Peter Stärkel
- Laboratory of Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium; Department of Gastroenterology, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Christine De Saeger
- Laboratory of Gastroenterology, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Margarita García de Bravo
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CONICET-UNLP, CCT-La Plata, Facultad de Ciencias Médicas, La Plata, Argentina
| | - Rosana Crespo
- Instituto de Investigaciones Bioquímicas de La Plata (INIBIOLP), CONICET-UNLP, CCT-La Plata, Facultad de Ciencias Médicas, La Plata, Argentina.
| |
Collapse
|
13
|
Shafran Y, Zurgil N, Ravid-Hermesh O, Sobolev M, Afrimzon E, Hakuk Y, Shainberg A, Deutsch M. Nitric oxide is cytoprotective to breast cancer spheroids vulnerable to estrogen-induced apoptosis. Oncotarget 2017; 8:108890-108911. [PMID: 29312577 PMCID: PMC5752490 DOI: 10.18632/oncotarget.21610] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 08/17/2017] [Indexed: 12/24/2022] Open
Abstract
Estrogen-induced apoptosis has become a successful treatment for postmenopausal metastatic, estrogen receptor-positive breast cancer. Nitric oxide involvement in the response to this endocrine treatment and its influence upon estrogen receptor-positive breast cancer progression is still unclear. Nitric oxide impact on the MCF7 breast cancer line, before and after estrogen-induced apoptosis, was investigated in 3D culture systems using unique live-cell imaging methodologies. Spheroids were established from MCF7 cells vulnerable to estrogen-induced apoptosis, before and after exposure to estrogen. Spheroids derived from estrogen-treated cells exhibited extensive apoptosis levels with downregulation of estrogen receptor expression, low proliferation rate and reduced metabolic activity, unlike spheroids derived from non-treated cells. In addition to basic phenotypic differences, these two cell cluster types are diverse in their reactions to exogenous nitric oxide. A dual effect of nitric oxide was observed in the breast cancer phenotype sensitive to estrogen-induced apoptosis. Nitric oxide, at the nanomolar level, induced cell proliferation, high metabolic activity, downregulation of estrogen receptor and enhanced collective invasion, contributing to a more aggressive phenotype. Following hormone supplementation, breast cancer 3D clusters were rescued from estrogen-induced apoptosis by these low nitric oxide-donor concentrations, since nitric oxide attenuates cell death levels, upregulates survivin expression and increases metabolic activity. Higher nitric oxide concentrations (100nM) inhibited cell growth, metabolism and promoted apoptosis. These results suggest that nitric oxide, in nanomolar concentrations, may inhibit estrogen-induced apoptosis, playing a major role in hormonal therapy. Inhibiting nitric oxide activity may benefit breast cancer patients and ultimately reduce tumor recurrence.
Collapse
Affiliation(s)
- Yana Shafran
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar Ilan University, Ramat Gan 52900, Israel
| | - Naomi Zurgil
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar Ilan University, Ramat Gan 52900, Israel
| | - Orit Ravid-Hermesh
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar Ilan University, Ramat Gan 52900, Israel
| | - Maria Sobolev
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar Ilan University, Ramat Gan 52900, Israel
| | - Elena Afrimzon
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar Ilan University, Ramat Gan 52900, Israel
| | - Yaron Hakuk
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar Ilan University, Ramat Gan 52900, Israel
| | - Asher Shainberg
- The Mina and Everard Goodman Faculty of Life Sciences, Bar Ilan University, Ramat Gan 52900, Israel
| | - Mordechai Deutsch
- The Biophysical Interdisciplinary Jerome Schottenstein Center for the Research and Technology of the Cellome, Physics Department, Bar Ilan University, Ramat Gan 52900, Israel
| |
Collapse
|
14
|
Tandon M, Othman AH, Ashok V, Stein GS, Pratap J. The role of Runx2 in facilitating autophagy in metastatic breast cancer cells. J Cell Physiol 2017; 233:559-571. [PMID: 28345763 DOI: 10.1002/jcp.25916] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 03/17/2017] [Indexed: 02/06/2023]
Abstract
Breast cancer metastases cause significant patient mortality. During metastases, cancer cells use autophagy, a catabolic process to recycle nutrients via lysosomal degradation, to overcome nutritional stress for their survival. The Runt-related transcription factor, Runx2, promotes cell survival under metabolic stress, and regulates breast cancer progression and bone metastases. Here, we identify that Runx2 enhances autophagy in metastatic breast cancer cells. We defined Runx2 function in cellular autophagy by monitoring microtubule-associated protein light chain (LC3B-II) levels, an autophagy-specific marker. The electron and confocal microscopic analyses were utilized to identify alterations in autophagic vesicles. The Runx2 knockdown cells accumulate LC3B-II protein and autophagic vesicles due to reduced turnover. Interestingly, Runx2 promotes autophagy by enhancing trafficking of LC3B vesicles. Our mechanistic studies revealed that Runx2 promotes autophagy by increasing acetylation of α-tubulin sub-units of microtubules. Inhibiting autophagy decreased cell adhesion and survival of Runx2 knockdown cells. Furthermore, analysis of LC3B protein in clinical breast cancer specimens and tumor xenografts revealed significant association between high Runx2 and low LC3B protein levels. Our studies reveal a novel regulatory mechanism of autophagy via Runx2 and provide molecular insights into the role of autophagy in metastatic cancer cells.
Collapse
Affiliation(s)
- Manish Tandon
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, Illinois
| | - Ahmad H Othman
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, Illinois
| | - Vivek Ashok
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, Illinois
| | - Gary S Stein
- University of Vermont Cancer Center and Department of Biochemistry, Larner College of Medicine, University of Vermont, Burlington, Vermont
| | - Jitesh Pratap
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, Illinois
| |
Collapse
|
15
|
Liu M, Li R, Tang Y, Chang J, Han R, Zhang S, Jiang N, Ma F. New applications of the acridine orange fluorescence staining method: Screening for circulating tumor cells. Oncol Lett 2017; 13:2221-2229. [PMID: 28454384 PMCID: PMC5403171 DOI: 10.3892/ol.2017.5724] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Accepted: 11/24/2016] [Indexed: 12/14/2022] Open
Abstract
The aim of the present study was to explore use of the acridine orange fluorescence (AO-F) staining method for screening of circulating tumor cells (CTCs) in renal cell carcinoma (RCC) patients. The AO-F positive staining rate of live and dead tumor cells was calculated. The positive staining rate in the live group was 93.4±3.0%, while the dead group failed to emit specific fluorescence. A known number of tumor cells were added to peripheral blood, and the detection sensitivity of the four groups (50, 100, 200 and 500 cells/tube) was 10.2±3.8, 9.2±2.3, 10.8±2.6 and 10.5±1.9%, respectively. The average detection sensitivity of the four groups was 10.16±2.73%. There was a positive correlation between the number of cells that was positively stained with AO-F and the total number cells in the system (χ2=0.959; P<0.001). Subsequently, the AO-F staining method was used to detect positive staining cells in 8 healthy volunteers (control group), and 112 non-metastatic and 27 metastatic RCC patients. The positive staining rate was 13.67% (19/139) in RCC patients, while none of the control group was positive. The AO-F positive staining rate was not significantly different between the metastatic and non-metastatic patients according to age, gender, the pathological pattern, T2/3 (according to the Tumor-Node-Metastasis classification) or Fuhrman grade, while there was a significant difference according to T1. The positive staining rate was 8.93% (10/112) for non-metastatic patients and 33.33% (9/27) for metastatic patients, which showed a significant difference (P<0.05). In 112 non-metastatic and 27 metastatic patients, the positive staining rate was not significantly associated with gender, age, tumor size, the pathological pattern, T classification, Fuhrman grade, the presence of a lesion or metastasis to the lungs. The present study demonstrated that the method of CTC staining with AO-F, which has high reproducibility and specificity, was feasible for identifying CTCs and warrants further study.
Collapse
Affiliation(s)
- Min Liu
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Ruizhe Li
- Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Yang Tang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Jiwu Chang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Rong Han
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Shumin Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Ning Jiang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| | - Fuling Ma
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, P.R. China
| |
Collapse
|
16
|
Feng J, Chen X, Shen J. Reactive nitrogen species as therapeutic targets for autophagy: implication for ischemic stroke. Expert Opin Ther Targets 2017; 21:305-317. [DOI: 10.1080/14728222.2017.1281250] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Jinghan Feng
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), Shenzhen, China
| | - Xingmiao Chen
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), Shenzhen, China
| | - Jiangang Shen
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), Shenzhen, China
| |
Collapse
|
17
|
Lewinska A, Adamczyk-Grochala J, Kwasniewicz E, Deregowska A, Wnuk M. Diosmin-induced senescence, apoptosis and autophagy in breast cancer cells of different p53 status and ERK activity. Toxicol Lett 2016; 265:117-130. [PMID: 27890807 DOI: 10.1016/j.toxlet.2016.11.018] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 11/23/2016] [Accepted: 11/24/2016] [Indexed: 11/17/2022]
Abstract
Relatively low bioavailability of plant-derived nutraceuticals with anticancer properties may limit their usefulness for prevention and therapy of cancer. In the present study, we have screened for nutraceuticals (n=30) that would act at low micromolar range against phenotypically distinct breast cancer cell lines, namely MCF-7 (ER+, PR+/-, HER2-), MDA-MB-231 (ER-, PR-, HER2-) and SK-BR-3 (ER-, PR-, HER2+), and diosmin, a citrus fruit flavonoid belonging to a flavone subclass, was selected. MCF-7 cell line was found to be the most sensitive to diosmin treatment. Diosmin caused G2/M cell cycle arrest, elevation in p53, p21 and p27 levels and stress-induced premature senescence when used at lower concentrations (5 and 10μM). Diosmin (20μM) also promoted apoptosis that was not observed in normal human mammary epithelial cells (HMEC). Diosmin stimulated oxidative and nitrosative stress, DNA damage and changes in global DNA methylation patterns. The status of p53 (wild type versus mutant) and the levels of phosphorylated ERK1/2 in a steady state, and diosmin-induced autophagy may reflect diverse response to diosmin treatment in MCF-7, MDA-MB-231 and SK-BR-3 cells, which in turn results in different cell fates. Taken together, diosmin that acts at low micromolar range against breast cancer cells may be considered as a promising candidate for anticancer therapy.
Collapse
Affiliation(s)
- Anna Lewinska
- Department of Genetics, University of Rzeszow, Werynia 502, 36-100 Kolbuszowa, Poland.
| | | | - Ewa Kwasniewicz
- Department of Genetics, University of Rzeszow, Werynia 502, 36-100 Kolbuszowa, Poland
| | - Anna Deregowska
- Department of Genetics, University of Rzeszow, Werynia 502, 36-100 Kolbuszowa, Poland; Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Maciej Wnuk
- Department of Genetics, University of Rzeszow, Werynia 502, 36-100 Kolbuszowa, Poland
| |
Collapse
|
18
|
p53-regulated autophagy is controlled by glycolysis and determines cell fate. Oncotarget 2016; 6:23135-56. [PMID: 26337205 PMCID: PMC4695109 DOI: 10.18632/oncotarget.5218] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 08/07/2015] [Indexed: 12/15/2022] Open
Abstract
The tumor suppressor p53 regulates downstream targets that determine cell fate. Canonical p53 functions include inducing apoptosis, growth arrest, and senescence. Non-canonical p53 functions include its ability to promote or inhibit autophagy and its ability to regulate metabolism. The extent to which autophagy and/or metabolic regulation determines cell fate by p53 is unclear. To address this, we compared cells resistant or sensitive to apoptosis by the p53 activator Nutlin-3a. In resistant cells, glycolysis was maintained upon Nutlin-3a treatment, and activated p53 promoted prosurvival autophagy. In contrast, in apoptosis sensitive cells activated p53 increased superoxide levels and inhibited glycolysis through repression of glycolytic pathway genes. Glycolysis inhibition and increased superoxide inhibited autophagy by repressing ATG genes essential for autophagic vesicle maturation. Inhibiting glycolysis increased superoxide and blocked autophagy in apoptosis-resistant cells, causing p62-dependent caspase-8 activation. Finally, treatment with 2-DG or the autophagy inhibitors chloroquine or bafilomycin A1 sensitized resistant cells to Nutlin-3a-induced apoptosis. Together, these findings reveal novel links between glycolysis and autophagy that determine apoptosis-sensitivity in response to p53. Specifically, the findings indicate 1) that glycolysis plays an essential role in autophagy by limiting superoxide levels and maintaining expression of ATG genes required for autophagic vesicle maturation, 2) that p53 can promote or inhibit autophagy depending on the status of glycolysis, and 3) that inhibiting protective autophagy can expand the breadth of cells susceptible to Nutlin-3a induced apoptosis.
Collapse
|
19
|
Ganji-Harsini S, Khazaei M, Rashidi Z, Ghanbari A. Thymoquinone Could Increase The Efficacy of Tamoxifen Induced Apoptosis in Human Breast Cancer Cells: An In Vitro Study. CELL JOURNAL 2016; 18:245-54. [PMID: 27540530 PMCID: PMC4988424 DOI: 10.22074/cellj.2016.4320] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 11/24/2015] [Indexed: 12/16/2022]
Abstract
Objective Thymoquinone (TQ), as the main component of Nigella Sativa plant, shows anticancer properties. This study was aimed to evaluate the combined effect of TQ and Tamoxifen
(TAM) on viability and apoptosis of human breast cancer cell lines.
Materials and Methods In this experimental study, estrogen positive MCF-7 and estrogen
negative MDA-MB-231 human breast cancer cell lines were induced by TAM (2 µM) or different doses of TQ (50, 75, 100, 150 µM), individually or in combination. Cell viability and
apoptosis were investigated by MTT assay and TdT-mediated deoxy-uracil nick end labeling
(TUNEL) assay; Acridine orange (AO)/Ethidium bromide (EB) staining respectively. Data
were analyzed by one way ANOVA and P<0.05 was considered significant.
Results In 24 hours treatment, TAM and all doses of TQ, solely or in combination,
significantly reduced cell viability of both cell lines, except in MCF-7 cells treated with 50 µM TQ,
and MDA-MB-231 cells treated with 50 or 75 µM TQ (P<0.01). After 48 hours treatment,
cell viability of both cell lines was reduced in all treated groups (P<0.05). Remarkable apoptotic index was observed in combination treatment of MCF-7 or MDA-MB-231 cell lines
with TAM and TQ (P<0.001). Conclusion The synergistic effect of TQ and TAM on human breast cancer cell lines
showed cell viability reduction as well as apoptosis induction, independent to estrogen.
Collapse
Affiliation(s)
- Sedigheh Ganji-Harsini
- Fertility and Infertility Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zahra Rashidi
- Fertility and Infertility Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ali Ghanbari
- Fertility and Infertility Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
20
|
Enhanced nitric oxide-mediated autophagy contributes to the hepatoprotective effects of ischemic preconditioning during ischemia and reperfusion. Nitric Oxide 2016; 58:10-9. [PMID: 27246638 DOI: 10.1016/j.niox.2016.05.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 05/25/2016] [Accepted: 05/27/2016] [Indexed: 12/19/2022]
Abstract
Ischemic preconditioning (IPC) protects against liver ischemia/reperfusion (I/R) injury. Autophagy is an essential cytoprotective system that is rapidly activated by multiple stressors. Nitric oxide (NO) acts as an inducer of IPC. We examined the impact of autophagy in liver IPC and its regulation by NO. Male C57BL/6 mice were subjected to 60 min of hepatic ischemia followed by 6 h of reperfusion. IPC was achieved for 10 min of ischemia followed by 10 min of reperfusion prior to sustained ischemia. N(ω)-Nitro-l-arginine methyl ester (L-NAME, 15 mg/kg, i.v., all NOS inhibitor) and aminoguanidine (AG, 10 mg/kg, i.v., iNOS inhibitor) were injected 10 min before IPC. SB203580 (10 mg/kg, i.p., p38 inhibitor) was injected 30 min before IPC. I/R increased serum alanine aminotransferase activity. IPC attenuated this increase, which was abolished by L-NAME, but not AG. Microtubule-associated protein-1 light chain 3-II levels increased and p62 protein levels decreased after I/R; these changes were augmented by IPC and abolished by L-NAME. I/R increased liver protein expression of autophagy-related protein (Atg)12-Atg5 complex and lysosome-associated membrane protein-2. IPC augmented the expression of these proteins, which were abolished by L-NAME, but not AG. IPC also augmented the level of phosphorylated p38 MAPK induced by I/R and this phosphorylation was abolished by L-NAME. Our findings suggest that IPC-mediated NO protects against I/R-induced liver injury by enhancing autophagic flux.
Collapse
|
21
|
Vahora H, Khan MA, Alalami U, Hussain A. The Potential Role of Nitric Oxide in Halting Cancer Progression Through Chemoprevention. J Cancer Prev 2016; 21:1-12. [PMID: 27051643 PMCID: PMC4819660 DOI: 10.15430/jcp.2016.21.1.1] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/10/2016] [Accepted: 03/14/2016] [Indexed: 12/30/2022] Open
Abstract
Nitric oxide (NO) in general plays a beneficial physiological role as a vasorelaxant and the role of NO is decided by its concentration present in physiological environments. NO either facilitates cancer-promoting characters or act as an anti-cancer agent. The dilemma in this regard still remains unanswered. This review summarizes the recent information on NO and its role in carcinogenesis and tumor progression, as well as dietary chemopreventive agents which have NO-modulating properties with safe cytotoxic profile. Understanding the molecular mechanisms and cross-talk modulating NO effect by these chemopreventive agents can allow us to develop better therapeutic strategies for cancer treatment.
Collapse
Affiliation(s)
- Huzefa Vahora
- School of Life Sciences, Manipal University, Zayed University, Dubai, United Arab Emirates
| | - Munawwar Ali Khan
- Department of Natural Science and Public Health, College of Sustainability Sciences and Humanities, Zayed University, Dubai, United Arab Emirates
| | - Usama Alalami
- Department of Natural Science and Public Health, College of Sustainability Sciences and Humanities, Zayed University, Dubai, United Arab Emirates
| | - Arif Hussain
- School of Life Sciences, Manipal University, Zayed University, Dubai, United Arab Emirates
| |
Collapse
|
22
|
Vahora H, Khan MA, Alalami U, Hussain A. The Potential Role of Nitric Oxide in Halting Cancer Progression Through Chemoprevention. J Cancer Prev 2016. [DOI: 10.15430/jcp.2016.21.1.1\] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
- Huzefa Vahora
- School of Life Sciences, Manipal University, Dubai, United Arab Emirates
| | - Munawwar Ali Khan
- Department of Natural Science and Public Health, College of Sustainability Sciences and Humanities, Zayed University, Dubai, United Arab Emirates
| | - Usama Alalami
- Department of Natural Science and Public Health, College of Sustainability Sciences and Humanities, Zayed University, Dubai, United Arab Emirates
| | - Arif Hussain
- School of Life Sciences, Manipal University, Dubai, United Arab Emirates
| |
Collapse
|
23
|
Yu X, Luo A, Liu Y, Wang S, Li Y, Shi W, Liu Z, Qu X. MiR-214 increases the sensitivity of breast cancer cells to tamoxifen and fulvestrant through inhibition of autophagy. Mol Cancer 2015; 14:208. [PMID: 26666173 PMCID: PMC4678692 DOI: 10.1186/s12943-015-0480-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 12/09/2015] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Tamoxifen (TAM) and fulvestrant (FUL) are the major drugs for patients with estrogen receptor-positive (ER(+)) breast cancers. However, the development of endocrine resistance is the impediment for successful treatment. We aimed to explore the mechanisms of endocrine resistance and therapeutic strategy for overcoming resistance against TAM and FUL. METHODS Experiments were performed in ER(+) and estrogen/TAM-sensitive MCF7 cells and antiestrogen-resistant MCF7/LCC9 cells. The expression of miR-214 and uncoupling protein 2 (UCP2) was determined by RT-qPCR and Western blot in breast cancer cells and human breast cancer tissue specimens. Cell autophagy was examined by fluorescent probe monodansyl cadaverine (MDC) and GFP-LC3-II-positive punctate identified by confocal microscopy. Apoptotic cells were determined by Annexin V-FITC/PI staining. The potential regulatory target of miR-214 was determined by prediction tool, target protein expression and luciferase reporter assay. RESULTS 4-OHT/FUL treatment resulted in induction of apoptosis as well as autophagy in breast cancer cells. Autophagy might be the major cause of endocrine resistance to 4-OHT or FUL. MiR-214 increased the sensitivity of breast cancer cells to the 4-OHT/FUL-induced apoptosis through inhibition of autophagy. Importantly, a negative correlation was established between miR-214 and UCP2 in human breast cancer tissue specimens assayed by RT-qPCR. UCP2 was identified to be a direct target of miR-214. Further study in MCF7/LCC9 cells indicated that endocrine resistance might arise from activation of the PI3K-Akt-mTOR pathway, thereby inducing autophagy by overexpression of UCP2. CONCLUSION MiR-214 increased the sensitivity of breast cancer cells to TAM and FUL through inhibition of autophagy by targeting UCP2. MiR-214 shows potential as a novel therapeutic strategy for overcoming endocrine resistance in ER(+) breast cancers.
Collapse
Affiliation(s)
- Xinfeng Yu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, No.10, Xitoutiao, Youanmenwai Avenue, 100069, Beijing, China
| | - Aiping Luo
- State Key Lab of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yicong Liu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, No.10, Xitoutiao, Youanmenwai Avenue, 100069, Beijing, China
| | - Shuqing Wang
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, No.10, Xitoutiao, Youanmenwai Avenue, 100069, Beijing, China
| | - Ye Li
- Department of Pharmacology, School of Chemical Biology & Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Wenna Shi
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, No.10, Xitoutiao, Youanmenwai Avenue, 100069, Beijing, China
| | - Zhihua Liu
- State Key Lab of Molecular Oncology, Cancer Institute & Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Xianjun Qu
- Department of Pharmacology, School of Basic Medical Sciences, Capital Medical University, No.10, Xitoutiao, Youanmenwai Avenue, 100069, Beijing, China.
| |
Collapse
|