51
|
Ismail T, Kim Y, Lee H, Lee DS, Lee HS. Interplay Between Mitochondrial Peroxiredoxins and ROS in Cancer Development and Progression. Int J Mol Sci 2019; 20:ijms20184407. [PMID: 31500275 PMCID: PMC6770548 DOI: 10.3390/ijms20184407] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/04/2019] [Accepted: 09/05/2019] [Indexed: 12/14/2022] Open
Abstract
Mitochondria are multifunctional cellular organelles that are major producers of reactive oxygen species (ROS) in eukaryotes; to maintain the redox balance, they are supplemented with different ROS scavengers, including mitochondrial peroxiredoxins (Prdxs). Mitochondrial Prdxs have physiological and pathological significance and are associated with the initiation and progression of various cancer types. In this review, we have focused on signaling involving ROS and mitochondrial Prdxs that is associated with cancer development and progression. An upregulated expression of Prdx3 and Prdx5 has been reported in different cancer types, such as breast, ovarian, endometrial, and lung cancers, as well as in Hodgkin's lymphoma and hepatocellular carcinoma. The expression of Prdx3 and Prdx5 in different types of malignancies involves their association with different factors, such as transcription factors, micro RNAs, tumor suppressors, response elements, and oncogenic genes. The microenvironment of mitochondrial Prdxs plays an important role in cancer development, as cancerous cells are equipped with a high level of antioxidants to overcome excessive ROS production. However, an increased production of Prdx3 and Prdx5 is associated with the development of chemoresistance in certain types of cancers and it leads to further complications in cancer treatment. Understanding the interplay between mitochondrial Prdxs and ROS in carcinogenesis can be useful in the development of anticancer drugs with better proficiency and decreased resistance. However, more targeted studies are required for exploring the tumor microenvironment in association with mitochondrial Prdxs to improve the existing cancer therapies and drug development.
Collapse
Affiliation(s)
- Tayaba Ismail
- KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Youni Kim
- KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Hongchan Lee
- KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Dong-Seok Lee
- KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea.
| | - Hyun-Shik Lee
- KNU-Center for Nonlinear Dynamics, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, College of Natural Sciences, Kyungpook National University, Daegu 41566, Korea.
| |
Collapse
|
52
|
Prooxidative Activity of Celastrol Induces Apoptosis, DNA Damage, and Cell Cycle Arrest in Drug-Resistant Human Colon Cancer Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6793957. [PMID: 31485297 PMCID: PMC6710751 DOI: 10.1155/2019/6793957] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/12/2019] [Accepted: 07/18/2019] [Indexed: 01/02/2023]
Abstract
Cancer resistance to chemotherapy is closely related to tumor heterogeneity, i.e., the existence of distinct subpopulations of cancer cells in a tumor mass. An important role is assigned to cancer stem cells (CSCs), a small subset of cancer cells with high tumorigenic potential and capacity of self-renewal and differentiation. These properties of CSCs are sustained by the ability of those cells to maintain a low intracellular reactive oxygen species (ROS) levels, via upregulation of ROS scavenging systems. However, the accumulation of ROS over a critical threshold disturbs CSCs—redox homeostasis causing severe cytotoxic consequences. In the present study, we investigated the capacity of celastrol, a natural pentacyclic triterpenoid, to induce the formation of ROS and, consequently, cell death of the colon cancer cells with acquired resistant to cytotoxic drugs (LOVO/DX cell line). LOVO/DX cells express several important stem-like cell features, including a higher frequency of side population (SP) cells, higher expression of multidrug resistant proteins, overexpression of CSC-specific cell surface marker (CD44), increased expression of DNA repair gene (PARP1), and low intracellular ROS level. We found that celastrol, at higher concentrations (above 1 μM), significantly increased ROS amount in LOVO/DX cells at both cytoplasmic and mitochondrial levels. This prooxidant activity was associated with the induction of DNA double-strand breaks (DSBs) and apoptotic/necrotic cell death, as well as with inhibition of cell proliferation by S phase cell cycle arrest. Coincubation with NAC, a ROS scavenger, completely reversed the above effects. In summary, our results provide evidence that celastrol exhibits effective cytotoxic effects via ROS-dependent mechanisms on drug-resistant colon cancer cells. These findings strongly suggest the potential of celastrol to effectively kill cancer stem-like cells, and thus, it is a promising agent to treat severe, resistant to conventional therapy, colon cancers.
Collapse
|
53
|
Lee BWL, Ghode P, Ong DST. Redox regulation of cell state and fate. Redox Biol 2019; 25:101056. [PMID: 30509603 PMCID: PMC6859564 DOI: 10.1016/j.redox.2018.11.014] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 10/29/2018] [Accepted: 11/20/2018] [Indexed: 02/07/2023] Open
Abstract
The failure in effective cancer treatment is thought to be attributed to a subpopulation of tumor cells with stem cell-like properties. These cancer stem cells (CSCs) are intimately linked to tumor initiation, heterogeneity, maintenance, recurrence and metastasis. Increasing evidence supports the view that a tight redox regulation is crucial for CSC proliferation, tumorigenicity, therapy resistance and metastasis in many cancer types. Since the distinct metabolic and epigenetic states of CSCs may influence ROS levels, and hence their malignancy, ROS modulating agents hold promise in their utility as anti-CSC agents that may improve the durability of current cancer treatments. This review will focus on (i) how ROS levels are regulated for CSCs to elicit their hallmark features; (ii) the link between ROS and metabolic plasticity of CSCs; and (iii) how ROS may interface with epigenetics that would enable CSCs to thrive in a stressful tumor microenvironment and survive therapeutic insults.
Collapse
Affiliation(s)
- Bernice Woon Li Lee
- Department of Physiology, National University of Singapore, Singapore 117593, Singapore
| | - Pramila Ghode
- Department of Physiology, National University of Singapore, Singapore 117593, Singapore
| | - Derrick Sek Tong Ong
- Department of Physiology, National University of Singapore, Singapore 117593, Singapore; Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A⁎STAR), Singapore.
| |
Collapse
|
54
|
Schulz A, Meyer F, Dubrovska A, Borgmann K. Cancer Stem Cells and Radioresistance: DNA Repair and Beyond. Cancers (Basel) 2019; 11:cancers11060862. [PMID: 31234336 PMCID: PMC6627210 DOI: 10.3390/cancers11060862] [Citation(s) in RCA: 188] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/17/2019] [Accepted: 06/18/2019] [Indexed: 12/12/2022] Open
Abstract
The current preclinical and clinical findings demonstrate that, in addition to the conventional clinical and pathological indicators that have a prognostic value in radiation oncology, the number of cancer stem cells (CSCs) and their inherent radioresistance are important parameters for local control after radiotherapy. In this review, we discuss the molecular mechanisms of CSC radioresistance attributable to DNA repair mechanisms and the development of CSC-targeted therapies for tumor radiosensitization. We also discuss the current challenges in preclinical and translational CSC research including the high inter- and intratumoral heterogeneity, plasticity of CSCs, and microenvironment-stimulated tumor cell reprogramming.
Collapse
Affiliation(s)
- Alexander Schulz
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
| | - Felix Meyer
- Laboratory of Radiobiology & Experimental Radiooncology, Department of Radiotherapy and Radiooncology, Center of Oncology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.
| | - Anna Dubrovska
- OncoRay-National Center for Radiation Research in Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany.
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiooncology-OncoRay, 01328 Dresden, Germany.
- German Cancer Consortium (DKTK), Partner Site Dresden, 01307 Dresden, Germany.
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| | - Kerstin Borgmann
- Laboratory of Radiobiology & Experimental Radiooncology, Department of Radiotherapy and Radiooncology, Center of Oncology, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany.
| |
Collapse
|
55
|
Tataranni T, Agriesti F, Pacelli C, Ruggieri V, Laurenzana I, Mazzoccoli C, Sala GD, Panebianco C, Pazienza V, Capitanio N, Piccoli C. Dichloroacetate Affects Mitochondrial Function and Stemness-Associated Properties in Pancreatic Cancer Cell Lines. Cells 2019; 8:cells8050478. [PMID: 31109089 PMCID: PMC6562462 DOI: 10.3390/cells8050478] [Citation(s) in RCA: 33] [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: 02/21/2019] [Revised: 05/14/2019] [Accepted: 05/15/2019] [Indexed: 12/14/2022] Open
Abstract
Targeting metabolism represents a possible successful approach to treat cancer. Dichloroacetate (DCA) is a drug known to divert metabolism from anaerobic glycolysis to mitochondrial oxidative phosphorylation by stimulation of PDH. In this study, we investigated the response of two pancreatic cancer cell lines to DCA, in two-dimensional and three-dimension cell cultures, as well as in a mouse model. PANC-1 and BXPC-3 treated with DCA showed a marked decrease in cell proliferation and migration which did not correlate with enhanced apoptosis indicating a cytostatic rather than a cytotoxic effect. Despite PDH activation, DCA treatment resulted in reduced mitochondrial oxygen consumption without affecting glycolysis. Moreover, DCA caused enhancement of ROS production, mtDNA, and of the mitophagy-marker LC3B-II in both cell lines but reduced mitochondrial fusion markers only in BXPC-3. Notably, DCA downregulated the expression of the cancer stem cells markers CD24/CD44/EPCAM only in PANC-1 but inhibited spheroid formation/viability in both cell lines. In a xenograft pancreatic cancer mouse-model DCA treatment resulted in retarding cancer progression. Collectively, our results clearly indicate that the efficacy of DCA in inhibiting cancer growth mechanistically depends on the cell phenotype and on multiple off-target pathways. In this context, the novelty that DCA might affect the cancer stem cell compartment is therapeutically relevant.
Collapse
Affiliation(s)
- Tiziana Tataranni
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (Pz), Italy.
| | - Francesca Agriesti
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (Pz), Italy.
| | - Consiglia Pacelli
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy.
| | - Vitalba Ruggieri
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (Pz), Italy.
| | - Ilaria Laurenzana
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (Pz), Italy.
| | - Carmela Mazzoccoli
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (Pz), Italy.
| | - Gerardo Della Sala
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (Pz), Italy.
| | - Concetta Panebianco
- Division of Gastroenterology, IRCCS "Casa Sollievo della Sofferenza" Hospital, 71013 San Giovanni Rotondo, Italy.
| | - Valerio Pazienza
- Division of Gastroenterology, IRCCS "Casa Sollievo della Sofferenza" Hospital, 71013 San Giovanni Rotondo, Italy.
| | - Nazzareno Capitanio
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy.
| | - Claudia Piccoli
- Laboratory of Pre-Clinical and Translational Research, IRCCS-CROB, Referral Cancer Center of Basilicata, 85028 Rionero in Vulture (Pz), Italy.
- Department of Clinical and Experimental Medicine, University of Foggia, 71100 Foggia, Italy.
| |
Collapse
|
56
|
Higd1a Protects Cells from Lipotoxicity under High-Fat Exposure. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6051262. [PMID: 31089410 PMCID: PMC6476072 DOI: 10.1155/2019/6051262] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2018] [Revised: 01/07/2019] [Accepted: 01/16/2019] [Indexed: 02/06/2023]
Abstract
Hypoxia-inducible gene domain family member 1A (Higd1a) has recently been reported to protect cells from hypoxia by helping to maintain normal mitochondrial function. The potential induction of Higd1a under high-fat exposure and whether it could protect cells from oxidative stress attracted our attention. Initially, 0.4 mM oleic acid and 0.2 mM palmitate were added to the growth media of HepG2 and LO2 cells for 72 hours. We discovered increased Higd1a expression, and knocking down Higd1a impaired mitochondrial transmembrane potential and induced cell apoptosis. We then identified that elevated reactive oxygen species (ROS) is responsible for increased Higd1a expression. Furthermore, we found that ROS promoted Higd1a expression by upregulating HIF-1a and PGC-1a expressions, and these two proteins could exert synergistic effects in inducing Higd1a expression. Taken together, these data suggest that Higd1a plays positive roles in protecting cells from oxidative stress, and ROS could induce Higd1a expression by upregulating PGC-1a and HIF-1a expressions.
Collapse
|
57
|
Mohamed RH, Abu-Shahba N, Mahmoud M, Abdelfattah AMH, Zakaria W, ElHefnawi M. Co-regulatory Network of Oncosuppressor miRNAs and Transcription Factors for Pathology of Human Hepatic Cancer Stem Cells (HCSC). Sci Rep 2019; 9:5564. [PMID: 30944375 PMCID: PMC6447552 DOI: 10.1038/s41598-019-41978-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Accepted: 03/21/2019] [Indexed: 12/11/2022] Open
Abstract
Hepatic cancer stem cells (HCSCs) are considered as main players for the hepatocellular carcinoma (HCC) initiation, metastasis, drug resistance and recurrence. There is a growing evidence supporting the down-regulated miRNAs in HCSCs as key suppressors for the stemness traits, but still more details are vague about how these miRNAs modulate the HCC development. To uncover some of these miRNA regulatory aspects in HCSC, we compiled 15 down-regulated miRNA and their validated and predicted up-regulated targets in HCSC. The targets were enriched for several cancer cell stemness hallmarks and CSC pre-metastatic niche, which support these miRNAs role in suppression of HCSCs neoplastic transformation. Further, we constructed miRNA-Transcription factor (TF) regulatory networks, which provided new insights on the role of the proposed miRNA-TF co-regulation in the cancer stemness axis and its cross talk with the surrounding microenvironment. Our analysis revealed HCSC important hubs as candidate regulators for targeting hepatic cancer stemness such as, miR-148a, miR-214, E2F family, MYC and SLC7A5. Finally, we proposed a possible model for miRNA and TF co-regulation of HCSC signaling pathways. Our study identified an HCSC signature and set bridges between the reported results to give guide for future validation of HCC therapeutic strategies avoiding drug resistance.
Collapse
Affiliation(s)
- Rania Hassan Mohamed
- Department of Biochemistry, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Nourhan Abu-Shahba
- Stem Cell Research Group, Centre of Excellence for Advanced Sciences, Department of Medical Molecular Genetics, National Research Centre, Cairo, Egypt
| | - Marwa Mahmoud
- Stem Cell Research Group, Centre of Excellence for Advanced Sciences, Department of Medical Molecular Genetics, National Research Centre, Cairo, Egypt
| | - Ahmed M H Abdelfattah
- Department of Mathematics, Faculty of Science, Ain Shams University, Cairo, Egypt.,VAP, CS Department, SUNY, Oswego, NY, USA
| | - Wael Zakaria
- Department of Mathematics, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Mahmoud ElHefnawi
- Biomedical informatics and Chemoinformatics group, Centre of Excellence for Advanced Sciences, Informatics and Systems Department, National Research Centre, Cairo, Egypt. .,Informatics and systems Department, Division of Engineering research, National Research Centre, Cairo, Egypt.
| |
Collapse
|
58
|
Liu J, Wang Y, Liu X, Yuan Q, Zhang Y, Li Y. Novel molecularly imprinted polymer (MIP) multiple sensors for endogenous redox couples determination and their applications in lung cancer diagnosis. Talanta 2019; 199:573-580. [PMID: 30952300 DOI: 10.1016/j.talanta.2019.03.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 02/16/2019] [Accepted: 03/02/2019] [Indexed: 12/27/2022]
Abstract
Multiplex electrochemical sensors for amperometric detection of glutathione disulfide (GSSG), glutathione (GSH), cysteine (Cys), cystine (Cyss), β-nicotinamide adenine dinucleotide phosphate (NADP+) and coenzyme II reduced tetrasodium salt (NADPH) were developed, in which analysis of Cyss, NADP+ and NADPH are the first report using this sensing system. Specificity of these sensors were ensured by a layer of molecularly imprinted polymer (MIP) which was electropolymerized in situ with the analytes as template. All the sensors were tested with standard buffers and mouse blood samples, showing satisfactory performance towards the corresponding analytes. Dynamic concentration for the six analytes was in the range of 10-11-10-8 mol/L with the detection limit down to 20 pmol/L. In addition, artificially synthesized MIP film on the electrodes allowed for good selectivity and stability. Real blood sample measurement proved that the sensors owned decent accuracy with recovery value ranging from 92%~112%. More importantly, blood samples from lung cancer patients and healthy donors were assayed by using the proposed sensors. Redox potentials (Ehc) were calculated based on the contents of these endogenic substances, which were utilized to reflect the health status of human body and help diagnose lung cancer. The levels of GSH, NADPH and the absolute value of Ehc(GSH/GSSG) in patients with lung cancer are significantly lower (P < 0.01) than those in healthy people, while the contents of GSSG (P < 0.01) are higher. The blood test results suggested that the content of GSH, NADPH, NADP+ and Ehc(GSH/GSSG) might serve as biomarkers for lung cancer prediagnosis. These novel sensors for liquid biospy of cancer have cost-benefit and scalability advantage over current techniques, potentially enabling broader clinical access and efficient population screening.
Collapse
Affiliation(s)
- Jie Liu
- College of Science, Harbin Institute of Technology, Shenzhen 518055, China; Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi 832000, China
| | - Yuli Wang
- The first affiliated hospital of the medical college of Shihezi University, Shihezi 832000, China
| | - Xiaoxue Liu
- College of Science, Harbin Institute of Technology, Shenzhen 518055, China
| | - Qunhui Yuan
- College of Materials Science and Engineering, Harbin Institute of Technology, Shenzhen 518055, China
| | - Yang Zhang
- College of Science, Harbin Institute of Technology, Shenzhen 518055, China.
| | - Yingchun Li
- College of Science, Harbin Institute of Technology, Shenzhen 518055, China; Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, School of Pharmacy, Shihezi University, Shihezi 832000, China.
| |
Collapse
|
59
|
Recalcati S, Gammella E, Cairo G. Dysregulation of iron metabolism in cancer stem cells. Free Radic Biol Med 2019; 133:216-220. [PMID: 30040994 DOI: 10.1016/j.freeradbiomed.2018.07.015] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 07/18/2018] [Accepted: 07/20/2018] [Indexed: 12/17/2022]
Abstract
Cancer stem cells (CSCs) are a distinct subpopulation of tumor cells endowed with stem-like properties. Importantly, CSCs can survive current standard therapies, resulting in metastatic disease and tumor recurrence. Here we describe the alterations of iron homeostasis occurring in CSCs, which in general are characterized by high intracellular iron content. Importantly, abnormalities of iron metabolism correlate with faster tumor growth and adverse prognosis in cancer patients. In line with the dependence of cancer on iron, we also discuss iron-dependent mechanisms as druggable pathways, as iron chelators have been considered for tumor therapy and new molecules currently proposed and studied as antineoplastic drugs may impinge on iron and its capacity to promote oxidative stress to have therapeutic value in cancer.
Collapse
Affiliation(s)
- Stefania Recalcati
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milano, Italy
| | - Elena Gammella
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milano, Italy
| | - Gaetano Cairo
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milano, Italy.
| |
Collapse
|
60
|
Di Rocco G, Baldari S, Pani G, Toietta G. Stem cells under the influence of alcohol: effects of ethanol consumption on stem/progenitor cells. Cell Mol Life Sci 2019; 76:231-244. [PMID: 30306211 PMCID: PMC6339663 DOI: 10.1007/s00018-018-2931-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 09/10/2018] [Accepted: 10/01/2018] [Indexed: 12/13/2022]
Abstract
Stem cells drive embryonic and fetal development. In several adult tissues, they retain the ability to self-renew and differentiate into a variety of specialized cells, thus contributing to tissue homeostasis and repair throughout life span. Alcohol consumption is associated with an increased risk for several diseases and conditions. Growing and developing tissues are particularly vulnerable to alcohol's influence, suggesting that stem- and progenitor-cell function could be affected. Accordingly, recent studies have revealed the possible relevance of alcohol exposure in impairing stem-cell properties, consequently affecting organ development and injury response in different tissues. Here, we review the main studies describing the effects of alcohol on different types of progenitor/stem cells including neuronal, hepatic, intestinal and adventitial progenitor cells, bone-marrow-derived stromal cell, dental pulp, embryonic and hematopoietic stem cells, and tumor-initiating cells. A better understanding of the nature of the cellular damage induced by chronic and episodic heavy (binge) drinking is critical for the improvement of current therapeutic strategies designed to treat patients suffering from alcohol-related disorders.
Collapse
Affiliation(s)
- Giuliana Di Rocco
- Department of Research, Advanced Diagnostic, and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Silvia Baldari
- Department of Research, Advanced Diagnostic, and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy
| | - Giovambattista Pani
- Institute of General Pathology, Laboratory of Cell Signaling, Catholic University Medical School, Largo F. Vito 1, 00168, Rome, Italy
| | - Gabriele Toietta
- Department of Research, Advanced Diagnostic, and Technological Innovation, Translational Research Area, IRCCS Regina Elena National Cancer Institute, Via E. Chianesi 53, 00144, Rome, Italy.
| |
Collapse
|
61
|
JIAPAER S, FURUTA T, TANAKA S, KITABAYASHI T, NAKADA M. Potential Strategies Overcoming the Temozolomide Resistance for Glioblastoma. Neurol Med Chir (Tokyo) 2018; 58:405-421. [PMID: 30249919 PMCID: PMC6186761 DOI: 10.2176/nmc.ra.2018-0141] [Citation(s) in RCA: 218] [Impact Index Per Article: 31.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Accepted: 07/31/2018] [Indexed: 12/14/2022] Open
Abstract
Glioblastoma (GBM) is a highly malignant type of primary brain tumor with a high mortality rate. Although the current standard therapy consists of surgery followed by radiation and temozolomide (TMZ), chemotherapy can extend patient's post-operative survival but most cases eventually demonstrate resistance to TMZ. O6-methylguanine-DNA methyltransferase (MGMT) repairs the main cytotoxic lesion, as O6-methylguanine, generated by TMZ, can be the main mechanism of the drug resistance. In addition, mismatch repair and BER also contribute to TMZ resistance. TMZ treatment can induce self-protective autophagy, a mechanism by which tumor cells resist TMZ treatment. Emerging evidence also demonstrated that a small population of cells expressing stem cell markers, also identified as GBM stem cells (GSCs), contributes to drug resistance and tumor recurrence owing to their ability for self-renewal and invasion into neighboring tissue. Some molecules maintain stem cell properties. Other molecules or signaling pathways regulate stemness and influence MGMT activity, making these GCSs attractive therapeutic targets. Treatments targeting these molecules and pathways result in suppression of GSCs stemness and, in highly resistant cases, a decrease in MGMT activity. Recently, some novel therapeutic strategies, targeted molecules, immunotherapies, and microRNAs have provided new potential treatments for highly resistant GBM cases. In this review, we summarize the current knowledge of different resistance mechanisms, novel strategies for enhancing the effect of TMZ, and emerging therapeutic approaches to eliminate GSCs, all with the aim to produce a successful GBM treatment and discuss future directions for basic and clinical research to achieve this end.
Collapse
Affiliation(s)
| | - Takuya FURUTA
- Department of Pathology, Kurume University, Kurume, Fukuoka, Japan
| | - Shingo TANAKA
- Department of Neurosurgery, Kanazawa University, Kanazawa, Ishikawa, Japan
| | | | - Mitsutoshi NAKADA
- Department of Neurosurgery, Kanazawa University, Kanazawa, Ishikawa, Japan
| |
Collapse
|
62
|
Purushothaman B, Arumugam P, Ju H, Kulsi G, Samson AAS, Song JM. Novel ruthenium(II) triazine complex [Ru(bdpta)(tpy)]2+ co-targeting drug resistant GRP78 and subcellular organelles in cancer stem cells. Eur J Med Chem 2018; 156:747-759. [DOI: 10.1016/j.ejmech.2018.07.048] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/14/2018] [Accepted: 07/17/2018] [Indexed: 12/17/2022]
|
63
|
Metabolic influence of walnut phenolic extract on mitochondria in a colon cancer stem cell model. Eur J Nutr 2018; 58:1635-1645. [PMID: 29740695 DOI: 10.1007/s00394-018-1708-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 05/01/2018] [Indexed: 12/18/2022]
Abstract
PURPOSE Walnut phenolic extract (WPE) reduces proliferation and enhances differentiation of colon cancer stem cells (CSCs). The present study investigated the metabolic influence of WPE on the mitochondrial function of colon CSCs to determine its underlying mechanism. METHODS CD133+CD44+ HCT116 colon cancer cells were selected by fluorescence-activated cell sorting and were treated with or without 40 µg/mL WPE. RNA-sequencing (RNA-Seq) was performed to identify differentially expressed genes (DEGs), which were further validated with RT-PCR. WPE-induced alterations in mitochondrial function were investigated through a mitochondrial stress test by determining cellular oxygen consumption rate (OCR), an indicator of mitochondrial respiration, and extracellular acidification rate (ECAR), an indicator of glycolysis, which were further confirmed by glucose uptake and lactate production tests. RESULTS RNA-Seq analysis identified two major functional clusters: metabolic and mitochondrial clusters. WPE treatment shifted the metabolic profile of cells towards the glycolysis pathway (ΔECAR = 36.98 mpH/min/ptn, p = 0.02) and oxidative pathway (ΔOCR = 29.18 pmol/min/ptn, p = 0.00001). Serial mitochondrial stimulations using respiration modulators, oligomycin, carbonyl cyanide-4 (trifluoromethoxy) phenylhydrazone, and rotenone/antimycin A, found an increased potential of mitochondrial respiration (ΔOCR = 111.5 pmol/min/ptn, p = 0.0006). WPE treatment also increased glucose uptake (Δ = 0.39 pmol/µL, p = 0.002) and lactate production (Δ = 0.08 nmol/µL, p = 0.005). CONCLUSIONS WPE treatment shifts the mitochondrial metabolism of colon CSC towards more aerobic glycolysis, which might be associated with the alterations in the characteristics of colon CSC.
Collapse
|
64
|
Samimi A, Kalantari H, Lorestani MZ, Shirzad R, Saki N. Oxidative stress in normal hematopoietic stem cells and leukemia. APMIS 2018; 126:284-294. [PMID: 29575200 DOI: 10.1111/apm.12822] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Accepted: 01/22/2018] [Indexed: 12/19/2022]
Abstract
Leukemia is developed following the abnormal proliferation of immature hematopoietic cells in the blood when hematopoietic stem cells lose the ability to turn into mature cells at different stages of maturation and differentiation. Leukemia initiating cells are specifically dependent upon the suppression of oxidative stress in the hypoglycemic bone marrow (BM) environment to be able to start their activities. Relevant literature was identified by a PubMed search (2000-2017) of English-language literature using the terms 'oxidative stress,' 'reactive oxygen species,' 'hematopoietic stem cell,' and 'leukemia.' The generation and degradation of free radicals is a main component of the metabolism in aerobic organisms. A certain level of ROS is required for proper cellular function, but values outside this range will result in oxidative stress (OS). Long-term overactivity of reactive oxygen species (ROS) has harmful effects on the function of cells and their vital macromolecules, including the transformation of proteins into autoantigens and increased degradation of protein/DNA, which eventually leads to the change in pathways involved in the development of cancer and several other disorders. According to the metabolic disorders of cancer, the relationship between OS changes, the viability of cancer cells, and their response to chemotherapeutic agents affecting this pathway are undeniable. Recently, studies have been conducted to determine the effect of herbal agents and cancer chemotherapy drugs on oxidative stress pathways. By emphasizing the role of oxidative stress on stem cells in the incidence of leukemia, this paper attempts to state and summarize this subject.
Collapse
Affiliation(s)
- Azin Samimi
- Department of Pharmacology and Toxicology, School of Pharmacy, Ahvaz Jundishpur University of Medical Sciences, Ahvaz, Iran
| | - Heybatullah Kalantari
- Department of Pharmacology and Toxicology, School of Pharmacy, Ahvaz Jundishpur University of Medical Sciences, Ahvaz, Iran
| | - Marzieh Zeinvand Lorestani
- Department of Pharmacology and Toxicology, School of Pharmacy, Ahvaz Jundishpur University of Medical Sciences, Ahvaz, Iran
| | - Reza Shirzad
- WHO-Collaborating Centre for Reference and Research on Rabies, Pasteur Institute of Iran, Tehran, Iran
| | - Najmaldin Saki
- Research Center of Thalassemia & Hemoglobinopathy, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
65
|
Shamsi TN, Parveen R, Fatima S. Trypsin inhibitors demonstrate antioxidant activities, inhibit A549 cell proliferation, and increase activities of reactive oxygen species scavenging enzymes. Indian J Pharmacol 2018; 49:155-160. [PMID: 28706328 PMCID: PMC5497437 DOI: 10.4103/ijp.ijp_553_16] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVES: Protease inhibitors are one of the most promising and investigated subjects for their role in pharmacognostical and pharmacological studies. This study aimed to investigate antineoplastic and antioxidant activity of trypsin inhibitors (TIs) isolated from three plant sources and their inhibitory role in the cell line. MATERIALS AND METHODOLOGY: TIs were obtained from different plant sources. Antineoplastic potential on adenocarcinoma human alveolar basal epithelial cell line (A549) and normal Human Embryonic Kidney (HEK) was determined using MTT assay. Activities of antioxidant enzyme, nitric oxide scavenger, superoxide dismutase, glutathione S-transferase, and glutathione peroxidase were assessed in cell lines incubated with and without TIs. The outcome was analyzed by spectrophotometer. RESULTS: TIs showed the higher cytotoxicity on A549 cells as compared to normal HEK cell line. TIs exhibited fair increase in antioxidant enzyme activity in A549 cells as compared to control. This might be one of the strategies of antineoplastic effect in cancer cells. CONCLUSIONS: This study has reported the antioxidant and antineoplastic properties of these TIs for the first time in A549 cells (to the best of our knowledge). The results show that TIs possess ability to prevent cancer and diseases caused due to oxidative stress. Therefore, we conclude that TIs can be used as supplements along with the conventional drugs for increased efficacy in the treatment of diseases such as cardiovascular disease, atherosclerosis, and cancer.
Collapse
Affiliation(s)
- Tooba Naz Shamsi
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Romana Parveen
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Sadaf Fatima
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
66
|
Lleonart ME, Abad E, Graifer D, Lyakhovich A. Reactive Oxygen Species-Mediated Autophagy Defines the Fate of Cancer Stem Cells. Antioxid Redox Signal 2018; 28:1066-1079. [PMID: 28683561 DOI: 10.1089/ars.2017.7223] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Significance: A fraction of tumorigenic cells, also known as tumor initiating or cancer stem cells (CSCs), is thought to drive tumor growth, metastasis, and chemoresistance. However, little is known regarding mechanisms that convey relevant pathways contributing to their self-renewal, proliferation, and differentiation abilities. Recent Advances: Recent works on CSCs provide evidence on the role of redox disruption and regulation of autophagic flux. This has been linked to increased DNA repair capacity and chemoresistance. Critical Issues: The current review summarizes the most recent studies assessing the role of redox homeostasis, autophagy, and chemoresistance in CSCs, including some novel findings on microRNAs and their role in horizontal transfer within cancer cell populations. Future Directions: Rational anticancer therapy and prevention should rely on the fact that cancer is a redox disease with the CSCs being the apex modulated by redox-mediated autophagy. Antioxid. Redox Signal. 28, 1066-1079.
Collapse
Affiliation(s)
- Matilde E Lleonart
- Biomedical Research in Cancer Stem Cells, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Etna Abad
- Biomedical Research in Cancer Stem Cells, Vall d'Hebron Research Institute, Barcelona, Spain
| | - Dmitry Graifer
- Faculty of Natural Sciences, Novosibirsk State University, Novosibirsk, Russia
| | - Alex Lyakhovich
- Biomedical Research in Cancer Stem Cells, Vall d'Hebron Research Institute, Barcelona, Spain.,Institute of Molecular Biology and Biophysics, Novosibirsk, Russia.,ICRC-FNUSA, International Clinical Research Center and St. Anne's University Hospital Brno, Brno, Czech Republic
| |
Collapse
|
67
|
Chittiboyina S, Bai Y, Lelièvre SA. Microenvironment-Cell Nucleus Relationship in the Context of Oxidative Stress. Front Cell Dev Biol 2018; 6:23. [PMID: 29594114 PMCID: PMC5854663 DOI: 10.3389/fcell.2018.00023] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 02/21/2018] [Indexed: 12/17/2022] Open
Abstract
The microenvironment is a source of reactive oxygen species (ROS) that influence cell phenotype and tissue homeostasis. The impact of ROS on redox pathways as well as directly on epigenetic mechanisms and the DNA illustrate communication with the cell nucleus. Changes in gene transcription related to redox conditions also influence the content and structure of the extracellular matrix. However, the importance of microenvironmental ROS for normal progression through life and disease development still needs to be thoroughly understood. We illustrate how different ROS concentration levels trigger various intracellular pathways linked to nuclear functions and determine processes necessary for the differentiation of stem cells. The abnormal predominance of ROS that leads to oxidative stress is emphasized in light of its impact on aging and diseases related to aging. These phenomena are discussed in the context of the possible contribution of extracellular ROS via direct diffusion into cells responsible for organ function, but also via an impact on stromal cells that triggers extracellular modifications and influences mechanotransduction. Finally, we argue that organs-on-a-chip with controlled microenvironmental conditions can help thoroughly grasp whether ROS production is readily a cause or a consequence of certain disorders, and better understand the concentration levels of extracellular ROS that are necessary to induce a switch in phenotype.
Collapse
Affiliation(s)
- Shirisha Chittiboyina
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States
- 3D Cell Culture Core, Birck Nanotechnology Center, Purdue University Discovery Park, West Lafayette, IN, United States
| | - Yunfeng Bai
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States
| | - Sophie A. Lelièvre
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, United States
- 3D Cell Culture Core, Birck Nanotechnology Center, Purdue University Discovery Park, West Lafayette, IN, United States
- Center for Cancer Research, West Lafayette, IN, United States
| |
Collapse
|
68
|
Choi YJ, Gurunathan S, Kim JH. Graphene Oxide-Silver Nanocomposite Enhances Cytotoxic and Apoptotic Potential of Salinomycin in Human Ovarian Cancer Stem Cells (OvCSCs): A Novel Approach for Cancer Therapy. Int J Mol Sci 2018; 19:E710. [PMID: 29494563 PMCID: PMC5877571 DOI: 10.3390/ijms19030710] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 02/22/2018] [Accepted: 02/26/2018] [Indexed: 01/06/2023] Open
Abstract
The use of graphene to target and eliminate cancer stem cells (CSCs) is an alternative approach to conventional chemotherapy. We show the biomolecule-mediated synthesis of reduced graphene oxide-silver nanoparticle nanocomposites (rGO-Ag) using R-phycoerythrin (RPE); the resulting RPE-rGO-Ag was evaluated in human ovarian cancer cells and ovarian cancer stem cells (OvCSCs). The synthesized RPE-rGO-Ag nanocomposite (referred to as rGO-Ag) was characterized using various analytical techniques. rGO-Ag showed significant toxicity towards both ovarian cancer cells and OvCSCs. After 3 weeks of incubating OvCSCs with rGO-Ag, the number of A2780 and ALDH⁺CD133⁺ colonies was significantly reduced. rGO-Ag was toxic to OvCSCs and reduced cell viability by mediating the generation of reactive oxygen species, leakage of lactate dehydrogenase, reduced mitochondrial membrane potential, and enhanced expression of apoptotic genes, leading to mitochondrial dysfunction and possibly triggering apoptosis. rGO-Ag showed significant cytotoxic potential towards highly tumorigenic ALDH⁺CD133⁺ cells. The combination of rGO-Ag and salinomycin induced 5-fold higher levels of apoptosis than each treatment alone. A combination of rGO-Ag and salinomycin at very low concentrations may be suitable for selectively killing OvCSCs and sensitizing tumor cells. rGO-Ag may be a novel nano-therapeutic molecule for specific targeting of highly tumorigenic ALDH⁺CD133⁺ cells and eliminating CSCs. This study highlights the potential for targeted therapy of tumor-initiating cells.
Collapse
Affiliation(s)
- Yun-Jung Choi
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea.
| | - Sangiliyandi Gurunathan
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea.
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 05029, Korea.
| |
Collapse
|
69
|
Abstract
Resistance to chemotherapy and cancer relapse are major clinical challenges attributed to a sub population of cancer stem cells (CSCs). The concept of CSCs has been the subject of intense research by the oncology community since evidence for their existence was first published over twenty years ago. Emerging data indicates that they are also able to evade novel therapies such as targeted agents, immunotherapies and anti-angiogenics. The inability to appropriately identify and isolate CSCs is a major hindrance to the field and novel technologies are now being utilized. Agents that target CSC-associated cell surface receptors and signaling pathways have generated promising pre-clinical results and are now entering clinical trial. Here we discuss and evaluate current therapeutic strategies to target CSCs.
Collapse
Affiliation(s)
- Stephanie Annett
- Molecular and Cellular Therapeutics, Royal College of Surgeons Ireland, Ireland
| | - Tracy Robson
- Molecular and Cellular Therapeutics, Royal College of Surgeons Ireland, Ireland.
| |
Collapse
|
70
|
Dagnell M, Schmidt EE, Arnér ESJ. The A to Z of modulated cell patterning by mammalian thioredoxin reductases. Free Radic Biol Med 2018; 115:484-496. [PMID: 29278740 PMCID: PMC5771652 DOI: 10.1016/j.freeradbiomed.2017.12.029] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 12/16/2017] [Accepted: 12/21/2017] [Indexed: 12/12/2022]
Abstract
Mammalian thioredoxin reductases (TrxRs) are selenocysteine-containing proteins (selenoproteins) that propel a large number of functions through reduction of several substrates including the active site disulfide of thioredoxins (Trxs). Well-known enzymatic systems that in turn are supported by Trxs and TrxRs include deoxyribonucleotide synthesis through ribonucleotide reductase, antioxidant defense through peroxiredoxins and methionine sulfoxide reductases, and redox modulation of a number of transcription factors. Although these functions may be essential for cells due to crucial roles in maintenance of cell viability and proliferation, findings during the last decade reveal that mammals have major redundancy in their cellular reductive systems. The synthesis of glutathione (GSH) and reductive functions of GSH-dependent pathways typically act in parallel with Trx-dependent pathways, with only one of these systems often being sufficient to support viability. Importantly, this does not imply that a modulation of the Trx system will remain without consequences, even when GSH-dependent pathways remain functional. As suggested by several recent findings, the Trx system in general and the TrxRs in particular, function as key regulators of signaling pathways. In this review article we will discuss findings that collectively suggest that modulation in mammalian systems of cytosolic TrxR1 (TXNRD1) or mitochondrial TrxR2 (TXNRD2) influence cell patterning and cellular stress responses. Effects of lower activities include increased adipogenesis, insulin responsiveness, glycogen accumulation, hyperproliferation, and distorted embryonic development, while increased activities correlate with decreased proliferation and extended lifespan, as well as worse cancer prognosis. The molecular mechanisms that underlie these diverse effects, involving regulation of protein phosphorylation cascades and of key transcription factors that guide cellular differentiation pathways, will be discussed. We conclude that the selenium-dependent oxidoreductases TrxR1 and TrxR2 should be considered as key components of signaling pathways that control cell differentiation and cellular stress responses.
Collapse
Affiliation(s)
- Markus Dagnell
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77, Stockholm, Sweden
| | - Edward E Schmidt
- Microbiology & Immunology, Montana State University, Bozeman, MT 59718, USA
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, SE-171 77, Stockholm, Sweden.
| |
Collapse
|
71
|
He F, Wei L, Luo W, Liao Z, Li B, Zhou X, Xiao X, You J, Chen Y, Zheng S, Li P, Murata M, Huang G, Zhang Z. Glutaredoxin 3 promotes nasopharyngeal carcinoma growth and metastasis via EGFR/Akt pathway and independent of ROS. Oncotarget 2018; 7:37000-37012. [PMID: 27203742 PMCID: PMC5095054 DOI: 10.18632/oncotarget.9454] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 04/16/2016] [Indexed: 12/14/2022] Open
Abstract
Glutaredoxin 3 (GLRX3) is antioxidant enzyme, maintaining a low level of ROS, thus contributing to the survival and metastasis of several types of cancer. However, the expression and functions of GLRX3 have not been addressed in nasopharyngeal carcinoma (NPC). In this study, we found that GLRX3 was overexpressed in NPC. Knockdown of GLRX3 in NPC cell lines inhibited proliferation in vitro, tumorignesis in vivo, and colony formation. In addition, GLRX3 knockdown decreased the migration and invasion capacity of NPC cells by reversing the epithelial-mesenchymal transition (EMT). Furthermore, stabilization of GLRX3 was positively related to with epidermal growth factor receptor (EGFR) expression and negatively with ROS generation. Phosphorylation of Akt, a key downstream effector, was induced by EGFR signaling but did not rely on increasing ROS level in NPC cells. GLRX3 might be an oncoprotein in NPC, playing important roles in increasing redox reaction and activating EGFR/ Akt signals, so it may be a therapeutic target for NPC.
Collapse
Affiliation(s)
- Feng He
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lili Wei
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Wenqi Luo
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhipeng Liao
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Bo Li
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaoying Zhou
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xue Xiao
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Jingping You
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Yufeng Chen
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shixing Zheng
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Ping Li
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Mariko Murata
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine, Mie, Japan
| | - Guangwu Huang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhe Zhang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
72
|
Miran T, Vogg ATJ, Drude N, Mottaghy FM, Morgenroth A. Modulation of glutathione promotes apoptosis in triple-negative breast cancer cells. FASEB J 2018; 32:2803-2813. [PMID: 29301945 DOI: 10.1096/fj.201701157r] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Triple-negative breast cancer has an extremely high rate of relapse. This is particularly due to the existence and survival of cancer stem cells (CSCs) characterized by increased amounts of glutathione (GSH). In this study, we evaluated the potential of pharmacological GSH depletion to sensitize CSCs to ionizing radiotherapy with an I-125-labeled nucleoside analog, 5-iodo-4'-thio-2'-deoxyuridine (ITdU). CSCs were isolated using CD24-- and CD44+-specific microbeads. GSH and reactive oxygen species (ROS) were evaluated by fluorescence-activated cell sorting. GSH synthesis was inhibited with buthionine sulfoximine (BSO). Apoptotic cells were identified with propidium iodide and double-strand DNA breaks were detected by γ-H2AX staining. For therapy study, BSO treated and untreated mice xenografted with breast CSCs received weekly I-125-ITdU. Therapy efficiency was monitored by fluorodeoxyglucose-18-µ-positron emission tomography. We showed that GSH modulation sensitizes CD24- and CD44+ breast cancer cells to endogenous nanoradiotherapy. BSO synergistically affects ROS generation induced by I-125-ITdU. In an in vivo study, we demonstrated a complete tumor regression as a consequence of preconditioning with a GSH-synthesis inhibitor prior to treatment with I-125-ITdU. GSH modulation in combination with an oxidative stress-generating treatment such as endogenous radiotherapy using an Auger emitter offers an extraordinary opportunity for selective and efficient eradication of drug-resistant CSCs.-Miran, T., Vogg, A. T. J., Drude, N., Mottaghy, F. M., Morgenroth, A. Modulation of glutathione promotes apoptosis in triple-negative breast cancer cells.
Collapse
Affiliation(s)
- Tara Miran
- Department of Nuclear Medicine, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Andreas T J Vogg
- Department of Nuclear Medicine, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Natascha Drude
- Department of Nuclear Medicine, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany.,Department of Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Agnieszka Morgenroth
- Department of Nuclear Medicine, University Hospital Aachen, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| |
Collapse
|
73
|
Jayakumar S, Patwardhan RS, Pal D, Singh B, Sharma D, Kutala VK, Sandur SK. Mitochondrial targeted curcumin exhibits anticancer effects through disruption of mitochondrial redox and modulation of TrxR2 activity. Free Radic Biol Med 2017; 113:530-538. [PMID: 29080841 DOI: 10.1016/j.freeradbiomed.2017.10.378] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/06/2017] [Accepted: 10/23/2017] [Indexed: 11/21/2022]
Abstract
Mitocurcumin is a derivative of curcumin, which has been shown to selectively enter mitochondria. Here we describe the anti-tumor efficacy of mitocurcumin in lung cancer cells and its mechanism of action. Mitocurcumin, showed 25-50 fold higher efficacy in killing lung cancer cells as compared to curcumin as demonstrated by clonogenic assay, flow cytometry and high throughput screening assay. Treatment of lung cancer cells with mitocurcumin significantly decreased the frequency of cancer stem cells. Mitocurcumin increased the mitochondrial reactive oxygen species (ROS), decreased the mitochondrial glutathione levels and induced strand breaks in the mitochondrial DNA. As a result, we observed increased BAX to BCL-2 ratio, cytochrome C release into the cytosol, loss of mitochondrial membrane potential and increased caspase-3 activity suggesting that mitocurcumin activates the intrinsic apoptotic pathway. Docking studies using mitocurcumin revealed that it binds to the active site of the mitochondrial thioredoxin reductase (TrxR2) with high affinity. In corroboration with the above finding, mitocurcumin decreased TrxR activity in cell free as well as the cellular system. The anti-cancer activity of mitocurcumin measured in terms of apoptotic cell death and the decrease in cancer stem cell frequency was accentuated by TrxR2 overexpression. This was due to modulation of TrxR2 activity to NADPH oxidase like activity by mitocurcumin, resulting in higher ROS accumulation and cell death. Thus, our findings reveal mitocurcumin as a potent anticancer agent with better efficacy than curcumin. This study also demonstrates the role of TrxR2 and mitochondrial DNA damage in mitocurcumin mediated killing of cancer cells.
Collapse
Affiliation(s)
- Sundarraj Jayakumar
- Radiation Biology & Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai, India
| | - Raghavendra S Patwardhan
- Radiation Biology & Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai, India
| | - Debojyoti Pal
- Radiation Biology & Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai, India
| | - Babita Singh
- Radiation Biology & Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai, India
| | - Deepak Sharma
- Radiation Biology & Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai, India; Homi Bhabha National Institute, Anushakti Nagar, Mumbai, India.
| | - Vijay Kumar Kutala
- Department of Clinical Pharmacology & Therapeutics, Nizam's Institute of Medical Sciences, Hyderabad, India
| | - Santosh Kumar Sandur
- Radiation Biology & Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre, Trombay, Mumbai, India; Homi Bhabha National Institute, Anushakti Nagar, Mumbai, India.
| |
Collapse
|
74
|
Jung N, Kwon HJ, Jung HJ. Downregulation of mitochondrial UQCRB inhibits cancer stem cell-like properties in glioblastoma. Int J Oncol 2017; 52:241-251. [PMID: 29115404 DOI: 10.3892/ijo.2017.4191] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 10/10/2017] [Indexed: 11/06/2022] Open
Abstract
Glioblastoma stem cell targeted therapies have become a powerful strategy for the treatment of this deadliest brain tumor. We demonstrate for the first time that downregulation of mitochondrial ubiquinol-cytochrome c reductase binding protein (UQCRB) inhibits the cancer stem cell-like properties in human glioblastoma cells. The synthetic small molecules targeting UQCRB significantly suppressed not only the self-renewal capacity such as growth and neurosphere formation, but also the metastatic potential such as migration and invasion of glioblastoma stem‑like cells (GSCs) derived from U87MG and U373MG at subtoxic concentrations. Notably, the UQCRB inhibitors repressed c‑Met-mediated downstream signal transduction and hypoxia‑inducible factor‑1α (HIF‑1α) activation, thereby reducing the expression levels of GSC markers including CD133, Nanog, Oct4 and Sox2 in the GSCs. Furthermore, the UQCRB inhibitors decreased mitochondrial ROS generation and mitochondrial membrane potential in the GSCs, indicating that they regulate the mitochondrial function in GSCs. Indeed, the knockdown of UQCRB gene by UQCRB siRNA significantly inhibited the cancer stem cell-like phenotypes as well as the expression of stemness markers by blocking mitochondrial ROS/HIF‑1α/c‑Met pathway in U87MG GSCs. These findings suggest that UQCRB and its inhibitors could be a new therapeutic target and lead compounds for eliminating cancer stem cells in glioblastoma.
Collapse
Affiliation(s)
- Narae Jung
- Department of BT-Convergent Pharmaceutical Engineering, Sun Moon University, Tangjeong-myeon, Asan-si, Chungnam 336-708, Republic of Korea
| | - Ho Jeong Kwon
- Department of Biotechnology, Yonsei University, Seodaemun-gu, Seoul 120-749, Republic of Korea
| | - Hye Jin Jung
- Department of BT-Convergent Pharmaceutical Engineering, Sun Moon University, Tangjeong-myeon, Asan-si, Chungnam 336-708, Republic of Korea
| |
Collapse
|
75
|
Murdolo G, Bartolini D, Tortoioli C, Piroddi M, Torquato P, Galli F. Selenium and Cancer Stem Cells. Adv Cancer Res 2017; 136:235-257. [PMID: 29054420 DOI: 10.1016/bs.acr.2017.07.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Selenium (Se) is an essential micronutrient that functions as "redox gatekeeper" and homeostasis factor of normal and cancer cells. Epidemiology and experimental studies, in the last years suggested that both inorganic and organic forms of Se may have favorable health effects. In this regard, a protective action of Se on cellular systems that may help preventing cancer cell differentiation has been demonstrated, while the hypothesis that Se compounds may cure cancer and its metastatic diffusion appears speculative and is still a matter of investigation. Indeed, the overall actions of Se compounds in carcinogenesis are controversial. The recognition that cancer is a stem cell disease instigated major paradigm shifts in our basic understanding of cancer and attracted a great deal of interest. Although current treatment approaches in cancer are grounded in the need to kill the majority of cancer cells, targeting cancer stem cells (CSCs) may hold great potential in improving cancer treatment. In this respect, Se compounds have been demonstrated modulating numerous signaling pathways involved in CSC biology and these findings are now stimulating further research on optimal Se concentrations, most effective and cancer-specific Se compounds, and inherent pathways involved in redox and metabolic regulation of CSCs. In this review, we summarize the current knowledge about the effects of Se compounds on CSCs, by focusing on redox-dependent pathways and main gene regulation checkpoints that affect self-renewal, differentiation, and migration responses in this subpopulation of cancer cells.
Collapse
Affiliation(s)
- Giuseppe Murdolo
- Section of Internal Medicine, Endocrine and Metabolic Sciences, University of Perugia, Perugia, Italy.
| | | | - Cristina Tortoioli
- Section of Internal Medicine, Endocrine and Metabolic Sciences, University of Perugia, Perugia, Italy
| | | | | | | |
Collapse
|
76
|
Polewski MD, Reveron-Thornton RF, Cherryholmes GA, Marinov GK, Aboody KS. SLC7A11 Overexpression in Glioblastoma Is Associated with Increased Cancer Stem Cell-Like Properties. Stem Cells Dev 2017; 26:1236-1246. [PMID: 28610554 DOI: 10.1089/scd.2017.0123] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
System xc- is a sodium-independent electroneutral transporter, comprising a catalytic subunit xCT (SLC7A11), which is involved in importing cystine. Certain cancers such as gliomas upregulate the expression of system xc-, which confers a survival advantage against the detrimental effects of reactive oxygen species (ROS) by increasing generation of the antioxidant glutathione. However, ROS have also been shown to function as targeted, intracellular second messengers in an array of physiological processes such as proliferation. Several studies have implicated ROS in important cancer features such as migration, invasion, and contribution to a cancer stem cell (CSC)-like phenotype. The role of system xc- in regulating these ROS-sensitive processes in glioblastoma multiforme (GBM), the most aggressive malignant primary brain tumor in adults, remains unknown. Stable SLC7A11 knockdown and overexpressing U251 glioma cells were generated and characterized to understand the role of redox and system xc- in glioma progression. SLC7A11 knockdown resulted in higher endogenous ROS levels and enhanced invasive properties. On the contrary, overexpression of SLC7A11 resulted in decreased endogenous ROS levels as well as decreased migration and invasion. However, SLC7A11-overexpressing cells displayed actin cytoskeleton changes reminiscent of epithelial-like cells and exhibited an increased CSC-like phenotype. The enhanced CSC-like phenotype may contribute to increased chemoresistance and suggests that overexpression of SLC7A11 in the context of GBM may contribute to tumor progression. These findings have important implications for cancer management where targeting system xC- in combination with other chemotherapeutics can reduce cancer resistance and recurrence and improve GBM patient survival.
Collapse
Affiliation(s)
- Monika D Polewski
- 1 Department of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute , Duarte, California.,2 Irell and Manella Graduate School of Biological Sciences, City of Hope National Medical Center and Beckman Research Institute , Duarte, California
| | - Rosyli F Reveron-Thornton
- 1 Department of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute , Duarte, California.,3 Department of Biological Sciences, California State University , San Bernardino, California
| | - Gregory A Cherryholmes
- 2 Irell and Manella Graduate School of Biological Sciences, City of Hope National Medical Center and Beckman Research Institute , Duarte, California.,4 Department of Cancer Immunotherapeutics and Tumor Immunology, City of Hope National Medical Center and Beckman Research Institute , Duarte, California
| | - Georgi K Marinov
- 5 Division of Biology, California Institute of Technology , Pasadena, California
| | - Karen S Aboody
- 1 Department of Developmental and Stem Cell Biology, City of Hope National Medical Center and Beckman Research Institute , Duarte, California.,6 Division of Neurosurgery, City of Hope National Medical Center and Beckman Research Institute , Duarte, California
| |
Collapse
|
77
|
Woo Y, Oh J, Kim JS. Suppression of Nrf2 Activity by Chestnut Leaf Extract Increases Chemosensitivity of Breast Cancer Stem Cells to Paclitaxel. Nutrients 2017; 9:nu9070760. [PMID: 28718813 PMCID: PMC5537874 DOI: 10.3390/nu9070760] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 07/11/2017] [Accepted: 07/13/2017] [Indexed: 02/07/2023] Open
Abstract
Due to metastatic potential and drug resistance, cancer stem cells (CSCs) have become a critical target for the development of chemotherapeutic agents. Recent studies showed that CSCs highly express NF-E2-related factor 2 (Nrf2)-mediated antioxidant enzymes and thereby retain relatively low levels of reactive oxygen species (ROS). Since anticancer agents usually utilize ROS as an arsenal for killing cancer cells, we hypothesized that inhibition of Nrf2 activity could increase the sensitivity of CSCs to anticancer drugs, and thus enhancing their therapeutic efficacy. We found that MCF-7-derived CSCs with a CD44high/CD24low phenotype formed mammospheres and highly expressed Nrf2 compared to the adherent parental MCF-7 cells. In a separate experiment, we screened 89 different edible plant extracts for inhibitory activity against the Nrf2 signaling pathway by using an antioxidant response element (ARE)-luciferase assay system. Among those extracts, Castanea crenata (chestnut) leaf extract significantly decreased the nuclear translocation of Nrf2 and protein expression of antioxidant enzymes in MCF-7-derived CSCs. The combined treatment of the CSCs with chestnut leaf extract and paclitaxel resulted in more effective cell death than the treatment with paclitaxel alone. These findings suggest that the chestnut leaf extract or its constituents could increase the susceptibility of breast CSCs to an anticancer drug, paclitaxel, through inhibition of the Nrf2 signaling pathway, and could be utilized as an adjuvant for chemotherapy.
Collapse
Affiliation(s)
- Yaejin Woo
- School of Food Science and Biotechnology (BK21 Plus), Kyungpook National University, Daegu 41566, Korea.
| | - Jisun Oh
- School of Food Science and Biotechnology (BK21 Plus), Kyungpook National University, Daegu 41566, Korea.
| | - Jong-Sang Kim
- School of Food Science and Biotechnology (BK21 Plus), Kyungpook National University, Daegu 41566, Korea.
- Institute of Agricultural Science & Technology, Kyungpook National University, Daegu 41566, Korea.
| |
Collapse
|
78
|
188Re-Liposome Can Induce Mitochondrial Autophagy and Reverse Drug Resistance for Ovarian Cancer: From Bench Evidence to Preliminary Clinical Proof-of-Concept. Int J Mol Sci 2017; 18:ijms18050903. [PMID: 28441355 PMCID: PMC5454816 DOI: 10.3390/ijms18050903] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 04/17/2017] [Accepted: 04/20/2017] [Indexed: 12/13/2022] Open
Abstract
Despite standard treatment, about 70% of ovarian cancer will recur. Cancer stem cells (CSCs) have been implicated in the drug-resistance mechanism. Several drug resistance mechanisms have been proposed, and among these, autophagy plays a crucial role for the maintenance and tumorigenicity of CSCs. Compared to their differentiated counterparts, CSCs have been demonstrated to display a significantly higher level of autophagy flux. Moreover, mitophagy, a specific type of autophagy that selectively degrades excessive or damaged mitochondria, is shown to contribute to cancer progression and recurrence in several types of tumors. Nanomedicine has been shown to tackle the CSCs problem by overcoming drug resistance. In this work, we developed a nanomedicine, 188Re-liposome, which was demonstrated to target autophagy and mitophagy in the tumor microenvironment. Of note, the inhibition of autophagy and mitophagy could lead to significant tumor inhibition in two xenograft animal models. Lastly, we presented two cases of recurrent ovarian cancer, both in drug resistance status that received a level I dose from a phase I clinical trial. Both cases developing drug resistance showed drug sensitivity to 188Re-liposome. These results suggest that inhibition of autophagy and mitophagy by a nanomedicine may be a novel strategy to overcome drug resistance in ovarian cancer.
Collapse
|
79
|
Time- and cell-resolved dynamics of redox-sensitive Nrf2, HIF and NF-κB activities in 3D spheroids enriched for cancer stem cells. Redox Biol 2017; 12:403-409. [PMID: 28319891 PMCID: PMC5357678 DOI: 10.1016/j.redox.2017.03.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/06/2017] [Accepted: 03/09/2017] [Indexed: 12/18/2022] Open
Abstract
Cancer cells have an altered redox status, with changes in intracellular signaling pathways. The knowledge of how such processes are regulated in 3D spheroids, being well-established tumor models, is limited. To approach this question we stably transfected HCT116 cells with a pTRAF reporter that enabled time- and cell-resolved activity monitoring of three redox-regulated transcription factors Nrf2, HIF and NF-κB in spheroids enriched for cancer stem cells. At the first day of spheroid formation, these transcription factors were activated and thereafter became repressed. After about a week, both HIF and Nrf2 were reactivated within the spheroid cores. Further amplifying HIF activation in spheroids by treatment with DMOG resulted in a dominant quiescent stem-cell-like phenotype, with high resistance to stress-inducing treatments. Auranofin, triggering oxidative stress and Nrf2 activation, had opposite effects with increased differentiation and proliferation. These novel high-resolution insights into spatiotemporal activation patterns demonstrate a striking coordination of redox regulated transcription factors within spheroids not occurring in conventional cell culture models.
Collapse
|
80
|
Abstract
SIGNIFICANCE There are a number of redox-active anticancer agents currently in development based on the premise that altered redox homeostasis is necessary for cancer cell's survival. Recent Advances: This review focuses on the relatively few agents that target cellular redox homeostasis to have entered clinical trial as anticancer drugs. CRITICAL ISSUES The success rate of redox anticancer drugs has been disappointing compared to other classes of anticancer agents. This is due, in part, to our incomplete understanding of the functions of the redox targets in normal and cancer tissues, leading to off-target toxicities and low therapeutic indexes of the drugs. The field also lags behind in the use biomarkers and other means to select patients who are most likely to respond to redox-targeted therapy. FUTURE DIRECTIONS If we wish to derive clinical benefit from agents that attack redox targets, then the future will require a more sophisticated understanding of the role of redox targets in cancer and the increased application of personalized medicine principles for their use. Antioxid. Redox Signal. 26, 262-273.
Collapse
Affiliation(s)
| | - Garth Powis
- 2 Sanford Burnham Prebys Medical Discovery Institute Cancer Center , La Jolla, California
| |
Collapse
|
81
|
Qureshi-Baig K, Ullmann P, Haan S, Letellier E. Tumor-Initiating Cells: a criTICal review of isolation approaches and new challenges in targeting strategies. Mol Cancer 2017; 16:40. [PMID: 28209178 PMCID: PMC5314476 DOI: 10.1186/s12943-017-0602-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/20/2017] [Indexed: 02/07/2023] Open
Abstract
Most cancers contain a subpopulation of highly tumorigenic cells, known as cancer stem cells (CSCs) or tumor-initiating cells (TICs). Targeting TICs may be essential to achieve cure, because of their self-renewal and tumorigenic properties as well as their resistance to conventional therapies. Despite significant advances in TIC biology, their isolation and identification remain largely disputed and incompletely established. In this review, we discuss the latest developments in isolation and culturing approaches of TICs, with focus on colorectal cancer (CRC). We feature recent findings on TIC-relevant signaling pathways and the metabolic identity of TICs, as well as their current clinical implications. Lastly, we highlight the influence of inter- and intra-tumoral heterogeneity on TIC function and targeting approaches.
Collapse
Affiliation(s)
- Komal Qureshi-Baig
- Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, 6, Avenue du Swing, L-4367, Campus Belval, Belvaux, Luxembourg
| | - Pit Ullmann
- Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, 6, Avenue du Swing, L-4367, Campus Belval, Belvaux, Luxembourg
| | - Serge Haan
- Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, 6, Avenue du Swing, L-4367, Campus Belval, Belvaux, Luxembourg
| | - Elisabeth Letellier
- Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, 6, Avenue du Swing, L-4367, Campus Belval, Belvaux, Luxembourg.
| |
Collapse
|
82
|
Choi YJ, Park JH, Han JW, Kim E, Jae-Wook O, Lee SY, Kim JH, Gurunathan S. Differential Cytotoxic Potential of Silver Nanoparticles in Human Ovarian Cancer Cells and Ovarian Cancer Stem Cells. Int J Mol Sci 2016; 17:E2077. [PMID: 27973444 PMCID: PMC5187877 DOI: 10.3390/ijms17122077] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 11/17/2016] [Accepted: 11/30/2016] [Indexed: 01/13/2023] Open
Abstract
The cancer stem cell (CSC) hypothesis postulates that cancer cells are composed of hierarchically-organized subpopulations of cells with distinct phenotypes and tumorigenic capacities. As a result, CSCs have been suggested as a source of disease recurrence. Recently, silver nanoparticles (AgNPs) have been used as antimicrobial, disinfectant, and antitumor agents. However, there is no study reporting the effects of AgNPs on ovarian cancer stem cells (OvCSCs). In this study, we investigated the cytotoxic effects of AgNPs and their mechanism of causing cell death in A2780 (human ovarian cancer cells) and OvCSCs derived from A2780. In order to examine these effects, OvCSCs were isolated and characterized using positive CSC markers including aldehyde dehydrogenase (ALDH) and CD133 by fluorescence-activated cell sorting (FACS). The anticancer properties of the AgNPs were evaluated by assessing cell viability, leakage of lactate dehydrogenase (LDH), reactive oxygen species (ROS), and mitochondrial membrane potential (mt-MP). The inhibitory effect of AgNPs on the growth of ovarian cancer cells and OvCSCs was evaluated using a clonogenic assay. Following 1-2 weeks of incubation with the AgNPs, the numbers of A2780 (bulk cells) and ALDH⁺/CD133⁺ colonies were significantly reduced. The expression of apoptotic and anti-apoptotic genes was measured by real-time quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). Our observations showed that treatment with AgNPs resulted in severe cytotoxicity in both ovarian cancer cells and OvCSCs. In particular, AgNPs showed significant cytotoxic potential in ALDH⁺/CD133⁺ subpopulations of cells compared with other subpopulation of cells and also human ovarian cancer cells (bulk cells). These findings suggest that AgNPs can be utilized in the development of novel nanotherapeutic molecules for the treatment of ovarian cancers by specific targeting of the ALDH⁺/CD133⁺ subpopulation of cells.
Collapse
Affiliation(s)
- Yun-Jung Choi
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 143-701, Korea.
| | - Jung-Hyun Park
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 143-701, Korea.
| | - Jae Woong Han
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 143-701, Korea.
| | - Eunsu Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 143-701, Korea.
| | - Oh Jae-Wook
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 143-701, Korea.
| | - Seung Yoon Lee
- Swine Consulting Group, HanByol Farm Tech, Gyeonggi 463-785, Korea.
| | - Jin-Hoi Kim
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 143-701, Korea.
| | - Sangiliyandi Gurunathan
- Department of Stem Cell and Regenerative Biotechnology, Konkuk University, Seoul 143-701, Korea.
| |
Collapse
|
83
|
Li Z, Qin B, Qi X, Mao J, Wu D. Isoalantolactone induces apoptosis in human breast cancer cells via ROS-mediated mitochondrial pathway and downregulation of SIRT1. Arch Pharm Res 2016; 39:1441-1453. [DOI: 10.1007/s12272-016-0815-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 08/09/2016] [Indexed: 02/06/2023]
|
84
|
Cancer Stem Cell Quiescence and Plasticity as Major Challenges in Cancer Therapy. Stem Cells Int 2016; 2016:1740936. [PMID: 27418931 PMCID: PMC4932171 DOI: 10.1155/2016/1740936] [Citation(s) in RCA: 257] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/15/2016] [Indexed: 02/06/2023] Open
Abstract
Cells with stem-like properties, tumorigenic potential, and treatment-resistant phenotypes have been identified in many human malignancies. Based on the properties they share with nonneoplastic stem cells or their ability to initiate and propagate tumors in vivo, such cells were designated as cancer stem (stem-like) or tumor initiating/propagating cells. Owing to their implication in treatment resistance, cancer stem cells (CSCs) have been the subject of intense investigation in past years. Comprehension of CSCs' intrinsic properties and mechanisms they develop to survive and even enhance their aggressive phenotype within the hostile conditions of the tumor microenvironment has reoriented therapeutic strategies to fight cancer. This report provides selected examples of malignancies in which the presence of CSCs has been evidenced and briefly discusses methods to identify, isolate, and functionally characterize the CSC subpopulation of cancer cells. Relevant biological targets in CSCs, their link to treatment resistance, proposed targeting strategies, and limitations of these approaches are presented. Two major aspects of CSC physiopathology, namely, relative in vivo quiescence and plasticity in response to microenvironmental cues or treatment, are highlighted. Implications of these findings in the context of the development of new therapies are discussed.
Collapse
|
85
|
Somasundaram V, Hemalatha SK, Pal K, Sinha S, Nair AS, Mukhopadhyay D, Srinivas P. Selective mode of action of plumbagin through BRCA1 deficient breast cancer stem cells. BMC Cancer 2016; 16:336. [PMID: 27229859 PMCID: PMC4882782 DOI: 10.1186/s12885-016-2372-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 05/19/2016] [Indexed: 12/16/2022] Open
Abstract
Background Studies over the past decade and half have identified cancer stem cells (CSCs) to be responsible for tumorigenesis, invasion, sustenance of metastatic disease, radio- and chemo-resistance and tumor relapse. Recent reports have described the plasticity of breast CSCs (BCSCs) to shift between the epithelial and mesenchymal phenotypes via Epithelial-Mesenchymal Transition (EMT) and Mesenchymal-Epithelial Transition (MET) states as the reason for their invasive capabilities. Additionally, BRCA1 has been found to be a mammary stem cell fate determinant. However, it is not clear what would be the best marker that can be used for identifying CSCs in BRCA1 mutated cancers. Also, anticancer agents that can reduce CSC population in a BRCA1 defective condition have not been addressed so far. Methods Putative BCSCs were identified based on Hoechst exclusion, CD44+/24–/low expression and Aldehyde Dehydrogenase 1 (ALDH1) positivity using flow cytometry. The ‘stemness’ of the isolated ALDH1+ cells were analysed by immunofluorescence, western blotting for stem cell and EMT markers as well as in vitro mammosphere assays. Induction of Reactive Oxygen Species (ROS) by Plumbagin (PB) in BCSCs was assayed by Dichloro-dihydro-fluorescein diacetate (DCF-DA) staining. Ovarian cancer xenografts treated with PB were subjected to immunohistochemical analysis to study the ability of PB to target CSCs. Results We have confirmed that ALDH1 positivity is the best marker for the identification of BCSCs in BRCA1-defective breast cancer cell lines when compared to the CD marker profile and Side Population (SP) analysis. BRCA1 status was observed to be a determinant of the abundance of epithelial-like (ALDH1+) or mesenchymal-like (CD44+/24–/low) BCSCs, and the reconstitution of a full length, wild type BRCA1 in HCC1937 breast cancer cells possessing a mutated BRCA1, transforms them from ‘stem-like’ to more ‘mesenchymal’. For the first time we have shown that Plumbagin (PB), a naturally occurring naphthoquinone which is predominantly a ROS inducer, could reduce BCSCs specifically in BRCA1-defective, basal-like cancer cells. Conclusions The best marker for identifying BCSCs in BRCA1 defective condition could be ALDH1 and that BRCA1 mutated BCSCs would be mostly ‘stem like’ than ‘mesenchymal’. Also ROS inducers like PB could reduce BCSCs in BRCA1 defective cancers. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2372-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Veena Somasundaram
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India.,Present address: Center for Cancer Research, National Cancer Institute, Building 567, Room 254, Frederick, MD, 21702-1201, USA
| | - Sreelatha K Hemalatha
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - Krishnendu Pal
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| | - Sutapa Sinha
- Department of Hematology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| | - Asha S Nair
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - Debabrata Mukhopadhyay
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| | - Priya Srinivas
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India.
| |
Collapse
|
86
|
Lang M, Wang X, Wang H, Dong J, Lan C, Hao J, Huang C, Li X, Yu M, Yang Y, Yang S, Ren H. Arsenic trioxide plus PX-478 achieves effective treatment in pancreatic ductal adenocarcinoma. Cancer Lett 2016; 378:87-96. [PMID: 27212442 DOI: 10.1016/j.canlet.2016.05.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Revised: 05/12/2016] [Accepted: 05/17/2016] [Indexed: 02/06/2023]
Abstract
Arsenic trioxide (ATO) has been selected as a promising treatment not only in leukemia but also in solid tumors. Previous studies showed that the cytotoxicity of ATO mainly depends on the induction of reactive oxygen species. However, ATO has only achieved a modest effect in pancreatic ductal adenocarcinoma, suggesting that the existing radical scavenging proteins, such as hypoxia inducible factor-1, attenuate the effect. The goal of this study is to investigate the effect of combination treatment of ATO plus PX-478 (hypoxia-inducible factor-1 inhibitor) and its underlying mechanism. Here, we showed that PX-478 robustly strengthened the anti-growth and pro-apoptosis effect of ATO on Panc-1 and BxPC-3 pancreatic cancer cells in vitro. Meanwhile, in vivo mouse xenograft models also showed the synergistic effect of ATO plus PX-478 compared with any single agent. Further studies showed that the anti-tumor effect of ATO plus PX-478 was derived from the reactive oxygen species-induced apoptosis. We next confirmed that Hypoxia-inducible factor-1 cleared reactive oxygen species by its downstream target, forkhead box O transcription factors, and this effect may justify the strategy of ATO plus PX-478 in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Mingxiao Lang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xiuchao Wang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Hongwei Wang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jie Dong
- School of Public Health, Tianjin Medical University, Tianjin, China
| | - Chungen Lan
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Jihui Hao
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Chongbiao Huang
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xin Li
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Ming Yu
- School of Public Health, Tianjin Medical University, Tianjin, China
| | - Yanhui Yang
- 2011 Collaborative Innovation Center of Tianjin for Medical Epigenetics, Key Laboratory of Hormones and Development (Ministry of Health), Metabolic Diseases Hospital & Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Shengyu Yang
- Department of Tumor Biology and Comprehensive Melanoma Research Center, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - He Ren
- National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Department of Pancreatic Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
| |
Collapse
|
87
|
Pacini N, Borziani F. Oncostatic-Cytoprotective Effect of Melatonin and Other Bioactive Molecules: A Common Target in Mitochondrial Respiration. Int J Mol Sci 2016; 17:341. [PMID: 26959015 PMCID: PMC4813203 DOI: 10.3390/ijms17030341] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2015] [Revised: 11/27/2015] [Accepted: 12/03/2015] [Indexed: 12/27/2022] Open
Abstract
For several years, oncostatic and antiproliferative properties, as well as thoses of cell death induction through 5-methoxy-N-acetiltryptamine or melatonin treatment, have been known. Paradoxically, its remarkable scavenger, cytoprotective and anti-apoptotic characteristics in neurodegeneration models, such as Alzheimer’s disease and Parkinson’s disease are known too. Analogous results have been confirmed by a large literature to be associated to the use of many other bioactive molecules such as resveratrol, tocopherol derivatives or vitamin E and others. It is interesting to note that the two opposite situations, namely the neoplastic pathology and the neurodegeneration, are characterized by deep alterations of the metabolome, of mitochondrial function and of oxygen consumption, so that the oncostatic and cytoprotective action can find a potential rationalization because of the different metabolic and mitochondrial situations, and in the effect that these molecules exercise on the mitochondrial function. In this review we discuss historical and general aspects of melatonin, relations between cancers and the metabolome and between neurodegeneration and the metabolome, and the possible effects of melatonin and of other bioactive molecules on metabolic and mitochondrial dynamics. Finally, we suggest a common general mechanism as responsible for the oncostatic/cytoprotective effect of melatonin and of other molecules examined.
Collapse
Affiliation(s)
- Nicola Pacini
- Laboratorio Privato di Biochimica F. Pacini, via trabocchetto 10, 89126 Reggio Calabria, Italy.
| | - Fabio Borziani
- Laboratorio Privato di Biochimica F. Pacini, via trabocchetto 10, 89126 Reggio Calabria, Italy.
| |
Collapse
|
88
|
Di Francesco AM, Toesca A, Cenciarelli C, Giordano A, Gasbarrini A, Puglisi MA. Metabolic Modification in Gastrointestinal Cancer Stem Cells: Characteristics and Therapeutic Approaches. J Cell Physiol 2016; 231:2081-7. [PMID: 26791139 DOI: 10.1002/jcp.25318] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Accepted: 01/20/2016] [Indexed: 12/19/2022]
Abstract
Currently, there is much interest in the characterization of metabolic profiling of cancer stem cells (CSCs), a small subset of tumor cells with self-renewal capacity. Indeed, ever-growing evidence indicate that metabolism and stemness are highly intertwined processes in tumor tissue. In this review, we analyze the potential metabolic targeting strategies for eradicating CSCs that could help to develop a more effective therapeutic approach for gastrointestinal cancers. Indeed, the successful elimination of a tumor requires an anticancer therapy that affects both cancer cells and CSCs. The observation that gastrointestinal CSCs possess higher inducible nitric oxide sinthase (iNOS) expression, lower reactive oxygen species (ROS) production, and a different metabolism respect to no-CSCs tumor cells has paved the way to develop drugs targeting CSC specific signaling. In particular, several studies have highlighted that metformin, aldehyde dehydrogenase 1, and iNOS inhibitors selectively suppressed CSC growth and that combinatorial therapy of them with standard chemotherapeutic drugs had a synergistic effect resulting in reduced tumor burden and delayed tumor recurrence. Thus, the possibility of combining specific CSC metabolism inhibitors with existing therapeutic approaches could have profound anticancer effects, changing the conventional treatment approaches to gastrointestinal cancers. J. Cell. Physiol. 231: 2081-2087, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
| | - Amelia Toesca
- Institute of Human Anatomy and Cell Biology, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Carlo Cenciarelli
- Institute of Translational Pharmacology-National Research Council, Rome, Italy
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, Temple University, Philadelphia, Pennsylvania
| | - Antonio Gasbarrini
- Department of Internal Medicine and Gastroenterology, Gemelli Hospital, Rome, Italy
| | | |
Collapse
|
89
|
Emulsions Made of Oils from Seeds of GM Flax Protect V79 Cells against Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:7510759. [PMID: 26779302 PMCID: PMC4686677 DOI: 10.1155/2016/7510759] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 08/21/2015] [Accepted: 08/31/2015] [Indexed: 01/12/2023]
Abstract
Polyunsaturated fatty acids, sterols, and hydrophilic phenolic compounds are components of flax oil that act as antioxidants. We investigated the impact of flax oil from transgenic flax in the form of emulsions on stressed Chinese hamster pulmonary fibroblasts. We found that the emulsions protect V79 cells against the H2O2 and the effect is dose dependent. They reduced the level of intracellular reactive oxygen species and protected genomic DNA against damage. The rate of cell proliferation increased upon treatment with the emulsions at a low concentration, while at a high concentration it decreased significantly, accompanied by increased frequency of apoptotic cell death. Expression analysis of selected genes revealed the upregulatory impact of the emulsions on the histones, acetylases, and deacetylases. Expression of apoptotic, proinflammatory, and anti-inflammatory genes was also altered. It is thus suggested that flax oil emulsions might be useful as a basis for biomedical products that actively protect cells against inflammation and degeneration. The beneficial effect on fibroblast resistance to oxidative damage was superior in the emulsion made of oil from transgenic plants which was correlated with the quantity of antioxidants and squalene. The emulsions from transgenic flax are promising candidates for skin protection against oxidative damage.
Collapse
|
90
|
Lyakhovich A, Lleonart ME. Bypassing Mechanisms of Mitochondria-Mediated Cancer Stem Cells Resistance to Chemo- and Radiotherapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2016:1716341. [PMID: 26697128 PMCID: PMC4677234 DOI: 10.1155/2016/1716341] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 08/24/2015] [Accepted: 08/25/2015] [Indexed: 01/03/2023]
Abstract
Cancer stem cells (CSCs) are highly resistant to conventional chemo- and radiotherapeutic regimes. Therefore, the multiple drug resistance (MDR) of cancer is most likely due to the resistance of CSCs. Such resistance can be attributed to some bypassing pathways including detoxification mechanisms of reactive oxygen and nitrogen species (RO/NS) formation or enhanced autophagy. Unlike in normal cells, where RO/NS concentration is maintained at certain threshold required for signal transduction or immune response mechanisms, CSCs may develop alternative pathways to diminish RO/NS levels leading to cancer survival. In this minireview, we will focus on elaborated mechanisms developed by CSCs to attenuate high RO/NS levels. Gaining a better insight into the mechanisms of stem cell resistance to chemo- or radiotherapy may lead to new therapeutic targets thus serving for better anticancer strategies.
Collapse
Affiliation(s)
- Alex Lyakhovich
- International Clinical Research Center, St. Anne's University Hospital, Masaryk University, Kamenice 5/A7, 625 00 Brno, Czech Republic
- Institute of Molecular Biology and Biophysics, Novosibirsk, Russia
| | - Matilde E. Lleonart
- Oncology and Pathology Group, Institut de Recerca Hospital Vall d'Hebron, Passeig Vall d'Hebron 119-129, 08035 Barcelona, Spain
| |
Collapse
|
91
|
Manda G, Isvoranu G, Comanescu MV, Manea A, Debelec Butuner B, Korkmaz KS. The redox biology network in cancer pathophysiology and therapeutics. Redox Biol 2015; 5:347-357. [PMID: 26122399 PMCID: PMC4501561 DOI: 10.1016/j.redox.2015.06.014] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 06/22/2015] [Accepted: 06/23/2015] [Indexed: 12/21/2022] Open
Abstract
The review pinpoints operational concepts related to the redox biology network applied to the pathophysiology and therapeutics of solid tumors. A sophisticated network of intrinsic and extrinsic cues, integrated in the tumor niche, drives tumorigenesis and tumor progression. Critical mutations and distorted redox signaling pathways orchestrate pathologic events inside cancer cells, resulting in resistance to stress and death signals, aberrant proliferation and efficient repair mechanisms. Additionally, the complex inter-cellular crosstalk within the tumor niche, mediated by cytokines, redox-sensitive danger signals (HMGB1) and exosomes, under the pressure of multiple stresses (oxidative, inflammatory, metabolic), greatly contributes to the malignant phenotype. The tumor-associated inflammatory stress and its suppressive action on the anti-tumor immune response are highlighted. We further emphasize that ROS may act either as supporter or enemy of cancer cells, depending on the context. Oxidative stress-based therapies, such as radiotherapy and photodynamic therapy, take advantage of the cytotoxic face of ROS for killing tumor cells by a non-physiologically sudden, localized and intense oxidative burst. The type of tumor cell death elicited by these therapies is discussed. Therapy outcome depends on the differential sensitivity to oxidative stress of particular tumor cells, such as cancer stem cells, and therefore co-therapies that transiently down-regulate their intrinsic antioxidant system hold great promise. We draw attention on the consequences of the damage signals delivered by oxidative stress-injured cells to neighboring and distant cells, and emphasize the benefits of therapeutically triggered immunologic cell death in metastatic cancer. An integrative approach should be applied when designing therapeutic strategies in cancer, taking into consideration the mutational, metabolic, inflammatory and oxidative status of tumor cells, cellular heterogeneity and the hypoxia map in the tumor niche, along with the adjoining and systemic effects of oxidative stress-based therapies. Critical point mutations and distorted redox-sensitive signaling pathways underlie the tumorigenic phenotype. Inter-cellular crosstalk under stress conditions in the tumor niche drives the behavior of tumor cells. ROS may act as either as supporter or enemy of tumor cells, depending on the context. Oxidative stress-injured cells deliver danger signals to neighboring and distant cells, hence dictating the outcome of therapy in cancer.
Collapse
Affiliation(s)
- Gina Manda
- Cellular and Molecular Medicine Department, Radiobiology Laboratory, "Victor Babes" National Institute of Pathology, Bucharest, Romania.
| | - Gheorghita Isvoranu
- Cellular and Molecular Medicine Department, Radiobiology Laboratory, "Victor Babes" National Institute of Pathology, Bucharest, Romania
| | - Maria Victoria Comanescu
- Cellular and Molecular Medicine Department, Radiobiology Laboratory, "Victor Babes" National Institute of Pathology, Bucharest, Romania
| | - Adrian Manea
- Cellular and Molecular Pharmacology Laboratory, Institute of Cellular Biology and Pathology "Nicolae Simionescu", Bucharest, Romania
| | - Bilge Debelec Butuner
- Faculty of Pharmacy, Department of Pharmaceutical Biotechnology, Ege University, Izmir, Turkey
| | - Kemal Sami Korkmaz
- Department of Bioengineering, Cancer Biology Laboratory, Ege University, İzmir, Turkey
| |
Collapse
|