1
|
Ochiai Y, Suzuki-Karasaki M, Ando T, Suzuki-Karasaki M, Nakayama H, Suzuki-Karasaki Y. Nitric oxide-dependent cell death in glioblastoma and squamous cell carcinoma via prodeath mitochondrial clustering. Eur J Cell Biol 2024; 103:151422. [PMID: 38795505 DOI: 10.1016/j.ejcb.2024.151422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 05/16/2024] [Accepted: 05/17/2024] [Indexed: 05/28/2024] Open
Abstract
Besides the fission-fusion dynamics, the cellular distribution of mitochondria has recently emerged as a critical biological parameter in regulating mitochondrial function and cell survival. We previously found that mitochondrial clustering on the nuclear periphery, or monopolar perinuclear mitochondrial clustering (MPMC), accompanies the anticancer activity of air plasma-activated medium (APAM) against glioblastoma and human squamous cell carcinoma, which is closely associated with oxidant-dependent tubulin remodeling and mitochondrial fragmentation. Accordingly, this study investigated the regulatory roles of nitric oxide (NO) in the anticancer activity of APAM. Time-lapse analysis revealed a time-dependent increase in NO accompanied by MPMC. In contrast, APAM caused minimal increases in MPMC and NO levels in nontransformed cells. NO, hydroxyl radicals, and lipid peroxide levels increased near the damaged nuclear periphery, possibly within mitochondria. NO scavenging prevented tubulin remodeling, MPMC, perinuclear oxidant production, nuclear damage, and cell death. Conversely, synthetic NO donors augmented all the prodeath events and acted synergistically with APAM. Salinomycin, an emerging drug against multidrug-resistant cancers, had similar NO-dependent effects. These results suggest that APAM and salinomycin induce NO-dependent cell death, where MPMC and oxidative mitochondria play critical roles. Our findings encourage further investigations on MPMC as a potential target for NO-driven anticancer agents against drug-resistant cancers.
Collapse
Affiliation(s)
- Yushi Ochiai
- Department of Research and Development, Plasma ChemiBio Laboratory, Nasushiobara, Tochigi Japan
| | - Manami Suzuki-Karasaki
- Department of Research and Development, Plasma ChemiBio Laboratory, Nasushiobara, Tochigi Japan; Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takashi Ando
- Department of Research and Development, Plasma ChemiBio Laboratory, Nasushiobara, Tochigi Japan
| | - Miki Suzuki-Karasaki
- Department of Research and Development, Plasma ChemiBio Laboratory, Nasushiobara, Tochigi Japan
| | - Hideki Nakayama
- Department of Oral and Maxillofacial Surgery, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | | |
Collapse
|
2
|
Khizar H, Hu Y, Wu Y, Yang J. The role and implication of autophagy in cholangiocarcinoma. Cell Death Discov 2023; 9:332. [PMID: 37666811 PMCID: PMC10477247 DOI: 10.1038/s41420-023-01631-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/13/2023] [Accepted: 08/24/2023] [Indexed: 09/06/2023] Open
Abstract
Cholangiocarcinoma (CCA) is a malignant tumor that originates from the biliary epithelial cells. It is characterized by a difficult diagnosis and limited treatment options. Autophagy is a cellular survival mechanism that maintains nutrient and energy homeostasis and eliminates intracellular pathogens. It is involved in various physiological and pathological processes, including the development of cancer. However, the role, mechanism, and potential therapeutic targets of autophagy in CCA have not been thoroughly studied. In this review, we introduce the classification, characteristics, process, and related regulatory genes of autophagy. We summarize the regulation of autophagy on the progression of CCA and collect the latest research progress on some autophagy modulators with clinical potential in CCA. In conclusion, combining autophagy modulators with immunotherapy, chemotherapy, and targeted therapy has great potential in the treatment of CCA. This combination may be a potential therapeutic target for CCA in the future.
Collapse
Affiliation(s)
- Hayat Khizar
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of medicine, 310006, Hangzhou, Zhejiang, China
- Department of Oncology, The Fourth Affiliated Hospital, International Institute of Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Yufei Hu
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of medicine, 310006, Hangzhou, Zhejiang, China
- Department of Gastroenterology, The Fourth School of Clinical medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Yanhua Wu
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of medicine, 310006, Hangzhou, Zhejiang, China
- Department of Gastroenterology, The Fourth School of Clinical medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jianfeng Yang
- Department of Gastroenterology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of medicine, 310006, Hangzhou, Zhejiang, China.
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, 310006, Hangzhou, Zhejiang, China.
- Key Laboratory of Integrated Traditional Chinese and Western Medicine for Biliary and Pancreatic Diseases of Zhejiang Province, 310006, Hangzhou, Zhejiang, China.
- Hangzhou Institute of Digestive Diseases, 310006, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Xing L, Tang Y, Li L, Tao X. ROS in hepatocellular carcinoma: What we know. Arch Biochem Biophys 2023:109699. [PMID: 37499994 DOI: 10.1016/j.abb.2023.109699] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/07/2023] [Accepted: 07/21/2023] [Indexed: 07/29/2023]
Abstract
Hepatocellular carcinoma (HCC), which is a primary liver cancer subtype, has a poor prognosis due to its high degree of malignancy. The lack of early diagnosis makes systemic therapy the only hope for HCC patients with advanced disease; however, resistance to drugs is a major obstacle. In recent years, targeted molecular therapy has gained popularity as a potential treatment for HCC. An increase in reactive oxygen species (ROS), which are cancer markers and a potential target for HCC therapy, can both promote and inhibit the disease. At present, many studies have examined targeted regulation of ROS in the treatment of HCC. Here, we reviewed the latest drugs that are still in the experimental stage, including nanocarrier drugs, exosome drugs, antibody drugs, aptamer drugs and polysaccharide drugs, to provide new hope for the clinical treatment of HCC patients.
Collapse
Affiliation(s)
- Lin Xing
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China; School of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Yuting Tang
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China; School of Pharmacy, Dalian Medical University, Dalian, 116044, China
| | - Lu Li
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| | - Xufeng Tao
- Department of Pharmacy, First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| |
Collapse
|
4
|
de Ávila Narciso Gomes R, Marmolejo-Garza A, Haan FJ, García TM, Chen T, Mauthe M, Moreira Franco Parisotto YE, Murakami MM, Marie SKN, Baptista MS, Dolga AM, Trombetta-Lima M. Mitochondrial dysfunction mediates neuronal cell response to DMMB photodynamic therapy. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2023; 1870:119429. [PMID: 36608805 DOI: 10.1016/j.bbamcr.2022.119429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 12/20/2022] [Accepted: 12/29/2022] [Indexed: 01/05/2023]
Abstract
Photodynamic therapy (PDT) is a process in which a photosensitizer (PS) is exposed to specific wavelengths and generates reactive oxygen species (ROS) which act within nanometers. The low invasive nature and directed cytotoxicity of this approach render it attractive to the treatment of different conditions, including the ones that affect the central nervous system (CNS). The effect of PDT on healthy neurons is one main concern over its use in the CNS, since neuronal-like cells were shown to be particularly sensitive to certain PSs. Among available PSs, 1,9-dimethyl-methylene blue (DMMB) stands out as being resistant to reduction to its inactive leuco form and by being able to produce high levels of singlet‑oxygen. In this study, we aimed to investigate DMMB photodamage mechanisms in the hippocampal cell line HT22. Our results demonstrate that DMMB-PDT decrease in cell viability was linked with an increase in cell death and overall ROS production. Besides, it resulted in a significant increase in mitochondrial ROS production and decreased mitochondria membrane potential. Furthermore, DMMB-PDT significantly increased the presence of acidic autolysosomes, which was accompanied by an increase in ATG1 and ATG8 homologue GaBarap1 expression, and decreased DRAM1 expression. Taken together our results indicated that mitochondrial and autophagic dysfunction underlie DMMB-PDT cytotoxicity in neuronal cells.
Collapse
Affiliation(s)
- Raphael de Ávila Narciso Gomes
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, the Netherlands; Chemistry Institute, Biochemistry Department, University of São Paulo (USP), 05508-000 São Paulo, Brazil
| | - Alejandro Marmolejo-Garza
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, the Netherlands; Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, the Netherlands
| | - Floris-Jan Haan
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, the Netherlands
| | - Teresa Mitchell García
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, the Netherlands
| | - Tingting Chen
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, the Netherlands
| | - Mario Mauthe
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Cell Biology, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, the Netherlands
| | | | - Mario Minor Murakami
- Medical School, Neurology Department, University of São Paulo (USP), 01246903 São Paulo, Brazil
| | | | - Maurício S Baptista
- Chemistry Institute, Biochemistry Department, University of São Paulo (USP), 05508-000 São Paulo, Brazil
| | - Amalia M Dolga
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, the Netherlands.
| | - Marina Trombetta-Lima
- Faculty of Science and Engineering, Department of Molecular Pharmacology, Groningen Research Institute of Pharmacy (GRIP), University of Groningen, 9713 AV Groningen, the Netherlands; Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, 9713 AV Groningen, the Netherlands.
| |
Collapse
|
5
|
Shah H, Stankov M, Panayotova-Dimitrova D, Yazdi A, Budida R, Klusmann JH, Behrens GMN. Autolysosomal activation combined with lysosomal destabilization efficiently targets myeloid leukemia cells for cell death. Front Oncol 2023; 13:999738. [PMID: 36816923 PMCID: PMC9931186 DOI: 10.3389/fonc.2023.999738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 01/09/2023] [Indexed: 02/04/2023] Open
Abstract
Introduction Current cancer research has led to a renewed interest in exploring lysosomal membrane permeabilization and lysosomal cell death as a targeted therapeutic approach for cancer treatment. Evidence suggests that differences in lysosomal biogenesis between cancer and normal cells might open a therapeutic window. Lysosomal membrane stability may be affected by the so-called 'busy lysosomal behaviour' characterized by higher lysosomal abundance and activity and more intensive fusion or interaction with other vacuole compartments. Methods We used a panel of multiple myeloid leukemia (ML) cell lines as well as leukemic patient samples and updated methodology to study auto-lysosomal compartment, lysosomal membrane permeabilization and lysosomal cell death. Results Our analyses demonstrated several-fold higher constitutive autolysosomal activity in ML cells as compared to human CD34+ hematopoietic cells. Importantly, we identified mefloquine as a selective activator of ML cells' lysosomal biogenesis, which induced a sizeable increase in ML lysosomal mass, acidity as well as cathepsin B and L activity. Concomitant mTOR inhibition synergistically increased lysosomal activity and autolysosomal fusion and simultaneously decreased the levels of key lysosomal stabilizing proteins, such as LAMP-1 and 2. Discussion In conclusion, mefloquine treatment combined with mTOR inhibition synergistically induced targeted ML cell death without additional toxicity. Taken together, these data provide a molecular mechanism and thus a rationale for a therapeutic approach for specific targeting of ML lysosomes.
Collapse
Affiliation(s)
- Harshit Shah
- Department for Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Metodi Stankov
- Department for Rheumatology and Immunology, Hannover Medical School, Hannover, Germany
| | - Diana Panayotova-Dimitrova
- Department of Dermatology and Allergology, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH), Aachen, Germany
| | - Amir Yazdi
- Department of Dermatology and Allergology, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH), Aachen, Germany
| | | | - Jan-Henning Klusmann
- Pediatric Hematology and Oncology, Department of Pediatrics, Goethe University Frankfurt, Frankfurt (Main), Germany
| | - Georg M. N. Behrens
- Department for Rheumatology and Immunology, Hannover Medical School, Hannover, Germany,*Correspondence: Georg M. N. Behrens,
| |
Collapse
|
6
|
Ando T, Suzuki-Karasaki M, Suzuki-Karasaki M, Ichikawa J, Ochiai T, Yoshida Y, Haro H, Suzuki-Karasaki Y. Combined Anticancer Effect of Plasma-Activated Infusion and Salinomycin by Targeting Autophagy and Mitochondrial Morphology. Front Oncol 2021; 11:593127. [PMID: 34150606 PMCID: PMC8212785 DOI: 10.3389/fonc.2021.593127] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 05/11/2021] [Indexed: 01/10/2023] Open
Abstract
Non-thermal atmospheric pressure plasma (NTAPP)-activated liquids have emerged as new promising anticancer agents because they preferentially injure malignant cells. Here, we report plasma-activated infusion (PAI) as a novel NTAPP-based anti-neoplastic agent. PAI was prepared by irradiating helium NTAP to form a clinically approved infusion fluid. PAI dose-dependently killed malignant melanoma and osteosarcoma cell lines while showing much lower cytotoxic effects on dermal and lung fibroblasts. We found that PAI and salinomycin (Sal), an emerging anticancer stem cell agent, mutually operated as adjuvants. The combined administration of PAI and Sal was much more effective than single-agent application in reducing the growth and lung metastasis of osteosarcoma allografts with minimal adverse effects. Mechanistically, PAI explicitly induced necroptosis and increased the phosphorylation of receptor-interacting protein 1/3 rapidly and transiently. PAI also suppressed the ambient autophagic flux by activating the mammalian target of the rapamycin pathway. PAI increased the phosphorylation of Raptor, Rictor, and p70-S6 kinase, along with decreased LC3-I/II expression. In contrast, Sal promoted autophagy. Moreover, Sal exacerbated the mitochondrial network collapse caused by PAI, resulting in aberrant clustering of fragmented mitochondrial in a tumor-specific manner. Our findings suggest that combined administration of PAI and Sal is a promising approach for treating these apoptosis-resistant cancers.
Collapse
Affiliation(s)
- Takashi Ando
- Department of Orthopaedic Surgery, Yamanashi University School of Medicine, Yamanashi, Japan
| | - Manami Suzuki-Karasaki
- Department of Research and Development, Plasma ChemiBio Laboratory, Plasma ChemiBio Laboratory, Nasushiobara, Tochigi, Japan
| | - Miki Suzuki-Karasaki
- Department of Research and Development, Plasma ChemiBio Laboratory, Plasma ChemiBio Laboratory, Nasushiobara, Tochigi, Japan
| | - Jiro Ichikawa
- Department of Orthopaedic Surgery, Yamanashi University School of Medicine, Yamanashi, Japan
| | - Toyoko Ochiai
- Department of Research and Development, Plasma ChemiBio Laboratory, Plasma ChemiBio Laboratory, Nasushiobara, Tochigi, Japan.,Department of Dermatology, Nihon University Hospital, Tokyo, Japan
| | - Yukihiro Yoshida
- Department of Orthopaedic Surgery, Nihon University School of Medicine, Nihon University Orthopaedic Surgery, Tokyo, Japan
| | - Hirotaka Haro
- Department of Orthopaedic Surgery, Yamanashi University School of Medicine, Yamanashi, Japan
| | - Yoshihiro Suzuki-Karasaki
- Department of Research and Development, Plasma ChemiBio Laboratory, Plasma ChemiBio Laboratory, Nasushiobara, Tochigi, Japan
| |
Collapse
|
7
|
Sundarraj K, Raghunath A, Panneerselvam L, Perumal E. Fisetin Inhibits Autophagy in HepG2 Cells via PI3K/Akt/mTOR and AMPK Pathway. Nutr Cancer 2020; 73:2502-2514. [DOI: 10.1080/01635581.2020.1836241] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Kiruthika Sundarraj
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamilnadu, India
| | - Azhwar Raghunath
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamilnadu, India
| | - Lakshmikanthan Panneerselvam
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamilnadu, India
| | - Ekambaram Perumal
- Molecular Toxicology Laboratory, Department of Biotechnology, Bharathiar University, Coimbatore, Tamilnadu, India
| |
Collapse
|
8
|
Shang XY, Guo R, Yu XQ, Lin B, Huang XX, Yao GD, Song SJ. Enantiomeric 8-O-4'-type neolignans from Crataegus pinnatifida exhibit cytotoxic effect via apoptosis and autophagy in Hep3B cells. Bioorg Chem 2020; 104:104267. [PMID: 32920350 DOI: 10.1016/j.bioorg.2020.104267] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 02/08/2023]
Abstract
Crataegus pinnatifida has been famous for its nutritional purpose. However, systematic investigation on the bioactive constituents is still lacking, although this fruit has been reported for its cytotoxic effect before. In this study, two pairs of new lignan enantiomers (1a/1b, 2a/2b), which isolated using chiral chromatographic column from the fruits of C. pinnatifida were studied. The absolute configurations of enantiomers were determined by comparison between the experimental electronic circular dichroism (ECD) and calculated ECD spectra. Among them, 1a/1b exhibited a better cytotoxic effect in hepatocellular carcinoma Hep3B cells with an IC50 value of 34.97 ± 2.74 and 17.42 ± 0.71 μM, respectively. In addition, 1b induced much more apoptotic, autophagic cells than 1a in Hep3B cells. Furthermore, the underlying mechanism was demonstrated that p38 activation could promote 1b-induced apoptosis and autophagy. Moreover, 1b-induced apoptosis was significantly decreased in the presence of autophagic inhibitor Bafilomycin A1 (Baf A1), suggesting that the induction of autophagy enhanced apoptotic cell death in 1b-treated cells. In general, these findings provide a valuable basis for further understanding the effect of 8-O-4' lignans in C. pinnatifida on cytotoxic effect.
Collapse
Affiliation(s)
- Xin-Yue Shang
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Rui Guo
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Xiao-Qi Yu
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Bin Lin
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, People's Republic of China
| | - Xiao-Xiao Huang
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China.
| | - Guo-Dong Yao
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China.
| | - Shao-Jiang Song
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Liaoning Province, School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China.
| |
Collapse
|
9
|
Radko L, Olejnik M, Posyniak A. Primary Human Hepatocytes, but Not HepG2 or Balb/c 3T3 Cells, Efficiently Metabolize Salinomycin and Are Resistant to Its Cytotoxicity. Molecules 2020; 25:E1174. [PMID: 32151009 PMCID: PMC7179450 DOI: 10.3390/molecules25051174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/03/2020] [Accepted: 03/03/2020] [Indexed: 11/16/2022] Open
Abstract
Salinomycin is a polyether antibiotic showing anticancer activity. There are many reports of its toxicity to animals but little is known about the potential adverse effects in humans. The action of the drug may be connected to its metabolism. That is why we investigated the cytotoxicity of salinomycin and pathways of its biotransformation using human primary hepatocytes, human hepatoma cells (HepG2), and the mouse fibroblast cell line (Balb/c 3T3). The cytotoxicity of salinomycin was time-dependent, concentration-dependent, and cell-dependent with primary hepatocytes being the most resistant. Among the studied models, primary hepatocytes were the only ones to efficiently metabolize salinomycin but even they were saturated at higher concentrations. The main route of biotransformation was monooxygenation leading to the formation of monohydroxysalinomycin, dihydroxysalinomycin, and trihydroxysalinomycin. Tiamulin, which is a known inhibitor of CYP450 izoenzymes, synergistically induced cytotoxicity of salinomycin in all cell types, including non-metabolising fibroblasts. Therefore, the pharmacokinetic interaction cannot fully explain tiamulin impact on salinomycin toxicity.
Collapse
Affiliation(s)
| | - Małgorzata Olejnik
- Department of Pharmacology and Toxicology, National Veterinary Research Institute, 57 Partyzantow Avenue, 24-100 Pulawy, Poland; (L.R.); (A.P.)
| | | |
Collapse
|
10
|
Misaponin B Induces G2/M Arrest, Cytokinesis Failure and Impairs Autophagy. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5925094. [PMID: 32090100 PMCID: PMC7029305 DOI: 10.1155/2020/5925094] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 01/04/2020] [Indexed: 12/14/2022]
Abstract
Saponins are a group of naturally occurring plant glycosides with the features of their strong foam-forming properties and multibiological effects such as antitumor activity. Though Misaponin B, one of the triterpenoid saponins from Madhuca longifolia, is known to have spermicidal and antioxidant activity, the other biological activities have been never reported so far. Thus, in the present study, the antitumor mechanism of Misaponin B was investigated in A549 and AsPC-1 cancer cells. Misaponin B exerted significant cytotoxicity in A549, H460, SKOV3, and AsPC-1 cancer cells. Among them, A549 and AsPC-1 cells were more susceptible to Misaponin B. Misaponin B induced G2/M arrest and cytokinesis failure and increased the expression of LC3B and p62 with autophagic vacuoles and GFP-LC3 punctae in A549 and AsPC-1 cells. Furthermore, Misaponin B suppressed autophagy flux in A549 cells transfected by GFP-mRFP-LC3 constructs by showing merged yellow color by autophagy flux assay. Overall, our findings provide evidences that Misaponin B induces G2M arrest and impairs autophagy in A549 and AsPC-1 cells.
Collapse
|
11
|
Yeon M, Lee S, Lee JE, Jung HS, Kim Y, Jeoung D. CAGE-miR-140-5p-Wnt1 Axis Regulates Autophagic Flux, Tumorigenic Potential of Mouse Colon Cancer Cells and Cellular Interactions Mediated by Exosomes. Front Oncol 2019; 9:1240. [PMID: 31799196 PMCID: PMC6868029 DOI: 10.3389/fonc.2019.01240] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 10/28/2019] [Indexed: 12/12/2022] Open
Abstract
Although the cancer/testis antigen CAGE has been implicated in tumorigenesis, the molecular mechanisms of CAGE-promoted tumorigenesis remain largely unknown. CT26Flag−CAGE cells, CT26 (mouse colon cancer cells) cells stably expressing CAGE, were established to investigate CAGE-promoted tumorigenesis. Down-regulation of CAGE led to decreased autophagic flux in CT26Flag−CAGE cells. CAGE interacted with Beclin1, a mediator of autophagy. The CT26Flag−CAGE cells showed enhanced autophagosome formation and displayed greater tumor spheroid-forming potential than CT26 cells. MicroRNA array analysis revealed that CAGE decreased the expression of various microRNAs, including miR-140-5p, in CT26 cells. CAGE was shown to bind to the promoter sequences of miR-140-5p. MiR-140-5p inhibition increased the tumorigenic potential of and autophagic flux in CT26 cells. A miR-140-5p mimic exerted negative effects on the tumorigenic potential of CT26Flag−CAGE cells and autophagic flux in CT26Flag−CAGE cells. MiR-140-5p was predicted to bind to the 3′-UTR of Wnt1. CT26Flag−CAGE cells showed higher expression of Wnt1 than CT26 cells. Down-regulation of Wnt1 decreased autophagic flux. Luciferase activity assays showed the direct regulation of wnt1 by miR-140-5p. Tumor tissue derived from the CT26Flag−CAGE cells revealed higher expressions of factors associated with activated mast cells and tumor-associated macrophages than tumor tissue derived from CT26 cells. Culture medium from the CT26Flag−CAGE cells increased autophagic flux in CT26 cells, mast cells and macrophages. Culture medium from the CT26Flag−CAGE cells increased CD163 and autophagic flux in CT26 cells, mast cells, and macrophages in a Wnt1-dependent manner. Exosomes from CT26Flag−CAGE cells increased autophagc flux in CT26 cells, mast cells, and macrophages. Exosomes from CT26Flag−CAGE cells increased the tumorigenic potential of CT26 cells. Wnt1 was shown to be present within the exosomes. Recombinant Wnt1 protein increased autophagic flux in CT26, mast cells, and macrophages. Recombinant wnt1 protein mediated interactions between the CT26 cells, mast cells, and macrophages. Our results showed novel roles for the CAGE-miR-140-5p-Wnt1 axis in autophagic flux and cellular interactions mediated by exosomes.
Collapse
Affiliation(s)
- Minjeong Yeon
- Department of Biochemistry, Kangwon National University, Chuncheon-si, South Korea
| | - Seungheon Lee
- Department of Biochemistry, Kangwon National University, Chuncheon-si, South Korea
| | - Joo-Eun Lee
- Department of Biochemistry, Kangwon National University, Chuncheon-si, South Korea
| | - Hyun Suk Jung
- Department of Biochemistry, Kangwon National University, Chuncheon-si, South Korea
| | - Youngmi Kim
- College of Medicine, Institute of New Frontier Research, Hallym University, Chuncheon-si, South Korea
| | - Dooil Jeoung
- Department of Biochemistry, Kangwon National University, Chuncheon-si, South Korea
| |
Collapse
|
12
|
Hong JM, Kim JH, Kim H, Lee WJ, Hwang YI. SB365, Pulsatilla Saponin D Induces Caspase-Independent Cell Death and Augments the Anticancer Effect of Temozolomide in Glioblastoma Multiforme Cells. Molecules 2019; 24:molecules24183230. [PMID: 31491945 PMCID: PMC6766801 DOI: 10.3390/molecules24183230] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/02/2019] [Accepted: 09/04/2019] [Indexed: 12/28/2022] Open
Abstract
SB365, a saponin D extracted from the roots of Pulsatilla koreana, has been reported to show cytotoxicity in several cancer cell lines. We investigated the effects of SB365 on U87-MG and T98G glioblastoma multiforme (GBM) cells, and its efficacy in combination with temozolomide for treating GBM. SB365 exerted a cytotoxic effect on GBM cells not by inducing apoptosis, as in other cancer cell lines, but by triggering caspase-independent cell death. Inhibition of autophagic flux and neutralization of the lysosomal pH occurred rapidly after application of SB365, followed by deterioration of mitochondrial membrane potential. A cathepsin B inhibitor and N-acetyl cysteine, an antioxidant, partially recovered cell death induced by SB365. SB365 in combination with temozolomide exerted an additive cytotoxic effect in vitro and in vivo. In conclusion, SB365 inhibits autophagic flux and induces caspase-independent cell death in GBM cells in a manner involving cathepsin B and mainly reactive oxygen species, and its use in combination with temozolomide shows promise for the treatment of GBM.
Collapse
Affiliation(s)
- Jun-Man Hong
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Korea.
| | - Jin-Hee Kim
- Department of Biomedical Laboratory Science, Cheongju University, Cheongju 28503, Korea.
| | - Hyemin Kim
- Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, Korea.
| | - Wang Jae Lee
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Korea.
| | - Young-Il Hwang
- Department of Anatomy and Cell Biology, Seoul National University College of Medicine, Seoul 03080, Korea.
| |
Collapse
|
13
|
Hochmair M, Rath B, Klameth L, Ulsperger E, Weinlinger C, Fazekas A, Plangger A, Zeillinger R, Hamilton G. Effects of salinomycin and niclosamide on small cell lung cancer and small cell lung cancer circulating tumor cell lines. Invest New Drugs 2019; 38:946-955. [PMID: 31446534 PMCID: PMC7340652 DOI: 10.1007/s10637-019-00847-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023]
Abstract
Tumor dissemination and recurrence is attributed to highly resistant cancer stem cells (CSCs) which may constitute a fraction of circulating tumor cells (CTCs). Small cell lung cancer (SCLC) constitutes a suitable model to investigate the relation of CTCs and CSCs due to rapid tumor spread and a high number of CTCs. Expansion of five SCLC CTC lines (BHGc7, 10, 16, 26 and UHGc5) in vitro at our institution allowed for the analysis of CSC markers and cytotoxicity of the CSC-selective drugs salinomycin and niclosamide against CTC single cell suspensions or CTC spheroids/ tumorospheres (TOS). Salinomycin exerted dose-dependent cytotoxicity against the SCLC lines but, with exception of BHGc7 TOS, there was no markedly enhanced activity against TOS. Similarly, niclosamide exhibits high activity against BHGc7 TOS and UHGc5 TOS but not against the other CTC spheroids. High expression of the CSC marker CD133 was restricted to three SCLC tumor lines and the BHGc10 CTC line. All SCLC CTCs are CD24-positive but lack expression of CD44 and ABCG2 in contrast to the SCLC tumor lines which show a phenotype more similar to that of CSCs. The stem cell marker SOX2 was found in all CTC lines and SCLC GLC14/16, whereas elevated expression of Oct-3/4 and Nanog was restricted to BHGc26 and UHGc5. In conclusion, the SCLC CTCs established from patients with relapsed disease lack a typical CSC phenotype in respect to chemosensitivity to CSC-selective drugs, surface markers, expression of pluripotent stem cell and transcription factors.
Collapse
Affiliation(s)
- Maximilian Hochmair
- Respiratory Oncology Unit, Otto Wagner Hospital, Baumgartner Höhe, Vienna, Austria
| | - Barbara Rath
- Department of Surgery, Medical University of Vienna, Spitalgasse, Vienna, Austria
| | - Lukas Klameth
- Department of Pathophysiology and Allergy Research, Medical University of Vienna, Vienna, Austria
| | | | - Christoph Weinlinger
- Respiratory Oncology Unit, Otto Wagner Hospital, Baumgartner Höhe, Vienna, Austria
| | - Andreas Fazekas
- Respiratory Oncology Unit, Otto Wagner Hospital, Baumgartner Höhe, Vienna, Austria
| | - Adelina Plangger
- Department of Surgery, Medical University of Vienna, Spitalgasse, Vienna, Austria
| | - Robert Zeillinger
- Department of Gynecology and Obstetrics, Molecular Oncology Group, Medical University of Vienna, Vienna, Austria
| | - Gerhard Hamilton
- Department of Surgery, Medical University of Vienna, Spitalgasse, Vienna, Austria.
| |
Collapse
|
14
|
Yang S, Yang L, Li X, Li B, Li Y, Zhang X, Ma Y, Peng X, Jin H, Li H. New insights into autophagy in hepatocellular carcinoma: mechanisms and therapeutic strategies. Am J Cancer Res 2019; 9:1329-1353. [PMID: 31392073 PMCID: PMC6682711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 06/13/2019] [Indexed: 06/10/2023] Open
Abstract
Autophagy is a mechanism by which cellular substances are transported to lysosomes for degradation, allowing the basic transformation of cellular components, and providing energy and macromolecular precursors. In cancer, the contradictory role of autophagy in tumor suppression and promotion has been widely acknowledged. Activation and suppression of autophagy have been proposed as cancer therapies, resulting in targeted treatment of cancer by autophagy being considered ambiguous. The dynamic effect of autophagy can also be applied to hepatocellular carcinoma (HCC), a malignant tumor with high incidence and a low survival rate. In this review, we introduce characteristics of different types of autophagy and summarize which genes, non-coding RNAs, and related signaling pathways are involved in autophagy and the regulation of the formation and progress of HCC. More importantly, we discuss the role of autophagy in the treatment of HCC, such as in traditional chemotherapy, molecular targeted drugs, and natural products.
Collapse
Affiliation(s)
- Shuo Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Liang Yang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Xinyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Bowen Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Yan Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Xiaodong Zhang
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Yingbo Ma
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Xueqiang Peng
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Hongyuan Jin
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| | - Hangyu Li
- Department of General Surgery, The Fourth Affiliated Hospital, China Medical University Shenyang 110000, P. R. China
| |
Collapse
|
15
|
Khan N, Yılmaz S, Aksoy S, Uzel A, Tosun Ç, Kirmizibayrak PB, Bedir E. Polyethers isolated from the marine actinobacterium Streptomyces cacaoi inhibit autophagy and induce apoptosis in cancer cells. Chem Biol Interact 2019; 307:167-178. [DOI: 10.1016/j.cbi.2019.04.035] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Revised: 04/20/2019] [Accepted: 04/30/2019] [Indexed: 12/26/2022]
|
16
|
Antoszczak M, Huczyński A. Salinomycin and its derivatives - A new class of multiple-targeted "magic bullets". Eur J Med Chem 2019; 176:208-227. [PMID: 31103901 DOI: 10.1016/j.ejmech.2019.05.031] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 05/06/2019] [Accepted: 05/08/2019] [Indexed: 12/23/2022]
Abstract
The history of drug development clearly shows the scale of painstaking effort leading to a finished product - a highly biologically active agent that would be at the same time no or little toxic to human organism. Moreover, the aim of modern drug discovery can move from "one-molecule one-target" concept to more promising "one-molecule multiple-targets" one, particularly in the context of effective fight against cancer and other complex diseases. Gratifyingly, natural compounds are excellent source of potential drug leads. One of such promising naturally-occurring drug candidates is a polyether ionophore - salinomycin (SAL). This compound should be identified as multi-target agent for two reasons. Firstly, SAL combines a broad spectrum of bioactivity, including antibacterial, antifungal, antiviral, antiparasitic and anticancer activity, with high selectivity of action, proving its significant therapeutic potential. Secondly, the multimodal mechanism of action of SAL has been shown to be related to its interactions with multiple molecular targets and signalling pathways that are synergistic for achieving a therapeutic anticancer effect. On the other hand, according to the Paul Ehrlich's "magic bullet" concept, invariably inspiring the scientists working on design of novel target-selective molecules, a very interesting direction of research is rational chemical modification of SAL. Importantly, many of SAL derivatives have been found to be more promising as chemotherapeutics than the native structure. This concise review article is focused both on the possible role of SAL and its selected analogues in future antimicrobial and/or cancer therapy, and on the potential use of SAL as a new class of multiple-targeted "magic bullet" because of its multimodal mechanism of action.
Collapse
Affiliation(s)
- Michał Antoszczak
- Department of Bioorganic Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Umultowska 89b, 61‒614, Poznań, Poland
| | - Adam Huczyński
- Department of Bioorganic Chemistry, Faculty of Chemistry, Adam Mickiewicz University, Umultowska 89b, 61‒614, Poznań, Poland.
| |
Collapse
|
17
|
Oxidative Stress-Driven Autophagy acROSs Onset and Therapeutic Outcome in Hepatocellular Carcinoma. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6050123. [PMID: 31205585 PMCID: PMC6530208 DOI: 10.1155/2019/6050123] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/28/2019] [Indexed: 12/22/2022]
Abstract
Reactive oxygen species- (ROS-) mediated autophagy physiologically contributes to management of cell homeostasis in response to mild oxidative stress. Cancer cells typically engage autophagy downstream of ROS signaling derived from hypoxia and starvation, which are harsh environmental conditions that need to be faced for cancer development and progression. Hepatocellular carcinoma (HCC) is a solid tumor for which several environmental risk factors, particularly viral infections and alcohol abuse, have been shown to promote carcinogenesis via augmentation of oxidative stress. In addition, ROS burst in HCC cells frequently takes place after administration of therapeutic compounds that promote apoptotic cell death or even autophagic cell death. The interplay between ROS and autophagy (i) in the disposal of dysfunctional mitochondria via mitophagy, as a tumor suppressor mechanism, or (ii) in the cell survival adaptive response elicited by chemotherapeutic interventions, as a tumor-promoting event, will be depicted in this review in relation to HCC development and progression.
Collapse
|
18
|
A comprehensive review of salinomycin derivatives as potent anticancer and anti-CSCs agents. Eur J Med Chem 2019; 166:48-64. [DOI: 10.1016/j.ejmech.2019.01.034] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/02/2019] [Accepted: 01/14/2019] [Indexed: 02/08/2023]
|
19
|
Klose J, Trefz S, Wagner T, Steffen L, Preißendörfer Charrier A, Radhakrishnan P, Volz C, Schmidt T, Ulrich A, Dieter SM, Ball C, Glimm H, Schneider M. Salinomycin: Anti-tumor activity in a pre-clinical colorectal cancer model. PLoS One 2019; 14:e0211916. [PMID: 30763370 PMCID: PMC6375586 DOI: 10.1371/journal.pone.0211916] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/22/2019] [Indexed: 12/19/2022] Open
Abstract
Objectives Salinomycin is a polyether antibiotic with selective activity against human cancer stem cells. The impact of salinomycin on patient-derived primary human colorectal cancer cells has not been investigated so far. Thus, here we aimed to investigate the activity of salinomycin against tumor initiating cells isolated from patients with colorectal cancer. Methods Primary tumor-initiating cells (TIC) isolated from human patients with colorectal liver metastases or from human primary colon carcinoma were exposed to salinomycin and compared to treatment with 5-FU and oxaliplatin. TICs were injected subcutaneously into NOD/SCID mice to induce a patient-derived mouse xenograft model of colorectal cancer. Animals were treated either with salinomycin, FOLFOX regimen, or salinomycin and FOLFOX. Human colorectal cancer cells were used to delineate an underlying molecular mechanism of salinomycin in this tumor entity. Results Applying TICs isolated from human patients with colorectal liver metastases or from human primary colon carcinoma, we demonstrated that salinomycin exerts increased antiproliferative activity compared to 5-fluorouracil and oxaliplatin treatment. Consistently, salinomycin alone or in combination with FOLFOX exerts superior antitumor activity compared to FOLFOX therapy in a patient-derived mouse xenograft model of colorectal cancer. Salinomycin induces apoptosis of human colorectal cancer cells, accompanied by accumulation of dysfunctional mitochondria and reactive oxygen species. These effects are associated with expressional down-regulation of superoxide dismutase-1 (SOD1) in response to salinomycin treatment. Conclusion Collectively, the results of this pre-clinical study indicate that salinomycin alone or in combination with 5-fluorouracil and oxaliplatin exerts increased antitumoral activity compared to common chemotherapy.
Collapse
Affiliation(s)
- Johannes Klose
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
- * E-mail:
| | - Stefan Trefz
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Tobias Wagner
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Luca Steffen
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | | | - Praveen Radhakrishnan
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Claudia Volz
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Thomas Schmidt
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Alexis Ulrich
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Sebastian M. Dieter
- Translational Functional Cancer Genomics, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Claudia Ball
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Dresden and German Cancer Research Center (DKFZ), Dresden, Germany
| | - Hanno Glimm
- Translational Functional Cancer Genomics, National Center for Tumor Diseases (NCT) Heidelberg and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Translational Medical Oncology, National Center for Tumor Diseases (NCT) Dresden and German Cancer Research Center (DKFZ), Dresden, Germany
- Center for Personalized Oncology, University Hospital Carl Gustav Carus Dresden at TU Dresden, Dresden, Germany
- German Consortium for Translational Cancer Research (DKTK) Dresden, Dresden, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
20
|
Martins WK, Santos NF, Rocha CDS, Bacellar IOL, Tsubone TM, Viotto AC, Matsukuma AY, Abrantes ABDP, Siani P, Dias LG, Baptista MS. Parallel damage in mitochondria and lysosomes is an efficient way to photoinduce cell death. Autophagy 2019; 15:259-279. [PMID: 30176156 PMCID: PMC6333451 DOI: 10.1080/15548627.2018.1515609] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 07/30/2018] [Accepted: 08/15/2018] [Indexed: 01/12/2023] Open
Abstract
Cells challenged by photosensitized oxidations face strong redox stresses and rely on autophagy to either survive or die. However, the use of macroautophagy/autophagy to improve the efficiency of photosensitizers, in terms of inducing cell death, remains unexplored. Here, we addressed the concept that a parallel damage in the membranes of mitochondria and lysosomes leads to a scenario of autophagy malfunction that can greatly improve the efficiency of the photosensitizer to cause cell death. Specific damage to these organelles was induced by irradiation of cells pretreated with 2 phenothiazinium salts, methylene blue (MB) and 1,9-dimethyl methylene blue (DMMB). At a low concentration level (10 nM), only DMMB could induce mitochondrial damage, leading to mitophagy activation, which did not progress to completion because of the parallel damage in lysosome, triggering cell death. MB-induced photodamage was perceived almost instantaneously after irradiation, in response to a massive and nonspecific oxidative stress at a higher concentration range (2 µM). We showed that the parallel damage in mitochondria and lysosomes activates and inhibits mitophagy, leading to a late and more efficient cell death, offering significant advantage (2 orders of magnitude) over photosensitizers that cause unspecific oxidative stress. We are confident that this concept can be used to develop better light-activated drugs. Abbreviations: ΔΨm: mitochondrial transmembrane inner potential; AAU: autophagy arbitrary units; ATG5, autophagy related 5; ATG7: autophagy related 7; BAF: bafilomycin A1; BSA: bovine serum albumin; CASP3: caspase 3; CF: carboxyfluorescein; CTSB: cathepsin B; CVS: crystal violet staining; DCF: dichlorofluorescein; DCFH2: 2',7'-dichlorodihydrofluorescein; DMMB: 1,9-dimethyl methylene blue; ER: endoplasmic reticulum; HaCaT: non-malignant immortal keratinocyte cell line from adult human skin; HP: hydrogen peroxide; LC3B-II: microtubule associated protein 1 light chain 3 beta-II; LMP: lysosomal membrane permeabilization; LTG: LysoTracker™ Green DND-26; LTR: LysoTracker™ Red DND-99; 3-MA: 3-methyladenine; MB: methylene blue; mtDNA: mitochondrial DNA; MitoSOX™: red mitochondrial superoxide probe; MTDR: MitoTracker™ Deep Red FM; MTO: MitoTracker™ Orange CMTMRos; MT-ND1: mitochondrially encoded NADH:ubiquinone oxidoreductase core subunit 1; MTT: methylthiazolyldiphenyl-tetrazolium bromide; 1O2: singlet oxygen; OH. hydroxil radical; PRKN/parkin: parkin RBR E3 ubiquitin protein ligase; PBS: phosphate-buffered saline; PI: propidium iodide; PDT: photodynamic therapy; PS: photosensitizer; QPCR: gene-specific quantitative PCR-based; Rh123: rhodamine 123; ROS: reactive oxygen species RTN: rotenone; SQSTM1/p62: sequestosome 1; SUVs: small unilamellar vesicles; TBS: Tris-buffered saline.
Collapse
Affiliation(s)
- Waleska K. Martins
- Instituto de Química, Departamento de Bioquímica, Universidade de São Paulo, São Paulo, Brazil
- Programa de Pós-graduação Stricto Sensue Pesquisa, Universidade Anhanguera de São Paulo, São Paulo, Brazil
| | - Nayra Fernandes Santos
- Instituto de Química, Departamento de Bioquímica, Universidade de São Paulo, São Paulo, Brazil
| | - Cleidiane de Sousa Rocha
- Instituto de Química, Departamento de Bioquímica, Universidade de São Paulo, São Paulo, Brazil
- Programa de Pós-graduação Stricto Sensue Pesquisa, Universidade Anhanguera de São Paulo, São Paulo, Brazil
| | - Isabel O. L. Bacellar
- Instituto de Química, Departamento de Bioquímica, Universidade de São Paulo, São Paulo, Brazil
| | - Tayana Mazin Tsubone
- Instituto de Química, Departamento de Bioquímica, Universidade de São Paulo, São Paulo, Brazil
| | - Ana Cláudia Viotto
- Instituto de Química, Departamento de Bioquímica, Universidade de São Paulo, São Paulo, Brazil
| | | | - Aline B. de P. Abrantes
- Instituto de Química, Departamento de Bioquímica, Universidade de São Paulo, São Paulo, Brazil
| | - Paulo Siani
- FFCLRP, Departamento de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Luís Gustavo Dias
- FFCLRP, Departamento de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Mauricio S. Baptista
- Instituto de Química, Departamento de Bioquímica, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|
21
|
Antoszczak M. A medicinal chemistry perspective on salinomycin as a potent anticancer and anti-CSCs agent. Eur J Med Chem 2019; 164:366-377. [DOI: 10.1016/j.ejmech.2018.12.057] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 12/20/2018] [Accepted: 12/24/2018] [Indexed: 01/30/2023]
|
22
|
To reduce premature drug release while ensuring burst intracellular drug release of solid lipid nanoparticle-based drug delivery system with clathrin modification. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2019; 15:108-118. [DOI: 10.1016/j.nano.2018.05.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 04/13/2018] [Accepted: 05/20/2018] [Indexed: 12/12/2022]
|
23
|
Zidovudine-Mediated Autophagy Inhibition Enhances Mitochondrial Toxicity in Muscle Cells. Antimicrob Agents Chemother 2018; 63:AAC.01443-18. [PMID: 30373793 DOI: 10.1128/aac.01443-18] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 10/15/2018] [Indexed: 11/20/2022] Open
Abstract
Nucleoside reverse transcriptase inhibitors (NRTI), such as zidovudine (AZT), are constituents of HIV-1 therapy and are used for the prevention of mother-to-child transmission. Prolonged thymidine analogue exposure has been associated with mitochondrial toxicities to heart, liver, and skeletal muscle. We hypothesized that the thymidine analogue AZT might interfere with autophagy in myocytes, a lysosomal degradation pathway implicated in the regulation of mitochondrial recycling, cell survival, and the pathogenesis of myodegenerative diseases. The impact of AZT and lamivudine (3TC) on C2C12 myocyte autophagy was studied using various methods based on LC3-green fluorescent protein overexpression or LC3 staining in combination with Western blotting, flow cytometry, and confocal and electron microscopy. Lysosomal and mitochondrial functions were studied using appropriate staining for lysosomal mass, acidity, cathepsin activity, as well as mitochondrial mass and membrane potential in combination with flow cytometry and confocal microscopy. AZT, but not 3TC, exerted a significant dose- and time-dependent inhibitory effect on late stages of autophagosome maturation, which was reversible upon mTOR inhibition. Inhibition of late autophagy at therapeutic drug concentrations led to dysfunctional mitochondrial accumulation with membrane hyperpolarization and increased reactive oxygen species (ROS) generation and, ultimately, compromised cell viability. These AZT effects could be readily replicated by pharmacological and genetic inhibition of myocyte autophagy and, most importantly, could be rescued by pharmacological stimulation of autophagolysosomal biogenesis. Our data suggest that the thymidine analogue AZT inhibits autophagy in myocytes, which in turn leads to the accumulation of dysfunctional mitochondria with increased ROS generation and compromised cell viability. This novel mechanism could contribute to our understanding of the long-term side effects of antiviral agents.
Collapse
|
24
|
Fu C, Wang L, Tian G, Zhang C, Zhao Y, Xu H, Su M, Wang Y. Enhanced anticancer effect of oncostatin M combined with salinomycin in CD133 + HepG2 liver cancer cells. Oncol Lett 2018; 17:1798-1806. [PMID: 30675240 PMCID: PMC6341778 DOI: 10.3892/ol.2018.9796] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Accepted: 11/02/2018] [Indexed: 12/24/2022] Open
Abstract
Oncostatin M (OSM) induces the differentiation of liver cancer stem cells (LCSCs) and increases sensitivity to the chemotherapeutic agent 5-fluorouracil, whereas salinomycin (Sal) induces apoptosis in cancer stem cells and inhibits the proliferation of liver cancer cells. However, there have been no studies investigating the anticancer effects of combination treatment with OSM and Sal. In the present study, we investigated the synergistic effects of OSM and Sal on LCSCs, the CD133+ subpopulations from HepG2 human liver cancer cells. CD133+ LCSCs were isolated using an immunomagnetic bead technique and identified through colony formation. After incubating with OSM and Sal, the ability of LCSC proliferation and invasion, as well as apoptosis rates were evaluated, and the expression of stemness-related genes was examined by quantitative real-time polymerase chain reaction. Additionally, the secretion of α-fetoprotein (AFP) and albumin (ALB) were analyzed by enzyme-linked immunosorbent assay. Our results indicated that OSM combined with Sal significantly suppressed LCSC proliferation and invasion and induced apoptosis, as determined by flow cytometry and increases in cleaved caspase-3 levels detected by western blotting. The results of the JC-1 staining assay indicated that this effect involved the mitochondrial pathway. Moreover, combination treatment reduced the expression of CD133 in LCSCs and suppressed stemness-related gene expression. Furthermore, the LCSCs produced lower levels of AFP and higher levels of ALB following combination treatment. In all experiments, combination treatment elicited more efficient anticancer effects on LCSCs as compared with single-drug treatment; therefore, our results demonstrated that combined treatment with OSM and Sal inhibited proliferation and induced differentiation and apoptosis in LCSCs, suggesting combined use of OSM and Sal as a therapeutic strategy for liver cancer.
Collapse
Affiliation(s)
- Changhao Fu
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Lu Wang
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Geer Tian
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Chen Zhang
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China.,Xiamen Institute of Rare Earth Materials, Chinese Academy of Sciences, Xiamen, Fujian 361021, P.R. China
| | - Yuanyuan Zhao
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hao Xu
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Manman Su
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yi Wang
- Department of Regenerative Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
25
|
Akkoç Y, Gözüaçık D. Autophagy and liver cancer. TURKISH JOURNAL OF GASTROENTEROLOGY 2018; 29:270-282. [PMID: 29755011 DOI: 10.5152/tjg.2018.150318] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Autophagy is a key biological phenomenon conserved from yeast to mammals. Under basal conditions, activation of autophagy leads to the protein degradation as well as damaged organelles for maintaining cellular homeostasis. Deregulation of autophagy has been identified as a key mechanism contributing to the pathogenesis and progression of several liver diseases, including hepatocellular carcinoma (HCC), one of the most common and mortal types of cancer. Currently used treatment strategies in patients with HCC result in variable success rates. Therefore, novel early diagnosis and treatment techniques should be developed. Manipulation of autophagy may improve responses of cancer cell to treatments and provide novel targeted therapy options for HCC. In this review, we summarized how our understanding of autophagy-cell death connection may have an impact on HCC therapy.
Collapse
Affiliation(s)
- Yunus Akkoç
- Department of Molecular Biology, Genetics and Bioengineering, Sabancı University School of Engineering and Natural Sciences, İstanbul, Turkey; Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, İstanbul, Turkey
| | - Devrim Gözüaçık
- Department of Molecular Biology, Genetics and Bioengineering, Sabancı University School of Engineering and Natural Sciences, İstanbul, Turkey; Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, İstanbul, Turkey
| |
Collapse
|
26
|
Kaushik V, Yakisich JS, Kumar A, Azad N, Iyer AKV. Ionophores: Potential Use as Anticancer Drugs and Chemosensitizers. Cancers (Basel) 2018; 10:E360. [PMID: 30262730 PMCID: PMC6211070 DOI: 10.3390/cancers10100360] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 09/13/2018] [Accepted: 09/21/2018] [Indexed: 01/08/2023] Open
Abstract
Ion homeostasis is extremely important for the survival of both normal as well as neoplastic cells. The altered ion homeostasis found in cancer cells prompted the investigation of several ionophores as potential anticancer agents. Few ionophores, such as Salinomycin, Nigericin and Obatoclax, have demonstrated potent anticancer activities against cancer stem-like cells that are considered highly resistant to chemotherapy and responsible for tumor relapse. The preclinical success of these compounds in in vitro and in vivo models have not been translated into clinical trials. At present, phase I/II clinical trials demonstrated limited benefit of Obatoclax alone or in combination with other anticancer drugs. However, future development in targeted drug delivery may be useful to improve the efficacy of these compounds. Alternatively, these compounds may be used as leading molecules for the development of less toxic derivatives.
Collapse
Affiliation(s)
- Vivek Kaushik
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University, Hampton, VA 23668, USA.
| | - Juan Sebastian Yakisich
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University, Hampton, VA 23668, USA.
| | - Anil Kumar
- Great Plains Health, North Platte, NE 69101, USA.
| | - Neelam Azad
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University, Hampton, VA 23668, USA.
| | - Anand K V Iyer
- Department of Pharmaceutical Sciences, School of Pharmacy, Hampton University, Hampton, VA 23668, USA.
| |
Collapse
|
27
|
Yuan X, Wang B, Yang L, Zhang Y. The role of ROS-induced autophagy in hepatocellular carcinoma. Clin Res Hepatol Gastroenterol 2018; 42:306-312. [PMID: 29544680 DOI: 10.1016/j.clinre.2018.01.005] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 01/11/2018] [Accepted: 01/19/2018] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is a main cause of cancer-related mortality and its etiology is not fully understood. As prominent factors that regulate cellular homeostasis, both reactive oxygen species (ROS) and autophagy are considered to play an essential role in the liver carcinogenesis. However, the crosstalk between ROS and autophagy is not well characterized in the pathogenesis of HCC. This review summarizes the roles of autophagy in ROS-mediated hepatocarcinogenesis and discusses the role of ROS-induced autophagy in HCC cell fate decision following treatment with chemotherapeutic agents in preclinical settings, which may allow the identification of novel strategies for the treatment of HCC.
Collapse
Affiliation(s)
- Xingxing Yuan
- Department of Gastroenterology, Nangang branch of Heilongjiang Academy of Traditional Chinese Medicine, No. 33 West Dazhi Road, Nangang District, Harbin, Heilongjiang 150006, China
| | - Bingyu Wang
- Department of Gastroenterology, Nangang branch of Heilongjiang Academy of Traditional Chinese Medicine, No. 33 West Dazhi Road, Nangang District, Harbin, Heilongjiang 150006, China
| | - Lei Yang
- Department of Gastroenterology, Nangang branch of Heilongjiang Academy of Traditional Chinese Medicine, No. 33 West Dazhi Road, Nangang District, Harbin, Heilongjiang 150006, China
| | - Yali Zhang
- Department of Gastroenterology, Nangang branch of Heilongjiang Academy of Traditional Chinese Medicine, No. 33 West Dazhi Road, Nangang District, Harbin, Heilongjiang 150006, China.
| |
Collapse
|
28
|
Versini A, Saier L, Sindikubwabo F, Müller S, Cañeque T, Rodriguez R. Chemical biology of salinomycin. Tetrahedron 2018. [DOI: 10.1016/j.tet.2018.07.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
29
|
Vu HT, Kobayashi M, Hegazy AM, Tadokoro Y, Ueno M, Kasahara A, Takase Y, Nomura N, Peng H, Ito C, Ino Y, Todo T, Nakada M, Hirao A. Autophagy inhibition synergizes with calcium mobilization to achieve efficient therapy of malignant gliomas. Cancer Sci 2018; 109:2497-2508. [PMID: 29902340 PMCID: PMC6113445 DOI: 10.1111/cas.13695] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 06/03/2018] [Accepted: 06/08/2018] [Indexed: 01/02/2023] Open
Abstract
Autophagy plays a critical role in tumorigenesis, but how autophagy contributes to cancer cells' responses to chemotherapeutics remains controversial. To investigate the roles of autophagy in malignant gliomas, we used CRISPR/CAS9 to knock out the ATG5 gene, which is essential for autophagosome formation, in tumor cells derived from patients with glioblastoma. While ATG5 disruption inhibited autophagy, it did not change the phenotypes of glioma cells and did not alter their sensitivity to temozolomide, an agent used for glioblastoma patient therapy. Screening of an anticancer drug library identified compounds that showed greater efficacy to ATG5-knockout glioma cells compared to control. While several selected compounds, including nigericin and salinomycin, remarkably induced autophagy, potent autophagy inducers by mTOR inhibition did not exhibit the ATG5-dependent cytoprotective effects. Nigericin in combination with ATG5 deficiency synergistically suppressed spheroid formation by glioma cells in a manner mitigated by Ca2+ chelation or CaMKK inhibition, indicating that, in combination with autophagy inhibition, calcium-mobilizing compounds contribute to efficient anticancer therapeutics. ATG5-knockout cells treated with nigericin showed increased mitochondria-derived reactive oxygen species and apoptosis compared to controls, indicating that autophagy protects glioma cells from mitochondrial reactive oxygen species-mediated damage. Finally, using a patient-derived xenograft model, we demonstrated that chloroquine, a pharmacological autophagy inhibitor, dramatically enhanced the efficacy of compounds selected in this study. Our findings propose a novel therapeutic strategy in which calcium-mobilizing compounds are combined with autophagy inhibitors to treat patients with glioblastoma.
Collapse
Affiliation(s)
- Ha Thi Vu
- Cancer and Stem Cell Research ProgramDivision of Molecular GeneticsCancer Research InstituteKanazawa UniversityKanazawaIshikawaJapan
| | - Masahiko Kobayashi
- Cancer and Stem Cell Research ProgramDivision of Molecular GeneticsCancer Research InstituteKanazawa UniversityKanazawaIshikawaJapan
- WPI Nano Life Science Institute (WPI‐Nano LSI)Kanazawa UniversityKanazawaIshikawaJapan
| | - Ahmed M. Hegazy
- Cancer and Stem Cell Research ProgramDivision of Molecular GeneticsCancer Research InstituteKanazawa UniversityKanazawaIshikawaJapan
| | - Yuko Tadokoro
- Cancer and Stem Cell Research ProgramDivision of Molecular GeneticsCancer Research InstituteKanazawa UniversityKanazawaIshikawaJapan
- WPI Nano Life Science Institute (WPI‐Nano LSI)Kanazawa UniversityKanazawaIshikawaJapan
| | - Masaya Ueno
- Cancer and Stem Cell Research ProgramDivision of Molecular GeneticsCancer Research InstituteKanazawa UniversityKanazawaIshikawaJapan
- WPI Nano Life Science Institute (WPI‐Nano LSI)Kanazawa UniversityKanazawaIshikawaJapan
| | - Atsuko Kasahara
- Cancer and Stem Cell Research ProgramDivision of Molecular GeneticsCancer Research InstituteKanazawa UniversityKanazawaIshikawaJapan
- Institute for Frontier Science InitiativeKanazawa UniversityKanazawaIshikawaJapan
| | - Yusuke Takase
- Cancer and Stem Cell Research ProgramDivision of Molecular GeneticsCancer Research InstituteKanazawa UniversityKanazawaIshikawaJapan
| | - Naho Nomura
- Cancer and Stem Cell Research ProgramDivision of Molecular GeneticsCancer Research InstituteKanazawa UniversityKanazawaIshikawaJapan
| | - Hui Peng
- Cancer and Stem Cell Research ProgramDivision of Molecular GeneticsCancer Research InstituteKanazawa UniversityKanazawaIshikawaJapan
| | - Chiaki Ito
- Cancer and Stem Cell Research ProgramDivision of Molecular GeneticsCancer Research InstituteKanazawa UniversityKanazawaIshikawaJapan
| | - Yasushi Ino
- Division of Innovative Cancer TherapyInstitute of Medical ScienceUniversity of TokyoTokyoJapan
| | - Tomoki Todo
- Division of Innovative Cancer TherapyInstitute of Medical ScienceUniversity of TokyoTokyoJapan
| | - Mitsutoshi Nakada
- Department of NeurosurgeryGraduate School of Medical ScienceKanazawa UniversityKanazawaIshikawaJapan
| | - Atsushi Hirao
- Cancer and Stem Cell Research ProgramDivision of Molecular GeneticsCancer Research InstituteKanazawa UniversityKanazawaIshikawaJapan
- WPI Nano Life Science Institute (WPI‐Nano LSI)Kanazawa UniversityKanazawaIshikawaJapan
| |
Collapse
|
30
|
Sommer AK, Hermawan A, Mickler FM, Ljepoja B, Knyazev P, Bräuchle C, Ullrich A, Wagner E, Roidl A. Salinomycin co-treatment enhances tamoxifen cytotoxicity in luminal A breast tumor cells by facilitating lysosomal degradation of receptor tyrosine kinases. Oncotarget 2018; 7:50461-50476. [PMID: 27409163 PMCID: PMC5226596 DOI: 10.18632/oncotarget.10459] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 06/17/2016] [Indexed: 11/25/2022] Open
Abstract
Luminal A breast cancer is the most common breast cancer subtype which is usually treated with selective estrogen receptor modulators (SERMS) like tamoxifen. Nevertheless, one third of estrogen receptor positive breast cancer patients initially do not respond to endocrine therapy and about 40% of luminal A breast tumors recur in five years. In this study, we investigated an alternative treatment approach by combining tamoxifen and salinomycin in luminal A breast cancer cell lines. We have found that salinomycin induces an additional cytotoxic effect by inhibiting the ligand independent activation of ERα. Thereby salinomycin increases the intracellular calcium level. This leads to a premature fusion of endosomes with lysosomes and thus to the degradation of Egfr family members. Since this process is essential for luminal A breast cancer cells to circumvent tamoxifen treatment, the combination of both drugs induces cytotoxicity in tamoxifen sensitive as well as resistant luminal A breast cancer cell lines.
Collapse
Affiliation(s)
- Ann-Katrin Sommer
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany.,Department of Molecular Biology, Max-Planck-Institute of Biochemistry, 82152 Martinsried, Germany
| | - Adam Hermawan
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Frauke Martina Mickler
- Physical Chemistry, Department of Chemistry, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Bojan Ljepoja
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Pjotr Knyazev
- Department of Molecular Biology, Max-Planck-Institute of Biochemistry, 82152 Martinsried, Germany
| | - Christoph Bräuchle
- Physical Chemistry, Department of Chemistry, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Axel Ullrich
- Department of Molecular Biology, Max-Planck-Institute of Biochemistry, 82152 Martinsried, Germany
| | - Ernst Wagner
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Andreas Roidl
- Pharmaceutical Biotechnology, Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| |
Collapse
|
31
|
Identification of DNA-PKcs as a primary resistance factor of salinomycin in osteosarcoma cells. Oncotarget 2018; 7:79417-79427. [PMID: 27765904 PMCID: PMC5346724 DOI: 10.18632/oncotarget.12712] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/26/2016] [Indexed: 01/01/2023] Open
Abstract
Malignant osteosarcoma (OS) is still a deadly disease for many affected patients. The search for the novel anti-OS agent is extremely urgent and important. Our previous study has proposed that salinomycin is a novel anti-OS agent. Here we characterized DNA-dependent protein kinase catalytic subunit (DNA-PKcs) as a primary salinomycin resistance factor in OS cells. DNA-PKcs inhibitors (NU7026, NU7441 and LY294002) or DNA-PKcs shRNA knockdown dramatically potentiated salinomycin-induced death and apoptosis of OS cells (U2OS and MG-63 lines). Further, forced-expression of microRNA-101 (“miR-101”) downregulated DNA-PKcs and augmented salinomycin's cytotoxicity against OS cells. Reversely, over-expression of DNA-PKcs in OS cells inhibited salinomycin's lethality. For the mechanism study, we show that DNA-PKcs is required for salinomycin-induced pro-survival autophagy activation. DNA-PKcs inhibition (by NU7441), shRNA knockdown or miR-101 expression inhibited salinomycin-induced Beclin-1 expression and autophagy induction. Meanwhile, knockdown of Beclin-1 by shRNA significantly sensitized salinomycin-induced OS cell lethality. In vivo, salinomycin administration suppressed U2OS xenograft tumor growth in severe combined immuno-deficient (SCID) mice, and its anti-tumor activity was dramatically potentiated with co-administration of the DNA-PKcs inhibitor NU7026. Together, these results suggest that DNA-PKcs could be a primary resistance factor of salinomycin in OS cells. DNA-PKcs inhibition or silence may thus significantly increase salinomycin's sensitivity in OS cells.
Collapse
|
32
|
Jiang J, Li H, Qaed E, Zhang J, Song Y, Wu R, Bu X, Wang Q, Tang Z. Salinomycin, as an autophagy modulator-- a new avenue to anticancer: a review. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:26. [PMID: 29433536 PMCID: PMC5809980 DOI: 10.1186/s13046-018-0680-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 01/15/2018] [Indexed: 12/25/2022]
Abstract
Since Salinomycin (Sal) emerged its ability to target breast cancer stem cells in 2009, numerous experiments have been carried out to test Sal’s anticancer effects. What deserve to be mentioned is that Sal can efficiently induce proliferation inhibition, cell death and metastasis suppression against human cancers from different origins both in vivo and in vitro without causing serious side effects as the conventional chemotherapeutical drugs on the body. There may be novel cell death pathways involving the anticancer effects of Sal except the conventional pathways, such as autophagic pathway. This review is focused on how autophagy involves the effects of Sal, trying to describe clearly and systematically why autophagy plays a vital role in predominant anticancer effects of Sal, including its distinctive characteristic. Based on recent advances, we present evidence that a dual role of Sal involving in autophagy may account for its unique anticancer effects - the preference for cancer cells. Further researches are required to confirm the authenticity of this suppose in order to develop an ideal anticancer drug.
Collapse
Affiliation(s)
- Jiang Jiang
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Hailong Li
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Eskandar Qaed
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Jing Zhang
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Yushu Song
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Rong Wu
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Xinmiao Bu
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Qinyan Wang
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| | - Zeyao Tang
- Department of Pharmacology, Dalian Medical University, 9 west section, south road of Lvshun, Dalian, 116044, China
| |
Collapse
|
33
|
Pellegrini P, Dyczynski M, Sbrana FV, Karlgren M, Buoncervello M, Hägg-Olofsson M, Ma R, Hartman J, Bajalica-Lagercrantz S, Grander D, Kharaziha P, De Milito A. Tumor acidosis enhances cytotoxic effects and autophagy inhibition by salinomycin on cancer cell lines and cancer stem cells. Oncotarget 2018; 7:35703-35723. [PMID: 27248168 PMCID: PMC5094956 DOI: 10.18632/oncotarget.9601] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Accepted: 04/20/2016] [Indexed: 01/07/2023] Open
Abstract
Sustained autophagy contributes to the metabolic adaptation of cancer cells to hypoxic and acidic microenvironments. Since cells in such environments are resistant to conventional cytotoxic drugs, inhibition of autophagy represents a promising therapeutic strategy in clinical oncology. We previously reported that the efficacy of hydroxychloroquine (HCQ), an autophagy inhibitor under clinical investigation is strongly impaired in acidic tumor environments, due to poor uptake of the drug, a phenomenon widely associated with drug resistance towards many weak bases. In this study we identified salinomycin (SAL) as a potent inhibitor of autophagy and cytotoxic agent effective on several cancer cell lines under conditions of transient and chronic acidosis. Since SAL has been reported to specifically target cancer-stem cells (CSC), we used an established model of breast CSC and CSC derived from breast cancer patients to examine whether this specificity may be associated with autophagy inhibition. We indeed found that CSC-like cells are more sensitive to autophagy inhibition compared to cells not expressing CSC markers. We also report that the ability of SAL to inhibit mammosphere formation from CSC-like cells was dramatically enhanced in acidic conditions. We propose that the development and use of clinically suitable SAL derivatives may result in improved autophagy inhibition in cancer cells and CSC in the acidic tumor microenvironment and lead to clinical benefits.
Collapse
Affiliation(s)
- Paola Pellegrini
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| | - Matheus Dyczynski
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| | | | - Maria Karlgren
- Department of Pharmacy and Uppsala University Drug Optimization and Pharmaceutical Profiling Platform (UDOPP) - Science for Life Laboratory, Department of Pharmacy, Uppsala Biomedical Center, Uppsala University, Sweden
| | | | - Maria Hägg-Olofsson
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| | - Ran Ma
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| | - Johan Hartman
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| | | | - Dan Grander
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| | - Pedram Kharaziha
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| | - Angelo De Milito
- Department of Oncology-Pathology, Cancer Center Karolinska, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
34
|
Semi-synthetic salinomycin analogs exert cytotoxic activity against human colorectal cancer stem cells. Biochem Biophys Res Commun 2018; 495:53-59. [DOI: 10.1016/j.bbrc.2017.10.147] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 10/27/2017] [Indexed: 12/16/2022]
|
35
|
Klose J, Guerlevik E, Trostel T, Kühnel F, Schmidt T, Schneider M, Ulrich A. Salinomycin inhibits cholangiocarcinoma growth by inhibition of autophagic flux. Oncotarget 2017; 9:3619-3630. [PMID: 29423070 PMCID: PMC5790487 DOI: 10.18632/oncotarget.23339] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 11/26/2017] [Indexed: 12/21/2022] Open
Abstract
Introduction Cholangiocarcinoma is characterized by aggressive tumor growth, high recurrence rates, and resistance against common chemotherapeutical regimes. The polyether-antibiotic Salinomycin is a promising drug in cancer therapy because of its ability to overcome apoptosis resistance of cancer cells and its selectivity against cancer stem cells. Here, we investigated the effectiveness of Salinomycin against cholangiocarcinoma in vivo, and analyzed interference of Salinomycin with autophagic flux in human cholangiocarcinoma cells. Results Salinomycin reduces tumor cell viability, proliferation, migration, invasion, and induced apoptosis in vitro. Subcutaneous and intrahepatic cholangiocarcinoma growth in vivo was inhibited upon Salinomycin treatment. Analysis of autophagy reveals inhibition of autophagic activity. This was accompanied by accumulation of mitochondrial mass and increased generation of reactive oxygen species. Conclusions This study demonstrates the effectiveness of Salinomycin against cholangiocarcinoma in vivo. Inhibition of autophagic flux represents an underlying molecular mechanism of Salinomycin against cholangiocarcinoma. Methods The two murine cholangiocarcinoma cell lines p246 and p254 were used to analyze tumor cell proliferation, viability, migration, invasion, and apoptosis in vitro. For in vivo studies, murine cholangiocarcinoma cells were injected into syngeneic C57-BL/6-mice to initiate subcutaneous cholangiocarcinoma growth. Intrahepatic tumor growth was induced by electroporation of oncogenic transposon-plasmids into the left liver lobe. For mechanistic studies in human cells, TFK-1 and EGI-1 were used, and activation of autophagy was analyzed after exposure to Salinomycin.
Collapse
Affiliation(s)
- Johannes Klose
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg 69120, Germany
| | - Engin Guerlevik
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover 30625, Germany
| | - Tina Trostel
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg 69120, Germany
| | - Florian Kühnel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover 30625, Germany
| | - Thomas Schmidt
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg 69120, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg 69120, Germany
| | - Alexis Ulrich
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg 69120, Germany
| |
Collapse
|
36
|
Han L, Zhang Y, Liu S, Zhao Q, Liang X, Ma Z, Gupta PK, Zhao M, Wang A. Autophagy flux inhibition, G2/M cell cycle arrest and apoptosis induction by ubenimex in glioma cell lines. Oncotarget 2017; 8:107730-107743. [PMID: 29296201 PMCID: PMC5746103 DOI: 10.18632/oncotarget.22594] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 11/03/2017] [Indexed: 12/21/2022] Open
Abstract
This study aimed to investigate whether ubenimex could work as an anti-tumor drug alone in glioma cells and figure out the underlying potential mechanisms. Ubenimex is widely used as an adjunct therapy in multiple solid cancers. However, it is rarely used to treat glioblastoma. The function of ubenimex in enhancing JQ1 treatment sensitivity of glioma cells by blocking autophagic degradation of HEXIM1 was previously studied. However, the detailed mechanism of autophagy regulation by ubenimex remains unclear. The U87 and U251 cell lines were treated with different doses of ubenimex. Cell viability was measured by using the WST-8 assay. Cell death was assessed using trypan blue staining and flow cytometry. The migration and invasive ability of glioma cells were examined by transwell migration/invasion assay. LC3-GFP-RFP was used to measure autophagic flux. Protein expression was assessed by Western blot analysis. Autophagosomes were evaluated using the transmission electron microscopy. Moreover, cell cycle arrest (PI Staining) was measured by flow cytometry. Results revealed that ubenimex inhibited cell proliferation as well as migration/invasion in glioma cells. Besides, ubenimex increased glioma cell death via autophagic flux inhibition. Meanwhile, ubenimex induced G2/M phase arrest and apoptosis, and this effect was accompanied by the decreased levels of p-Akt, indicating the role of ubenimex in the regulation of glioma cell proliferation and metastasis. To sum up, this study concluded that ubenimex could work as an anti-tumor drug alone in the glioma cells via inhibiting autophagic flux and inducing G2/M arrest as well as apoptosis.
Collapse
Affiliation(s)
- Liping Han
- Department of Neurology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, P.R. China.,Department of Neurology, Shandong Police Hospital, Jinan, P.R. China
| | - Yongfei Zhang
- Department of Dermatology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, P.R. China
| | - Shuai Liu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, P.R. China
| | - Qingwei Zhao
- Department of Neurology, Shandong Police Hospital, Jinan, P.R. China
| | - Xianhong Liang
- Department of Neurology, Shandong Police Hospital, Jinan, P.R. China
| | - Zhiguo Ma
- Department of Neurology, Shandong Police Hospital, Jinan, P.R. China
| | | | - Miaoqing Zhao
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, P.R. China
| | - Aihua Wang
- Department of Neurology, Qianfoshan Hospital Affiliated to Shandong University, Jinan, P.R. China
| |
Collapse
|
37
|
Therapeutic effects of the euglenoid ichthyotoxin, euglenophycin, in colon cancer. Oncotarget 2017; 8:104347-104358. [PMID: 29262645 PMCID: PMC5732811 DOI: 10.18632/oncotarget.22238] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Accepted: 10/13/2017] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) remains one of the most commonly diagnosed cancers and the 3rd leading cause of cancer-related mortality. The emergence of drug resistance poses a major challenge in CRC care or treatment. This can be addressed by determining cancer mechanisms, discovery of druggable targets, and development of new drugs. In search for novel agents, aquatic microorganisms offer a vastly untapped pharmacological source that can be developed for cancer therapeutics. In this study, we characterized the anti-colorectal cancer potential of euglenophycin, a microalgal toxin from Euglena sanguinea. The toxin (49.1-114.6 μM) demonstrated cytotoxic, anti-proliferative, anti-clonogenic, and anti-migration effects against HCT116, HT29, and SW620 CRC cells. We identified G1 cell cycle arrest and cell type - dependent modulation of autophagy as mechanisms of growth inhibition. We validated euglenophycin’s anti-tumorigenic activity in vivo using CRL:Nu(NCr)Foxn1nu athymic nude mouse CRC xenograft models. Intraperitoneal toxin administration (100 mg/kg; 5 days) decreased HCT116 and HT29 xenograft tumor volumes (n=10 each). Tumor inhibition was associated with reduced expression of autophagy negative regulator mechanistic target of rapamycin (mTOR) and decreased trend of serum pro-inflammatory cytokines. Together, these results provide compelling evidence that euglenophycin can be a promising anti-colorectal cancer agent targeting multiple cancer-promoting processes. Furthermore, this study supports expanding natural products drug discovery to freshwater niches as prospective sources of anti-cancer compounds.
Collapse
|
38
|
Chen X, Chen L, Jiang S, Huang S. Maduramicin induces apoptosis and necrosis, and blocks autophagic flux in myocardial H9c2 cells. J Appl Toxicol 2017; 38:366-375. [PMID: 29047155 DOI: 10.1002/jat.3546] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 08/30/2017] [Accepted: 09/07/2017] [Indexed: 12/18/2022]
Abstract
Maduramicin, a polyether ionophore antibiotic, is widely used as an anticoccidial agent in the poultry industry. It has been reported that maduramicin may cause heart and skeletal muscle cell damage, resulting in heart failure, skeletal muscle degeneration and even death in animals and humans, if improperly used. However, the molecular mechanism behind its capability to cause death of cardiac cells is not known. Here, we show that maduramicin induced apoptosis and necrosis in rat myocardial cells (H9c2). Maduramicin did not apparently upregulate the expression of pro-apoptotic proteins (e.g., BAD, BAK and BAX) or downregulate the expression of anti-apoptotic proteins (e.g. Bcl-2, Bcl-xL, Mcl-1 and survivin). Interestingly, maduramicin increased the expression of DR4 and TRAIL, activating caspases 8/3 and triggering cleavage of poly ADP ribose polymerase (PARP). In addition, maduramicin induced nuclear translocation of apoptosis inducing factor. Furthermore, maduramicin blocked autophagic flux, as evidenced by inducing accumulation of both LC3-II and p62/SQSTM1. Taken together, the above results suggest that maduramicin executes its toxicity in the myocardial cells at least by inducing caspase-dependent cell death through TRAIL/DR4-mediated extrinsic pathway and caspase-independent cell death by inducing apoptosis inducing factor nuclear translocation and blocking autophagic flux. Our findings provide a new insight into the molecular mechanism of maduramicin's toxicity in myocardial cells.
Collapse
Affiliation(s)
- Xin Chen
- Postdoctoral Mobile Station of Biology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu Province, 210023, People's Republic of China.,Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, People's Republic of China.,Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130-3932, USA
| | - Long Chen
- Postdoctoral Mobile Station of Biology, College of Life Sciences, Nanjing Normal University, Nanjing, Jiangsu Province, 210023, People's Republic of China
| | - Shanxiang Jiang
- Laboratory of Veterinary Pharmacology and Toxicology, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu Province, 210095, People's Republic of China
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130-3932, USA.,Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA, 71130-3932, USA
| |
Collapse
|
39
|
Regulation of SIRT1/AMPK axis is critically involved in gallotannin-induced senescence and impaired autophagy leading to cell death in hepatocellular carcinoma cells. Arch Toxicol 2017; 92:241-257. [PMID: 28676953 DOI: 10.1007/s00204-017-2021-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 06/28/2017] [Indexed: 12/20/2022]
|
40
|
Klose J, Eissele J, Volz C, Schmitt S, Ritter A, Ying S, Schmidt T, Heger U, Schneider M, Ulrich A. Salinomycin inhibits metastatic colorectal cancer growth and interferes with Wnt/β-catenin signaling in CD133 + human colorectal cancer cells. BMC Cancer 2016; 16:896. [PMID: 27855654 PMCID: PMC5114842 DOI: 10.1186/s12885-016-2879-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 10/24/2016] [Indexed: 12/22/2022] Open
Abstract
Background The polyether antibiotic Salinomycin (Sal) is regarded as an inhibitor of cancer stem cells. Its effectiveness on human colorectal cancer (CRC) cells in vitro has been demonstrated before. The aim of this study was to establish a murine model to investigate the effectiveness of Sal in vivo. Furthermore, we investigated the impact of Sal on Wnt/β-catenin signaling in human CD133+ CRC cells. Methods The two murine CRC cell lines MC38 and CT26 were used to analyze the impact of Sal on tumor cell proliferation, viability, migration, cell cycle progression and cell death in vitro. For in vivo studies, CT26 cells were injected into syngeneic BALB/c mice to initiate (i) subcutaneous, (ii) orthotopic, or (iii) metastatic CRC growth. Sal was administered daily, 5-Fluoruracil served as a control. For mechanistic studies, the CD133+and CD133- subpopulations of human CRC cells were separated by flow cytometry and separately exposed to increasing concentrations of Sal. The impact on Wnt/β-catenin signaling was determined by Western blotting and quantitative PCR. Results Sal markedly impaired tumor cell viability, proliferation and migration, and induced necrotic cell death in vitro. CRC growth in vivo was likewise inhibited upon Sal treatment. Interference with Wnt signaling and reduced expression of the Wnt target genes Fibronectin and Lgr5 indicates a novel molecular mechanism, mediating anti-tumoral effects of Sal in CRC. Conclusion Sal effectively impairs CRC growth in vivo. Furthermore, Sal acts as an inhibitor of Wnt/β-catenin signaling. Thus, Salinomycin represents a promising candidate for clinical CRC treatment. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2879-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Johannes Klose
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany.
| | - Jana Eissele
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Claudia Volz
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Steffen Schmitt
- German Cancer Research Center, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Alina Ritter
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Shen Ying
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Thomas Schmidt
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Ulrike Heger
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| | - Alexis Ulrich
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Im Neuenheimer Feld 110, 69120, Heidelberg, Germany
| |
Collapse
|
41
|
Zhang XF, Gurunathan S. Combination of salinomycin and silver nanoparticles enhances apoptosis and autophagy in human ovarian cancer cells: an effective anticancer therapy. Int J Nanomedicine 2016; 11:3655-75. [PMID: 27536105 PMCID: PMC4977082 DOI: 10.2147/ijn.s111279] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Ovarian cancer is one of the most important malignancies, and the origin, detection, and pathogenesis of epithelial ovarian cancer remain elusive. Although many cancer drugs have been developed to dramatically reduce the size of tumors, most cancers eventually relapse, posing a critical problem to overcome. Hence, it is necessary to identify possible alternative therapeutic approaches to reduce the mortality rate of this devastating disease. To identify alternative approaches, we first synthesized silver nanoparticles (AgNPs) using a novel bacterium called Bacillus clausii. The synthesized AgNPs were homogenous and spherical in shape, with an average size of 16–20 nm, which are known to cause cytotoxicity in various types of human cancer cells, whereas salinomycin (Sal) is able to kill cancer stem cells. Therefore, we selected both Sal and AgNPs to study their combined effect on apoptosis and autophagy in ovarian cancer cells. The cells treated with either Sal or AgNPs showed a dose-dependent effect with inhibitory concentration (IC)-50 values of 6.0 µM and 8 µg/mL for Sal and AgNPs, respectively. To determine the combination effect, we measured the IC25 values of both Sal and AgNPs (3.0 µM and 4 µg/mL), which showed a more dramatic inhibitory effect on cell viability and cell morphology than either Sal or AgNPs alone. The combination of Sal and AgNPs had more pronounced effect on cytotoxicity and expression of apoptotic genes and also significantly induced the accumulation of autophagolysosomes, which was associated with mitochondrial dysfunction and loss of cell viability. Our data show a strong synergistic interaction between Sal and AgNPs in tested cancer cells. The combination treatment increased the therapeutic potential and demonstrated the relevant targeted therapy for the treatment of ovarian cancer. Furthermore, we provide, for the first time, a mode of action for Sal and AgNPs in ovarian cancer cells: enhanced apoptosis and autophagy.
Collapse
Affiliation(s)
- Xi-Feng Zhang
- College of Biological and Pharmaceutical Engineering, Wuhan Polytechnic University, Wuhan, People's Republic of China
| | | |
Collapse
|
42
|
Glucose starvation-mediated inhibition of salinomycin induced autophagy amplifies cancer cell specific cell death. Oncotarget 2016; 6:10134-45. [PMID: 25912307 PMCID: PMC4496345 DOI: 10.18632/oncotarget.3548] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 02/13/2015] [Indexed: 12/30/2022] Open
Abstract
Salinomycin has been used as treatment for malignant tumors in a small number of humans, causing far less side effects than standard chemotherapy. Several studies show that Salinomycin targets cancer-initiating cells (cancer stem cells, or CSC) resistant to conventional therapies. Numerous studies show that Salinomycin not only reduces tumor volume, but also decreases tumor recurrence when used as an adjuvant to standard treatments. In this study we show that starvation triggered different stress responses in cancer cells and primary normal cells, which further improved the preferential targeting of cancer cells by Salinomycin. Our in vitro studies further demonstrate that the combined use of 2-Fluoro 2-deoxy D-glucose, or 2-deoxy D-glucose with Salinomycin is lethal in cancer cells while the use of Oxamate does not improve cell death-inducing properties of Salinomycin. Furthermore, we show that treatment of cancer cells with Salinomycin under starvation conditions not only increases the apoptotic caspase activity, but also diminishes the protective autophagy normally triggered by the treatment with Salinomycin alone. Thus, this study underlines the potential use of Salinomycin as a cancer treatment, possibly in combination with short-term starvation or starvation-mimicking pharmacologic intervention.
Collapse
|
43
|
Unravelling the relationship between macroautophagy and mitochondrial ROS in cancer therapy. Apoptosis 2016; 21:517-31. [DOI: 10.1007/s10495-016-1236-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
44
|
Phosphoethanolamine Modification of Neisseria gonorrhoeae Lipid A Reduces Autophagy Flux in Macrophages. PLoS One 2015; 10:e0144347. [PMID: 26641098 PMCID: PMC4671640 DOI: 10.1371/journal.pone.0144347] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 11/17/2015] [Indexed: 12/14/2022] Open
Abstract
Autophagy, an ancient homeostasis mechanism for macromolecule degradation, performs an important role in host defense by facilitating pathogen elimination. To counteract this host defense strategy, bacterial pathogens have evolved a variety of mechanisms to avoid or otherwise dysregulate autophagy by phagocytic cells so as to enhance their survival during infection. Neisseria gonorrhoeae is a strictly human pathogen that causes the sexually transmitted infection, gonorrhea. Phosphoethanolamine (PEA) addition to the 4' position of the lipid A (PEA-lipid A) moiety of the lipooligosaccharide (LOS) produced by gonococci performs a critical role in this pathogen’s ability to evade innate defenses by conferring decreased susceptibility to cationic antimicrobial (or host-defense) peptides, complement-mediated killing by human serum and intraleukocytic killing by human neutrophils compared to strains lacking this PEA decoration. Heretofore, however, it was not known if gonococci can evade autophagy and if so, whether PEA-lipid A contributes to this ability. Accordingly, by using murine macrophages and human macrophage-like phagocytic cell lines we investigated if PEA decoration of gonococcal lipid A modulates autophagy formation. We report that infection with PEA-lipid A-producing gonococci significantly reduced autophagy flux in murine and human macrophages and enhanced gonococcal survival during their association with macrophages compared to a PEA-deficient lipid A mutant. Our results provide further evidence that PEA-lipid A produced by gonococci is a critical component in the ability of this human pathogen to evade host defenses.
Collapse
|
45
|
Cell type-dependent ROS and mitophagy response leads to apoptosis or necroptosis in neuroblastoma. Oncogene 2015; 35:3839-53. [PMID: 26640148 DOI: 10.1038/onc.2015.455] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 10/05/2015] [Indexed: 02/06/2023]
Abstract
A limiting factor in the therapeutic outcome of children with high-risk neuroblastoma is the intrinsic and acquired resistance to common chemotherapeutic treatments. Here we investigated the molecular mechanisms by which the hemisynthetic cardiac glycoside UNBS1450 overcomes this limitation and induces differential cell death modalities in both neuroblastic and stromal neuroblastoma through stimulation of a cell-type-specific autophagic response eventually leading to apoptosis or necroptosis. In neuroblastic SH-SY5Y cells, we observed a time-dependent production of reactive oxygen species that affects lysosomal integrity inducing lysosome-associated membrane protein 2 degradation and cathepsin B and L activation. Subsequent mitochondrial membrane depolarization and accumulation of mitochondria in phagophores occurred after 8h of UNBS1450 treatment. Results were confirmed by mitochondrial mass analysis, electron microscopy and co-localization of mitochondria with GFP-LC3, suggesting the impaired clearance of damaged mitochondria. Thus, a stress-induced defective autophagic flux and the subsequent lack of clearance of damaged mitochondria sensitized SH-SY5Y cells to UNBS1450-induced apoptosis. Inhibition of autophagy with small inhibitory RNAs against ATG5, ATG7 and Beclin-1 protected SH-SY5Y cells against the cytotoxic effect of UNBS1450 by inhibiting apoptosis. In contrast, autophagy progression towards the catabolic state was observed in stromal SK-N-AS cells: here reactive oxygen species (ROS) generation remained undetectable preserving intact lysosomes and engulfing damaged mitochondria after UNBS1450 treatment. Moreover, autophagy inhibition determined sensitization of SK-N-AS to apoptosis. We identified efficient mitophagy as the key mechanism leading to failure of activation of the apoptotic pathway that increased resistance of SK-N-AS to UNBS1450, triggering rather necroptosis at higher doses. Altogether we characterize here the differential modulation of ROS and mitophagy as a main determinant of neuroblastoma resistance with potential relevance for personalized anticancer therapeutic approaches.
Collapse
|
46
|
Early effects of the antineoplastic agent salinomycin on mitochondrial function. Cell Death Dis 2015; 6:e1930. [PMID: 26492365 PMCID: PMC4632293 DOI: 10.1038/cddis.2015.263] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Revised: 07/10/2015] [Accepted: 08/03/2015] [Indexed: 02/07/2023]
Abstract
Salinomycin, isolated from Streptomyces albus, displays antimicrobial activity. Recently, a large-scale screening approach identified salinomycin and nigericin as selective apoptosis inducers of cancer stem cells. Growing evidence suggests that salinomycin is able to kill different types of non-stem tumor cells that usually display resistance to common therapeutic approaches, but the mechanism of action of this molecule is still poorly understood. Since salinomycin has been suggested to act as a K(+) ionophore, we explored its impact on mitochondrial bioenergetic performance at an early time point following drug application. In contrast to the K(+) ionophore valinomycin, salinomycin induced a rapid hyperpolarization. In addition, mitochondrial matrix acidification and a significant decrease of respiration were observed in intact mouse embryonic fibroblasts (MEFs) and in cancer stem cell-like HMLE cells within tens of minutes, while increased production of reactive oxygen species was not detected. By comparing the chemical structures and cellular effects of this drug with those of valinomycin (K(+) ionophore) and nigericin (K(+)/H(+) exchanger), we conclude that salinomycin mediates K(+)/H(+) exchange across the inner mitochondrial membrane. Compatible with its direct modulation of mitochondrial function, salinomycin was able to induce cell death also in Bax/Bak-less double-knockout MEF cells. Since at the concentration range used in most studies (around 10 μM) salinomycin exerts its effect at the level of mitochondria and alters bioenergetic performance, the specificity of its action on pathologic B cells isolated from patients with chronic lymphocytic leukemia (CLL) versus B cells from healthy subjects was investigated. Mesenchymal stromal cells (MSCs), proposed to mimic the tumor environment, attenuated the apoptotic effect of salinomycin on B-CLL cells. Apoptosis occurred to a significant extent in healthy B cells as well as in MSCs and human primary fibroblasts. The results indicate that salinomycin, when used above μM concentrations, exerts direct, mitochondrial effects, thus compromising cell survival.
Collapse
|
47
|
Geno- and cytotoxicity of salinomycin in human nasal mucosa and peripheral blood lymphocytes. Toxicol In Vitro 2015; 29:813-8. [PMID: 25769976 DOI: 10.1016/j.tiv.2015.01.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 12/23/2014] [Accepted: 01/18/2015] [Indexed: 01/03/2023]
Abstract
Salinomycin is usually applied in stock breading but has also been described as a promising agent against cancer stem cells (CSC). However, knowledge about the toxicity of this ionophor substance is incomplete. The aim of this study was to investigate cyto- and genotoxic effects of salinomycin in human non-malignant cells. Primary human nasal mucosa cells (monolayer and mini organ cultures) and peripheral blood lymphocytes from 10 individuals were used to study the cytotoxic effects of salinomycin (0.1-175 μM) by annexin-propidiumiodide- and MTT-test. The comet assay was performed to evaluate DNA damage. Additionally, the secretion of interleukin-8 was analyzed by ELISA. Flow cytometry and MTT assay revealed significant cytotoxic effects in nasal mucosa cells and lymphocytes at low salinomycin concentrations of 10-20 μM. No genotoxic effects could be observed. IL-8 secretion was elevated at 5 μM. Salinomycin-induced cytotoxic and pro-inflammatory effects were seen at concentrations relevant for anti-cancer treatment. Concurrent to the evaluation of salinomycin application in experimental oncology, adverse effects in non-malignant cells need to be monitored and reduced as much as possible. Further studies are also warranted to evaluate the toxic effects in a variety of human cell systems, e.g., liver, kidney and muscle cells.
Collapse
|
48
|
Skiera I, Antoszczak M, Trynda J, Wietrzyk J, Boratyński P, Kacprzak K, Huczyński A. Antiproliferative Activity of Polyether Antibiotic -CinchonaAlkaloid Conjugates ObtainedviaClick Chemistry. Chem Biol Drug Des 2015; 86:911-7. [DOI: 10.1111/cbdd.12523] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 12/12/2014] [Accepted: 01/13/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Iwona Skiera
- Faculty of Chemistry; Adam Mickiewicz University; Umultowska 89b 61-614 Poznan Poland
| | - Michał Antoszczak
- Faculty of Chemistry; Adam Mickiewicz University; Umultowska 89b 61-614 Poznan Poland
| | - Justyna Trynda
- Ludwik Hierszfeld Institute of Immunology and Experimental Therapy; Polish Academy of Sciences; Rudolfa Weigla 12 53-114 Wroclaw Poland
| | - Joanna Wietrzyk
- Ludwik Hierszfeld Institute of Immunology and Experimental Therapy; Polish Academy of Sciences; Rudolfa Weigla 12 53-114 Wroclaw Poland
| | - Przemysław Boratyński
- Faculty of Chemistry; Wrocław University of Technology; Wyspiańskiego 27 50-370 Wroclaw Poland
| | - Karol Kacprzak
- Faculty of Chemistry; Adam Mickiewicz University; Umultowska 89b 61-614 Poznan Poland
| | - Adam Huczyński
- Faculty of Chemistry; Adam Mickiewicz University; Umultowska 89b 61-614 Poznan Poland
| |
Collapse
|