1
|
Świderska-Kołacz G, Madej M, Zmorzynski S, Styk W, Surowiec I, Witek B, Wojciechowska A, Czerwik-Marcinkowska J, Nowakowska A. Effects of bortezomib on intracellular antioxidant and apoptosis in HepG2cells. PeerJ 2025; 13:e19235. [PMID: 40313384 PMCID: PMC12045286 DOI: 10.7717/peerj.19235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 03/10/2025] [Indexed: 05/03/2025] Open
Abstract
Bortezomib, as a proteasome inhibitor, is used in clinical trials related to solid cancers. However, its use is not always associated with a good response to treatment. Taking into account the above, we decided to analyze the effect of the time-dependency (24 vs. 48 h) and the dose-dependency of bortezomib (2, 4, 8 and 16 nM) on apoptosis and activities of antioxidant enzymes such as catalase (CAT), superoxide dismutase (SOD), glutathione reductase (GR), glutathione peroxidase (GPx) and glutathione transferase (GST), as well as concentrations of reduced glutathione (GSH) and malondialdehyde (MDA) in hepatoblastoma cell line (HepG2) cells. We have shown that increasing concentrations of bortezomib caused (I) a gradual decrease in the levels of GSH; (II) changes in MDA concentrations and antioxidant enzymes activities; (III) increase in apoptosis levels in HepG2 cells. We did not find significant association between antioxidant parameters and number of apoptotic cells. Our study showed that the analyzed parameters (such as: CAT, SOD, GR, GPx, GST, GSH, MDA) changed after bortezomib treatment. It is important to search for new anti-cancer therapies based on next-generation proteasome inhibitors. It is possible that the use of proteins associated with oxidative stress will help enhance the action of these inhibitors and will provide a better treatment effect.
Collapse
Affiliation(s)
| | - Magdalena Madej
- Institute of Biology, Jan Kochanowski University, Kielce, Poland
| | | | - Wojciech Styk
- Academic Laboratory of Psychological Tests, Medical University of Lublin, Lublin, Poland
| | - Iwona Surowiec
- Institute of Biology, Jan Kochanowski University, Kielce, Poland
| | - Bożena Witek
- Institute of Biology, Jan Kochanowski University, Kielce, Poland
| | - Anna Wojciechowska
- Department of Geobotany and Landscape Planning, Nicolaus Copernicus University of Torun, Toruń, Poland
| | | | - Anna Nowakowska
- Department of Animal Physiology and Neurobiology, Nicolaus Copernicus University of Torun, Toruń, Poland
| |
Collapse
|
2
|
Matić M, Obradović A, Paunović M, Ognjanović B, Mihailović V, Srećković N, Stanković M. Green-Synthesized Silver Nanoparticles Using Filipendula ulmaria (L.) Maxim. and Salvia verticillata L. Extracts Inhibit Migration and Modulate Redox Homeostasis in Human Breast Cancer Cells via Nrf-2 Signaling Pathway. Antioxidants (Basel) 2025; 14:469. [PMID: 40298802 PMCID: PMC12024124 DOI: 10.3390/antiox14040469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 03/31/2025] [Accepted: 04/08/2025] [Indexed: 04/30/2025] Open
Abstract
Breast cancer is a leading cancer diagnosis for women around the world, with a variable degree of curability. Conventional chemotherapeutic treatments often induce toxicity and damage to healthy tissues, as well as the development of drug resistance, which is why an increasing number of new therapeutic regimens focus on the use of natural products and various modifications of their delivery to target tissues. Silver nanoparticles possess unique physicochemical characteristics, notably their increased surface area, suggesting that they hold significant potential for biomedical applications. This research evaluates the capacity of silver nanoparticles green synthesized with aqueous extracts of Filipendula ulmaria (FUAgNPs) and Salvia verticillata (SVAgNPs) to alter migration and redox homeostasis in the human breast cancer cell line MDA-MB-231. To determine the values of redox homeostasis parameters, the cells were treated with five different concentrations (5, 10, 20, 50, and 100 μg/mL) for 24 h and 72 h, while to test the migratory potential and concentrations of matrix metalloproteinase-9 (MMP-9) and nuclear factor erythroid 2-related factor 2 (Nrf-2), the cells were treated at two concentrations (5 and 50 µg/mL) for 72 h. The obtained results indicate increased production of superoxide anion radicals, malondialdehyde (MDA), and nitrites after the investigated treatment on MDA-MB-231 cells. The treatments induced only a slight elevation in Nrf-2 levels, which correlates with weak de novo synthesis of reduced glutathione (GSH), suggesting that the tested nanoparticles weaken the inherent antioxidative systems of the tested cells. The migration potential of cells was significantly reduced, and MMP-9 concentration was significantly inhibited. Based on the demonstrated antitumor effect, confirmed by the reduced migratory potential of the examined cells and disrupted redox balance, these nanoparticles have potential for additional investigation with the aim of improving the efficacy of antitumor therapy. Also, FUAgNPs and SVAgNPs possess the capacity to be potentially promising novel chemotherapeutic agents against breast cancer progression and metastasis.
Collapse
Affiliation(s)
- Miloš Matić
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (M.M.); (M.P.); (B.O.); (M.S.)
| | - Ana Obradović
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (M.M.); (M.P.); (B.O.); (M.S.)
| | - Milica Paunović
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (M.M.); (M.P.); (B.O.); (M.S.)
| | - Branka Ognjanović
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (M.M.); (M.P.); (B.O.); (M.S.)
| | - Vladimir Mihailović
- Department of Chemistry, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (V.M.); (N.S.)
| | - Nikola Srećković
- Department of Chemistry, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (V.M.); (N.S.)
| | - Milan Stanković
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia; (M.M.); (M.P.); (B.O.); (M.S.)
| |
Collapse
|
3
|
Pulliam CF, Fath MA, Sho S, Johnson ST, Wagner BA, Singhania M, Kalen AL, Bayanbold K, Solst SR, Allen BG, George BN, Caster JM, Buettner GR, Riley DP, Keene JL, Beardsley RA, Spitz DR. Pharmacological ascorbate combined with rucosopasem selectively radio-chemo-sensitizes NSCLC via generation of H 2O 2. Redox Biol 2025; 80:103505. [PMID: 39884000 PMCID: PMC11830350 DOI: 10.1016/j.redox.2025.103505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 02/01/2025] Open
Abstract
Differences in cancer and normal cell oxidative metabolism provide a unique therapeutic opportunity for developing combined modality approaches with redox-active small molecules as radio-chemosensitizers that are well-tolerated by normal tissues. Pentaazamacrocyclic Mn (II)-containing (MnPAM) superoxide dismutase (SOD) mimetics and pharmacological ascorbate given IV to achieve [mM] plasma levels (pharmacological ascorbate: P-AscH‾) have been shown to act individually as cancer cell radio- and chemosensitizers via the generation of H2O2in vivo. The current study shows that the combination of newly developed MnPAM dismutase mimetic, rucosopasem manganese (RUC) with P-AscH‾ radio-sensitizes non-small cell lung cancer cells (NSCLC) and increases steady state levels of intracellular H2O2 with no additional toxicity to normal human bronchial epithelial cells (HBECs). Conditional over expression of catalase (CAT) in H1299T CATc15 cells demonstrates that the combination of RUC and P-AscH‾ causes radio-sensitization through an H2O2-dependent mechanism. Interestingly, RUC combined with P-AscH‾ demonstrates more than additive cytotoxicity in both H1299T and A549 NSCLC cells, but conditional over-expression of ferritin heavy chain (FtH) protected only the H1299T, and not the A549, from this toxicity. Most importantly, the combination of RUC + P-AscH‾ was found to sensitize both H1299T and A549 cell types to radio-chemotherapy with cisplatin (CIS) + etoposide (ETOP). Finally, in H1299T NSCLC xenografts the combination of RUC + P-AscH‾ with CIS + ETOP and 12 × 2 Gy radiation significantly inhibits tumor growth and increased median overall over survival. These results support the hypothesis that selective MnPAM dismutase mimetic + P-AscH‾ enhances the efficacy of radio-chemotherapy in NSCLC through a mechanism governed by redox active metals and H2O2 production.
Collapse
Affiliation(s)
- C F Pulliam
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA; Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA, 52242, USA.
| | - M A Fath
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - S Sho
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - S T Johnson
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - B A Wagner
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - M Singhania
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - A L Kalen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - K Bayanbold
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - S R Solst
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - B G Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA; Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA, 52242, USA.
| | - B N George
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - J M Caster
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA.
| | - G R Buettner
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA; Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA, 52242, USA.
| | - D P Riley
- Galera Therapeutics, Malvern, PA, 19355, USA.
| | - J L Keene
- Galera Therapeutics, Malvern, PA, 19355, USA.
| | | | - D R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa College of Medicine, Iowa City, IA 52242, USA; Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA, 52242, USA
| |
Collapse
|
4
|
Mohapatra P, Ghosh S, Patra S, Mandal M, Das S. Comparing Ag-O coordinated AgMOF-5 and Ag-N coordinated Ag nanosphere catalytic polymers for real time monitoring of H 2O 2 level in cancer cells. Biosens Bioelectron 2025; 271:117056. [PMID: 39719814 DOI: 10.1016/j.bios.2024.117056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 12/06/2024] [Accepted: 12/08/2024] [Indexed: 12/26/2024]
Abstract
Monitoring H2O2 levels in live cells is essential due to its superior stability and possible severity inside the cell. The quest for a superior platform capable of detecting cellular-level hydrogen peroxide (H2O2) concentrations without necessitating the use of high-cost enzymes is of utmost importance. Here, the quantification of intracellular H2O2 concentrations has been performed using silver metal polymer-based nonenzymatic electrochemical detection. Two forms of silver metal-organic polymers are synthesized to explore its suitability as catalytic nanozyme activity for cell-level H2O2 detection with good accuracy. An Ag-O coordination interaction-based silver metal-organic framework, AgMOF-5 was synthesized and then transformed into a compact silver nanosphere (Ag nanosphere), rich in Ag-N coordination bond, to find the potential of individual coordination arrangement of each material in favour of the electrical conductivity, catalytic property, and sensing ability. AgMOF-5 modified Glassy Carbon Electrode (AgMOF/GCE) was found to be highly conductive while also demonstrating exceeding catalytic potential towards both commercial H2O2 and cell-secreted H2O2 with a LOD of 0.7 nM and sensitivity of 1300 μA mM-1cm-2. AgMOF/GCE demonstrated superior sensor characteristics like superconductivity, high stability, high reproducibility, and good selectivity upon investigation. Among many cellular byproducts of metabolism, H2O2 and other ROS species indicate the oxidative stress and are a key differentiator between healthy and diseased conditions. The modified GCE was used for live monitoring of H2O2 levels in L929, HeLa, T98G, and LN18 cell lines. The detection of higher H2O2 levels in glial cells established its predisposition towards increased oxidative stress. The electrochemical results of AgMOF/GCE were validated against a Phenol Red, HRPO-based live cell compatible Colourimetric detection for applicability and acceptance of AgMOF/GCE as H2O2 detection platform.
Collapse
Affiliation(s)
- Pratyusa Mohapatra
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur, 721302, West Bengal, India
| | - Swachhatoa Ghosh
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur, 721302, West Bengal, India
| | - Sucharita Patra
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur, 721302, West Bengal, India
| | - Mahitosh Mandal
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur, 721302, West Bengal, India
| | - Soumen Das
- School of Medical Science and Technology, Indian Institute of Technology, Kharagpur, 721302, West Bengal, India.
| |
Collapse
|
5
|
Lu C, Yu D, Wang X, Li J, Zhang Y, Wang C, Jia Q, Tan J, Zheng W, Sun H, Meng Z. Selective Therapeutic Potential of a H2O2-Inducible DNA Interstrand Cross-linker in Anaplastic Thyroid Carcinoma. Endocrinology 2025; 166:bqaf029. [PMID: 39950985 DOI: 10.1210/endocr/bqaf029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Indexed: 03/15/2025]
Abstract
We aimed to investigate hydrogen peroxide-inducible DNA interstrand cross-link (HP-ICL) as a targeted therapy for anaplastic thyroid cancer (ATC) due to its higher H2O2 content than normal cells. In vitro analysis included fluorescence microscopy for H2O2 levels and exposure of ATC cells to various HP-ICL concentrations followed by assessment of cell viability, apoptosis, cell cycle, and DNA damage using methyl thiazolyl tetrazolium (MTT), flow cytometry, and a γH2AX assay. Protein levels related to apoptosis and the phosphatidylinositol-3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway were measured by Western blotting. An ATC xenograft mouse model was used to evaluate the HP-ICL's in vivo effects. ATC cells had higher H2O2 levels than normal thyroid cells. HP-ICL treatment caused a dose-dependent decrease in cell viability and an increase in apoptosis, with a slight G2/M phase arrest. A 30 µM HP-ICL treatment doubled γH2AX foci. Bcl-2 levels decreased, while Bax, cleaved-Caspase 3, and PARP increased in a dose-dependent manner. It also inhibited p-PI3K, p-AKT, and p-mTOR. In vivo, the HP-ICL significantly inhibited tumor growth while maintaining body weight and without causing organ damage or altering thyroid hormone levels. Additionally, tumor sections exhibited increased TUNEL staining, decreased Ki67 expression, and reduced levels of p-PI3K, p-AKT, and p-mTOR. The HP-ICL significantly inhibited ATC both in vitro and in vivo, suggesting its potential as an effective therapy for ATC.
Collapse
Affiliation(s)
- Chenghui Lu
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, P. R. China
| | - Dehao Yu
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics School of Pharmacy, Tianjin Medical University, Tianjin 300070, P. R. China
| | - Xufu Wang
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, P. R. China
| | - Jiao Li
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, P. R. China
| | - Yingying Zhang
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, P. R. China
| | - Congcong Wang
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, Qingdao 266003, P. R. China
| | - Qiang Jia
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Jian Tan
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Wei Zheng
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| | - Huabing Sun
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics School of Pharmacy, Tianjin Medical University, Tianjin 300070, P. R. China
| | - Zhaowei Meng
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin 300052, P. R. China
| |
Collapse
|
6
|
Wang Y, Wang H, Hu J, Chai J, Luan J, Li J, Xu Q. FLASH radiotherapy: mechanisms, nanotherapeutic strategy and future development. NANOSCALE ADVANCES 2025; 7:711-721. [PMID: 39781242 PMCID: PMC11705069 DOI: 10.1039/d4na00753k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/21/2024] [Indexed: 01/12/2025]
Abstract
Ultra-high dose-rate (FLASH) radiotherapy serves as an ideal procedure to treat tumors efficiently without harming normal tissues and has demonstrated satisfactory antitumor effects in multiple animal tumor models. However, the biological mechanisms of FLASH radiotherapy have not yet been fully elucidated, and the small number of devices delivering FLASH dose rate has limited its wide application. This review summarizes the possible biological mechanisms and antitumor effects of FLASH radiotherapy, its application in nanotherapeutic strategy, as well as its challenges and future development. Furthermore, some valuable guidance for promoting the progress of FLASH radiotherapy in nanotherapeutic strategies are provided.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College Wuhu China
| | - Huifang Wang
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College Wuhu China
| | - Jiawei Hu
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College Wuhu China
| | - Jingjing Chai
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College Wuhu China
| | - Jiajie Luan
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College Wuhu China
| | - Jie Li
- Mianyang Central Hospital, School of Medicine, University of Electronic Science and Technology of China Mianyang China
| | - Qingwen Xu
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College Wuhu China
| |
Collapse
|
7
|
Zandieh A, Shariatpanahi SP, Ravassipour AA, Azadipour J, Nezamtaheri MS, Habibi-Kelishomi Z, Ghanizadeh M, Same-Majandeh A, Majidzadeh-A K, Taheri A, Ansari AM, Javidi MA, Pirnia MM, Goliaei B. An amplification mechanism for weak ELF magnetic fields quantum-bio effects in cancer cells. Sci Rep 2025; 15:2964. [PMID: 39849096 PMCID: PMC11757740 DOI: 10.1038/s41598-025-87235-w] [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: 09/01/2024] [Accepted: 01/17/2025] [Indexed: 01/25/2025] Open
Abstract
Observing quantum mechanical characteristics in biological processes is a surprising and important discovery. One example, which is gaining more experimental evidence and practical applications, is the effect of weak magnetic fields with extremely low frequencies on cells, especially cancerous ones. In this study, we use a mathematical model of ROS dynamics in cancer cells to show how ROS oscillatory patterns can act as a resonator to amplify the small effects of the magnetic fields on the radical pair dynamics in mitochondrial Complex III. We suggest such a resonator can act in two modes for distinct states in cancer cells: (1) cells at the edge of mitochondrial oscillation and (2) cells with local oscillatory patches. When exposed to magnetic fields, the first group exhibits high-amplitude oscillations, while the second group synchronizes to reach a whole-cell oscillation. Both types of amplification are frequency-dependent in the range of hertz and sub-hertz. We use UV radiation as a positive control to observe the two states of cells in DU and HELA cell lines. Application of magnetic fields shows frequency-dependent results on both the ROS and mitochondrial potential which agree with the model for both type of cells. We also observe the oscillatory behavior in the time-lapse fluorescence microscopy for 0.02 and 0.04 Hz magnetic fields. Finally, we investigate the dependence of the results on the field strength and propose a quantum spin-forbidden mechanism for the effect of magnetic fields on superoxide production in QO site of mitochondrial Complex III.
Collapse
Affiliation(s)
- Amirali Zandieh
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | | | | | - Javad Azadipour
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | | | | | - Mojtaba Ghanizadeh
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Ali Same-Majandeh
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Keivan Majidzadeh-A
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Amir Taheri
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Alireza Madjid Ansari
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Mohammad Amin Javidi
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | | | - Bahram Goliaei
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
8
|
Ivanova D, Semkova S, Grigorov B, Tzanova M, Georgieva A, Danchev D, Nikolova B, Yaneva Z. The General Principle of the Warburg Effect as a Possible Approach for Cancer Immunotherapy: The Regulatory Effect of Plant Extracts Could Change the Game. Molecules 2025; 30:393. [PMID: 39860262 PMCID: PMC11767411 DOI: 10.3390/molecules30020393] [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: 10/15/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
The interpretation of the biochemistry of immune metabolism could be considered an attractive scientific field of biomedicine research. In this review, the role of glycolysis in macrophage polarization is discussed together with mitochondrial metabolism in cancer cells. In the first part, the focus is on the Warburg effect and redox metabolism during macrophage polarization, cancer development, and management of the immune response by the cancer cells. The second part addresses the possibility of impacts on the Warburg effect through targeting peroxisome proliferator-activated receptors (PPARs). This could be an activator of native immune responses. Because of the reported serious adverse effects of using synthetic ligands for PPARs in combination with chemotherapeutics, searches for less toxic and more active PPAR inhibitors, as well as blocking undesirable cellular PPAR-dependent processes, are in progress. On the other hand, recent research in modern immunotherapy has focused on the search for gentle immune-modulating natural compounds with harmless synergistic chemotherapeutic efficacy that can be used as an adjuvant. It is a well-known fact that the plant kingdom is a source of important therapeutic agents with multifaceted effectiveness. One of these is the known association with PPAR activities. In this regard, the secondary metabolites extracted from plants could change the game.
Collapse
Affiliation(s)
- Donika Ivanova
- Department of Pharmacology, Animal Physiology Biochemistry and Chemistry, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria;
- Department of Chemistry and Biochemistry, Faculty of Medicine, Trakia University, 6000 Stara Zagora, Bulgaria
| | - Severina Semkova
- Department of Electroinduced and Adhesive Properties, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| | - Boncho Grigorov
- Department of Molecular Biology, Immunology and Medical Genetics, Faculty of Medicine, Trakia University, 6000 Stara Zagora, Bulgaria;
| | - Milena Tzanova
- Department of Biological Sciences, Faculty of Agriculture, Trakia University, 6000 Stara Zagora, Bulgaria;
| | | | | | - Biliana Nikolova
- Department of Electroinduced and Adhesive Properties, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, 1113 Sofia, Bulgaria;
| | - Zvezdelina Yaneva
- Department of Pharmacology, Animal Physiology Biochemistry and Chemistry, Faculty of Veterinary Medicine, Trakia University, 6000 Stara Zagora, Bulgaria;
| |
Collapse
|
9
|
Ibrahem E, Osman A, Taha H, Abo El-Maati MF, Sitohy B, Sitohy M. Anticarcinogenic cationic peptides derived from tryptic hydrolysis of β-lactoglobulin. Front Mol Biosci 2025; 11:1444457. [PMID: 39866644 PMCID: PMC11757936 DOI: 10.3389/fmolb.2024.1444457] [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: 06/05/2024] [Accepted: 12/09/2024] [Indexed: 01/28/2025] Open
Abstract
Introduction This study investigated the tryptic hydrolysis of β-lactoglobulin (BLG) for 30, 60, 90, and 120 min at 1/200 E/S (enzyme/substrate ratio, w/w) to prepare potentially anticarcinogenic peptides. Methods The properties of hydrolysates were characterized, including degree of hydrolysis, free amino acids, SDS-PAGE, FTIR, and antioxidant activity employing DPPH-assay, β-carotene/linoleic acid, and FRAP assay. Results BLG tryptic hydrolysate produced after 60 min hydrolysis recorded the highest antioxidant activity, and LCMS analysis revealed 162 peptides of molecular masses ranging from 800 to 5671Da, most of them are of hydrophobic nature. Within the low-MW peptide group (24 peptides), there were nine hydrophobic basic (HB) and seven hydrophobic acidic (HA), representing 38% and 29%, respectively. The HB peptides may be responsible for the considerable biological activity of the hydrolysate. With dominant basic character supporting the carcinogenic activity of this hydrolysate. The in vitro anticancer activity against Mcf-7, Caco-2, and A-549 human cancer cell lines proliferation by MTT assay recorded IC50% at 42.8, 76.92, and 45.93 μg/mL, respectively. Treating each cell line with IC50% of the hydrolysate for 24 h increased the apoptosis by enhancing the expression of caspase-9 by 5.66, 7.97, and 3.28 folds over the untreated control and inhibited angiogenesis by reducing VEGFR-2 expression by about 56, 76, and 70%, respectively, indicating strong anticancer and antiangiogenic actions on human cancer cells. BLG tryptic hydrolysate may serve as a natural anticarcinogenic agent. The results of this study demonstrated that BLG hydrolysates have direct anticancer and antiangiogenic effects on human cancer cells. The chemical composition and characteristics of the BLG tryptic hydrolysate influence these biological and anticancer activities. The tryptic hydrolysates were generally effective against the three cancer cell lines studied (Mcf-7, Caco-2, and A-549). This effectiveness was assessed by measuring cell proliferation using the MTT assay and by evaluating their impact on angiogenesis through inhibition of VEGFR-2 activity. Discussion Future studies may focus on enhancing the anticarcinogenic effectiveness of the peptides by isolating and evaluating the most prominent individual peptide and varying the treatment conditions.
Collapse
Affiliation(s)
- Eman Ibrahem
- Department of Biochemistry, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Ali Osman
- Department of Biochemistry, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Hefnawy Taha
- Department of Biochemistry, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | | | - Basel Sitohy
- Department of Clinical Microbiology, Infection and Immunology, Umeå University, Umeå, Sweden
- Department of Diagnostics and Intervention, Oncology, Umeå University, Umeå, Sweden
| | - Mahmoud Sitohy
- Department of Biochemistry, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| |
Collapse
|
10
|
He L, Zhang L, Peng Y, He Z. Selenium in cancer management: exploring the therapeutic potential. Front Oncol 2025; 14:1490740. [PMID: 39839762 PMCID: PMC11746096 DOI: 10.3389/fonc.2024.1490740] [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: 09/03/2024] [Accepted: 12/16/2024] [Indexed: 01/23/2025] Open
Abstract
Selenium (Se) is important and plays significant roles in many biological processes or physiological activities. Prolonged selenium deficiency has been conclusively linked to an elevated risk of various diseases, including but not limited to cancer, cardiovascular disease, inflammatory bowel disease, Keshan disease, and acquired immunodeficiency syndrome. The intricate relationship between selenium status and health outcomes is believed to be characterized by a non-linear U-shaped dose-response curve. This review delves into the significance of maintaining optimal selenium levels and the detrimental effects that can arise from selenium deficiency. Of particular interest is the important role that selenium plays in both prevention and treatment of cancer. Finally, this review also explores the diverse classes of selenium entities, encompassing selenoproteins, selenium compounds and selenium nanoparticles, while examining the mechanisms and molecular targets of their anticancer efficacy.
Collapse
Affiliation(s)
- Lingwen He
- Department of Oncology, Dongguan Songshan Lake Tungwah Hospital, Dongguan, China
| | - Lu Zhang
- Department of Oncology, Dongguan Songshan Lake Tungwah Hospital, Dongguan, China
| | - Yulong Peng
- Department of Oncology, Dongguan Tungwah Hospital, Dongguan, China
| | - Zhijun He
- School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan, China
- Shenzhen Key Laboratory of Marine Biotechnology and Ecology, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, Guangdong, China
| |
Collapse
|
11
|
Zuo Q, Kang Y. Metabolic Reprogramming and Adaption in Breast Cancer Progression and Metastasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:347-370. [PMID: 39821033 DOI: 10.1007/978-3-031-70875-6_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Recent evidence has revealed that cancer is not solely driven by genetic abnormalities but also by significant metabolic dysregulation. Cancer cells exhibit altered metabolic demands and rewiring of cellular metabolism to sustain their malignant characteristics. Metabolic reprogramming has emerged as a hallmark of cancer, playing a complex role in breast cancer initiation, progression, and metastasis. The different molecular subtypes of breast cancer exhibit distinct metabolic genotypes and phenotypes, offering opportunities for subtype-specific therapeutic approaches. Cancer-associated metabolic phenotypes encompass dysregulated nutrient uptake, opportunistic nutrient acquisition strategies, altered utilization of glycolysis and TCA cycle intermediates, increased nitrogen demand, metabolite-driven gene regulation, and metabolic interactions with the microenvironment. The tumor microenvironment, consisting of stromal cells, immune cells, blood vessels, and extracellular matrix components, influences metabolic adaptations through modulating nutrient availability, oxygen levels, and signaling pathways. Metastasis, the process of cancer spread, involves intricate steps that present unique metabolic challenges at each stage. Successful metastasis requires cancer cells to navigate varying nutrient and oxygen availability, endure oxidative stress, and adapt their metabolic processes accordingly. The metabolic reprogramming observed in breast cancer is regulated by oncogenes, tumor suppressor genes, and signaling pathways that integrate cellular signaling with metabolic processes. Understanding the metabolic adaptations associated with metastasis holds promise for identifying therapeutic targets to disrupt the metastatic process and improve patient outcomes. This chapter explores the metabolic alterations linked to breast cancer metastasis and highlights the potential for targeted interventions in this context.
Collapse
Affiliation(s)
- Qianying Zuo
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, USA
| | - Yibin Kang
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
- Ludwig Institute for Cancer Research Princeton Branch, Princeton, NJ, USA.
| |
Collapse
|
12
|
King SA, Solst SR, Graham CH, Fiore LZ, Rheem R, Tomanek-Chalkley A, Fath MA, Caster JM, Spitz DR, Howard ME. Additive Effects of Cu-ATSM and Radiation on Survival of Diffuse Intrinsic Pontine Glioma Cells. Radiat Res 2025; 203:10-17. [PMID: 39492578 PMCID: PMC11815956 DOI: 10.1667/rade-24-00076.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024]
Abstract
Diffuse intrinsic pontine gliomas (DIPG) are highly aggressive and treatment-resistant childhood primary brainstem tumors with a median survival of less than one year after diagnosis. The prevailing standard of care for DIPG, radiation therapy, does not prevent fatal disease progression, with most patients succumbing to this disease 3-8 months after completion of radiation therapy. This underscores the urgent need for novel combined-modality approaches for enhancing therapy responses. This study demonstrates that the cellular redox modulating drug, copper (II)-diacetyl-bis(N4-methylthiosemicarbazone) (Cu-ATSM) dose-dependently (1-3 μM) decreased clonogenic cell survival in SU-DIPG50 and SU-DIPG36 cell lines during 6 h of exposure but had no significant effect on survival in normal human astrocytes (NHA). Additional significant (>90%) decreases in DIPG clonogenic survival were observed at 24 h of Cu-ATSM exposure. However, NHAs also began to show dose-dependent 10-70% survival decreases at this point. Notably, 3 μM Cu-ATSM for 6 h resulted in additive clonogenic cell killing of DIPG lines when combined with radiation, which was not seen in NHAs and was partially inhibited by the copper chelator, bathocuproinedisulfonic acid. Cu-ATSM toxicity in DIPG cells was also inhibited by overexpression of mitochondrial-targeted catalase. These results support the hypothesis that Cu-ATSM is selectively cytotoxic to DIPGs by a mechanism involving H2O2 generation and copper and being additively cytotoxic with ionizing radiation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Michelle E. Howard
- Free Radical and Radiation Biology Program, B180 Medical Laboratories, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa 52242
| |
Collapse
|
13
|
Johnson SS, Liu D, Ewald JT, Robles-Planells C, Pulliam C, Christensen KA, Bayanbold K, Wels BR, Solst SR, O’Dorisio MS, Menda Y, Spitz DR, Fath MA. Auranofin inhibition of thioredoxin reductase sensitizes lung neuroendocrine tumor cells (NETs) and small cell lung cancer (SCLC) cells to sorafenib as well as inhibiting SCLC xenograft growth. Cancer Biol Ther 2024; 25:2382524. [PMID: 39054566 PMCID: PMC11275529 DOI: 10.1080/15384047.2024.2382524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/23/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024] Open
Abstract
Thioredoxin Reductase (TrxR) functions to recycle thioredoxin (Trx) during hydroperoxide metabolism mediated by peroxiredoxins and is currently being targeted using the FDA-approved anti-rheumatic drug, auranofin (AF), to selectively sensitize cancer cells to therapy. AF treatment decreased TrxR activity and clonogenic survival in small cell lung cancer (SCLC) cell lines (DMS273 and DMS53) as well as the H727 atypical lung carcinoid cell line. AF treatment also significantly sensitized DMS273 and H727 cell lines in vitro to sorafenib, an FDA-approved multi-kinase inhibitor that depleted intracellular glutathione (GSH). The pharmacokinetic, pharmacodynamic, and safety profile of AF was examined in nude mice with DMS273 xenografts administered AF intraperitoneally at 2 mg/kg or 4 mg/kg (IP) once (QD) or twice daily (BID) for 1-5 d. Plasma levels of AF were 10-20 μM (determined by mass spectrometry of gold), and the optimal inhibition of TrxR activity was obtained at 4 mg/kg once daily, with no effect on glutathione peroxidase 1 activity. This AF treatment extended for 14 d, inhibited TrxR (>75%), and resulted in a significant prolongation of median overall survival from 19 to 23 d (p = .04, N = 30 controls, 28 AF). In this experiment, there were no observed changes in animal bodyweight, complete blood counts (CBCs), bone marrow toxicity, blood urea nitrogen, or creatinine. These results support the hypothesis that AF effectively inhibits TrxR both in vitro and in vivo in SCLC, sensitizes NETs and SCLC to sorafenib, and could be repurposed as an adjuvant therapy with targeted agents that induce disruptions in thiol metabolism.
Collapse
Affiliation(s)
- Spenser S. Johnson
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, Free Radical and Radiation Biology Program, University of Iowa Hospitals and Clinics, IA, USA
| | - Dijie Liu
- Department Pediatrics, University of Iowa Hospitals and Clinics, IA, USA
| | - Jordan T. Ewald
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, Free Radical and Radiation Biology Program, University of Iowa Hospitals and Clinics, IA, USA
| | | | - Casey Pulliam
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, Free Radical and Radiation Biology Program, University of Iowa Hospitals and Clinics, IA, USA
| | - Keegan A. Christensen
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, Free Radical and Radiation Biology Program, University of Iowa Hospitals and Clinics, IA, USA
| | - Khaliunaa Bayanbold
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, Free Radical and Radiation Biology Program, University of Iowa Hospitals and Clinics, IA, USA
| | - Brian R. Wels
- State Hygienic Laboratory, University of Iowa, IA, USA
| | - Shane R. Solst
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, Free Radical and Radiation Biology Program, University of Iowa Hospitals and Clinics, IA, USA
| | - M. Sue O’Dorisio
- Department Pediatrics, University of Iowa Hospitals and Clinics, IA, USA
| | - Yusuf Menda
- Department of Radiology, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, IA, USA
| | - Douglas R. Spitz
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, Free Radical and Radiation Biology Program, University of Iowa Hospitals and Clinics, IA, USA
| | - Melissa A. Fath
- Department of Radiation Oncology, Holden Comprehensive Cancer Center, Free Radical and Radiation Biology Program, University of Iowa Hospitals and Clinics, IA, USA
| |
Collapse
|
14
|
Aisu Y, Oshima N, Hyodo F, Elhelaly AE, Masuo A, Okada T, Hisamori S, Tsunoda S, Hida K, Morimoto T, Miyoshi H, Taketo MM, Matsuo M, Neckers LM, Sakai Y, Obama K. Dual inhibition of oxidative phosphorylation and glycolysis exerts a synergistic antitumor effect on colorectal and gastric cancer by creating energy depletion and preventing metabolic switch. PLoS One 2024; 19:e0309700. [PMID: 39666615 PMCID: PMC11637386 DOI: 10.1371/journal.pone.0309700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 08/18/2024] [Indexed: 12/14/2024] Open
Abstract
Pyruvate is situated at the intersection of oxidative phosphorylation (OXPHOS) and glycolysis, which are the primary energy-producing pathways in cells. Cancer therapies targeting these pathways have been previously documented, indicating that inhibiting one pathway may lead to functional compensation by the other, resulting in an insufficient antitumor effect. Thus, effective cancer treatment necessitates concurrent and comprehensive suppression of both. However, whether a metabolic switch between the metabolic pathways occurs in colorectal and gastric cancer cells and whether blocking it by inhibiting both pathways has an antitumor effect remain to be determined. In the present study, we used two small molecules, namely OXPHOS and glycolysis inhibitors, to target pyruvate metabolic pathways as a cancer treatment in these cancer cells. OXPHOS and glycolysis inhibition each augmented the other metabolic pathway in vitro and in vivo. OXPHOS inhibition alone enhanced glycolysis and showed antitumor effects on colorectal and gastric cancer cells in vitro and in vivo. Moreover, glycolysis inhibition in addition to OXPHOS inhibition blocked the metabolic switch from OXPHOS to glycolysis, causing an energy depletion and deterioration of the tumor microenvironment that synergistically enhanced the antitumor effect of OXPHOS inhibitors. In addition, using hyperpolarized 13C-magnetic resonance spectroscopic imaging (HP-MRSI), which enables real-time and in vivo monitoring of molecules containing 13C, we visualized how the inhibitors shifted the flux of pyruvate and how this dual inhibition in colorectal and gastric cancer mouse models altered the two pathways. Integrating dual inhibition of OXPHOS and glycolysis with HP-MRSI, this therapeutic model shows promise as a future "cancer theranostics" treatment option.
Collapse
Affiliation(s)
- Yuki Aisu
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Nobu Oshima
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Surgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Fuminori Hyodo
- Department of Radiology, Gifu University Hospital, Gifu, Japan
- Department of Radiology, Frontier Science for Imaging, Gifu University, Gifu, Japan
| | | | - Akihiko Masuo
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomoaki Okada
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shigeo Hisamori
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Shigeru Tsunoda
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Koya Hida
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Tomonori Morimoto
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroyuki Miyoshi
- Colon Cancer Project, Kyoto University Hospital-iACT, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Makoto M. Taketo
- Colon Cancer Project, Kyoto University Hospital-iACT, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masayuki Matsuo
- Department of Radiology, Gifu University Hospital, Gifu, Japan
| | - Leonard M. Neckers
- National Cancer Institute, Urologic Oncology Branch, Center for Cancer Research, NIH, Bethesda, Maryland, United States of America
| | - Yoshiharu Sakai
- Department of Gastrointestinal Surgery, Osaka Red Cross Hospital, Osaka, Japan
| | - Kazutaka Obama
- Department of Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
15
|
Korns J, Wicker CA, Lehn M, Shyamsunder S, Thompson S, Lester C, Wise-Draper TM, Waltz SE, Takiar V. Telaglenastat as an alternative to cisplatin as a radiosensitizer in the treatment of head and neck squamous cell carcinoma. Cancer Lett 2024; 606:217320. [PMID: 39489210 PMCID: PMC11583984 DOI: 10.1016/j.canlet.2024.217320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024]
Abstract
The efficacy of radiation treatment (RT) of head and neck squamous cell carcinoma (HNSCC) is limited by radioresistance and the toxicity of FDA approved radiosensitizers. In extension to our previous research where we demonstrated that telaglenastat (CB839) increased efficacy of RT in in vitro and in vivo HNSCC models, here, we examine the radiosensitizing effects of telaglenastat in comparison to cisplatin's, as cisplatin is currently the standard of care for concurrent therapy. Combination of telaglenastat with RT reduced tumor volume in a HNSCC patient derived xenograft mouse model. The efficacy of telaglenastat with RT in reducing cell survival and increasing apoptosis was similar if not greater than that of cisplatin with RT in Cal27 and HN5 HNSCC cells. The addition of telaglenastat increased reactive oxygen species and reduced the antioxidant glutathione in both Cal27 and HN5 cells. Reverse Phase Protein Array analyses revealed alterations in cell death and DNA damage response proteins. This study provides the scientific underpinnings for the use of telaglenastat as a radiosensitizer in the treatment of HNSCC either as an alternative to cisplatin or in cisplatin-ineligible patients.
Collapse
Affiliation(s)
- Julianna Korns
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, USA; Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, USA
| | - Christina A Wicker
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Maria Lehn
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Shreya Shyamsunder
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, USA; Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, USA
| | - Samuel Thompson
- Cincinnati Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Carissa Lester
- Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, USA
| | - Trisha M Wise-Draper
- Department of Internal Medicine, Division of Hematology Oncology, University of Cincinnati, Cincinnati, OH, USA
| | - Susan E Waltz
- Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, USA; Research Service, Cincinnati Veteran's Affairs Medical Center, Cincinnati, OH, USA
| | - Vinita Takiar
- Department of Radiation Oncology, University of Cincinnati, Cincinnati, OH, USA; Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, USA; Research Service, Cincinnati Veteran's Affairs Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
16
|
Liao ZQ, Lv YF, Kang MD, Ji YL, Liu Y, Wang LR, Tang JL, Deng ZQ, Yi Y, Tang Q. Inhibition of XPR1-dependent phosphate efflux induces mitochondrial dysfunction: A potential molecular target therapy for hepatocellular carcinoma? Mol Carcinog 2024; 63:2332-2345. [PMID: 39136583 DOI: 10.1002/mc.23812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 11/16/2024]
Abstract
Xenotropic and polytropic retrovirus receptor 1 (XPR1) is the only known transporter associated with Pi efflux in mammals, and its impact on tumor progression is gradually being revealed. However, the role of XPR1 in hepatocellular carcinoma (HCC) is unknown. A bioinformatics screen for the phosphate exporter XPR1 was performed in HCC patients. The expression of XPR1 in clinical specimens was analyzed using quantitative real-time PCR, Western blot analysis, and immunohistochemical assays. Knockdown of the phosphate exporter XPR1 was performed by shRNA transfection to investigate the cellular phenotype and phosphate-related cytotoxicity of the Huh7 and HLF cell lines. In vivo tests were conducted to investigate the tumorigenicity of HCC cells xenografted into immunocompromised mice after silencing XPR1. Compared with that in paracancerous tissue, XPR1 expression in HCC tissues was markedly upregulated. High XPR1 expression significantly correlated with poor patient survival. Silencing of XPR1 leads to decreased proliferation, migration, invasion, and colony formation in HCC cells. Mechanistically, knockdown of XPR1 causes an increase in intracellular phosphate levels; mitochondrial dysfunction characterized by reduced mitochondrial membrane potential and adenosine triphosphate levels; increased reactive oxygen species levels; abnormal mitochondrial morphology; and downregulation of key mitochondrial fusion, fission, and inner membrane genes. This ultimately results in mitochondria-dependent apoptosis. These findings reveal the prognostic value of XPR1 in HCC progression and, more importantly, suggest that XPR1 might be a potential therapeutic target.
Collapse
Affiliation(s)
- Zi-Qiang Liao
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, College of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute for Advanced Study, Nanchang University, Nanchang, China
| | - Yang-Feng Lv
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, College of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute for Advanced Study, Nanchang University, Nanchang, China
| | - Mei-Diao Kang
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yu-Long Ji
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Yue Liu
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Le-Ran Wang
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang, China
| | | | - Zhi-Qiang Deng
- Department of Oncology, The First People's Hospital of Fuzhou, Fuzhou, China
| | - Yun Yi
- Biobank Center, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qun Tang
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, College of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, China
- Institute for Advanced Study, Nanchang University, Nanchang, China
| |
Collapse
|
17
|
Alhaddad L, Osipov AN, Leonov S. FLASH Radiotherapy: Benefits, Mechanisms, and Obstacles to Its Clinical Application. Int J Mol Sci 2024; 25:12506. [PMID: 39684218 DOI: 10.3390/ijms252312506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 12/18/2024] Open
Abstract
Radiotherapy (RT) has been shown to be a cornerstone of both palliative and curative tumor care. RT has generally been reported to be sharply limited by ionizing radiation (IR)-induced toxicity, thereby constraining the control effect of RT on tumor growth. FLASH-RT is the delivery of ultra-high dose rate (UHDR) several orders of magnitude higher than what is presently used in conventional RT (CONV-RT). The FLASH-RT clinical trials have been designed to examine the UHDR deliverability, the effectiveness of tumor control, the dose tolerance of normal tissue, and the reproducibility of treatment effects across several institutions. Although it is still in its infancy, FLASH-RT has been shown to have potential to rival current RT in terms of safety. Several studies have suggested that the adoption of FLASH-RT is very limited, and the incorporation of this new technique into routine clinical RT will require the use of accurate dosimetry methods and reproducible equipment that enable the reliable and robust measurements of doses and dose rates. The purpose of this review is to highlight the advantages of this technology, the potential mechanisms underpinning the FLASH-RT effect, and the major challenges that need to be tackled in the clinical transfer of FLASH-RT.
Collapse
Affiliation(s)
- Lina Alhaddad
- Department of Environmental Sciences, Faculty of Science, Damascus University, Damascus P.O. Box 30621, Syria
| | - Andreyan N Osipov
- N.N. Semenov Federal Research Center for Chemical Physics, Russian Academy of Sciences, Moscow 119991, Russia
- Moscow Center for Advanced Studies, Kulakova Str. 20, Moscow 123592, Russia
- State Research Center-Burnasyan Federal Medical Biophysical Center of Federal Medical Biological Agency (SRC-FMBC), Moscow 123098, Russia
- CANDLE Synchrotron Research Institute, 31 Acharyan, Yerevan 0040, Armenia
| | - Sergey Leonov
- Moscow Center for Advanced Studies, Kulakova Str. 20, Moscow 123592, Russia
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino 142290, Russia
| |
Collapse
|
18
|
Vaezi MA, Nekoufar S, Robati AK, Salimi V, Tavakoli-Yaraki M. Therapeutic potential of β-hydroxybutyrate in the management of pancreatic neoplasms: exploring novel diagnostic and treatment strategies. Lipids Health Dis 2024; 23:376. [PMID: 39543582 PMCID: PMC11562866 DOI: 10.1186/s12944-024-02368-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/09/2024] [Indexed: 11/17/2024] Open
Abstract
Pancreatic neoplasm, a highly aggressive and often fatal cancer, poses challenges due to late detection and nonspecific symptoms. Therefore, both early diagnosis and appropriate therapeutic approaches are necessary to augment the condition of these patients. Cancer cells undergo metabolic deregulation, which enables their proliferation, survival, and invasion. As a result, it is crucial to focus on the metabolic pathways in prevalent cancers and explore treatment strategies that target these pathways to control tumor growth effectively. This is particularly relevant in cancers like pancreatic cancer, which undergo numerous metabolic alterations. The ketogenic regimen, characterized by low carbohydrate and protein contents and high-fat sources, does not involve caloric restriction. This allows for the induction of ketogenesis and an increase in ketone bodies, while insulin and glucose levels remain low even after meals. This unique metabolic state may influence the tumor microenvironment. Given the lack of unanimous agreement on the precise role and mechanism of the ketogenic diet, this review aims to clarify the diagnostic value and accuracy of ketone bodies in various types of pancreatic tumors and explore the potential anti-cancer effects of the ketogenic diet when used alone or in conjunction with chemotherapy, also to determine the potential of the ketogenic diet to be used as adjuvant therapy. The outcomes of this study are instrumental in enhancing our understanding of the benefits and drawbacks associated with employing this diet for the management and diagnosis of pancreatic cancer.
Collapse
Affiliation(s)
- Mohammad Amin Vaezi
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran
| | - Samira Nekoufar
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran
| | - Ali Karami Robati
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran
| | - Vahid Salimi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Tavakoli-Yaraki
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, P.O. Box: 1449614535, Tehran, Iran.
- Finetech in Medicine Research Center, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
19
|
Cao D, Sun W, Li X, Jian L, Zhou X, Bode AM, Luo X. The role of novel protein acylations in cancer. Eur J Pharmacol 2024; 979:176841. [PMID: 39033839 DOI: 10.1016/j.ejphar.2024.176841] [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: 03/04/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 07/23/2024]
Abstract
Novel protein acylations are a class of protein post-translational modifications, such as lactylation, succinylation, crotonylation, palmitoylation, and β-hydroxybutyrylation. These acylation modifications are common in prokaryotes and eukaryotes and play pivotal roles in various key cellular processes by regulating gene transcription, protein subcellular localization, stability and activity, protein-protein interactions, and protein-DNA interactions. The diversified acylations are closely associated with various human diseases, especially cancer. In this review, we provide an overview of the distinctive characteristics, effects, and regulatory factors of novel protein acylations. We also explore the various mechanisms through which novel protein acylations are involved in the occurrence and progression of cancer. Furthermore, we discuss the development of anti-cancer drugs targeting novel acylations, offering promising avenues for cancer treatment.
Collapse
Affiliation(s)
- Dan Cao
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Wenxuan Sun
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Xinyi Li
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Lian Jian
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Xinran Zhou
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China
| | - Ann M Bode
- The Hormel Institute, University of Minnesota, Austin, MN, 55912, USA
| | - Xiangjian Luo
- Key Laboratory of Carcinogenesis and Invasion, Chinese Ministry of Education, Department of Nuclear Medicine, Xiangya Hospital, Central South University, Changsha, Hunan, 410078, China; Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, Hunan, 410078, China; Hunan Key Laboratory of Oncotarget Gene, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, China; Molecular Imaging Research Center of Central South University, Changsha, Hunan, 410078, China; Key Laboratory of Biological Nanotechnology of National Health Commission, Central South University, Changsha, Hunan, 410078, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410078, China.
| |
Collapse
|
20
|
Liu HF, Chou SC, Huang SC, Huang TY, Hsiao PY, Chou FP, Wu TK. Dehydroepiandrosterone-α-2-Deoxyglucoside Exhibits Enhanced Anticancer Effects in MCF-7 Breast Cancer Cells and Inhibits Glucose-6-Phosphate Dehydrogenase Activity. Chem Biol Drug Des 2024; 104:e14624. [PMID: 39317696 DOI: 10.1111/cbdd.14624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 08/05/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024]
Abstract
In the pentose phosphate pathway, dehydroepiandrosterone (DHEA) uncompetitively inhibits glucose-6-phosphate dehydrogenase (G6PD), reducing NADPH production and increasing oxidative stress, which can influence the onset and/or progression of several diseases, including cancer. 2-Deoxy-D-glucose (2-DG), a glucose mimetic, competes with glucose for cellular uptake, inhibiting glycolysis and competing with glucose-6-phosphate (G-6-P) for G6PD activity. In this study, we report that DHEA-α-2-DG (5), an α-covalent conjugate of DHEA and 2-DG, exhibits better anticancer activity than DHEA, 2-DG, DHEA +2-DG, and polydatin in MCF-7 cells, and reduces NADPH/NADP+ ratio in cellular assays. In vitro enzyme kinetics and molecular docking studies showed that 5 uncompetitively inhibits human G6PD activity and binds to the structural NADP+ site but not to the catalytic NADP+ site. Further combining 5 with the FDA-approved drug tamoxifen enhanced its cytotoxicity against MCF-7 cells, suggesting that it could serve as a candidate for combination of drug strategies.
Collapse
Affiliation(s)
- Hsu-Feng Liu
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsin-Chu, Taiwan
| | - Shen-Chieh Chou
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsin-Chu, Taiwan
| | - Sheng-Cih Huang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsin-Chu, Taiwan
| | - Tzu-Yu Huang
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsin-Chu, Taiwan
| | - Po-Yun Hsiao
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsin-Chu, Taiwan
| | - Feng-Pai Chou
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsin-Chu, Taiwan
| | - Tung-Kung Wu
- Department of Biological Science and Technology, National Yang Ming Chiao Tung University, Hsin-Chu, Taiwan
- Center for Emergent Functional Matter Science, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| |
Collapse
|
21
|
Glorieux C, Liu S, Trachootham D, Huang P. Targeting ROS in cancer: rationale and strategies. Nat Rev Drug Discov 2024; 23:583-606. [PMID: 38982305 DOI: 10.1038/s41573-024-00979-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2024] [Indexed: 07/11/2024]
Abstract
Reactive oxygen species (ROS) in biological systems are transient but essential molecules that are generated and eliminated by a complex set of delicately balanced molecular machineries. Disruption of redox homeostasis has been associated with various human diseases, especially cancer, in which increased ROS levels are thought to have a major role in tumour development and progression. As such, modulation of cellular redox status by targeting ROS and their regulatory machineries is considered a promising therapeutic strategy for cancer treatment. Recently, there has been major progress in this field, including the discovery of novel redox signalling pathways that affect the metabolism of tumour cells as well as immune cells in the tumour microenvironment, and the intriguing ROS regulation of biomolecular phase separation. Progress has also been made in exploring redox regulation in cancer stem cells, the role of ROS in determining cell fate and new anticancer agents that target ROS. This Review discusses these research developments and their implications for cancer therapy and drug discovery, as well as emerging concepts, paradoxes and future perspectives.
Collapse
Affiliation(s)
- Christophe Glorieux
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Shihua Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | | | - Peng Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China.
- Metabolic Innovation Center, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
22
|
Hata M, Kadoya Y, Ueno J, Taki M, Kodera M. Dicopper Complexes of p-Cresol-2,6-bis(amide-tether-dpa 4-X) (X = MeO and Cl): Selective ROS Generation and Cytotoxicity Enhancement Controlled by Electronic and Hydrophobic Effects of the MeO and Cl Groups. Inorg Chem 2024; 63:13893-13902. [PMID: 39011904 DOI: 10.1021/acs.inorgchem.4c01072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/17/2024]
Abstract
Two new p-cresol-2,6-bis(amide-tether-dpa4-X) ligands (HL4-X, X = MeO and Cl) and their dicopper complexes [Cu2(μ-1,1-OAc)(μ-1,3-OAc)(L4-MeO)]Y (Y = PF6 1a, OAc 1b) and [Cu2(μ-1,3-OAc)2(L4-Cl)]Y (Y = ClO4 2a, OAc 2b) were synthesized. The electronic and hydrophobic effects of the MeO and Cl groups were examined compared with nonsubstituted complex [Cu2(μ-1,1-OAc)(μ-1,3-OAc)(L)]+ (3). The electronic effects were found in crystal structures, spectroscopic characterization, and redox potentials of these complexes. 1b and 2b were reduced to Cu(I)Cu(I) with sodium ascorbate and reductively activated O2 to produce H2O2 and HO•. The H2O2 release and HO• generation are promoted by the electronic effects. The hydrophobic effects increased the lipophilicity of 1b and 2b. Cellular ROS generation of 1b, 2b, and 3 was visualized by DCFH-DA. To examine the intracellular behavior, boron dipyrromethene (Bodipy)-modified complexes 4B and 5B corresponding to 1b and 2b were synthesized. These support that 1b and 2b are localized at the ER and Golgi apparatus. The cytotoxicity of 1b and 2b against various cell lines was examined by MTT assay. 1b and 2b were 7- and 41-fold more cytotoxic than 3. 1b generated ROS selectively in cancer cell but 2b nonselectively in cancer and normal cells, causing cancer- and normal-cell-selective cytotoxicity, respectively.
Collapse
Affiliation(s)
- Machi Hata
- Department of Molecular Chemistry and Biochemistry, Doshisha University, Kyotanabe Kyoto 610-0321, Japan
| | - Yuki Kadoya
- Department of Molecular Chemistry and Biochemistry, Doshisha University, Kyotanabe Kyoto 610-0321, Japan
| | - Jin Ueno
- Department of Molecular Chemistry and Biochemistry, Doshisha University, Kyotanabe Kyoto 610-0321, Japan
| | - Masayasu Taki
- Institute of Transformative Bio-Molecules (WPI-ITbM), Nagoya University, Furo, Chikusa, Nagoya 464-8601, Japan
| | - Masahito Kodera
- Department of Molecular Chemistry and Biochemistry, Doshisha University, Kyotanabe Kyoto 610-0321, Japan
| |
Collapse
|
23
|
Liu D, Fang L. Oxidative stress-related genes score predicts prognosis and immune cell infiltration landscape characterization in breast cancer. Heliyon 2024; 10:e34046. [PMID: 39071696 PMCID: PMC11283013 DOI: 10.1016/j.heliyon.2024.e34046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/25/2024] [Accepted: 07/02/2024] [Indexed: 07/30/2024] Open
Abstract
Background The tumor microenvironment (TME) typically experiences oxidative stress (OS), marked by a high level of reactive oxygen species (ROS) that can impact tumor advancement and prognosis by modulating the behavior of tumor cells and various immune cells. Oxidative stress-related genes (OSRG) encompass a range of genes involved in ROS pathways, and their specific roles in breast cancer (BC) necessitate further investigation. Methods Univariate Cox analysis was performed on genes linked to the OS pathway in the Gene Set Enrichment Analysis (GSEA) database, leading to the identification of 29 significant OSRG in BC. OSRG was divided into three distinct clusters according to the expression and the OSRG score based on the differentially expressed genes (DEGs) was further calculated by principal component analysis (PCA). The correlation between OSRG score and BC clinical features, mutation characteristics, immune checkpoints and immune cell infiltration was analyzed. Establish a multiariable Cox regression model to predict OSRG score effects on clinical characteristics. Results Significant differences were observed in survival analysis, enriched pathways, and immune infiltration among the three OSRG clusters based on 29 genes. Gene clusters were identified through the final selected 395 DEGs, revealing three distinct OSRG expression patterns. An OSRG score model was constructed using DEGs, demonstrating a significant association between high OSRG score and poor prognosis. Significantly, immune checkpoint-related genes exhibited a notable upregulation in the high OSRG score cohort. Additionally, a positive correlation was observed between the OSRG score and tumor mutation burden (TMB) in BC. The OSRG score holds potential implications for clinical immunotherapy in BC patients, and a nomogram was constructed with robust predictive capability for evaluating patient prognosis. Conclusions This study elucidated the features of OSRG within BC TME and their possible prognostic significance, offering valuable insights for the development of more targeted immunotherapy approaches for individuals with BC.
Collapse
Affiliation(s)
- Diya Liu
- Department of Thyroid and Breast Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| | - Lin Fang
- Department of Thyroid and Breast Surgery, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China
| |
Collapse
|
24
|
Vo TTT, Peng TY, Nguyen TH, Bui TNH, Wang CS, Lee WJ, Chen YL, Wu YC, Lee IT. The crosstalk between copper-induced oxidative stress and cuproptosis: a novel potential anticancer paradigm. Cell Commun Signal 2024; 22:353. [PMID: 38970072 PMCID: PMC11225285 DOI: 10.1186/s12964-024-01726-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 06/25/2024] [Indexed: 07/07/2024] Open
Abstract
Copper is a crucial trace element that plays a role in various pathophysiological processes in the human body. Copper also acts as a transition metal involved in redox reactions, contributing to the generation of reactive oxygen species (ROS). Under prolonged and increased ROS levels, oxidative stress occurs, which has been implicated in different types of regulated cell death. The recent discovery of cuproptosis, a copper-dependent regulated cell death pathway that is distinct from other known regulated cell death forms, has raised interest to researchers in the field of cancer therapy. Herein, the present work aims to outline the current understanding of cuproptosis, with an emphasis on its anticancer activities through the interplay with copper-induced oxidative stress, thereby providing new ideas for therapeutic approaches targeting modes of cell death in the future.
Collapse
Affiliation(s)
- Thi Thuy Tien Vo
- Faculty of Dentistry, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam
| | - Tzu-Yu Peng
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Thi Hong Nguyen
- Faculty of Dentistry, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam
| | - Trang Ngoc Huyen Bui
- Faculty of Dentistry, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam
| | - Ching-Shuen Wang
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - Wei-Ju Lee
- School of Food Safety, College of Nutrition, Taipei Medical University, Taipei, 110301, Taiwan
| | - Yuh-Lien Chen
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei, 100233, Taiwan
| | - Yang-Che Wu
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan
| | - I-Ta Lee
- School of Dentistry, College of Oral Medicine, Taipei Medical University, Taipei, 110301, Taiwan.
| |
Collapse
|
25
|
Tada T, Mizuno Y, Shibata Y, Yasui H, Kuge Y. Application of copper (I) selective ligands for PET imaging of reactive oxygen species through metabolic trapping. Nucl Med Biol 2024; 134-135:108914. [PMID: 38733873 DOI: 10.1016/j.nucmedbio.2024.108914] [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: 10/17/2023] [Revised: 03/29/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024]
Abstract
INTRODUCTION Reactive oxygen species (ROS) are attractive targets for clinical PET imaging. In this study, we hypothesized that PET imaging of ROS would be possible by using chelating ligands (L) that form stable complexes with copper (I) but not with copper (II), based on metabolic trapping. Namely, when [64Cu][CuI(L)2]+ is oxidized by ROS, the oxidized complex will release [64Cu]Cu2+. Then, the released [64Cu]Cu2+ will be trapped inside the cell, resulting in PET signal depending on the redox potential of ROS. To examine the potential of this novel molecular design for ROS imaging, we synthesized copper (I) complexes with bicinchoninic acid (BCA) disodium salt and bathocuproinedisulfonic acid (BCS) disodium salt and evaluated their reactivity with several kinds of ROS. In addition, the cellular uptake of [64Cu][CuI(BCS)2]3- and the stability of [64Cu][CuI(BCS)2]3- in a biological condition were also evaluated. METHODS [64Cu]Cu2+ was reduced to [64Cu]Cu+ by ascorbic acid and coordinated with BCA and BCS in the acetate buffer to synthesize [64Cu][CuI(BCA)2]3- and [64Cu][CuI(BCS)2]3-. The radiochemical yields were determined by thin-layer chromatography (TLC). After [64Cu][CuI(BCS)2]3- was incubated with hydroxyl radical, lipid peroxide, superoxide, and hydrogen peroxide, the percentage of released [64Cu]Cu2+ from the parent complex was evaluated by TLC. HT-1080 human fibrosarcoma cells were treated with 0.1 % Dimethyl sulfoxide (control), imidazole ketone erastin (IKE), or IKE + ferrostatin-1 (Fer-1). Then, the uptake of [64Cu][CuI(BCS)2]3- to HT-1080 cells in each group was evaluated as %Dose/mg protein. Lastly, [64Cu][CuI(BCS)2]3- was incubated in human plasma, and its intact ratio was determined by TLC. RESULTS The radiochemical yield of [64Cu][CuI(BCS)2]3- (86 ± 1 %) was higher than that of [64Cu][CuI(BCA)2]3- (44 ± 3 %). [64Cu][CuI(BCA)2]3- was unstable and partially decomposed on TLC. After [64Cu][CuI(BCS)2]3- was reacted with hydroxyl radical, lipid peroxide, and superoxide, 67 ± 2 %, 44 ± 13 %, and 22 ± 3 % of total radioactivity was detected as [64Cu]Cu2+, respectively. On the other hand, the reaction with hydrogen peroxide did not significantly increase the ratio of [64Cu]Cu2+ (4 ± 1 %). These results suggest that [64Cu][CuI(BCS)2]3- could be used for detecting high-redox-potential ROS such as hydroxyl radical and lipid peroxide with high selectivity. The cellular uptake values of [64Cu][CuI(BCS)2]3- in the control, IKE, and Fer-1 group were 42 ± 2, 54 ± 2, and 47 ± 5 %Dose/mg protein (n = 3), respectively, suggesting the ROS specific uptake of [64Cu][CuI(BCS)2]3-. On the other hand, the intact ratio after the incubation of [64Cu][CuI(BCS)2]3- in human plasma was 9 ± 5 %. CONCLUSION PET imaging of ROS would be possible by using a copper (I) selective ligand, based on metabolic trapping. Although improvement of the membrane permeability and the stability of copper (I) complexes is required, the present results pave the way for the development of novel 64Cu-labeled complexes for PET imaging of ROS.
Collapse
Affiliation(s)
- Tetsuro Tada
- Graduate School of Biomedical Science and Engineering, Hokkaido University, Hokkaido 060-0815, Japan
| | - Yuki Mizuno
- Central Institute of Isotope Science, Hokkaido University, Hokkaido 060-0815, Japan; Global Center for Biomedical Science and Engineering, Hokkaido University, Hokkaido 060-0815, Japan.
| | - Yuki Shibata
- Central Institute of Isotope Science, Hokkaido University, Hokkaido 060-0815, Japan
| | - Hironobu Yasui
- Faculty of Veterinary Medicine, Hokkaido University, Hokkaido 060-0818, Japan; One Health Research Center, Hokkaido University, Hokkaido 060-0818, Japan
| | - Yuji Kuge
- Central Institute of Isotope Science, Hokkaido University, Hokkaido 060-0815, Japan; Global Center for Biomedical Science and Engineering, Hokkaido University, Hokkaido 060-0815, Japan
| |
Collapse
|
26
|
Wang Y. The interplay of exercise and polyphenols in cancer treatment: A focus on oxidative stress and antioxidant mechanisms. Phytother Res 2024; 38:3459-3488. [PMID: 38690720 DOI: 10.1002/ptr.8215] [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: 02/01/2024] [Revised: 04/12/2024] [Accepted: 04/14/2024] [Indexed: 05/02/2024]
Abstract
Exercise has been demonstrated to induce an elevated production of free radicals, leading to the onset of oxidative stress. Numerous studies highlight the positive impacts of aerobic exercise, primarily attributed to the increase in overall antioxidant capacity. The evidence suggests that engaging in aerobic exercise contributes to a reduction in the likelihood of advanced cancer and mortality. Oxidative stress occurs when there is an imbalance between the generation of free radicals and the collective antioxidant defense system, encompassing both enzymatic and nonenzymatic antioxidants. Typically, oxidative stress triggers the formation of reactive oxygen or nitrogen species, instigating or advancing various issues in cancers and other diseases. The pro-oxidant-antioxidant balance serves as a direct measure of this imbalance in oxidative stress. Polyphenols contain a variety of bioactive compounds, including flavonoids, flavanols, and phenolic acids, conferring antioxidant properties. Previous research highlights the potential of polyphenols as antioxidants, with documented effects on reducing cancer risk by influencing processes such as proliferation, angiogenesis, and metastasis. This is primarily attributed to their recognized antioxidant capabilities. Considering the extensive array of signaling pathways associated with exercise and polyphenols, this overview will specifically focus on oxidative stress, the antioxidant efficacy of polyphenols and exercise, and their intricate interplay in cancer treatment.
Collapse
Affiliation(s)
- Yubing Wang
- College of Physical Education, Qilu Normal University, Jinan, Shandong, China
| |
Collapse
|
27
|
Mukherjee A, Ghosh KK, Chakrabortty S, Gulyás B, Padmanabhan P, Ball WB. Mitochondrial Reactive Oxygen Species in Infection and Immunity. Biomolecules 2024; 14:670. [PMID: 38927073 PMCID: PMC11202257 DOI: 10.3390/biom14060670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024] Open
Abstract
Reactive oxygen species (ROS) contain at least one oxygen atom and one or more unpaired electrons and include singlet oxygen, superoxide anion radical, hydroxyl radical, hydroperoxyl radical, and free nitrogen radicals. Intracellular ROS can be formed as a consequence of several factors, including ultra-violet (UV) radiation, electron leakage during aerobic respiration, inflammatory responses mediated by macrophages, and other external stimuli or stress. The enhanced production of ROS is termed oxidative stress and this leads to cellular damage, such as protein carbonylation, lipid peroxidation, deoxyribonucleic acid (DNA) damage, and base modifications. This damage may manifest in various pathological states, including ageing, cancer, neurological diseases, and metabolic disorders like diabetes. On the other hand, the optimum levels of ROS have been implicated in the regulation of many important physiological processes. For example, the ROS generated in the mitochondria (mitochondrial ROS or mt-ROS), as a byproduct of the electron transport chain (ETC), participate in a plethora of physiological functions, which include ageing, cell growth, cell proliferation, and immune response and regulation. In this current review, we will focus on the mechanisms by which mt-ROS regulate different pathways of host immune responses in the context of infection by bacteria, protozoan parasites, viruses, and fungi. We will also discuss how these pathogens, in turn, modulate mt-ROS to evade host immunity. We will conclude by briefly giving an overview of the potential therapeutic approaches involving mt-ROS in infectious diseases.
Collapse
Affiliation(s)
- Arunima Mukherjee
- Department of Biological Sciences, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur 522502, Andhra Pradesh, India;
| | - Krishna Kanta Ghosh
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (B.G.)
| | - Sabyasachi Chakrabortty
- Department of Chemistry, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur 522502, Andhra Pradesh, India;
| | - Balázs Gulyás
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (B.G.)
- Cognitive Neuroimaging Centre, 59 Nanyang Drive, Nanyang Technological University, Singapore 636921, Singapore
- Department of Clinical Neuroscience, Karolinska Institute, 17176 Stockholm, Sweden
| | - Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, 59 Nanyang Drive, Singapore 636921, Singapore; (K.K.G.); (B.G.)
- Cognitive Neuroimaging Centre, 59 Nanyang Drive, Nanyang Technological University, Singapore 636921, Singapore
| | - Writoban Basu Ball
- Department of Biological Sciences, School of Engineering and Sciences, SRM University AP Andhra Pradesh, Guntur 522502, Andhra Pradesh, India;
| |
Collapse
|
28
|
Littleflower AB, Parambil ST, Antony GR, Subhadradevi L. The determinants of metabolic discrepancies in aerobic glycolysis: Providing potential targets for breast cancer treatment. Biochimie 2024; 220:107-121. [PMID: 38184121 DOI: 10.1016/j.biochi.2024.01.003] [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: 08/10/2023] [Revised: 12/22/2023] [Accepted: 01/03/2024] [Indexed: 01/08/2024]
Abstract
Altered aerobic glycolysis is the robust mechanism to support cancer cell survival and proliferation beyond the maintenance of cellular energy metabolism. Several investigators portrayed the important role of deregulated glycolysis in different cancers, including breast cancer. Breast cancer is the most ubiquitous form of cancer and the primary cause of cancer death in women worldwide. Breast cancer with increased glycolytic flux is hampered to eradicate with current therapies and can result in tumor recurrence. In spite of the low order efficiency of ATP production, cancer cells are highly addicted to glycolysis. The glycolytic dependency of cancer cells provides potential therapeutic strategies to preferentially kill cancer cells by inhibiting glycolysis using antiglycolytic agents. The present review emphasizes the most recent research on the implication of glycolytic enzymes, including glucose transporters (GLUTs), hexokinase (HK), phosphofructokinase (PFK), pyruvate kinase (PK), lactate dehydrogenase-A (LDHA), associated signalling pathways and transcription factors, as well as the antiglycolytic agents that target key glycolytic enzymes in breast cancer. The potential activity of glycolytic inhibitors impinges cancer prevalence and cellular resistance to conventional drugs even under worse physiological conditions such as hypoxia. As a single agent or in combination with other chemotherapeutic drugs, it provides the feasibility of new therapeutic modalities against a wide spectrum of human cancers.
Collapse
Affiliation(s)
- Ajeesh Babu Littleflower
- Division of Cancer Research, Regional Cancer Centre (Research Centre, University of Kerala), Thiruvananthapuram, Kerala, 695011, India
| | - Sulfath Thottungal Parambil
- Division of Cancer Research, Regional Cancer Centre (Research Centre, University of Kerala), Thiruvananthapuram, Kerala, 695011, India
| | - Gisha Rose Antony
- Division of Cancer Research, Regional Cancer Centre (Research Centre, University of Kerala), Thiruvananthapuram, Kerala, 695011, India
| | - Lakshmi Subhadradevi
- Division of Cancer Research, Regional Cancer Centre (Research Centre, University of Kerala), Thiruvananthapuram, Kerala, 695011, India.
| |
Collapse
|
29
|
Chen M, Tan J, Jin Z, Jiang T, Wu J, Yu X. Research progress on Sirtuins (SIRTs) family modulators. Biomed Pharmacother 2024; 174:116481. [PMID: 38522239 DOI: 10.1016/j.biopha.2024.116481] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 03/15/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024] Open
Abstract
Sirtuins (SIRTs) represent a class of nicotinamide adenine dinucleotide (NAD+)-dependent protein deacetylases that exert a crucial role in cellular signal transduction and various biological processes. The mammalian sirtuins family encompasses SIRT1 to SIRT7, exhibiting therapeutic potential in counteracting cellular aging, modulating metabolism, responding to oxidative stress, inhibiting tumors, and improving cellular microenvironment. These enzymes are intricately linked to the occurrence and treatment of diverse pathological conditions, including cancer, autoimmune diseases, and cardiovascular disorders. Given the significance of histone modification in gene expression and chromatin structure, maintaining the equilibrium of the sirtuins family is imperative for disease prevention and health restoration. Mounting evidence suggests that modulators of SIRTs play a crucial role in treating various diseases and maintaining physiological balance. This review delves into the molecular structure and regulatory functions of the sirtuins family, reviews the classification and historical evolution of SIRTs modulators, offers a systematic overview of existing SIRTs modulation strategies, and elucidates the regulatory mechanisms of SIRTs modulators (agonists and inhibitors) and their clinical applications. The article concludes by summarizing the challenges encountered in SIRTs modulator research and offering insights into future research directions.
Collapse
Affiliation(s)
- Mingkai Chen
- Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China; School of Medicine Jiangsu University, Zhenjiang, Jiangsu, China
| | - Junfei Tan
- School of Medicine Jiangsu University, Zhenjiang, Jiangsu, China
| | - Zihan Jin
- Changzhou Second People's Hospital Affiliated to Nanjing Medical University, Changzhou City, China
| | - Tingting Jiang
- Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
| | - Jiabiao Wu
- Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China
| | - Xiaolong Yu
- Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu, China; The Wujin Clinical College of Xuzhou Medical University, Changzhou, Jiangsu, China.
| |
Collapse
|
30
|
Palma FR, Coelho DR, Pulakanti K, Sakiyama MJ, Huang Y, Ogata FT, Danes JM, Meyer A, Furdui CM, Spitz DR, Gomes AP, Gantner BN, Rao S, Backman V, Bonini MG. Histone H3.1 is a chromatin-embedded redox sensor triggered by tumor cells developing adaptive phenotypic plasticity and multidrug resistance. Cell Rep 2024; 43:113897. [PMID: 38493478 PMCID: PMC11209755 DOI: 10.1016/j.celrep.2024.113897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 01/08/2024] [Accepted: 02/16/2024] [Indexed: 03/19/2024] Open
Abstract
Chromatin structure is regulated through posttranslational modifications of histone variants that modulate transcription. Although highly homologous, histone variants display unique amino acid sequences associated with specific functions. Abnormal incorporation of histone variants contributes to cancer initiation, therapy resistance, and metastasis. This study reports that, among its biologic functions, histone H3.1 serves as a chromatin redox sensor that is engaged by mitochondrial H2O2. In breast cancer cells, the oxidation of H3.1Cys96 promotes its eviction and replacement by H3.3 in specific promoters. We also report that this process facilitates the opening of silenced chromatin domains and transcriptional activation of epithelial-to-mesenchymal genes associated with cell plasticity. Scavenging nuclear H2O2 or amino acid substitution of H3.1(C96S) suppresses plasticity, restores sensitivity to chemotherapy, and induces remission of metastatic lesions. Hence, it appears that increased levels of H2O2 produced by mitochondria of breast cancer cells directly promote redox-regulated H3.1-dependent chromatin remodeling involved in chemoresistance and metastasis.
Collapse
Affiliation(s)
- Flavio R Palma
- Department of Medicine, Division of Hematology Oncology, Northwestern University Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center of Chicago, Chicago, IL 60611, USA
| | - Diego R Coelho
- Department of Medicine, Division of Hematology Oncology, Northwestern University Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center of Chicago, Chicago, IL 60611, USA
| | - Kirthi Pulakanti
- Versiti Blood Research Institute of Wisconsin, and Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Marcelo J Sakiyama
- Department of Medicine, Division of Hematology Oncology, Northwestern University Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center of Chicago, Chicago, IL 60611, USA
| | - Yunping Huang
- Department of Medicine, Division of Hematology Oncology, Northwestern University Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center of Chicago, Chicago, IL 60611, USA
| | - Fernando T Ogata
- Department of Medicine, Division of Hematology Oncology, Northwestern University Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center of Chicago, Chicago, IL 60611, USA
| | - Jeanne M Danes
- Department of Medicine, Division of Hematology Oncology, Northwestern University Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center of Chicago, Chicago, IL 60611, USA
| | - Alison Meyer
- Versiti Blood Research Institute of Wisconsin, and Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Cristina M Furdui
- Department of Internal Medicine, Section on Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52245, USA
| | - Ana P Gomes
- Molecular Oncology Program, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Benjamin N Gantner
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Sridhar Rao
- Versiti Blood Research Institute of Wisconsin, and Department of Cell Biology, Neurobiology, and Anatomy, Medical College of Wisconsin, Milwaukee, WI 53226, USA; Department of Medicine, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Vadim Backman
- Department of Biomedical Engineering, Northwestern University McCormick School of Engineering, Evanston, IL 60208, USA
| | - Marcelo G Bonini
- Department of Medicine, Division of Hematology Oncology, Northwestern University Feinberg School of Medicine and the Robert H. Lurie Comprehensive Cancer Center of Chicago, Chicago, IL 60611, USA; Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
31
|
Zmorzynski S, Popek-Marciniec S, Biernacka B, Szudy-Szczyrek A, Chocholska S, Styk W, Czerwik-Marcinkowska J, Swiderska-Kolacz G. In Vitro Low-Bortezomib Doses Induce Apoptosis and Independently Decrease the Activities of Glutathione S-Transferase and Glutathione Peroxidase in Multiple Myeloma, Taking into Account the GSTT1 and GSTM1 Gene Variants. Genes (Basel) 2024; 15:387. [PMID: 38540446 PMCID: PMC10970692 DOI: 10.3390/genes15030387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 03/13/2024] [Accepted: 03/19/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND Multiple myeloma (MM) is a malignancy derived from plasma cells. Bortezomib affects the concentration of reduced glutathione (GSH) and the activity of glutathione enzymes. The aim of our study was to analyze deletion (null/present) variants of GSTT1 and GSTM1 genes and their association with the levels of glutathione and its enzymes in bortezomib-treated cell cultures derived from MM patients. MATERIALS AND METHODS This study included 180 individuals (80 MM patients and 100 healthy blood donors) who were genotyped via multiplex PCR (for the GSTT1/GSTM1 genes). Under in vitro conditions, MM bone marrow cells were treated with bortezomib (1-4 nM) to determine apoptosis (via fluorescence microscopy), GSH concentration, and activity of glutathione enzymes (via ELISA). RESULTS Bortezomib increased the number of apoptotic cells and decreased the activity of S-glutathione transferase (GST) and glutathione peroxidase (GPx). We found significant differences in GST activity between 1 nM (GSTT1-null vs. GSTT1-present), 2 nM (GSTT1-null vs. GSTT1-present), and 4 nM (GSTM1-null vs. GSTM1-present) bortezomib: 0.07 vs. 0.12, p = 0.02; 0.06 vs. 0.10, p = 0.02; and 0.03 vs. 0.08, p = 0.01, respectively. CONCLUSIONS Bortezomib affects the activities of GST and GPx. GST activity was associated with GSTT1 and GSTM1 variants but only at some bortezomib doses.
Collapse
Affiliation(s)
| | | | - Beata Biernacka
- Institute of Nursing and Obstetrics, Academy of Zamosc, 22-400 Zamosc, Poland
| | - Aneta Szudy-Szczyrek
- Chair and Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-059 Lublin, Poland; (A.S.-S.); (S.C.)
| | - Sylwia Chocholska
- Chair and Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-059 Lublin, Poland; (A.S.-S.); (S.C.)
| | - Wojciech Styk
- Academic Laboratory of Psychological Tests, Medical University of Lublin, 20-059 Lublin, Poland;
| | | | | |
Collapse
|
32
|
Takasaki T, Hamabe Y, Touchi K, Khandakar GI, Ueda T, Okada H, Sakai K, Nishio K, Tanabe G, Sugiura R. ACA-28, an ERK MAPK Signaling Modulator, Exerts Anticancer Activity through ROS Induction in Melanoma and Pancreatic Cancer Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2024; 2024:7683793. [PMID: 38500550 PMCID: PMC10948229 DOI: 10.1155/2024/7683793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 02/19/2024] [Accepted: 02/27/2024] [Indexed: 03/20/2024]
Abstract
The extracellular signal-regulated kinase (ERK) MAPK pathway is dysregulated in various human cancers and is considered an attractive therapeutic target for cancer. Therefore, several inhibitors of this pathway are being developed, and some are already used in the clinic. We have previously identified an anticancer compound, ACA-28, with a unique property to preferentially induce ERK-dependent apoptosis in melanoma cells. To comprehensively understand the biological cellular impact induced by ACA-28, we performed a global gene expression analysis of human melanoma SK-MEL-28 cells exposed to ACA-28 using a DNA microarray. The transcriptome analysis identified nuclear factor erythroid 2-related factor 2 (Nrf2), a master transcription factor that combats oxidative stress, as the most upregulated genetic pathway after ACA-28 treatment. Consistently, ACA-28 showed properties to increase the levels of reactive oxygen species (ROS) as well as Nrf2 protein, which is normally repressed by proteasomal degradation and activated in response to oxidative stresses. Furthermore, the ROS scavenger N-acetyl cysteine significantly attenuated the anticancer activity of ACA-28. Thus, ACA-28 activates Nrf2 signaling and exerts anticancer activity partly via its ROS-stimulating property. Interestingly, human A549 cancer cells with constitutively high levels of Nrf2 protein showed resistance to ACA-28, as compared with SK-MEL-28. Transient overexpression of Nrf2 also increased the resistance of cells to ACA-28, while knockdown of Nrf2 exerted the opposite effect. Thus, upregulation of Nrf2 signaling protects cancer cells from ACA-28-mediated cell death. Notably, the Nrf2 inhibitor ML385 substantially enhanced the cell death-inducing property of ACA-28 in pancreatic cancer cells, T3M4 and PANC-1. Our data suggest that Nrf2 plays a key role in determining cancer cell susceptibility to ACA-28 and provides a novel strategy for cancer therapy to combine the Nrf2 inhibitor and ACA-28.
Collapse
Affiliation(s)
- Teruaki Takasaki
- Laboratory of Molecular Pharmacogenomics, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka 577-8502, Japan
| | - Yasuyuki Hamabe
- Laboratory of Molecular Pharmacogenomics, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka 577-8502, Japan
| | - Kenta Touchi
- Laboratory of Molecular Pharmacogenomics, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka 577-8502, Japan
| | - Golam Iftakhar Khandakar
- Laboratory of Molecular Pharmacogenomics, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka 577-8502, Japan
| | - Takeshi Ueda
- Department of Biochemistry, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan
- Anti-Aging Center, Kindai University, Osaka 577-8502, Japan
| | - Hitoshi Okada
- Department of Biochemistry, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan
- Anti-Aging Center, Kindai University, Osaka 577-8502, Japan
| | - Kazuko Sakai
- Department of Genome Biology, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Faculty of Medicine, Kindai University, Osaka 589-8511, Japan
| | - Genzoh Tanabe
- Laboratory of Organic Chemistry, Department of Pharmacy, Faculty of Pharmacy, Kindai University, Osaka 577-8502, Japan
| | - Reiko Sugiura
- Laboratory of Molecular Pharmacogenomics, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Kindai University, Osaka 577-8502, Japan
- Anti-Aging Center, Kindai University, Osaka 577-8502, Japan
| |
Collapse
|
33
|
Selvam R, Pearl WG, Perevedentseva E, Karmenyan A, Cheng CL. Fe-doped nanodiamond-based photo-Fenton catalyst for dual-modal fluorescence imaging and improved chemotherapeutic efficacy against tumor hypoxia. RSC Adv 2024; 14:4285-4300. [PMID: 38298935 PMCID: PMC10828792 DOI: 10.1039/d3ra08465e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 01/17/2024] [Indexed: 02/02/2024] Open
Abstract
The deficiency of oxygen in most solid tumors plays a profound role in their proliferation, metastasis, and invasion and contributes to their resistance to treatments such as radiation, chemotherapy, and photodynamic therapy (PDT). A therapeutic approach based on the Fenton reaction has received considerable interest as a means of treating cancer with ROS-based nano catalytic medicine, referred to as chemodynamic therapy (CDT). A range of modified treatment strategies are being explored to enhance both CDT and conventional methods of therapy. These include Fenton-like reactions, photo-enhanced Fenton reactions, and Fenton catalytic-enhanced synergistic therapies. In this article, we propose and demonstrate a photochemotherapy (PCT) strategy for cancer treatment utilizing near-infrared (NIR)-induced Fenton reactions using Fe-doped nanodiamond (FeND). When FeND is exposed to human lung cancer cells A549, it exhibits outstanding biocompatibility. However, when particle-treated cells are exposed to NIR laser radiation, the particle exhibits cytotoxicity to a certain degree. The anticancer medication doxorubicin (DOX) was adsorbed onto the FeND to address this issue. The conjugated DOX could undergo a redox cycle to generate excess H2O2 inside the cells, and in addition, DOX can also cause tumor cell apoptosis. Combining chemotherapy (via DOX) with a Fenton reaction results in enhanced therapeutic effectiveness. Moreover, the intrinsic fluorescence of the nanodiamond in FeND can be used to monitor the interaction of particles with cells as well as their localization, thus making it an excellent imaging probe. In our study, we found that FeND could serve as a CDT agent, biomarker, drug carrier, and potentially valuable candidate for CDT agents and contribute to the further development of more effective CDT platforms using nanodiamond.
Collapse
Affiliation(s)
- Rajakar Selvam
- Department of Physics, National Dong Hwa University Taiwan
| | | | - Elena Perevedentseva
- Department of Physics, National Dong Hwa University Taiwan
- P. N. Lebedev Institute of Physics, Russian Academy of Science Moscow Russia
| | | | | |
Collapse
|
34
|
Tian S, Wang R, Wang Y, Chen R, Lin T, Xiao X, Liu X, Ideozu JE, Geng H, Wang Y, Yue D. p32 regulates glycometabolism and TCA cycle to inhibit ccRCC progression via copper-induced DLAT lipoylation oligomerization. Int J Biol Sci 2024; 20:516-536. [PMID: 38169635 PMCID: PMC10758103 DOI: 10.7150/ijbs.84399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 11/17/2023] [Indexed: 01/05/2024] Open
Abstract
A key player in mitochondrial respiration, p32, often referred to as C1QBP, is mostly found in the mitochondrial matrix. Previously, we showed that p32 interacts with DLAT in the mitochondria. Here, we found that p32 expression was reduced in ccRCC and suppressed progression and metastasis in ccRCC animal models. We observed that increasing p32 expression led to an increase in oxidative phosphorylation by interacting with DLAT, thus, regulating the activation of the pyruvate dehydrogenase complex (PDHc). Mechanistically, reduced p32 expression, in concert with DLAT, suppresses PDHc activity and the TCA cycle. Furthermore, our research discovered that p32 has a direct binding affinity for copper, facilitating the copper-induced oligomerization of lipo-DLAT specifically in ccRCC cells. This finding reveals an innovative function of the p32/DLAT/copper complex in regulating glycometabolism and the TCA cycle in ccRCC. Importantly, our research provides important new understandings of the underlying molecular processes causing the abnormal mitochondrial metabolism linked to this cancer.
Collapse
Affiliation(s)
- Shaoping Tian
- Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300203, China
| | - Rui Wang
- Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300203, China
| | - Yiting Wang
- Department of Clinical Laboratory, Tianjin Children's Hospital/Tianjin University Children's Hospital, Tianjin 300134, China
| | - Ruibing Chen
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Tianyu Lin
- Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300203, China
| | - Xuesong Xiao
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Xinyu Liu
- Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300203, China
| | - Justin Eze Ideozu
- Genomic Medicine, Genomic Research Center, AbbVie, North Chicago, IL 60064, USA
| | - Hua Geng
- Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, USA
| | - Yong Wang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Dan Yue
- Department of Microbiology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300203, China
| |
Collapse
|
35
|
Lakpour N, Sadeghi MR, Jafarzadeh N, Henkel R, Hajiparvaneh A, Fathi Z, Ghods R, Gilany K, Madjd Z. Metabolic Fingerprinting of Serum and Seminal Plasma of Testicular Cancer Patients Using Raman Spectroscopy: A Pilot Study. J Reprod Infertil 2024; 25:3-11. [PMID: 39157284 PMCID: PMC11330202 DOI: 10.18502/jri.v25i1.15193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 03/13/2024] [Indexed: 08/20/2024] Open
Abstract
Background Testicular cancer (TC) is a relatively rare type of cancer in men. Early diagnosis of TC remains challenging. Metabolomics holds promise in offering valuable insights in this regard. In this study, a metabolic fingerprinting approach was employed to identify potential biomarkers in both serum and seminal plasma of TC patients. Methods A total of 9 patients with testicular cancer and 10 controls were included in the study. The metabolic fingerprinting approach was utilized as a rapid diagnostic tool to analyze the metabolome in serum and seminal plasma of TC patients in comparison to fertile men. Raman spectroscopy was applied for the analysis of metabolites in these biological samples. Results Principal component analysis (PCA) and functional group analysis showed that the differentiation between serum samples from healthy men and TC patients was not possible. However, when analyzing seminal plasma, a significant difference was found between the two groups (p<0.05). Functional group analysis of serum only showed an increase in tryptophan concentration ratio in TC patients as compared to healthy men (p=0.03). In contrast, in seminal plasma of TC patients, this increase was observed in all analyzed compounds, including phenylalanine, tyrosine, lipids, proteins, phenols (p<0.001). Conclusion Our study highlights the potential of metabolic fingerprinting as a fast diagnostic tool for screening TC patients, with seminal plasma serving as a valuable biological sample. Furthermore, several potential biomarkers, particularly phenylalanine, were identified in seminal plasma. This research contributes to our understanding of TC pathogenesis and has the potential to pave the way for early detection and personalized treatment approaches.
Collapse
Affiliation(s)
- Niknam Lakpour
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Reproductive Biotechnology Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Mohammad Reza Sadeghi
- Monoclonal Antibody Research Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Naser Jafarzadeh
- Department of Medical Physics, Tarbiat Modares University, Tehran, Iran
| | - Ralf Henkel
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Department of Medical Bioscience, University of the Western Cape, Bellville, South Africa
- LogixX Pharma, Theale, Berkshire, UK
| | | | - Zohreh Fathi
- Avicenna Fertility Center, Avicenna Research Institute, ACECR, Tehran, Iran
| | - Roya Ghods
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Kambiz Gilany
- Integrative Oncology Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Pal C. Small-molecule redox modulators with anticancer activity: A comprehensive mechanistic update. Free Radic Biol Med 2023; 209:211-227. [PMID: 37898387 DOI: 10.1016/j.freeradbiomed.2023.10.406] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 09/27/2023] [Accepted: 10/25/2023] [Indexed: 10/30/2023]
Abstract
The pursuit of effective anticancer therapies has led to a burgeoning interest in the realm of redox modulation. This review provides a comprehensive exploration of the intricate mechanisms by which diverse anticancer molecules leverage redox pathways for therapeutic intervention. Redox modulation, encompassing the fine balance of oxidation-reduction processes within cells, has emerged as a pivotal player in cancer treatment. This review delves into the multifaceted mechanisms of action employed by various anticancer compounds, including small molecules and natural products, to disrupt cancer cell proliferation and survival. Beginning with an examination of the role of redox signaling in cancer development and resistance, the review highlights how aberrant redox dynamics can fuel tumorigenesis. It then meticulously dissects the strategies employed by anticancer agents to induce oxidative stress, perturb redox equilibrium, and trigger apoptosis within cancer cells. Furthermore, the review explores the challenges and potential side effects associated with redox-based treatments, along with the development of novel redox-targeted agents. In summary, this review offers a profound understanding of the dynamic interplay between redox modulation and anticancer molecules, presenting promising avenues to revolutionize cancer therapy and enhance patient outcomes.
Collapse
Affiliation(s)
- Chinmay Pal
- Department of Chemistry, Gobardanga Hindu College, North 24 Parganas, West Bengal, 743273, India.
| |
Collapse
|
37
|
Karimpur Zahmatkesh A, Khalaj-Kondori M, Hosseinpour Feizi MA, Baradaran B. GLUL gene knockdown and restricted glucose level show synergistic inhibitory effect on the luminal subtype breast cancer MCF7 cells' proliferation and metastasis. EXCLI JOURNAL 2023; 22:847-861. [PMID: 37780942 PMCID: PMC10539544 DOI: 10.17179/excli2023-6287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 08/07/2023] [Indexed: 10/03/2023]
Abstract
The glutamine synthetase path is one of the most important metabolic pathways in luminal breast cancer cells, which plays a critical role in supplying glutamine as an intermediate in the biosynthesis of amino acids and nucleotides. On the other hand, glycolysis and its dominant substrate, glucose, are the most critical players in cancer metabolism. Accordingly, targeting these two critical paths might be more efficient in luminal-type breast cancer treatment. MCF7 cells were cultivated in media containing 4.5, 2, and 1 g/L glucose to study its effects on GLUL (Glutamate Ammonia Ligase) expression. Followingly, high and low glucose cell cultures were transfected with 220 pM of siGLUL and incubated for 48 h at 37 ºC. The cell cycle progression and apoptosis were monitored and assessed by flow cytometry. Expression of GLUL, known as glutamine synthetase, was evaluated in mRNA and protein levels by qRT-PCR and western blotting, respectively. To examine the migration and invasion capacity of studied cells exploited from wound healing assay and subsequent expression studies of glutathione-S-transferase Mu3 (GSTM3) and alfa-enolase (ENO1). Expression of GLUL significantly decreased in cells cultured at lower glucose levels compared to those at higher glucose levels. siRNA-mediated knockdown of GLUL expression in low glucose cultures significantly reduced growth, proliferation, migration, and invasion of the MCF7 cells and enhanced their apoptosis compared to the controls. Based on the results, GLUL suppression down-regulated GSTM3, a main detoxifying enzyme, and up-regulated Bax. According to the role of glycolysis as a ROS suppressor, decreased amounts of glucose could be associated with increased ROS; it can be considered an efficient involved mechanism in this study. Also, increased expression of Bax could be attributable to mTOR/AKT inhibition following GLUL repression. In conclusion, utilizing GLUL and glycolysis inhibitors might be a more effective strategy in luminal-type breast cancer therapy. See also Figure 1(Fig. 1).
Collapse
Affiliation(s)
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz
| |
Collapse
|
38
|
Hata M, Ueno J, Hitomi Y, Kodera M. Roles of DNA Target in Cancer Cell-Selective Cytotoxicity by Dicopper Complexes with DNA Target/Ligand Conjugates. ACS OMEGA 2023; 8:28690-28701. [PMID: 37576680 PMCID: PMC10413468 DOI: 10.1021/acsomega.3c03387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/18/2023] [Indexed: 08/15/2023]
Abstract
The DNA target/ligand conjugates (HLX, X = Pn and Mn, n = 1-3) were synthesized where various lengths of -CONH(CH2CH2O)nCH2CH2NHCO- linkers with a 9-phenanthrenyl (P) or methyl (M) terminal as DNA targets replace the methyl group of 2,6-di(amide-tether cyclen)-p-cresol ligand (HL). DNA binding, DNA cleavage, cellular uptake, and cytotoxicity of [Cu2(μ-OH)(LX)](ClO4)2 (1X) are examined and compared with those of [Cu2(μ-OH)(L)](ClO4)2 (1) to clarify roles of DNA targets. Upon reaction of 1X with H2O2, μ-1,1-O2H complexes are formed for DNA cleavage. 1P1, 1P2, and 1P3 are 22-, 11-, 3-fold more active for conversion of Form II to III in the cleavage of supercoiled plasmid DNA with H2O2 than 1, where the short P-linker may fix a dicopper moiety within a small number of base pairs to facilitate DNA double-strand breaks (dsb). This enhances the proapoptotic activity of 1P1, 1P2, and 1P3, which are 30-, 12-, and 9.9-fold cytotoxic against HeLa cells than 1. DNA dsb and cytotoxicity are 44% correlated in 1P1-3 but 5% in 1M1-3, suggesting specific DNA binding of P-linkers and nonspecific binding of M-linkers in biological cells. 1P1-3 exert cancer cell-selective cytotoxicity against lung and pancreas cancer and normal cells where the short P-linker enhances the selectivity, but 1M1-3 do not. Intracellular visualization, apoptosis assay, and caspase activity assay clarify mitochondrial apoptosis caused by 1P1-3. The highest cancer cell selectivity of 1P1 may be enabled by the short P-linker promoting dsb of mitochondrial DNA with H2O2 increased by mitochondrial dysfunction in cancer cells.
Collapse
Affiliation(s)
- Machi Hata
- Molecular Chemistry and Biochemistry, Doshisha University, Tatara-Miyakodani 1-3, Kyotanabe 610-0321, Japan
| | - Jin Ueno
- Molecular Chemistry and Biochemistry, Doshisha University, Tatara-Miyakodani 1-3, Kyotanabe 610-0321, Japan
| | - Yutaka Hitomi
- Molecular Chemistry and Biochemistry, Doshisha University, Tatara-Miyakodani 1-3, Kyotanabe 610-0321, Japan
| | - Masahito Kodera
- Molecular Chemistry and Biochemistry, Doshisha University, Tatara-Miyakodani 1-3, Kyotanabe 610-0321, Japan
| |
Collapse
|
39
|
Li X, Cao D, Sun S, Wang Y. Anticancer therapeutic effect of ginsenosides through mediating reactive oxygen species. Front Pharmacol 2023; 14:1215020. [PMID: 37564184 PMCID: PMC10411515 DOI: 10.3389/fphar.2023.1215020] [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: 05/01/2023] [Accepted: 07/10/2023] [Indexed: 08/12/2023] Open
Abstract
Dysregulation of reactive oxygen species (ROS) production and ROS-regulated pathways in cancer cells leads to abnormal accumulation of reactive oxygen species, displaying a double-edged role in cancer progression, either supporting transformation/proliferation and stimulating tumorigenesis or inducing cell death. Cancer cells can accommodate reactive oxygen species by regulating them at levels that allow the activation of pro-cancer signaling pathways without inducing cell death via modulation of the antioxidant defense system. Therefore, targeting reactive oxygen species is a promising approach for cancer treatment. Ginsenosides, their derivatives, and related drug carriers are well-positioned to modulate multiple signaling pathways by regulating oxidative stress-mediated cellular and molecular targets to induce apoptosis; regulate cell cycle arrest and autophagy, invasion, and metastasis; and enhance the sensitivity of drug-resistant cells to chemotherapeutic agents of different cancers depending on the type, level, and source of reactive oxygen species, and the type and stage of the cancer. Our review focuses on the pro- and anticancer effects of reactive oxygen species, and summarizes the mechanisms and recent advances in different ginsenosides that bring about anticancer effects by targeting reactive oxygen species, providing new ideas for designing further anticancer studies or conducting more preclinical and clinical studies.
Collapse
Affiliation(s)
- Xiaonan Li
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, China
| | - Donghui Cao
- Department of Clinical Research, The First Hospital of Jilin University, Changchun, China
| | - Siming Sun
- Department of Clinical Research, The First Hospital of Jilin University, Changchun, China
| | - Yuehui Wang
- Department of Geriatrics, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
40
|
Zhu H, Wang X, Lu S, Ou K. Metabolic reprogramming of clear cell renal cell carcinoma. Front Endocrinol (Lausanne) 2023; 14:1195500. [PMID: 37347113 PMCID: PMC10280292 DOI: 10.3389/fendo.2023.1195500] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 05/16/2023] [Indexed: 06/23/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is a malignancy that exhibits metabolic reprogramming as a result of genetic mutations. This reprogramming accommodates the energy and anabolic needs of the cancer cells, leading to changes in glucose, lipid, and bio-oxidative metabolism, and in some cases, the amino acid metabolism. Recent evidence suggests that ccRCC may be classified as a metabolic disease. The metabolic alterations provide potential targets for novel therapeutic interventions or biomarkers for monitoring tumor growth and prognosis. This literature review summarized recent discoveries of metabolic alterations in ccRCC, including changes in glucose, lipid, and amino acid metabolism. The development of metabolic drugs targeting these metabolic pathways was also discussed, such as HIF-2α inhibitors, fatty acid synthase (FAS) inhibitors, glutaminase (GLS) inhibitors, indoleamine 2,3-dioxygenase (IDO) inhibitors, and arginine depletion. Future trends in drug development are proposed, including the use of combination therapies and personalized medicine approaches. In conclusion, this review provides a comprehensive overview of the metabolic alterations in ccRCC and highlights the potential for developing new treatments for this disease.
Collapse
Affiliation(s)
- Haiyan Zhu
- Department of Geriatrics, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xin Wang
- Department of Neurology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Shihao Lu
- Orthopaedics, Changzheng Hospital Affiliated to Second Military Medical University, Shanghai, China
| | - Kongbo Ou
- Department of Urology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
41
|
Zhang B, Wang Q, Lin Z, Zheng Z, Zhou S, Zhang T, Zheng D, Chen Z, Zheng S, Zhang Y, Lin X, Dong R, Chen J, Qian H, Hu X, Zhuang Y, Zhang Q, Jin Z, Jiang S, Ma Y. A novel glycolysis-related gene signature for predicting the prognosis of multiple myeloma. Front Cell Dev Biol 2023; 11:1198949. [PMID: 37333985 PMCID: PMC10272536 DOI: 10.3389/fcell.2023.1198949] [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: 04/02/2023] [Accepted: 05/25/2023] [Indexed: 06/20/2023] Open
Abstract
Background: Metabolic reprogramming is an important hallmark of cancer. Glycolysis provides the conditions on which multiple myeloma (MM) thrives. Due to MM's great heterogeneity and incurability, risk assessment and treatment choices are still difficult. Method: We constructed a glycolysis-related prognostic model by Least absolute shrinkage and selection operator (LASSO) Cox regression analysis. It was validated in two independent external cohorts, cell lines, and our clinical specimens. The model was also explored for its biological properties, immune microenvironment, and therapeutic response including immunotherapy. Finally, multiple metrics were combined to construct a nomogram to assist in personalized prediction of survival outcomes. Results: A wide range of variants and heterogeneous expression profiles of glycolysis-related genes were observed in MM. The prognostic model behaved well in differentiating between populations with various prognoses and proved to be an independent prognostic factor. This prognostic signature closely coordinated with multiple malignant features such as high-risk clinical features, immune dysfunction, stem cell-like features, cancer-related pathways, which was associated with the survival outcomes of MM. In terms of treatment, the high-risk group showed resistance to conventional drugs such as bortezomib, doxorubicin and immunotherapy. The joint scores generated by the nomogram showed higher clinical benefit than other clinical indicators. The in vitro experiments with cell lines and clinical subjects further provided convincing evidence for our study. Conclusion: We developed and validated the utility of the MM glycolysis-related prognostic model, which provides a new direction for prognosis assessment, treatment options for MM patients.
Collapse
Affiliation(s)
- Bingxin Zhang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Quanqiang Wang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhili Lin
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ziwei Zheng
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shujuan Zhou
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tianyu Zhang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Dong Zheng
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zixing Chen
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Sisi Zheng
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yu Zhang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xuanru Lin
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Rujiao Dong
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jingjing Chen
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Honglan Qian
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xudong Hu
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yan Zhuang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qianying Zhang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhouxiang Jin
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Songfu Jiang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yongyong Ma
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Key Laboratory of Intelligent Treatment and Life Support for Critical Diseases of Zhejiang Province, Wenzhou, Zhejiang, China
- Zhejiang Engineering Research Center for Hospital Emergency and Process Digitization, Wenzhou, Zhejiang, China
| |
Collapse
|
42
|
Hatshan MR, Saquib Q, Siddiqui MA, Faisal M, Ahmad J, Al-Khedhairy AA, Shaik MR, Khan M, Wahab R, Matteis VD, Adil SF. Effectiveness of Nonfunctionalized Graphene Oxide Nanolayers as Nanomedicine against Colon, Cervical, and Breast Cancer Cells. Int J Mol Sci 2023; 24:9141. [PMID: 37298090 PMCID: PMC10252622 DOI: 10.3390/ijms24119141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/26/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023] Open
Abstract
Recent studies in nanomedicine have intensively explored the prospective applications of surface-tailored graphene oxide (GO) as anticancer entity. However, the efficacy of nonfunctionalized graphene oxide nanolayers (GRO-NLs) as an anticancer agent is less explored. In this study, we report the synthesis of GRO-NLs and their in vitro anticancer potential in breast (MCF-7), colon (HT-29), and cervical (HeLa) cancer cells. GRO-NLs-treated HT-29, HeLa, and MCF-7 cells showed cytotoxicity in the MTT and NRU assays via defects in mitochondrial functions and lysosomal activity. HT-29, HeLa, and MCF-7 cells treated with GRO-NLs exhibited substantial elevations in ROS, disturbances of the mitochondrial membrane potential, an influx of Ca2+, and apoptosis. The qPCR quantification showed the upregulation of caspase 3, caspase 9, bax, and SOD1 genes in GRO-NLs-treated cells. Western blotting showed the depletion of P21, P53, and CDC25C proteins in the above cancer cell lines after GRO-NLs treatment, indicating its function as a mutagen to induce mutation in the P53 gene, thereby affecting P53 protein and downstream effectors P21 and CDC25C. In addition, there may be a mechanism other than P53 mutation that controls P53 dysfunction. We conclude that nonfunctionalized GRO-NLs exhibit prospective biomedical application as a putative anticancer entity against colon, cervical, and breast cancers.
Collapse
Affiliation(s)
- Mohammad Rafe Hatshan
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.R.H.); (M.R.S.); (M.K.); (S.F.A.)
| | - Quaiser Saquib
- Chair for DNA Research, Zoology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.A.S.); (J.A.); (A.A.A.-K.); (R.W.)
| | - Maqsood A. Siddiqui
- Chair for DNA Research, Zoology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.A.S.); (J.A.); (A.A.A.-K.); (R.W.)
| | - Mohammad Faisal
- Botany and Microbiology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia;
| | - Javed Ahmad
- Chair for DNA Research, Zoology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.A.S.); (J.A.); (A.A.A.-K.); (R.W.)
| | - Abdulaziz A. Al-Khedhairy
- Chair for DNA Research, Zoology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.A.S.); (J.A.); (A.A.A.-K.); (R.W.)
| | - Mohammed Rafi Shaik
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.R.H.); (M.R.S.); (M.K.); (S.F.A.)
| | - Mujeeb Khan
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.R.H.); (M.R.S.); (M.K.); (S.F.A.)
| | - Rizwan Wahab
- Chair for DNA Research, Zoology Department, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.A.S.); (J.A.); (A.A.A.-K.); (R.W.)
| | - Valeria De Matteis
- Department of Mathematics and Physics “Ennio De Giorgi”, University of Salento, Via Arnesano, 73100 Lecce, Italy;
| | - Syed Farooq Adil
- Department of Chemistry, College of Sciences, King Saud University, P.O. Box 2455, Riyadh 11451, Saudi Arabia; (M.R.H.); (M.R.S.); (M.K.); (S.F.A.)
| |
Collapse
|
43
|
Li S, Wang D, Zheng X, Li Y, Ding C, Wang M, Ge X, Jiang J, Qiao Y, Wang Y. Combination of niclosamide and quinacrine inactivates Akt/HK2/Cyclin D1 axis mediated by glucose deprivation towards the inhibition of melanoma cell proliferation. Biomed Pharmacother 2023; 163:114865. [PMID: 37187020 DOI: 10.1016/j.biopha.2023.114865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/01/2023] [Accepted: 05/08/2023] [Indexed: 05/17/2023] Open
Abstract
Malignant melanoma is one of the most aggressive and lethal skin cancer. At present, the treatment methods for melanoma have shortcomings. Glucose is the primary energy source of cancer cells. However, it is unclear whether glucose deprivation can be used to treat melanoma. Herein, we first found glucose played an essential role in melanoma proliferation. We then further found a drug combination of niclosamide and quinacrine could inhibit melanoma proliferation and glucose intake. Thirdly, we revealed the mechanism of anti-melanoma effect of the drug combination, which suppressed the Akt pathway. In addition, the first-rate limiting enzyme HK2 of glucose metabolism was inhibited. This work also disclosed that the decrease of HK2 inhibited cyclin D1 by reducing the activity of transcription factor E2F3, which further suppressed the proliferation of melanoma cells. The drug combination treatment also resulted in significant tumor regression in the absence of obvious morphologic changes in primary organ in vivo. In summary, our study demonstrated that the drug combination treatment created glucose deprivation to inactive the Akt/HK2/cyclin D1 axis, thereby inhibited the proliferation of melanoma cells, providing a potential anti-melanoma strategy.
Collapse
Affiliation(s)
- Shuangting Li
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Diancan Wang
- Department of Oral and Maxillofacial Surgery, Peking University School and Hospital of Stomatology, No.22, Zhongguancun Avenue South, Haidian District, Beijing 100081, China
| | - Xuan Zheng
- Qingdao Stomatological Hospital Affiliated to Qingdao University, No.17 Dexian Road, Shinan District, Qingdao 266001, Shandong Province, China
| | - Yi Li
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China
| | - Chong Ding
- Central Laboratory, Peking University School and Hospital of Stomatology, No.22, Zhongguancun Avenue South, Haidian District, Beijing 100081, China
| | - Meng Wang
- Central Laboratory, Peking University School and Hospital of Stomatology, No.22, Zhongguancun Avenue South, Haidian District, Beijing 100081, China
| | - Xuejun Ge
- Shanxi Medical University School and Hospital of Stomatology, Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan 030001, China.
| | - Jiuhui Jiang
- Department of Orthodontics, Peking University School and Hospital of Stomatology, No.22, Zhongguancun Avenue South, Haidian District, Beijing 100081, China.
| | - Yan Qiao
- Beijing National Laboratory for Molecular Sciences (BNLMS), Laboratory of Polymer Physics and Chemistry, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Yixiang Wang
- Central Laboratory, Peking University School and Hospital of Stomatology, No.22, Zhongguancun Avenue South, Haidian District, Beijing 100081, China.
| |
Collapse
|
44
|
Callaghan CM, Abukhiran IM, Masaadeh A, Van Rheeden RV, Kalen AL, Rodman SN, Petronek MS, Mapuskar KA, George BN, Coleman MC, Goswami PC, Allen BG, Spitz DR, Caster JM. Manipulation of Redox Metabolism Using Pharmacologic Ascorbate Opens a Therapeutic Window for Radio-Sensitization by ATM Inhibitors in Colorectal Cancer. Int J Radiat Oncol Biol Phys 2023; 115:933-944. [PMID: 36228747 PMCID: PMC9974877 DOI: 10.1016/j.ijrobp.2022.10.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/30/2022] [Accepted: 10/05/2022] [Indexed: 11/11/2022]
Abstract
PURPOSE Ataxia telangiectasia mutated kinase (ATM) inhibitors are potent radiosensitizers that regulate DNA damage responses and redox metabolism, but they have not been translated clinically because of the potential for excess normal tissue toxicity. Pharmacologic ascorbate (P-AscH-; intravenous administration achieving mM plasma concentrations) selectively enhances H2O2-induced oxidative stress and radiosensitization in tumors while acting as an antioxidant and mitigating radiation damage in normal tissues including the bowel. We hypothesized that P-AscH- could enhance the therapeutic index of ATM inhibitor-based chemoradiation by simultaneously enhancing the intended effects of ATM inhibitors in tumors and mitigating off-target effects in adjacent normal tissues. METHODS AND MATERIALS Clonogenic survival was assessed in human (human colon tumor [HCT]116, SW480, HT29) and murine (CT26, MC38) colorectal tumor lines and normal cells (human umbilical vein endothelial cell, FHs74) after radiation ± DNA repair inhibitors ± P-AscH-. Tumor growth delay was assessed in mice with HCT116 or MC38 tumors after fractionated radiation (5 Gy × 3) ± the ATM inhibitor KU60019 ± P-AscH-. Intestinal injury, oxidative damage, and transforming growth factor β immunoreactivity were quantified using immunohistochemistry after whole abdominal radiation (10 Gy) ± KU60019 ± P-AscH-. Cell cycle distribution and ATM subcellular localization were assessed using flow cytometry and immunohistochemistry. The role of intracellular H2O2 fluxes was assessed using a stably expressed doxycycline-inducible catalase transgene. RESULTS KU60019 with P-AscH- enhanced radiosensitization in colorectal cancer models in vitro and in vivo by H2O2-dependent oxidative damage to proteins and enhanced DNA damage, abrogation of the postradiation G2 cell cycle checkpoint, and inhibition of ATM nuclear localization. In contrast, concurrent P-AscH- markedly reduced intestinal toxicity and oxidative damage with KU60019. CONCLUSIONS We provide evidence that redox modulating drugs, such as P-AscH-, may facilitate the clinical translation of ATM inhibitors by enhancing tumor radiosensitization while simultaneously protecting normal tissues.
Collapse
Affiliation(s)
- Cameron M Callaghan
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa
| | - Ibrahim M Abukhiran
- Department of Pathology, University of Iowa Hospitals and Clinics and Carver College of Medicine, Iowa City, Iowa
| | - Amr Masaadeh
- Department of Pathology, University of Iowa Hospitals and Clinics and Carver College of Medicine, Iowa City, Iowa
| | | | - Amanda L Kalen
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Samuel N Rodman
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Michael S Petronek
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Kranti A Mapuskar
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Benjamin N George
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa
| | - Mitchell C Coleman
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Prabhat C Goswami
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Bryan G Allen
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Douglas R Spitz
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa
| | - Joseph M Caster
- Department of Radiation Oncology, University of Iowa Hospital and Clinics, Iowa City, Iowa; Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa.
| |
Collapse
|
45
|
Liu Y, Liu K, Thorne RF, Shi R, Zhang Q, Wu M, Liu L. Mitochondrial SENP2 regulates the assembly of SDH complex under metabolic stress. Cell Rep 2023; 42:112041. [PMID: 36708515 DOI: 10.1016/j.celrep.2023.112041] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 10/17/2022] [Accepted: 01/13/2023] [Indexed: 01/28/2023] Open
Abstract
Succinate dehydrogenase (SDH) is a heterotetrameric enzyme complex belonging to the mitochondrial respiratory chain and uniquely links the tricarboxylic acid (TCA) cycle with oxidative phosphorylation. Cancer-related SDH mutations promote succinate accumulation, which is regarded as an oncometabolite. Post-translational modifications of SDH complex components are known to regulate SDH activity, although the contribution of SUMOylation remains unclear. Here, we show that SDHA is SUMOylated by PIAS3 and deSUMOylated by SENP2, events dictating the assembly and activity of the SDH complex. Moreover, CBP acetylation of SENP2 negatively regulates its deSUMOylation activity. Under glutamine deprivation, CBP levels decrease, and the ensuing SENP2 activation and SDHA deSUMOylation serve to concurrently dampen the TCA cycle and electron transport chain (ETC) activity. Along with succinate accumulation, this mechanism avoids excessive reactive oxygen species (ROS) production to promote cancer cell survival. This study elucidates a major function of mitochondrial-localized SENP2 and expands our understanding of the role of SUMOylation in resolving metabolic stress.
Collapse
Affiliation(s)
- Ying Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Kejia Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Rick F Thorne
- Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Zhengzhou 450053, China; School of Environmental and Life Sciences, University of Newcastle, Callaghan, NSW 2258, Australia
| | - Ronghua Shi
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Qingyuan Zhang
- Department of Radiation Oncology, Harbin Medical University Cancer Hospital, Harbin 150081, China.
| | - Mian Wu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Translational Research Institute, Henan Provincial People's Hospital, Academy of Medical Science, Zhengzhou University, Zhengzhou 450053, China.
| | - Lianxin Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei 230001, China.
| |
Collapse
|
46
|
Huntingtin-associated protein 1 is a potential tumor suppressor for gastric cancer. Mol Biol Rep 2023; 50:1517-1531. [PMID: 36509909 DOI: 10.1007/s11033-022-08090-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 11/04/2022] [Indexed: 12/14/2022]
Abstract
BACKGROUND Gastric cancer is heterogeneous cancer and the causes of this disease are complex. New diagnostic and therapeutic targets are urgently needed to explore. Huntingtin-associated protein 1 (HAP1) is directly related to Huntington's disease (HD). However, patients with Huntington's disease have a lower incidence of cancer. Therefore, we are committed to studying the correlation between HAP1 and gastric carcinogenesis and development. METHODS AND RESULTS Immunohistochemical staining, western blot analysis, and RT-qPCR were conducted to explore the localization and expression of HAP1 in gastric cancer. To study the biological significance of HAP1, we overexpressed HAP1 in both MKN28 and AGS cell lines by lentivirus infection. To explore the role of HAP1 in cell proliferation, the cells counting assay, EdU incorporation assay, and colony formation assay were carried out. We performed the wound healing assay and transwell assay to study the cell migration and invasion. To further investigate whether HAP1 could regulate gastric cancer cell death during glucose deprivation, Annexin V-FITC/PI staining was performed. In our study, we elucidated that HAP1 was downregulated in gastric cancer. What's more, overexpressing HAP1 inhibited cell proliferation, cell migration and invasion, and triggered apoptosis during glucose deprivation. More importantly, the antitumor properties and mechanisms of HAP1 have been elucidated further in gastric cancer. CONCLUSIONS Taken together, the available evidence implies that HAP1 may serve as a potential tumor suppressor, making it a significant target in preventing and treating gastric cancer. This research provides a theoretical basis for the early diagnosis, clinical targeted therapy, and prognosis evaluation of gastric cancer.
Collapse
|
47
|
Eaton L, Welch I, Halal AK, Bengtsson J, Pamenter ME. Apocynin reduces dihydroethidium fluorescence in naked mole-rat cortex independently of NADPH oxidase. Comp Biochem Physiol A Mol Integr Physiol 2023; 276:111342. [PMID: 36375753 DOI: 10.1016/j.cbpa.2022.111342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
Abstract
Pharmacological agents that modulate cellular targets offer a powerful approach to interrogate the role of a given component in cellular signalling cascades. However, such drugs are often nonspecific and/or have unexpected off-target effects. One cellular target of interest is the NADPH oxidase (NOX) enzyme family, which consume oxygen and produce reactive oxygen species. Among the most widely used inhibitors of NOX is apocynin, but apocynin also has off-target effects that may interfere with detection assays of hydrogen peroxide (H2O2) or directly scavenge H2O2 in some cell lines. Nonetheless, apocynin remains widely used for in vivo studies of brain function. Therefore, we used apocynin and another widely-used NOX inhibitor - diphenyleneiodonium (DPI) - to study the role of NOX in ROS homeostasis of hypoxia-tolerant naked mole-rat cortical brain slices during a normoxia➔hypoxia➔reoxygenation protocol. Using fluorescence microscopy, we found that apocynin decreased dihydroethidium fluorescence from naked mole-rat cortex in all treatment conditions by 65-75% of pre-drug normoxic control. This change was rapid, occurring within minutes of drug perfusion, and reversed equally rapidly upon washout. Conversely, apocynin had no effect on 5-(and-6)-chloromethyl-2',7'-dichlorodihydrofluorescein diacetate, acetyl ester (CM-H2DCFDA) fluorescence. DPI also had no effect on either fluorescence signal, suggesting that the effect of apocynin is due to indirect actions of the drug and not due to modulation of NOX. Taken together, our results highlight the pitfalls of pharmacological neuroscience and add to the body of evidence suggesting that apocynin is not a useful compound for targeting NOX.
Collapse
Affiliation(s)
- Liam Eaton
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Isabella Welch
- Department of Biology, University of Ottawa, Ottawa, ON, Canada. https://twitter.com/Isabellawel1998
| | | | - John Bengtsson
- Department of Biology, University of Ottawa, Ottawa, ON, Canada
| | - Matthew E Pamenter
- Department of Biology, University of Ottawa, Ottawa, ON, Canada; University of Ottawa Brain and Mind Research Institute, Ottawa, ON, Canada.
| |
Collapse
|
48
|
Mikolaskova I, Crnogorac-Jurcevic T, Smolkova B, Hunakova L. Nutraceuticals as Supportive Therapeutic Agents in Diabetes and Pancreatic Ductal Adenocarcinoma: A Systematic Review. BIOLOGY 2023; 12:158. [PMID: 36829437 PMCID: PMC9953002 DOI: 10.3390/biology12020158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 01/22/2023]
Abstract
The correlation between pancreatic ductal adenocarcinoma (PDAC) and diabetes-related mechanisms support the hypothesis that early therapeutic strategies targeting diabetes can contribute to PDAC risk reduction and treatment improvement. A systematic review was conducted, using PubMed, Embase and Cochrane Library databases, to evaluate the current evidence from clinical studies qualitatively examining the efficacy of four natural products: Curcumin-Curcuma longa L.; Thymoquinone-Nigella sativa L.; Genistein-Glycine max L.; Ginkgo biloba L.; and a low-carbohydrate ketogenic diet in type 2 diabetes (T2D) and PDAC treatment. A total of 28 clinical studies were included, showing strong evidence of inter-study heterogeneity. Used as a monotherapy or in combination with chemo-radiotherapy, the studied substances did not significantly improve the treatment response of PDAC patients. However, pronounced therapeutic efficacy was confirmed in T2D. The natural products and low-carbohydrate ketogenic diet, combined with the standard drugs, have the potential to improve T2D treatment and thus potentially reduce the risk of cancer development and improve multiple biological parameters in PDAC patients.
Collapse
Affiliation(s)
- Iveta Mikolaskova
- Institute of Immunology, Faculty of Medicine, Comenius University, Odborarske Namestie 14, 811 08 Bratislava, Slovakia
| | - Tatjana Crnogorac-Jurcevic
- Barts Cancer Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University, Charterhouse Square, London EC1M 6BQ, UK
| | - Bozena Smolkova
- Biomedical Research Center, Slovak Academy of Sciences, Cancer Research Institute, Dubravska Cesta 9, 845 05 Bratislava, Slovakia
| | - Luba Hunakova
- Institute of Immunology, Faculty of Medicine, Comenius University, Odborarske Namestie 14, 811 08 Bratislava, Slovakia
| |
Collapse
|
49
|
Mendes MIP, Arnaut LG. Redaporfin Development for Photodynamic Therapy and its Combination with Glycolysis Inhibitors. Photochem Photobiol 2022; 99:769-776. [PMID: 36564949 DOI: 10.1111/php.13770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 12/13/2022] [Indexed: 12/25/2022]
Abstract
Photodynamic therapy (PDT) remains an underutilized treatment modality in oncology. Many efforts have been dedicated to the development of better photosensitizers, better formulations and delivery methods, rigorous planning of light dose distribution in tissues, mechanistic insight, improvement of treatment protocols and combinations with other therapeutic agents. Hopefully, progress in all these fields will eventually expand the use of PDT. Here we offer a brief review of our own contribution to the development of a photosensitizer for PDT - redaporfin - currently in Phase II clinical trials, and present data on its combination with two glycolysis inhibitors: 2-deoxyglucose and 3-bromopyruvate. We show that 3-bromopyruvate is more cytotoxic to a carcinoma cell line (CT26) than to a normal fibroblast (3T3) cell line, and that this selectivity is maintained in the in vitro combination with redaporfin-PDT. This combination was investigated in BALB/c mice with large subcutaneous CT26 tumors and it is shown that the cure rate in the combination is higher (33% cures) than in PDT (11% cures) or in 3-bromopyruvate (no cures) alone. The combination of redaporfin-PDT with 3-bromopyruvate illustrates the potential of combination therapies and how PDT benefits can be enhanced by systemic drugs with complementary targets.
Collapse
Affiliation(s)
| | - Luis G Arnaut
- CQC-IMS, Department of Chemistry, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
50
|
Vukic MD, Vukovic NL, Obradovic AD, Galovičová L, Čmiková N, Kačániová M, Matic MM. Chemical Composition and Biological Activity of Tanacetum balsamita Essential Oils Obtained from Different Plant Organs. PLANTS (BASEL, SWITZERLAND) 2022; 11:plants11243474. [PMID: 36559586 PMCID: PMC9783112 DOI: 10.3390/plants11243474] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/02/2022] [Accepted: 12/03/2022] [Indexed: 05/25/2023]
Abstract
The aim of this study is to evaluate the chemical composition of Tanacetum balsamita L. essential oils (EOs) obtained from different plant organs, flowers (FEO), leaves (LEO), and stems (SEO), as well as to assess their biological properties. The results obtained by using GC and GC/MS analysis indicate that this plant belongs to the carvone chemotype. Moreover, we examined the oil's antimicrobial and antitumor potential. Antimicrobial effects were determined using minimum inhibitory concentrations assay and the vapor phase method. Obtained results indicate better antimicrobial activity of investigated EO samples compared to the commercially available antibiotics. On the treatment with FEO, Y. enterocolitica and H. influenzae showed high sensitivity, while treatment with LEO and SEO showed the highest effects against S. aureus. The vapor phase method, as an in situ antibacterial analysis, was performed using LEO. Obtained results showed that this EO has significant activity toward S. pneumoniae in the apple and carrot models, L. monocytogenes in the pear model, and Y. enterocolitica in the white radish model. The potential antitumor mechanisms of FEO, LEO, and SEO were determined by the means of cell viability, redox potential, and migratory capacity in the MDA-MB-231 and MDA-MB-468 cell lines. The results show that these EOs exert antiviability potential in a time- and dose-dependent manner. Moreover, treatments with these EOs decreased the levels of superoxide anion radical and increased the levels of nitric oxide in both tested cell lines. The results regarding total and reduced glutathione revealed, overall, an increase in the levels of total glutathione and a decrease in the levels of reduced glutathione, indicating strong antioxidative potential in tested cancer cells in response to the prooxidative effects of the tested EOs. The tested EOs also exerted a drop in migratory capacity, which indicates that they can be potentially used as chemotherapeutic agents.
Collapse
Affiliation(s)
- Milena D. Vukic
- Department of Chemistry, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Nenad L. Vukovic
- Department of Chemistry, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Ana D. Obradovic
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia
| | - Lucia Galovičová
- Institute of Horticulture, Faculty of Horticulture and Landscape Engineering, Slovak University of Agriculture, Tr. A. Hlinku 2, 94976 Nitra, Slovakia
| | - Natália Čmiková
- Institute of Horticulture, Faculty of Horticulture and Landscape Engineering, Slovak University of Agriculture, Tr. A. Hlinku 2, 94976 Nitra, Slovakia
| | - Miroslava Kačániová
- Institute of Horticulture, Faculty of Horticulture and Landscape Engineering, Slovak University of Agriculture, Tr. A. Hlinku 2, 94976 Nitra, Slovakia
- Department of Bioenergy, Food Technology and Microbiology, Institute of Food Technology and Nutrition, University of Rzeszow, 4 Zelwerowicza Str., 35-601 Rzeszow, Poland
| | - Milos M. Matic
- Department of Biology and Ecology, Faculty of Science, University of Kragujevac, 34000 Kragujevac, Serbia
| |
Collapse
|