1
|
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
|
2
|
Geirnaert F, Kerkhove L, Montay-Gruel P, Gevaert T, Dufait I, De Ridder M. Exploring the Metabolic Impact of FLASH Radiotherapy. Cancers (Basel) 2025; 17:133. [PMID: 39796760 PMCID: PMC11720285 DOI: 10.3390/cancers17010133] [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: 12/16/2024] [Revised: 12/30/2024] [Accepted: 12/31/2024] [Indexed: 01/13/2025] Open
Abstract
FLASH radiotherapy (FLASH RT) is an innovative modality in cancer treatment that delivers ultrahigh dose rates (UHDRs), distinguishing it from conventional radiotherapy (CRT). FLASH RT has demonstrated the potential to enhance the therapeutic window by reducing radiation-induced damage to normal tissues while maintaining tumor control, a phenomenon termed the FLASH effect. Despite promising outcomes, the precise mechanisms underlying the FLASH effect remain elusive and are a focal point of current research. This review explores the metabolic and cellular responses to FLASH RT compared to CRT, with particular focus on the differential impacts on normal and tumor tissues. Key findings suggest that FLASH RT may mitigate damage in healthy tissues via altered reactive oxygen species (ROS) dynamics, which attenuate downstream oxidative damage. Studies indicate the FLASH RT influences iron metabolism and lipid peroxidation pathways differently than CRT. Additionally, various studies indicate that FLASH RT promotes the preservation of mitochondrial integrity and function, which helps maintain apoptotic pathways in normal tissues, attenuating damage. Current knowledge of the metabolic influences following FLASH RT highlights its potential to minimize toxicity in normal tissues, while also emphasizing the need for further studies in biologically relevant, complex systems to better understand its clinical potential. By targeting distinct metabolic pathways, FLASH RT could represent a transformative advance in RT, ultimately improving the therapeutic window for cancer treatment.
Collapse
Affiliation(s)
- Febe Geirnaert
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (F.G.); (L.K.); (T.G.); (I.D.)
| | - Lisa Kerkhove
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (F.G.); (L.K.); (T.G.); (I.D.)
| | - Pierre Montay-Gruel
- Radiation Oncology Department, Iridium Netwerk, 2610 Antwerp, Belgium;
- Antwerp Research in Radiation Oncology (AreRO), Center for Oncological Research (CORE), University of Antwerp, 2020 Antwerp, Belgium
| | - Thierry Gevaert
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (F.G.); (L.K.); (T.G.); (I.D.)
| | - Inès Dufait
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (F.G.); (L.K.); (T.G.); (I.D.)
| | - Mark De Ridder
- Department of Radiotherapy, Universitair Ziekenhuis Brussel, Vrije Universiteit Brussel, 1090 Brussels, Belgium; (F.G.); (L.K.); (T.G.); (I.D.)
| |
Collapse
|
3
|
Vaishampayan P, Lee Y. Redox-active vitamin C suppresses human osteosarcoma growth by triggering intracellular ROS-iron-calcium signaling crosstalk and mitochondrial dysfunction. Redox Biol 2024; 75:103288. [PMID: 39083898 PMCID: PMC11342202 DOI: 10.1016/j.redox.2024.103288] [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: 07/01/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024] Open
Abstract
Pharmacological vitamin C (VC) has gained attention for its pro-oxidant characteristics and selective ability to induce cancer cell death. However, defining its role in cancer has been challenging due to its complex redox properties. In this study, using a human osteosarcoma (OS) model, we show that the redox-active property of VC is critical for inducing non-apoptotic cancer cell death via intracellular reactive oxygen species (ROS)-iron-calcium crosstalk and mitochondrial dysfunction. In both 2D and 3D OS cell culture models, only the oxidizable form of VC demonstrated potent dose-dependent cytotoxicity, while non-oxidizable and oxidized VC derivatives had minimal effects. Live-cell imaging showed that only oxidizable VC caused a surge in cytotoxic ROS, dependent on iron rather than copper. Inhibitors of ferroptosis, a form of iron-dependent cell death, along with classical apoptosis inhibitors, were unable to completely counteract the cytotoxic effects induced by VC. Further pharmacological and genetic inhibition analyses showed that VC triggers calcium release through inositol 1,4,5-trisphosphate receptors (IP3Rs), leading to mitochondrial ROS production and eventual cell death. RNA sequencing revealed down-regulation of genes involved in the mitochondrial electron transport chain and oxidative phosphorylation upon pharmacological VC treatment. Consistently, high-dose VC reduced mitochondrial membrane potential, oxidative phosphorylation, and ATP levels, with ATP reconstitution rescuing VC-induced cytotoxicity. In vivo OS xenograft studies demonstrated reduced tumor growth with high-dose VC administration, concomitant with the altered expression of mitochondrial ATP synthase (MT-ATP). These findings emphasize VC's potential clinical utility in osteosarcoma treatment by inducing mitochondrial metabolic dysfunction through a vicious intracellular ROS-iron-calcium cycle.
Collapse
Affiliation(s)
- Prajakta Vaishampayan
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, 99202, USA
| | - Yool Lee
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA, 99202, USA; Department of Integrative Physiology and Neuroscience, College of Veterinary Medicine, Washington State University, Pullman, WA, 99164, USA; Sleep and Performance Research Center, Washington State University, Spokane, WA, 99202, USA; Steve Gleason Institute for Neuroscience, Washington State University, Spokane, WA, 99202, USA.
| |
Collapse
|
4
|
Kittilukkana A, Carmona A, Pilapong C, Ortega R. TauSTED super-resolution imaging of labile iron in primary hippocampal neurons. Metallomics 2024; 16:mfad074. [PMID: 38148121 DOI: 10.1093/mtomcs/mfad074] [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: 10/27/2023] [Accepted: 12/25/2023] [Indexed: 12/28/2023]
Abstract
Iron dyshomeostasis is involved in many neurological disorders, particularly neurodegenerative diseases where iron accumulates in various brain regions. Identifying mechanisms of iron transport in the brain is crucial for understanding the role of iron in healthy and pathological states. In neurons, it has been suggested that iron can be transported by the axon to different brain regions in the form of labile iron; a pool of reactive and exchangeable intracellular iron. Here we report a novel approach to imaging labile ferrous iron, Fe(II), in live primary hippocampal neurons using confocal and TauSTED (stimulated emission depletion) microscopy. TauSTED is based on super-resolution STED nanoscopy, which combines high spatial resolution imaging (<40 nm) with fluorescence lifetime information, thus reducing background noise and improving image quality. We applied TauSTED imaging utilizing biotracker FerroFarRed Fe(II) and found that labile iron was present as submicrometric puncta in dendrites and axons. Some of these iron-rich structures are mobile and move along neuritic pathways, arguing for a labile iron transport mechanism in neurons. This super-resolution imaging approach offers a new perspective for studying the dynamic mechanisms of axonal and dendritic transport of iron at high spatial resolution in living neurons. In addition, this methodology could be transposed to the imaging of other fluorescent metal sensors.
Collapse
Affiliation(s)
- Aiyarin Kittilukkana
- Chiang Mai University, Faculty of Associated Medical Sciences, Department of Radiologic Technology, Center of Excellence for Molecular Imaging (CEMI), 50200 Chiang Mai, Thailand
- University Bordeaux, CNRS, LP2I Bordeaux, UMR 5797, Chemical Imaging and Speciation, F-33170 Gradignan, France
| | - Asuncion Carmona
- University Bordeaux, CNRS, LP2I Bordeaux, UMR 5797, Chemical Imaging and Speciation, F-33170 Gradignan, France
| | - Chalermchai Pilapong
- Chiang Mai University, Faculty of Associated Medical Sciences, Department of Radiologic Technology, Center of Excellence for Molecular Imaging (CEMI), 50200 Chiang Mai, Thailand
| | - Richard Ortega
- University Bordeaux, CNRS, LP2I Bordeaux, UMR 5797, Chemical Imaging and Speciation, F-33170 Gradignan, France
| |
Collapse
|
5
|
Wagner BA, Buettner GR. Stability of aqueous solutions of ascorbate for basic research and for intravenous administration. ADVANCES IN REDOX RESEARCH : AN OFFICIAL JOURNAL OF THE SOCIETY FOR REDOX BIOLOGY AND MEDICINE AND THE SOCIETY FOR FREE RADICAL RESEARCH-EUROPE 2023; 9:100077. [PMID: 37808406 PMCID: PMC10552410 DOI: 10.1016/j.arres.2023.100077] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
Ascorbate (vitamin C) can rapidly oxidize in many near-neutral pH, aqueous solutions. We report on the stability of ascorbate solutions prepared for infusion into patients using standard pharmacy protocols, for example, 75 g of ascorbate/L in water for infusion. The concentration of ascorbate was monitored for changes over time using direct UV-Vis spectroscopy. The pH of the solution was about 5.7 with no significant change over 24 h. There was only an approximate loss of 1% per day over the first 3 days of storage. This information allows decisions on how far ahead of need such preparations can be made. We also provide laboratory approaches to minimize or control the rate of oxidation of ascorbate solutions for use in chemical and biochemical studies as well as preclinical animal studies. The goal is to have the amount of ascorbate intended to be used in experiments be the actual amount available.
Collapse
Affiliation(s)
- Brett A. Wagner
- Free Radical and Radiation Biology Program, ESR Facility, The University of Iowa, Iowa City, IA 52242, USA
| | - Garry R. Buettner
- Free Radical and Radiation Biology Program, ESR Facility, The University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
6
|
Wang L, Shao J, Su C, Yang J. The application of optical technology in the diagnosis and therapy of oxidative stress-mediated hepatic ischemia-reperfusion injury. Front Bioeng Biotechnol 2023; 11:1133039. [PMID: 36890921 PMCID: PMC9986550 DOI: 10.3389/fbioe.2023.1133039] [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: 12/28/2022] [Accepted: 02/09/2023] [Indexed: 02/22/2023] Open
Abstract
Hepatic ischemia-reperfusion injury (HIRI) is defined as liver tissue damage and cell death caused by reperfusion during liver transplantation or hepatectomy. Oxidative stress is one of the important mechanisms of HIRI. Studies have shown that the incidence of HIRI is very high, however, the number of patients who can get timely and efficient treatment is small. The reason is not hard to explain that invasive ways of detection and lack of timely of diagnostic methods. Hence, a new detection method is urgently needed in clinic application. Reactive oxygen species (ROS), which are markers of oxidative stress in the liver, could be detected by optical imaging and offer timely and effective non-invasive diagnosis and monitoring. Optical imaging could become the most potential tool of diagnosis of HIRI in the future. In addition, optical technology can also be used in disease treatment. It found that optical therapy has the function of anti-oxidative stress. Consequently, it has possibility to treat HIRI caused by oxidative stress. In this review, we mainly summarized the application and prospect of optical techniques in oxidative stress-induced by HIRI.
Collapse
Affiliation(s)
- Lijuan Wang
- Department of Medicine, Hengyang Medical School, University of South China, Hengyang, China.,Department of Anesthesiology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jiali Shao
- Department of Anesthesiology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Chen Su
- Department of Anesthesiology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jinfeng Yang
- Department of Anesthesiology, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| |
Collapse
|
7
|
Gęgotek A, Skrzydlewska E. Antioxidative and Anti-Inflammatory Activity of Ascorbic Acid. Antioxidants (Basel) 2022; 11:1993. [PMID: 36290716 PMCID: PMC9598715 DOI: 10.3390/antiox11101993] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 09/30/2022] [Accepted: 10/05/2022] [Indexed: 07/24/2023] Open
Abstract
Ascorbic acid, as a one of the basic exogenous vitamins, occurs in the body in the form of ascorbate, known for its strong antioxidant and anti-inflammatory properties. The presented review shows not only the importance of ascorbate as a free radical scavenger but also summarizes its antioxidant action based on other mechanisms, including the activation of intracellular antioxidant systems and its effect on the NFκB/TNFα pathway and apoptosis. Ascorbate interacts with small-molecule antioxidants, including tocopherol, glutathione, and thioredoxin; it can also stimulate biosynthesis and the activation of antioxidant enzymes, such as superoxide dismutase, catalase, or glutathione peroxidase. Moreover, ascorbate promotes the activity of transcription factors (Nrf2, Ref-1, AP-1), which enables the expression of genes encoding antioxidant proteins. Additionally, it supports the action of other exogenous antioxidants, mainly polyphenols. In this regard, both DNA, proteins, and lipids are protected against oxidation, leading to an inflammatory reaction and even cell death. Although ascorbate has strong antioxidant properties, it can also have pro-oxidant effects in the presence of free transition metals. However, its role in the prevention of DNA mutation, inflammation, and cell apoptosis, especially in relation to cancer cells, is controversial.
Collapse
|
8
|
Role of Fe, Transferrin and Transferrin Receptor in Anti-Tumor Effect of Vitamin C. Cancers (Basel) 2022; 14:cancers14184507. [PMID: 36139668 PMCID: PMC9496724 DOI: 10.3390/cancers14184507] [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: 08/15/2022] [Revised: 09/14/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
High-dose vitamin C (VC) exhibits anti-tumor effects, and the cytotoxicity of VC is correlated with oxidative stress. However, iron, as a redox metal, plays an important effect in redox cycling and free radical formation in cells. This study addresses the role of iron ion in the cytotoxicity of VC. We found that iron supplementation increases the anti-tumor effect of VC, which was influenced by the cellular iron uptake pathway-transferrin (TF)/transferrin receptor (TFR) system. The TFR expression of tumors can be assessed by 68Ga-citrate PET imaging, and it would be helpful to screen out the tumor type which is more sensitive to VC combined with an iron supplementation treatment.
Collapse
|
9
|
Travaglini S, Gurnari C, Antonelli S, Silvestrini G, Noguera NI, Ottone T, Voso MT. The Anti-Leukemia Effect of Ascorbic Acid: From the Pro-Oxidant Potential to the Epigenetic Role in Acute Myeloid Leukemia. Front Cell Dev Biol 2022; 10:930205. [PMID: 35938170 PMCID: PMC9352950 DOI: 10.3389/fcell.2022.930205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/24/2022] [Indexed: 11/13/2022] Open
Abstract
Data derived from high-throughput sequencing technologies have allowed a deeper understanding of the molecular landscape of Acute Myeloid Leukemia (AML), paving the way for the development of novel therapeutic options, with a higher efficacy and a lower toxicity than conventional chemotherapy. In the antileukemia drug development scenario, ascorbic acid, a natural compound also known as Vitamin C, has emerged for its potential anti-proliferative and pro-apoptotic activities on leukemic cells. However, the role of ascorbic acid (vitamin C) in the treatment of AML has been debated for decades. Mechanistic insight into its role in many biological processes and, especially, in epigenetic regulation has provided the rationale for the use of this agent as a novel anti-leukemia therapy in AML. Acting as a co-factor for 2-oxoglutarate-dependent dioxygenases (2-OGDDs), ascorbic acid is involved in the epigenetic regulations through the control of TET (ten-eleven translocation) enzymes, epigenetic master regulators with a critical role in aberrant hematopoiesis and leukemogenesis. In line with this discovery, great interest has been emerging for the clinical testing of this drug targeting leukemia epigenome. Besides its role in epigenetics, ascorbic acid is also a pivotal regulator of many physiological processes in human, particularly in the antioxidant cellular response, being able to scavenge reactive oxygen species (ROS) to prevent DNA damage and other effects involved in cancer transformation. Thus, for this wide spectrum of biological activities, ascorbic acid possesses some pharmacologic properties attractive for anti-leukemia therapy. The present review outlines the evidence and mechanism of ascorbic acid in leukemogenesis and its therapeutic potential in AML. With the growing evidence derived from the literature on situations in which the use of ascorbate may be beneficial in vitro and in vivo, we will finally discuss how these insights could be included into the rational design of future clinical trials.
Collapse
Affiliation(s)
- S. Travaglini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - C. Gurnari
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, United States
| | - S. Antonelli
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - G. Silvestrini
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - N. I. Noguera
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Neuro-Oncohematology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - T. Ottone
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Neuro-Oncohematology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - M. T. Voso
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Neuro-Oncohematology Unit, IRCCS Fondazione Santa Lucia, Rome, Italy
- *Correspondence: M. T. Voso,
| |
Collapse
|
10
|
Chen HC, Tang HH, Hsu WH, Wu SY, Cheng WH, Wang BY, Su CL. Vulnerability of Triple-Negative Breast Cancer to Saponin Formosanin C-Induced Ferroptosis. Antioxidants (Basel) 2022; 11:antiox11020298. [PMID: 35204181 PMCID: PMC8868405 DOI: 10.3390/antiox11020298] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/27/2022] [Accepted: 01/30/2022] [Indexed: 01/25/2023] Open
Abstract
Targeting ferritin via autophagy (ferritinophagy) to induce ferroptosis, an iron- and reactive oxygen species (ROS)-dependent cell death, provides novel strategies for cancer therapy. Using a ferroptosis-specific inhibitor and iron chelator, the vulnerability of triple-negative breast cancer (TNBC) MDA-MB-231 cells to ferroptosis was identified and compared to that of luminal A MCF-7 cells. Saponin formosanin C (FC) was revealed as a potent ferroptosis inducer characterized by superior induction in cytosolic and lipid ROS formation as well as GPX4 depletion in MDA-MB-231 cells. The FC-induced ferroptosis was paralleled by downregulation of ferroportin and xCT expressions. Immunoprecipitation and electron microscopy demonstrated the involvement of ferritinophagy in FC-treated MDA-MB-231 cells. The association of FC with ferroptosis was strengthened by the results that observed an enriched pathway with differentially expressed genes from FC-treated cells. FC sensitized cisplatin-induced ferroptosis in MDA-MB-231 cells. Through integrated analysis of differentially expressed genes and pathways using the METABRIC patients’ database, we confirmed that autophagy and ferroptosis were discrepant between TNBC and luminal A and that TNBC was hypersensitive to ferroptosis. Our data suggest a therapeutic strategy by ferroptosis against TNBC, an aggressive subtype with a poor prognosis.
Collapse
Affiliation(s)
- Hsin-Chih Chen
- Department of Human Development and Family Studies, National Taiwan Normal University, Taipei 106, Taiwan;
| | - Han-Hsuan Tang
- Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei 116, Taiwan; (H.-H.T.); (W.-H.H.); (S.-Y.W.); (B.-Y.W.)
- Genome and Systems Biology Degree Program, Academia Sinica and National Taiwan University, Taipei 106, Taiwan
| | - Wei-Hsiang Hsu
- Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei 116, Taiwan; (H.-H.T.); (W.-H.H.); (S.-Y.W.); (B.-Y.W.)
| | - Shan-Ying Wu
- Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei 116, Taiwan; (H.-H.T.); (W.-H.H.); (S.-Y.W.); (B.-Y.W.)
- Department of Microbiology and Immunology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Wen-Hsing Cheng
- Department of Food Science, Nutrition, and Health Promotion, Mississippi State University, Starkville, MS 39762, USA;
| | - Bao-Yuan Wang
- Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei 116, Taiwan; (H.-H.T.); (W.-H.H.); (S.-Y.W.); (B.-Y.W.)
| | - Chun-Li Su
- Department of Human Development and Family Studies, National Taiwan Normal University, Taipei 106, Taiwan;
- Graduate Program of Nutrition Science, School of Life Science, National Taiwan Normal University, Taipei 116, Taiwan; (H.-H.T.); (W.-H.H.); (S.-Y.W.); (B.-Y.W.)
- Correspondence: ; Tel.: +886-2-7749-1436; Fax: +886-2-2931-2904
| |
Collapse
|
11
|
Abstract
Significance: Vitamin C (ascorbate), in regard to its effectiveness against malignancies, has had a controversial history in cancer treatment. It has been shown that in vitro and in vivo anticancer efficacy of ascorbate relies on its pro-oxidant effect mainly from an increased generation of reactive oxygen species (ROS). A growing understanding of its anticancer activities and pharmacokinetic properties has prompted scientists to re-evaluate the significance of ascorbate in cancer treatment. Recent Advances: A recent resurge in ascorbate research emerged after discovering that, at high doses, ascorbate preferentially kills Kirsten-Ras (K-ras)- and B-raf oncogene (BRAF)-mutant cancer cells. In addition, some of the main hallmarks of cancer cells, such as redox homeostasis and oxygen-sensing regulation (through inhibition of hypoxia-inducible factor-1 alpha [HIF-1α] activity), are affected by vitamin C. Critical Issues: Currently, there is no clear consensus from the literature in regard to the beneficial effects of antioxidants. Results from both human and animal studies provide no clear evidence about the benefit of antioxidant treatment in preventing or suppressing cancer development. Since pro-oxidants may affect both normal and tumor cells, the extremely low toxicity of ascorbate represents a main advantage. This guarantees the safe inclusion of ascorbate in clinical protocols to treat cancer patients. Future Directions: Current research could focus on elucidating the wide array of reactions between ascorbate and reactive species, namely ROS, reactive nitrogen species as well as reactive sulfide species, and their intracellular molecular targets. Unraveling these mechanisms could allow researchers to assess what could be the optimal combination of ascorbate with standard treatments.
Collapse
Affiliation(s)
- Christophe Glorieux
- Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Guangzhou, P. R. China
| | - Pedro Buc Calderon
- Química y Farmacia, Facultad de Ciencias de la Salud, Universidad Arturo Prat, Iquique, Chile.,Research Group in Metabolism and Nutrition, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
12
|
Guo H, Nomoto T, Muttaqien SE, Sun X, Komoto K, Matsui M, Miura Y, Nishiyama N. Polymeric Iron Chelators Enhancing Pro-Oxidant Antitumor Efficacy of Vitamin C by Inhibiting the Extracellular Fenton Reaction. Mol Pharm 2021; 18:4475-4485. [PMID: 34726400 DOI: 10.1021/acs.molpharmaceut.1c00673] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Intravenously injected high-dose vitamin C (VC) induces extracellular H2O2, which can penetrate into the tumor cells and suppress tumor growth. However, extracellular labile iron ions in the tumor decompose H2O2 via the Fenton reaction, limiting the therapeutic effect. In this regard, we recently developed a polymeric iron chelator that can inactivate the intratumoral labile iron ions. Here, we examined the effect of our polymeric iron chelator on the high-dose VC therapy in in vitro and in vivo. In the in vitro study, the polymeric iron chelator could inactivate the extracellular labile iron ions and prevent the unfavorable decomposition of VC-induced H2O2, augmenting pro-oxidative damage to DNA and inducing apoptosis in cultured cancer cells. Even in the in vivo study, the polymeric iron chelator significantly improved the antitumor effect of VC in subcutaneous DLD-1 and CT26 tumors in mice, while conventional iron chelators could not. This work indicates the importance of modulating tumor-associated iron ions in the high-dose VC therapy and should contribute to a better understanding of its mechanism.
Collapse
Affiliation(s)
- Haochen Guo
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Takahiro Nomoto
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Sjaikhurrizal El Muttaqien
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Center for Pharmaceutical and Medical Technology, Agency for the Assessment and Application of Technology (BPPT), LAPTIAB I, PUSPITEK, Serpong, Banten 15314, Indonesia
| | - Xiaohang Sun
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Kana Komoto
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Makoto Matsui
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Yutaka Miura
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan
| | - Nobuhiro Nishiyama
- Laboratory for Chemistry and Life Science, Institute of Innovative Research, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Department of Life Science and Technology, School of Life Science and Technology, Tokyo Institute of Technology, 4259 Nagatsuta-cho, Midori-ku, Yokohama, Kanagawa 226-8503, Japan.,Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa 210-0821, Japan
| |
Collapse
|
13
|
Böttger F, Vallés-Martí A, Cahn L, Jimenez CR. High-dose intravenous vitamin C, a promising multi-targeting agent in the treatment of cancer. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2021; 40:343. [PMID: 34717701 PMCID: PMC8557029 DOI: 10.1186/s13046-021-02134-y] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 10/07/2021] [Indexed: 12/21/2022]
Abstract
Mounting evidence indicates that vitamin C has the potential to be a potent anti-cancer agent when administered intravenously and in high doses (high-dose IVC). Early phase clinical trials have confirmed safety and indicated efficacy of IVC in eradicating tumour cells of various cancer types. In recent years, the multi-targeting effects of vitamin C were unravelled, demonstrating a role as cancer-specific, pro-oxidative cytotoxic agent, anti-cancer epigenetic regulator and immune modulator, reversing epithelial-to-mesenchymal transition, inhibiting hypoxia and oncogenic kinase signalling and boosting immune response. Moreover, high-dose IVC is powerful as an adjuvant treatment for cancer, acting synergistically with many standard (chemo-) therapies, as well as a method for mitigating the toxic side-effects of chemotherapy. Despite the rationale and ample evidence, strong clinical data and phase III studies are lacking. Therefore, there is a need for more extensive awareness of the use of this highly promising, non-toxic cancer treatment in the clinical setting. In this review, we provide an elaborate overview of pre-clinical and clinical studies using high-dose IVC as anti-cancer agent, as well as a detailed evaluation of the main known molecular mechanisms involved. A special focus is put on global molecular profiling studies in this respect. In addition, an outlook on future implications of high-dose vitamin C in cancer treatment is presented and recommendations for further research are discussed.
Collapse
Affiliation(s)
- Franziska Böttger
- Department of Medical Oncology, Cancer Center Amsterdam, OncoProteomics Laboratory, Amsterdam UMC, Location VU University Medical Center, 1081 HV, Amsterdam, the Netherlands
| | - Andrea Vallés-Martí
- Department of Medical Oncology, Cancer Center Amsterdam, OncoProteomics Laboratory, Amsterdam UMC, Location VU University Medical Center, 1081 HV, Amsterdam, the Netherlands
| | - Loraine Cahn
- Department of Medical Oncology, Cancer Center Amsterdam, OncoProteomics Laboratory, Amsterdam UMC, Location VU University Medical Center, 1081 HV, Amsterdam, the Netherlands
| | - Connie R Jimenez
- Department of Medical Oncology, Cancer Center Amsterdam, OncoProteomics Laboratory, Amsterdam UMC, Location VU University Medical Center, 1081 HV, Amsterdam, the Netherlands.
| |
Collapse
|
14
|
Chen L, Wu X, Yu H, Wu L, Wang Q, Zhang J, Liu X, Li Z, Yang XF. An Edaravone-Guided Design of a Rhodamine-Based Turn-on Fluorescent Probe for Detecting Hydroxyl Radicals in Living Systems. Anal Chem 2021; 93:14343-14350. [PMID: 34643369 DOI: 10.1021/acs.analchem.1c03877] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The hydroxyl radical (·OH), one of the reactive oxygen species (ROS) in biosystems, is found to be involved in many physiological and pathological processes. However, specifically detecting endogenous ·OH remains an outstanding challenge owing to the high reactivity and short lifetime of this radical. Herein, inspired by the scavenging mechanism of a neuroprotective drug edaravone toward ·OH, we developed a new ·OH-specific fluorescent probe RH-EDA. RH-EDA is a hybrid of rhodamine and edaravone and exploits a ·OH-specific 3-methyl-pyrazolone moiety to control its fluorescence behavior. RH-EDA itself is almost nonfluorescent in physiological conditions, which was attributed to the formation of a twisted intramolecular charge transfer (TICT) state upon photoexcitation and the acylation of its rhodamine nitrogen at the 3' position. However, upon a treatment with ·OH, its edaravone subunit was converted to the corresponding 2-oxo-3-(phenylhydrazono)-butanoic acid (OPB) derivative (to afford RH-OPB), thus leading to a significant fluorescence increase (ca. 195-fold). RH-EDA shows a high sensitivity and selectivity to ·OH without interference from other ROS. RH-EDA has been utilized for imaging endogenous ·OH production in living cells and zebrafishes under different stimuli. Moreover, RH-EDA allows a high-contrast discrimination of cancer cells from normal ones by monitoring their different ·OH levels upon stimulation with β-Lapachone (β-Lap), an effective ROS-generating anticancer therapeutic agent. The present study provides a promising methodology for the construction of probes through a drug-guided approach.
Collapse
Affiliation(s)
- Liqin Chen
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry & Materials Science, Northwest University, Xi'an 710127, P. R. China
| | - Xia Wu
- Singapore University of Technology and Design, 8 Somapah Road, Singapore 487372, Singapore
| | - Hanjie Yu
- College of Life Sciences, Northwest University, Xi'an 710069, P. R. China
| | - Lei Wu
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry & Materials Science, Northwest University, Xi'an 710127, P. R. China
| | - Qin Wang
- School of Chemistry and Environment Science, Shaanxi University of Technology, Hanzhong 723000, Shaanxi, P. R. China
| | - Jianjian Zhang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry & Materials Science, Northwest University, Xi'an 710127, P. R. China
| | - Xiaogang Liu
- Singapore University of Technology and Design, 8 Somapah Road, Singapore 487372, Singapore
| | - Zheng Li
- College of Life Sciences, Northwest University, Xi'an 710069, P. R. China
| | - Xiao-Feng Yang
- Key Laboratory of Synthetic and Natural Functional Molecule Chemistry of Ministry of Education, College of Chemistry & Materials Science, Northwest University, Xi'an 710127, P. R. China
| |
Collapse
|
15
|
Halliwell B. Commentary on "Ascorbate kills breast cancer cells by rewiring metabolism via redox imbalance and energy crisis" by Ghanem et al. [Free Radic. Biol. Med. 163 (2021) 196-209]. Free Radic Biol Med 2021; 171:124-125. [PMID: 33964397 DOI: 10.1016/j.freeradbiomed.2021.04.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/19/2021] [Accepted: 04/21/2021] [Indexed: 11/25/2022]
Affiliation(s)
- Barry Halliwell
- National University of Singapore, Department of Biochemistry, Centre for Life Sciences, #05-01A, 28 Medical Drive, 117456, Singapore.
| |
Collapse
|
16
|
Catalase Modulates the Radio-Sensitization of Pancreatic Cancer Cells by Pharmacological Ascorbate. Antioxidants (Basel) 2021; 10:antiox10040614. [PMID: 33923601 PMCID: PMC8073689 DOI: 10.3390/antiox10040614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 01/24/2023] Open
Abstract
Pancreatic cancer cells (PDACs) are more susceptible to an oxidative insult than normal cells, resulting in greater cytotoxicity upon exposure to agents that increase pro-oxidant levels. Pharmacological ascorbate (P-AscH-), i.e., large amounts given intravenously (IV), generates significant fluxes of hydrogen peroxide (H2O2), resulting in the killing of PDACs but not normal cells. Recent studies have demonstrated that P-AscH- radio-sensitizes PDAC but is a radioprotector to normal cells and tissues. Several mechanisms have been hypothesized to explain the dual roles of P-AscH- in radiation-induced toxicity including the activation of nuclear factor-erythroid 2-related factor 2 (Nrf2), RelB, as well as changes in bioenergetic profiles. We have found that P-AscH- in conjunction with radiation increases Nrf2 in both cancer cells and normal cells. Although P-AscH- with radiation decreases RelB in cancer cells vs. normal cells, the knockout of RelB does not radio-sensitize PDACs. Cellular bioenergetic profiles demonstrate that P-AscH- with radiation increases the ATP demand/production rate (glycolytic and oxidative phosphorylation) in both PDACs and normal cells. Knocking out catalase results in P-AscH- radio-sensitization in PDACs. In a phase I trial where P-AscH- was included as an adjuvant to the standard of care, short-term survivors had higher catalase levels in tumor tissue, compared to long-term survivors. These data suggest that P-AscH- radio-sensitizes PDACs through increased peroxide flux. Catalase levels could be a possible indicator for how tumors will respond to P-AscH-.
Collapse
|
17
|
Yin M, Liu Y, Chen Y. Iron metabolism: an emerging therapeutic target underlying the anti-cancer effect of quercetin. Free Radic Res 2021; 55:296-303. [PMID: 33818251 DOI: 10.1080/10715762.2021.1898604] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Iron, an essential micronutrient for all kinds of cells, is essential for the balance of body internal environment. Notably, cancer cells exhibit a strong dependence on iron and require a large amount of iron for proliferation. A growing number of studies suggested that iron metabolism imbalance and subsequent excess iron accumulation are closely related to the occurrence and progression of cancer. Precisely, excess iron promotes the development of cancer due to the pro-oxidative nature of iron and its damaging effects on DNA. Simultaneously, tumor cells acquire large amounts of iron to maintain rapid growth and proliferation. Therefore, targeting iron metabolism may provide a new way for the treatment of cancer. Quercetin, a natural flavonoid, has long been regarded as potential drug for cancer treatments owing to its anti-inflammatory, antioxidant and anti-tumor effects. It is proven that quercetin possesses a high iron-chelating capacity, depriving cancer cells of iron or altering iron metabolism. Herein, we conduct a review on the mechanisms of iron imbalance in tumors and the role of quercetin in iron chelation, which will provide insight into the potential for quercetin as an anti-cancer drug.
Collapse
Affiliation(s)
- Mingming Yin
- College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, PR China
| | - Yongping Liu
- School of Medicine, Hunan University of Chinese Medicine, Changsha, PR China
| | - Yi Chen
- School of Medicine, Hunan University of Chinese Medicine, Changsha, PR China
| |
Collapse
|
18
|
High-Dose Vitamin C: Preclinical Evidence for Tailoring Treatment in Cancer Patients. Cancers (Basel) 2021; 13:cancers13061428. [PMID: 33804775 PMCID: PMC8003833 DOI: 10.3390/cancers13061428] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/02/2021] [Accepted: 03/16/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Vitamin C is an indispensable micronutrient in the human diet due to the multiple functions it carries out in the body. Reports of clinical studies have indicated that, when administered at high dosage by the intravenous route, vitamin C may exert beneficial antitumor effects in patients with advanced stage cancers, including those refractory to previous treatment with chemotherapy. The aim of this article is to provide an overview of the current scientific evidence concerning the different mechanisms of action by which high-dose vitamin C may kill tumor cells. A special focus will be given to those mechanisms that provide the rationale basis for tailoring vitamin C treatment according to specific molecular alterations present in the tumor and for the selection of the most appropriate companion drugs. Abstract High-dose vitamin C has been proposed as a potential therapeutic approach for patients with advanced tumors who failed previous treatment with chemotherapy. Due to vitamin C complex pharmacokinetics, only intravenous administration allows reaching sufficiently high plasma concentrations required for most of the antitumor effects observed in preclinical studies (>0.250 mM). Moreover, vitamin C entry into cells is tightly regulated by SVCT and GLUT transporters, and is cell type-dependent. Importantly, besides its well-recognized pro-oxidant effects, vitamin C modulates TET enzymes promoting DNA demethylation and acts as cofactor of HIF hydroxylases, whose activity is required for HIF-1α proteasomal degradation. Furthermore, at pharmacological concentrations lower than those required for its pro-oxidant activity (<1 mM), vitamin C in specific genetic contexts may alter the DNA damage response by increasing 5-hydroxymethylcytosine levels. These more recently described vitamin C mechanisms offer new treatment opportunities for tumors with specific molecular defects (e.g., HIF-1α over-expression or TET2, IDH1/2, and WT1 alterations). Moreover, vitamin C action at DNA levels may provide the rationale basis for combination therapies with PARP inhibitors and hypomethylating agents. This review outlines the pharmacokinetic and pharmacodynamic properties of vitamin C to be taken into account in designing clinical studies that evaluate its potential use as anticancer agent.
Collapse
|
19
|
Carroll RS, Buettner GR, Cullen JJ. Pharmacological ascorbate and use in pancreatic cancer. Cancer 2021. [DOI: 10.1016/b978-0-12-819547-5.00046-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
20
|
New promising developments for potential therapeutic applications of high-dose ascorbate as an anticancer drug. Hematol Oncol Stem Cell Ther 2020; 14:179-191. [PMID: 33278349 DOI: 10.1016/j.hemonc.2020.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 11/11/2020] [Indexed: 12/25/2022] Open
Abstract
Vitamin C (ascorbate) is an essential dietary requirement, with fundamental redox, anti-oxidant functions at physiologic concentrations. Vitamin C is a cofactor for Fe2+ and 2-oxoglutarate-dependent dioxygenases, englobing large families of enzymes, including also epigenetic regulators of DNA and histone methylation. Importantly, vitamin C is involved in the control of the activity of TET (ten-eleven translocation) enzymes, key epigenetic regulators. For this spectrum of activities, often involving pathways deregulated in cancer cells, vitamin C possesses some pharmacologic activities that can be exploited in anticancer therapy. In particular, the capacity of pharmacological doses of vitamin C to target redox imbalance and to rescue deregulated epigenetic program observed in some cancer cells represents a consistent therapeutic potentiality. Several recent studies have identified some cancer subsets that could benefit from the pharmacological activities of vitamin C. The identification of these potentially responsive patients will help to carefully define controlled clinical trials aiming to evaluate the anticancer activity of Vitamin C.
Collapse
|
21
|
Cushing CM, Petronek MS, Bodeker KL, Vollstedt S, Brown HA, Opat E, Hollenbeck NJ, Shanks T, Berg DJ, Smith BJ, Smith MC, Monga V, Furqan M, Howard MA, Greenlee JD, Mapuskar KA, St-Aubin J, Flynn RT, Cullen JJ, Buettner GR, Spitz DR, Buatti JM, Allen BG, Magnotta VA. Magnetic resonance imaging (MRI) of pharmacological ascorbate-induced iron redox state as a biomarker in subjects undergoing radio-chemotherapy. Redox Biol 2020; 38:101804. [PMID: 33260088 PMCID: PMC7708874 DOI: 10.1016/j.redox.2020.101804] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 10/29/2020] [Accepted: 11/13/2020] [Indexed: 12/14/2022] Open
Abstract
Pharmacological ascorbate (P-AscH-) combined with standard of care (SOC) radiation and temozolomide is being evaluated in a phase 2 clinical trial (NCT02344355) in the treatment of glioblastoma (GBM). Previously published data demonstrated that paramagnetic iron (Fe3+) catalyzes ascorbate's oxidation to form diamagnetic iron (Fe2+). Because paramagnetic Fe3+ may influence relaxation times observed in MR imaging, quantitative MR imaging of P-AscH--induced changes in redox-active Fe was assessed as a biomarker for therapy response. Gel phantoms containing either Fe3+ or Fe2+ were imaged with T2* and quantitative susceptibility mapping (QSM). Fifteen subjects receiving P-AscH- plus SOC underwent T2* and QSM imaging four weeks into treatment. Subjects were scanned: pre-P-AscH- infusion, post-P-AscH- infusion, and post-radiation (3-4 h between scans). Changes in T2* and QSM relaxation times in tumor and normal tissue were calculated and compared to changes in Fe3+ and Fe2+ gel phantoms. A GBM mouse model was used to study the relationship between the imaging findings and the labile iron pool. Phantoms containing Fe3+ demonstrated detectable changes in T2* and QSM relaxation times relative to Fe2+ phantoms. Compared to pre-P-AscH-, GBM T2* and QSM imaging were significantly changed post-P-AscH- infusion consistent with conversion of Fe3+ to Fe2+. No significant changes in T2* or QSM were observed in normal brain tissue. There was moderate concordance between T2* and QSM changes in both progression free survival and overall survival. The GBM mouse model showed similar results with P-AscH- inducing greater changes in tumor labile iron pools compared to the normal tissue. CONCLUSIONS: T2* and QSM MR-imaging responses are consistent with P-AscH- reducing Fe3+ to Fe2+, selectively in GBM tumor volumes and represent a potential biomarker of response. This study is the first application using MR imaging in humans to measure P-AscH--induced changes in redox-active iron.
Collapse
Affiliation(s)
- Cameron M Cushing
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, USA
| | - Michael S Petronek
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, USA
| | - Kellie L Bodeker
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, USA
| | - Sandy Vollstedt
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, USA
| | - Heather A Brown
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, USA
| | - Emyleigh Opat
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, USA
| | - Nancy J Hollenbeck
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, USA
| | - Thomas Shanks
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, USA
| | - Daniel J Berg
- Division of Hematology and Oncology, Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa Hospitals & Clinics, Iowa City, IA, USA
| | - Brian J Smith
- Department of Biostatistics, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA, USA
| | - Mark C Smith
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, USA
| | - Varun Monga
- Division of Hematology and Oncology, Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa Hospitals & Clinics, Iowa City, IA, USA
| | - Muhammad Furqan
- Division of Hematology and Oncology, Department of Internal Medicine, Holden Comprehensive Cancer Center, University of Iowa Hospitals & Clinics, Iowa City, IA, USA
| | - Matthew A Howard
- Department of Neurosurgery, University of Iowa Hospitals & Clinics, Iowa City, IA, USA
| | - Jeremy D Greenlee
- Department of Neurosurgery, University of Iowa Hospitals & Clinics, Iowa City, IA, USA
| | - Kranti A Mapuskar
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, USA
| | - Joel St-Aubin
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, USA
| | - Ryan T Flynn
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, USA
| | - Joseph J Cullen
- Department of Surgery, University of Iowa College of Medicine, Iowa City, IA, USA; Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, IA, USA; Holden Comprehensive Cancer Center, Iowa City, IA, USA; Veterans Affairs Medical Center, Iowa City, IA, USA
| | - Garry R Buettner
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, USA
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, USA
| | - John M Buatti
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, USA
| | - Bryan G Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, USA.
| | - Vincent A Magnotta
- Department of Radiology, Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, USA.
| |
Collapse
|
22
|
Zhao Y, Li J, Guo W, Li H, Lei L. Periodontitis-level butyrate-induced ferroptosis in periodontal ligament fibroblasts by activation of ferritinophagy. Cell Death Discov 2020; 6:119. [PMID: 33298848 PMCID: PMC7655826 DOI: 10.1038/s41420-020-00356-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 10/08/2020] [Accepted: 10/21/2020] [Indexed: 02/06/2023] Open
Abstract
Loss of periodontal ligament fibroblasts (PDLFs) is one critical issue for regenerating lost periodontal tissues. A wide variety of regulated cell death pathways, such as apoptosis, pyroptosis, and necroptosis have been proposed in the periodontitis development. The aim of the present study was to explore whether long-term periodontitis-level butyrate may trigger ferroptosis, a newly characterized iron-dependent regulated cell death in PDLFs. Here, we showed that long-term treatment of butyrate, an important short-chain fatty acid in the periodontal pocket, induces the cargo receptor nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy and ferroptosis in PDLFs. Butyrate-induced iron accumulation, reactive oxygen species (ROS) generation, glutathione depletion and lipid peroxidation in PDLFs, and the butyrate-induced ferroptosis can be blocked by the lipid peroxide scavenger ferrostatin-1. The NCOA4-mediated ferritinophagy is dependent on p38/hypoxia inducible factor-1α (HIF-1α) pathway activation as well as Bromodomain-containing protein (BRD) 4 and cyclin-dependent kinase 9 (CDK9) coordination. These lines of evidence provide a new mechanistic insight into the mechanism of loss of PDLFs during periodontitis development, showing that periodontitis-level butyrate disrupted iron homeostasis by activation of NCOA4-mediated ferritinophagy, leading to ferroptosis in PDLFs.
Collapse
Affiliation(s)
- Yunhe Zhao
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 210008, Nanjing, China.,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, 210008, Nanjing, China
| | - Jiao Li
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 210008, Nanjing, China
| | - Wei Guo
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 210008, Nanjing, China.,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, 210008, Nanjing, China
| | - Houxuan Li
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 210008, Nanjing, China.
| | - Lang Lei
- Nanjing Stomatological Hospital, Medical School of Nanjing University, 210008, Nanjing, China. .,Central Laboratory of Stomatology, Nanjing Stomatological Hospital, Medical School of Nanjing University, 210008, Nanjing, China.
| |
Collapse
|
23
|
Dynasore Blocks Ferroptosis through Combined Modulation of Iron Uptake and Inhibition of Mitochondrial Respiration. Cells 2020; 9:cells9102259. [PMID: 33050207 PMCID: PMC7650611 DOI: 10.3390/cells9102259] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/25/2020] [Accepted: 10/03/2020] [Indexed: 12/20/2022] Open
Abstract
Ferroptosis is a form of regulated necrosis characterized by a chain-reaction of detrimental membrane lipid peroxidation following collapse of glutathione peroxidase 4 (Gpx4) activity. This lipid peroxidation is catalyzed by labile ferric iron. Therefore, iron import mediated via transferrin receptors and both, enzymatic and non-enzymatic iron-dependent radical formation are crucial prerequisites for the execution of ferroptosis. Intriguingly, the dynamin inhibitor dynasore, which has been shown to block transferrin receptor endocytosis, can protect from ischemia/reperfusion injury as well as neuronal cell death following spinal cord injury. Yet, it is unknown how dynasore exerts these cell death-protective effects. Using small interfering RNA suppression, lipid reactive oxygen species (ROS), iron tracers and bona fide inducers of ferroptosis, we find that dynasore treatment in lung adenocarcinoma and neuronal cell lines strongly protects these from ferroptosis. Surprisingly, while the dynasore targets dynamin 1 and 2 promote extracellular iron uptake, their silencing was not sufficient to block ferroptosis suggesting that this route of extracellular iron uptake is dispensable for acute induction of ferroptosis and dynasore must have an additional off-target activity mediating full ferroptosis protection. Instead, in intact cells, dynasore inhibited mitochondrial respiration and thereby mitochondrial ROS production which can feed into detrimental lipid peroxidation and ferroptotic cell death in the presence of labile iron. In addition, in cell free systems, dynasore showed radical scavenger properties and acted as a broadly active antioxidant which is superior to N-acetylcysteine (NAC) in blocking ferroptosis. Thus, dynasore can function as a highly active inhibitor of ROS-driven types of cell death via combined modulation of the iron pool and inhibition of general ROS by simultaneously blocking two routes required for ROS and lipid-ROS driven cell death, respectively. These data have important implications for the interpretation of studies observing tissue-protective effects of this dynamin inhibitor as well as raise awareness that off-target ROS scavenging activities of small molecules used to interrogate the ferroptosis pathway should be taken into consideration.
Collapse
|
24
|
|
25
|
Zhang S, Cao Y, Yang Q. Transferrin receptor 1 levels at the cell surface influence the susceptibility of newborn piglets to PEDV infection. PLoS Pathog 2020; 16:e1008682. [PMID: 32730327 PMCID: PMC7419007 DOI: 10.1371/journal.ppat.1008682] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 08/11/2020] [Accepted: 06/04/2020] [Indexed: 12/16/2022] Open
Abstract
Porcine epidemic diarrhea virus (PEDV) mainly infects the intestinal epithelial cells of newborn piglets causing acute, severe atrophic enteritis. The underlying mechanisms of PEDV infection and the reasons why newborn piglets are more susceptible than older pigs remain incompletely understood. Iron deficiency is common in newborn piglets. Here we found that high levels of transferrin receptor 1 (TfR1) distributed in the apical tissue of the intestinal villi of newborns, and intracellular iron levels influence the susceptibility of newborn piglets to PEDV. We show that iron deficiency induced by deferoxamine (DFO, an iron chelating agent) promotes PEDV infection while iron accumulation induced by ferric ammonium citrate (FAC, an iron supplement) impairs PEDV infection in vitro and in vivo. Besides, PEDV infection was inhibited by occluding TfR1 with antibodies or decreasing TfR1 expression. Additionally, PEDV infection was increased in PEDV-resistant Caco-2 and HEK 293T cells over-expressed porcine TfR1. Mechanistically, the PEDV S1 protein interacts with the extracellular region of TfR1 during PEDV entry, promotes TfR1 re-localization and clustering, then activates TfR1 tyrosine phosphorylation mediated by Src kinase, and heightens the internalization of TfR1, thereby promoting PEDV entry. Taken together, these data suggest that the higher expression of TfR1 in the apical tissue of the intestinal villi caused by iron deficiency, accounts for newborn piglets being acutely susceptible to PEDV. Newborn piglets are particularly susceptible to infection by PEDV, with 80–100% dying within days of infection. The reasons for newborns’ acute susceptibility to PEDV infection have not been elucidated clearly. The primarily target of PEDV is the porcine intestinal epithelial cells. Here, we show that the high expression of TfR1 in the apical tissue of intestinal villi in newborn piglets with iron deficiency is a reason for their susceptibility to PEDV. Further, we demonstrate that iron supplementation reduces PEDV infection. This study reveals that iron plays an important role in the susceptibility of newborn piglets to PEDV and provides insights into therapies for the prevention and treatment of PEDV infections.
Collapse
Affiliation(s)
- Shuai Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang, Nanjing, Jiangsu, PR China
| | - Yanan Cao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang, Nanjing, Jiangsu, PR China
| | - Qian Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang, Nanjing, Jiangsu, PR China
| |
Collapse
|
26
|
Delesderrier E, Curioni C, Omena J, Macedo CR, Cople-Rodrigues C, Citelli M. Antioxidant nutrients and hemolysis in sickle cell disease. Clin Chim Acta 2020; 510:381-390. [PMID: 32673671 DOI: 10.1016/j.cca.2020.07.020] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 07/09/2020] [Accepted: 07/09/2020] [Indexed: 01/29/2023]
Abstract
Hemolysis is one of the main pathophysiological characteristics of sickle cell disease (SCD) and might cause or could be the result of oxidative stress. Antioxidants are studied in SCD due to their potential to ensure redox balance and minimize deleterious effects on erythrocyte membranes. The objective of this systematic review was to evaluate the efficacy of antioxidant nutrient supplementation on reducing hemolysis in SCD patients through randomized clinical trials. We conducted our study according to the Preferred Reporting Items for Systematic Reviews and Meta-analyses and the Cochrane Handbook for Systematic Reviews of Interventions investigating whether antioxidants could improve the hemolytic status of SCD patients. This study included 587 articles published until April 2020. We reduced this pool to 12 articles by excluding duplicates, reviews, comments, and studies with non-human subjects. Omega-3 fatty acids, vitamin A, and zinc were the antioxidants that reportedly improved the indirect hemolysis parameters such as hemoglobin, hematocrit, mean corpuscular volume, or red blood cells. High-dose vitamin C and E supplementation worsened hemolysis, causing increased reticulocytes, lactate dehydrogenase, indirect bilirubin, and haptoglobin. More intervention studies especially high-quality controlled randomized clinical trials are needed to investigate the effects of antioxidant nutrients in reducing hemolysis in SCD.
Collapse
Affiliation(s)
- Emília Delesderrier
- Nutrition Institute, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Cíntia Curioni
- Nutrition Institute, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Juliana Omena
- Nutrition Institute, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | | | | | - Marta Citelli
- Nutrition Institute, Rio de Janeiro State University, Rio de Janeiro, Brazil.
| |
Collapse
|
27
|
Zhou L, Zhang L, Wang S, Zhao B, Lv H, Shang P. Labile iron affects pharmacological ascorbate-induced toxicity in osteosarcoma cell lines. Free Radic Res 2020; 54:385-396. [PMID: 32183598 DOI: 10.1080/10715762.2020.1744577] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Vitamin C and iron are both important nutrients for humans and involved in several physiological processes. The biological activities of vitamin C and iron are based on their abilities to accept or donate electrons. Although vitamin C is well known as an excellent electron donor in physiological conditions, it also has pro-oxidant properties, especially with catalytic metal iron. Cancer cells have a higher iron requirement than normal cells, which allows pharmacological ascorbate to kill cancer cells selectively. In this study, we demonstrated that the levels of H2O2 in cells were significantly raised after treated with pharmacological ascorbate, and intracellular labile iron could increase pharmacological ascorbate-mediated oxidative stress by Fenton reaction. Catalytic metal iron plays opposite roles in and outside cells. Intracellular excess labile iron improved ascorbate-induced toxicity, while the excess labile iron in the medium abolished ascorbate-induced toxicity. Fe3+ and Fe2+ have the same effect on ascorbate-induced toxicity, but Fe3+ chelator deferoxamine (DFO) has a profound inhibition effect than Fe2+ chelator 2,2'-bipyridyl (BIP) on ascorbate-induced toxicity. The influence of intracellular labile iron and ascorbate on the ferritin expression may cause selective sensitivity in osteosarcoma cell lines on pharmacological ascorbate. High iron requirement of many cancer cells facilitates pharmacological ascorbate on cancer treatment. In addition, increasing iron content in tumour tissue may be effective strategies to improve the effects of pharmacological ascorbate.
Collapse
Affiliation(s)
- Liangfu Zhou
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Lixiu Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Shenghang Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Bin Zhao
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China
| | - Huanhuan Lv
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, China.,Research and Development Institute, Northwestern Polytechnical University in Shenzhen, Shenzhen, China.,Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environment Biophysics, Northwestern Polytechnical University, Xi'an, China
| | - Peng Shang
- Research and Development Institute, Northwestern Polytechnical University in Shenzhen, Shenzhen, China.,Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environment Biophysics, Northwestern Polytechnical University, Xi'an, China
| |
Collapse
|
28
|
Riedelberger M, Kuchler K. Analyzing the Quenchable Iron Pool in Murine Macrophages by Flow Cytometry. Bio Protoc 2020; 10:e3552. [PMID: 33659526 DOI: 10.21769/bioprotoc.3552] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/08/2019] [Accepted: 10/17/2019] [Indexed: 02/04/2023] Open
Abstract
Tissue-resident macrophages are pivotal for a tightly-regulated iron metabolism at a cellular and systemic level, since subtle iron alterations increase the susceptibility for microbial infections or drive multiple diseases. However, research on cellular iron homeostasis in macrophages remains challenging due to the limited amount of available methods using radioactive 59Fe isotopes or strong iron chelators, which might be inapplicable in certain experimental settings. This protocol describes the analysis of the quenchable iron pool (QIP) in macrophages by loading these cells with exogenous iron-complexes. Thereby, the cytoplasmic iron pool can be determined, since the iron uptake ability of macrophages inversely correlates with intracellular iron levels. Thus, this assay enables the accurate analysis of even minor alterations in cytoplasmic iron fluxes and is applicable in almost every laboratory environment. In addition, the protocol can also be adopted for other immune cell types in vitro and in vivo.
Collapse
Affiliation(s)
- Michael Riedelberger
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, A-1030 Vienna, Austria
| | - Karl Kuchler
- Medical University of Vienna, Center for Medical Biochemistry, Max Perutz Labs Vienna, Campus Vienna Biocenter, A-1030 Vienna, Austria
| |
Collapse
|
29
|
Musser ML, Mahaffey AL, Fath MA, Buettner GR, Wagner BA, Schneider BK, Seo YJ, Mochel JP, Johannes CM. In vitro Cytotoxicity and Pharmacokinetic Evaluation of Pharmacological Ascorbate in Dogs. Front Vet Sci 2019; 6:385. [PMID: 31788483 PMCID: PMC6854015 DOI: 10.3389/fvets.2019.00385] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/21/2019] [Indexed: 11/21/2022] Open
Abstract
Background: High-dose, pharmacological ascorbate (P-AscH-) is preferentially cytotoxic to human cancer cells in vitro. Investigations on the efficacy of P-AscH- as an adjuvant treatment for multiple human cancers are on-going, but has yet to be formally investigated in dogs. The primary objectives of this study were to determine the pharmacokinetic (PK) profile of P-AscH- in healthy Beagle dogs and the effects of P-AscH- on canine osteosarcoma cells in vitro. Methods: Eight purpose-bred, healthy, spayed female Beagle dogs, between 20 and 21 months old, and 8-10 kg were administered two doses of P-AscH- (550 or 2,200 mg/kg) via intravenous infusion over 6 h, on separate days. Plasma ascorbate concentrations were measured at 12 time points during and after infusion for PK analysis using nonlinear mixed-effects (NLME) and non-compartmental analysis (NCA). Clonogenic assays were performed on 2 canine osteosarcoma cell lines (D-17 and OSCA-8) and canine primary dermal fibroblasts after exposure to high concentrations of ascorbate (75 pmoles/cell). Results: Plasma ascorbate levels in the dogs peaked at approximately 10 mM following 2,200 mg/kg and returned to baseline 6-8 h after dosing. Minor adverse effects were seen in two dogs. Ascorbate (75 pmoles/cell) significantly decreased survival in both the osteosarcoma cell lines (D-17 63% SD 0.010, P = 0.005; OSCA-8 50% SD 0.086, P = 0.026), compared to normal fibroblasts (27% SD 0.056). Conclusions: Pharmacological ascorbate is preferentially cytotoxic to canine-derived cancer cells. High levels of ascorbate can be safely administered to dogs. Further studies are needed to determine the effects of P-AscH- on canine patients.
Collapse
Affiliation(s)
- Margaret L. Musser
- Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States
| | - Alyssa L. Mahaffey
- Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States
| | - Melissa A. Fath
- Free Radical and Radiation Biology Program, The University of Iowa, Iowa City, IA, United States
| | - Garry R. Buettner
- Free Radical and Radiation Biology Program, The University of Iowa, Iowa City, IA, United States
| | - Brett A. Wagner
- Free Radical and Radiation Biology Program, The University of Iowa, Iowa City, IA, United States
| | | | - Yeon-Jung Seo
- Biomedical Sciences, Iowa State University, Ames, IA, United States
| | | | - Chad M. Johannes
- Veterinary Clinical Sciences, Iowa State University, Ames, IA, United States
| |
Collapse
|
30
|
Spitz DR, Buettner GR, Limoli CL. Response to letter regarding "An integrated physico-chemical approach for explaining the differential impact of FLASH versus conventional dose rate irradiation on cancer and normal tissue responses". Radiother Oncol 2019; 139:64-65. [PMID: 31427044 DOI: 10.1016/j.radonc.2019.07.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 07/02/2019] [Indexed: 11/16/2022]
Affiliation(s)
- Douglas R Spitz
- Department of Radiation Oncology, The University of Iowa, United States
| | - Garry R Buettner
- Department of Radiation Oncology, The University of Iowa, United States
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, United States
| |
Collapse
|
31
|
Buranasudja V, Doskey CM, Gibson AR, Wagner BA, Du J, Gordon DJ, Koppenhafer SL, Cullen JJ, Buettner GR. Pharmacologic Ascorbate Primes Pancreatic Cancer Cells for Death by Rewiring Cellular Energetics and Inducing DNA Damage. Mol Cancer Res 2019; 17:2102-2114. [PMID: 31337671 DOI: 10.1158/1541-7786.mcr-19-0381] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 06/21/2019] [Accepted: 07/17/2019] [Indexed: 01/21/2023]
Abstract
The clinical potential of pharmacologic ascorbate (P-AscH-; intravenous delivery achieving mmol/L concentrations in blood) as an adjuvant in cancer therapy is being reevaluated. At mmol/L concentrations, P-AscH- is thought to exhibit anticancer activity via generation of a flux of H2O2 in tumors, which leads to oxidative distress. Here, we use cell culture models of pancreatic cancer to examine the effects of P-AscH- on DNA damage, and downstream consequences, including changes in bioenergetics. We have found that the high flux of H2O2 produced by P-AscH- induces DNA damage. In response to this DNA damage, we observed that PARP1 is hyperactivated. Using our unique absolute quantitation, we found that P-AscH- mediated the overactivation of PARP1, which results in consumption of NAD+, and subsequently depletion of ATP leading to mitotic cell death. We have also found that Chk1 plays a major role in the maintenance of genomic integrity following treatment with P-AscH-. Hyperactivation of PARP1 and DNA repair are ATP-consuming processes. Using a Seahorse XF96 analyzer, we demonstrated that the severe decrease in ATP after challenging with P-AscH- is because of increased demand, not changes in the rate of production. Genetic deletion and pharmacologic inhibition of PARP1 preserved both NAD+ and ATP; however, the toxicity of P-AscH- remained. These data indicate that disruption of bioenergetics is a secondary factor in the toxicity of P-AscH-; damage to DNA appears to be the primary factor. IMPLICATIONS: Efforts to leverage P-AscH- in cancer therapy should first focus on DNA damage.
Collapse
Affiliation(s)
- Visarut Buranasudja
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, Iowa
| | - Claire M Doskey
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, Iowa
| | - Adrienne R Gibson
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, Iowa
| | - Brett A Wagner
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, Iowa
| | - Juan Du
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, Iowa.,Department of Surgery, The University of Iowa, Iowa City, Iowa
| | - David J Gordon
- Department of Pediatrics, The University of Iowa, Iowa City, Iowa
| | | | - Joseph J Cullen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, Iowa.,Department of Surgery, The University of Iowa, Iowa City, Iowa.,Veterans Affairs Medical Center, The University of Iowa, Iowa City, Iowa
| | - Garry R Buettner
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, Iowa. .,Free Radical and Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, Iowa
| |
Collapse
|
32
|
Bai X, Ng KKH, Hu JJ, Ye S, Yang D. Small-Molecule-Based Fluorescent Sensors for Selective Detection of Reactive Oxygen Species in Biological Systems. Annu Rev Biochem 2019; 88:605-633. [DOI: 10.1146/annurev-biochem-013118-111754] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Reactive oxygen species (ROS) encompass a collection of intricately linked chemical entities characterized by individually distinct physicochemical properties and biological reactivities. Although excessive ROS generation is well known to underpin disease development, it has become increasingly evident that ROS also play central roles in redox regulation and normal physiology. A major challenge in uncovering the relevant biological mechanisms and deconvoluting the apparently paradoxical roles of distinct ROS in human health and disease lies in the selective and sensitive detection of these transient species in the complex biological milieu. Small-molecule-based fluorescent sensors enable molecular imaging of ROS with great spatial and temporal resolution and have thus been appreciated as excellent tools for aiding discoveries in modern redox biology. We review a selection of state-of-the-art sensors with demonstrated utility in biological systems. By providing a systematic overview based on underlying chemical sensing mechanisms, we wish to highlight the strengths and weaknesses in prior sensor works and propose some guiding principles for the development of future probes.
Collapse
Affiliation(s)
| | | | - Jun Jacob Hu
- Morningside Laboratory for Chemical Biology, Department of Chemistry, The University of Hong Kong, Hong Kong, P. R. China;, , , ,
| | - Sen Ye
- Morningside Laboratory for Chemical Biology, Department of Chemistry, The University of Hong Kong, Hong Kong, P. R. China;, , , ,
| | - Dan Yang
- Morningside Laboratory for Chemical Biology, Department of Chemistry, The University of Hong Kong, Hong Kong, P. R. China;, , , ,
| |
Collapse
|
33
|
Spitz DR, Buettner GR, Petronek MS, St-Aubin JJ, Flynn RT, Waldron TJ, Limoli CL. An integrated physico-chemical approach for explaining the differential impact of FLASH versus conventional dose rate irradiation on cancer and normal tissue responses. Radiother Oncol 2019; 139:23-27. [PMID: 31010709 DOI: 10.1016/j.radonc.2019.03.028] [Citation(s) in RCA: 187] [Impact Index Per Article: 31.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/04/2019] [Accepted: 03/27/2019] [Indexed: 12/22/2022]
Abstract
For decades the field of radiation oncology has sought to improve the therapeutic ratio through innovations in physics, chemistry, and biology. To date, technological advancements in image guided beam delivery techniques have provided clinicians with their best options for improving this critical tool in cancer care. Medical physics has focused on the preferential targeting of tumors while minimizing the collateral dose to the surrounding normal tissues, yielding only incremental progress. However, recent developments involving ultra-high dose rate irradiation termed FLASH radiotherapy (FLASH-RT), that were initiated nearly 50 years ago, have stimulated a renaissance in the field of radiotherapy, long awaiting a breakthrough modality able to enhance therapeutic responses and limit normal tissue injury. Compared to conventional dose rates used clinically (0.1-0.2 Gy/s), FLASH can implement dose rates of electrons or X-rays in excess of 100 Gy/s. The implications of this ultra-fast delivery of dose are significant and need to be re-evaluated to appreciate the fundamental aspects underlying this seemingly unique radiobiology. The capability of FLASH to significantly spare normal tissue complications in multiple animal models, when compared to conventional rates of dose-delivery, while maintaining persistent growth inhibition of select tumor models has generated considerable excitement, as well as skepticism. Based on fundamental principles of radiation physics, radio-chemistry, and tumor vs. normal cell redox metabolism, this article presents a series of testable, biologically relevant hypotheses, which may help rationalize the differential effects of FLASH irradiation observed between normal tissue and tumors.
Collapse
Affiliation(s)
- Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Free Radical Metabolism and Imaging Program, Holden Comprehensive Cancer Center, The University of Iowa, United States.
| | - Garry R Buettner
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Free Radical Metabolism and Imaging Program, Holden Comprehensive Cancer Center, The University of Iowa, United States
| | - Michael S Petronek
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Free Radical Metabolism and Imaging Program, Holden Comprehensive Cancer Center, The University of Iowa, United States
| | - Joël J St-Aubin
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Free Radical Metabolism and Imaging Program, Holden Comprehensive Cancer Center, The University of Iowa, United States
| | - Ryan T Flynn
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Free Radical Metabolism and Imaging Program, Holden Comprehensive Cancer Center, The University of Iowa, United States
| | - Timothy J Waldron
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Free Radical Metabolism and Imaging Program, Holden Comprehensive Cancer Center, The University of Iowa, United States
| | - Charles L Limoli
- Department of Radiation Oncology, University of California, Irvine, United States
| |
Collapse
|
34
|
|
35
|
Worley BL, Kim YS, Mardini J, Zaman R, Leon KE, Vallur PG, Nduwumwami A, Warrick JI, Timmins PF, Kesterson JP, Phaëton R, Lee NY, Walter V, Endres L, Mythreye K, Aird KM, Hempel N. GPx3 supports ovarian cancer progression by manipulating the extracellular redox environment. Redox Biol 2018; 25:101051. [PMID: 30509602 PMCID: PMC6859581 DOI: 10.1016/j.redox.2018.11.009] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 11/13/2018] [Accepted: 11/15/2018] [Indexed: 12/31/2022] Open
Abstract
Ovarian cancer remains the most lethal gynecologic malignancy, and is primarily diagnosed at late stage when considerable metastasis has occurred in the peritoneal cavity. At late stage abdominal cavity ascites accumulation provides a tumor-supporting medium in which cancer cells gain access to growth factors and cytokines that promote survival and metastasis. However, little is known about the redox status of ascites, or whether antioxidant enzymes are required to support ovarian cancer survival during transcoelomic metastasis in this medium. Gene expression cluster analysis of antioxidant enzymes identified two distinct populations of high-grade serous adenocarcinomas (HGSA), the most common ovarian cancer subtype, which specifically separated into clusters based on glutathione peroxidase 3 (GPx3) expression. High GPx3 expression was associated with poorer overall patient survival and increased tumor stage. GPx3 is an extracellular glutathione peroxidase with reported dichotomous roles in cancer. To further examine a potential pro-tumorigenic role of GPx3 in HGSA, stable OVCAR3 GPx3 knock-down cell lines were generated using lentiviral shRNA constructs. Decreased GPx3 expression inhibited clonogenicity and anchorage-independent cell survival. Moreover, GPx3 was necessary for protecting cells from exogenous oxidant insult, as demonstrated by treatment with high dose ascorbate. This cytoprotective effect was shown to be due to GPx3-dependent removal of extracellular H2O2. Importantly, GPx3 was necessary for clonogenic survival when cells were cultured in patient-derived ascites fluid. While oxidation reduction potential (ORP) of malignant ascites was heterogeneous in our patient cohort, and correlated positively with ascites iron content, GPx3 was required for optimal survival regardless of ORP or iron content. Collectively, our data suggest that HGSA ovarian cancers cluster into distinct groups of high and low GPx3 expression. GPx3 is necessary for HGSA ovarian cancer cellular survival in the ascites tumor environment and protects against extracellular sources of oxidative stress, implicating GPx3 as an important adaptation for transcoelomic metastasis. High grade serous ovarian cancers cluster into distinct groups of antioxidant enzyme expression. High GPx3 expression is associated with decreased overall patient survival. GPx3 promotes cell viability by protecting cells from extracellular sources of oxidative stress. GPx3 enhances cell survival in ovarian cancer patient ascites fluid. Malignant ascites oxidation-reduction potential correlates with iron content.
Collapse
Affiliation(s)
- Beth L Worley
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Yeon Soo Kim
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Jennifer Mardini
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Rameez Zaman
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Kelly E Leon
- Department of Molecular and Cellular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Piyushi Gupta Vallur
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Asvelt Nduwumwami
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Joshua I Warrick
- Department of Pathology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | | | - Joshua P Kesterson
- Department of Obstetrics and Gynecology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Rébécca Phaëton
- Department of Obstetrics and Gynecology, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Nam Y Lee
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ, USA
| | - Vonn Walter
- Department of Public Health Sciences, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Biochemistry and Molecular Biology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Lauren Endres
- Department of Biology and Chemistry, SUNY Polytechnic Institute, Utica, NY, USA
| | - Karthikeyan Mythreye
- Department of Chemistry and Biochemistry, University of South Carolina, Columbia, SC, USA
| | - Katherine M Aird
- Department of Molecular and Cellular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Nadine Hempel
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA, USA; Department of Obstetrics and Gynecology, Pennsylvania State University College of Medicine, Hershey, PA, USA.
| |
Collapse
|
36
|
Alexander MS, Wilkes JG, Schroeder SR, Buettner GR, Wagner BA, Du J, Gibson-Corley K, O'Leary BR, Spitz DR, Buatti JM, Berg DJ, Bodeker KL, Vollstedt S, Brown HA, Allen BG, Cullen JJ. Pharmacologic Ascorbate Reduces Radiation-Induced Normal Tissue Toxicity and Enhances Tumor Radiosensitization in Pancreatic Cancer. Cancer Res 2018; 78:6838-6851. [PMID: 30254147 DOI: 10.1158/0008-5472.can-18-1680] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 08/07/2018] [Accepted: 09/20/2018] [Indexed: 12/23/2022]
Abstract
: Chemoradiation therapy is the mainstay for treatment of locally advanced, borderline resectable pancreatic cancer. Pharmacologic ascorbate (P-AscH-, i.e., intravenous infusions of ascorbic acid, vitamin C), but not oral ascorbate, produces high plasma concentrations capable of selective cytotoxicity to tumor cells. In doses achievable in humans, P-AscH- decreases the viability and proliferative capacity of pancreatic cancer via a hydrogen peroxide (H2O2)-mediated mechanism. In this study, we demonstrate that P-AscH- radiosensitizes pancreatic cancer cells but inhibits radiation-induced damage to normal cells. Specifically, radiation-induced decreases in clonogenic survival and double-stranded DNA breaks in tumor cells, but not in normal cells, were enhanced by P-AscH-, while radiation-induced intestinal damage, collagen deposition, and oxidative stress were also reduced with P-AscH- in normal tissue. We also report on our first-in-human phase I trial that infused P-AscH- during the radiotherapy "beam on." Specifically, treatment with P-AscH- increased median overall survival compared with our institutional average (21.7 vs. 12.7 months, P = 0.08) and the E4201 trial (21.7 vs. 11.1 months). Progression-free survival in P-AscH--treated subjects was also greater than our institutional average (13.7 vs. 4.6 months, P < 0.05) and the E4201 trial (6.0 months). Results indicated that P-AscH- in combination with gemcitabine and radiotherapy for locally advanced pancreatic adenocarcinoma is safe and well tolerated with suggestions of efficacy. Because of the potential effect size and minimal toxicity, our findings suggest that investigation of P-AscH- efficacy is warranted in a phase II clinical trial. SIGNIFICANCE: These findings demonstrate that pharmacologic ascorbate enhances pancreatic tumor cell radiation cytotoxicity in addition to offering potential protection from radiation damage in normal surrounding tissue, making it an optimal agent for improving treatment of locally advanced pancreatic adenocarcinoma.
Collapse
Affiliation(s)
- Matthew S Alexander
- Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Justin G Wilkes
- Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Samuel R Schroeder
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Garry R Buettner
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Brett A Wagner
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Juan Du
- Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Katherine Gibson-Corley
- The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Brianne R O'Leary
- Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - John M Buatti
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Daniel J Berg
- The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,Division of Hematology, Oncology, and Blood and Marrow Transplantation, Department of Internal Medicine, The University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Kellie L Bodeker
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Sandy Vollstedt
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Heather A Brown
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Bryan G Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa
| | - Joseph J Cullen
- Department of Surgery, University of Iowa Hospitals and Clinics, Iowa City, Iowa. .,Free Radical and Radiation Biology Program, Department of Radiation Oncology, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,The Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa.,The Veterans' Affairs Medical Center, Iowa City, Iowa
| |
Collapse
|
37
|
Erudaitius D, Mantooth J, Huang A, Soliman J, Doskey CM, Buettner GR, Rodgers VGJ. Calculated cell-specific intracellular hydrogen peroxide concentration: Relevance in cancer cell susceptibility during ascorbate therapy. Free Radic Biol Med 2018; 120:356-367. [PMID: 29601946 PMCID: PMC6160292 DOI: 10.1016/j.freeradbiomed.2018.03.044] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 03/13/2018] [Accepted: 03/25/2018] [Indexed: 01/24/2023]
Abstract
The high extracellular hydrogen peroxide (H2O2) concentrations generated during pharmacological ascorbate (P-AscH-) therapy has been shown to exhibit a high flux into susceptible cancer cells leading to a decrease in clonogenic survival. It is hypothesized that the intracellular H2O2 concentration for susceptibility is independent of cell type and that the variation observed in dosing is associated with differences in the cell-specific overall steady-state intracellular H2O2 concentration values. The steady-state variation in intracellular H2O2 concentration is coupled to a number of cellular specific transport and reaction factors including catalase activity and membrane permeability. Here a lumped-parameter mathematical modeling approach, assuming a catalase-dominant peroxide removal mechanism, is used to calculate intracellular H2O2 concentration for several cell lines. Experimental measurements of critical parameters pertaining to the model are obtained. The cell lines investigated are normal pancreatic cells, H6c7, the pancreatic cancer cell line, MIA PaCa-2 and the glioblastoma cell lines, LN-229, T98G, and U-87; all which vary in susceptibility. The intracellular H2O2 concentration estimates are correlated with the clonogenic surviving fraction for each cell line, in-vitro. The results showed that, despite the fact that the experimental parameters including catalase concentration and plasma membrane permeability demonstrated significant variability across cell lines, the calculated steady-state intracellular to extracellular H2O2 concentration ratio did not vary significantly across cell lines. Thus, the calculated intracellular H2O2 concentration is not unique in characterizing susceptibility. These results imply that, although intracellular H2O2 concentration plays a key role in cellular susceptibility to P-AscH- adjuvant therapy, its overall contribution in a unifying mechanism across cell types is complex.
Collapse
Affiliation(s)
- Dieanira Erudaitius
- Department of Bioengineering, University of California, Riverside, Riverside, CA 92521, USA
| | - Jacqueline Mantooth
- Department of Bioengineering, University of California, Riverside, Riverside, CA 92521, USA
| | - Andrew Huang
- Department of Neuroscience, University of California, Riverside, Riverside, CA 92521, USA
| | - Jesse Soliman
- Department of Bioengineering, University of California, Riverside, Riverside, CA 92521, USA
| | - Claire M Doskey
- Center for Integrative Toxicology, Michigan State University, East Lansing, MI 48823, USA
| | - Garry R Buettner
- Free Radical & Radiation Biology, Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Victor G J Rodgers
- Department of Bioengineering, University of California, Riverside, Riverside, CA 92521, USA.
| |
Collapse
|
38
|
O'Leary BR, Houwen FK, Johnson CL, Allen BG, Mezhir JJ, Berg DJ, Cullen JJ, Spitz DR. Pharmacological Ascorbate as an Adjuvant for Enhancing Radiation-Chemotherapy Responses in Gastric Adenocarcinoma. Radiat Res 2018; 189:456-465. [PMID: 29547353 DOI: 10.1667/rr14978.1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Gastric adenocarcinoma most often presents at an advanced stage and overall five-year survival of ∼30%. Pharmacological ascorbate (high-dose IV ascorbate) has been proposed as a promising nontoxic adjuvant to standard radio-chemotherapies in several cancer types. In the current study, pharmacological ascorbate (0.5-2 m M) caused a dose-dependent decrease (70-85% at 2 m M) in clonogenic survival of gastric adenocarcinoma cells (AGS and MNK-45), but was relatively nontoxic to a small intestinal epithelial nonimmortalized human cell isolate (FHs 74 Int). The addition of pharmacological ascorbate (1 m M) to standard radio-chemotherapies [i.e., 5-FU (5 μ M); cisplatin (0.5 μ M); irinotecan (2.5 μ M); carboplatin (5 μ M); paclitaxel (2-4 n M); and X rays (1.8 Gy)] also potentiated gastric cancer clonogenic cell killing [additional decreases were noted with: ascorbate plus 5-FU/radiation (1%); ascorbate plus cisplatin/irinotecan (9-19%); and ascorbate plus paclitaxel/carboplatin (6-7%)]. The gastric cancer cell toxicity and chemosensitization seen with pharmacological ascorbate was dependent on H2O2 and the presence of catalytic metal ions. In addition, pharmacological ascorbate dosing resulted in a concentration-dependent decrease (64% at 20 m M, P ≤ 0.0001) in cancer cell invasion and migration that was inhibited by catalase. Finally, pharmacological ascorbate significantly increased the overall survival of mice with gastric cancer xenografts when used in combination with paclitaxel, carboplatin and radiation ( P = 0.019). These results demonstrate that pharmacological ascorbate is selectively cytotoxic to gastric adenocarcinoma cells (relative to normal intestinal epithelial cells) by a mechanism involving H2O2 and redox active metal ions. Furthermore, pharmacological ascorbate significantly enhances gastric cancer xenograft responses to radio-chemotherapy as well as inhibiting tumor cell migration and invasiveness. Overall, these results support the hypothesis that pharmacological ascorbate can be used as an adjuvant with standard-of-care radio-chemotherapies for the treatment of gastric adenocarcinomas.
Collapse
Affiliation(s)
| | | | | | | | | | - Daniel J Berg
- c Internal Medicine, Free Radical and Radiation Biology Program, The Holden Comprehensive Cancer Center at the University of Iowa College of Medicine, Iowa City, Iowa
| | - Joseph J Cullen
- Departments of a Surgery.,b Radiation Oncology.,d The Veterans Affairs Medical Center, Iowa City, Iowa
| | | |
Collapse
|
39
|
Heer CD, Davis AB, Riffe DB, Wagner BA, Falls KC, Allen BG, Buettner GR, Beardsley RA, Riley DP, Spitz DR. Superoxide Dismutase Mimetic GC4419 Enhances the Oxidation of Pharmacological Ascorbate and Its Anticancer Effects in an H₂O₂-Dependent Manner. Antioxidants (Basel) 2018; 7:antiox7010018. [PMID: 29351198 PMCID: PMC5789328 DOI: 10.3390/antiox7010018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/12/2018] [Accepted: 01/16/2018] [Indexed: 01/17/2023] Open
Abstract
Lung cancer, together with head and neck cancer, accounts for more than one-fourth of cancer deaths worldwide. New, non-toxic therapeutic approaches are needed. High-dose IV vitamin C (aka, pharmacological ascorbate; P-AscH−) represents a promising adjuvant to radiochemotherapy that exerts its anti-cancer effects via metal-catalyzed oxidation to form H2O2. Mn(III)-porphyrins possessing superoxide dismutase (SOD) mimetic activity have been shown to increase the rate of oxidation of AscH−, enhancing the anti-tumor effects of AscH− in several cancer types. The current study demonstrates that the Mn(II)-containing pentaazamacrocyclic selective SOD mimetic GC4419 may serve as an AscH−/O2•− oxidoreductase as evidenced by the increased rate of oxygen consumption, steady-state concentrations of ascorbate radical, and H2O2 production in complete cell culture media. GC4419, but not CuZnSOD, was shown to significantly enhance the toxicity of AscH− in H1299, SCC25, SQ20B, and Cal27 cancer cell lines. This enhanced cancer cell killing was dependent upon the catalytic activity of the SOD mimetic and the generation of H2O2, as determined using conditional overexpression of catalase in H1299T cells. GC4419 combined with AscH− was also capable of enhancing radiation-induced cancer cell killing. Currently, AscH− and GC4419 are each being tested separately in clinical trials in combination with radiation therapy. Data presented here support the hypothesis that the combination of GC4419 and AscH− may provide an effective means by which to further enhance radiation therapy responses.
Collapse
Affiliation(s)
- Collin D Heer
- 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.
| | - Andrew B Davis
- 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.
| | - David B Riffe
- 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.
| | - Brett 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.
| | - Kelly C Falls
- 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.
| | - Bryan 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.
| | - Garry 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.
| | | | | | - Douglas 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.
| |
Collapse
|
40
|
Sun Y, Pham AN, Waite TD. The effect of vitamin C and iron on dopamine-mediated free radical generation: implications to Parkinson's disease. Dalton Trans 2018; 47:4059-4069. [DOI: 10.1039/c7dt04373b] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
While the application of Asc alone may aggravate the progression of PD in view of the possible peroxidation of Asc bound Fe(ii), a combination therapy of Asc and strong clinically appropriate iron chelator would appear to be a promising direction for the treatment of PD as a result of the enhanced iron chelation and attenuation in oxidative stress and toxicity induced by DA derived quinones.
Collapse
Affiliation(s)
- Yingying Sun
- School of Civil and Environmental Engineering
- The University of New South Wales
- Sydney
- Australia
| | - An Ninh Pham
- School of Civil and Environmental Engineering
- The University of New South Wales
- Sydney
- Australia
| | - T. David Waite
- School of Civil and Environmental Engineering
- The University of New South Wales
- Sydney
- Australia
| |
Collapse
|
41
|
Polireddy K, Dong R, Reed G, Yu J, Chen P, Williamson S, Violet PC, Pessetto Z, Godwin AK, Fan F, Levine M, Drisko JA, Chen Q. High Dose Parenteral Ascorbate Inhibited Pancreatic Cancer Growth and Metastasis: Mechanisms and a Phase I/IIa study. Sci Rep 2017; 7:17188. [PMID: 29215048 PMCID: PMC5719364 DOI: 10.1038/s41598-017-17568-8] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/28/2017] [Indexed: 12/18/2022] Open
Abstract
Pancreatic cancer is among the most lethal cancers with poorly tolerated treatments. There is increasing interest in using high-dose intravenous ascorbate (IVC) in treating this disease partially because of its low toxicity. IVC bypasses bioavailability barriers of oral ingestion, provides pharmacological concentrations in tissues, and exhibits selective cytotoxic effects in cancer cells through peroxide formation. Here, we further revealed its anti-pancreatic cancer mechanisms and conducted a phase I/IIa study to investigate pharmacokinetic interaction between IVC and gemcitabine. Pharmacological ascorbate induced cell death in pancreatic cancer cells with diverse mutational backgrounds. Pharmacological ascorbate depleted cellular NAD+ preferentially in cancer cells versus normal cells, leading to depletion of ATP and robustly increased α-tubulin acetylation in cancer cells. While ATP depletion led to cell death, over-acetylated tubulin led to inhibition of motility and mitosis. Collagen was increased, and cancer cell epithelial-mesenchymal transition (EMT) was inhibited, accompanied with inhibition in metastasis. IVC was safe in patients and showed the possibility to prolong patient survival. There was no interference to gemcitabine pharmacokinetics by IVC administration. Taken together, these data revealed a multi-targeting mechanism of pharmacological ascorbate's anti-cancer action, with minimal toxicity, and provided guidance to design larger definitive trials testing efficacy of IVC in treating advanced pancreatic cancer.
Collapse
Affiliation(s)
- Kishore Polireddy
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Integrative Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Ruochen Dong
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Integrative Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Gregory Reed
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Jun Yu
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Integrative Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Ping Chen
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA
- Integrative Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Stephen Williamson
- Department of Internal Medicine, Hematology and Oncology Division, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Pierre-Christian Violet
- National Institute of Diabetes, Digestive and Kidney Diseases, the National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ziyan Pessetto
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Andrew K Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Fang Fan
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Mark Levine
- National Institute of Diabetes, Digestive and Kidney Diseases, the National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jeanne A Drisko
- Integrative Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| | - Qi Chen
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
- Integrative Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA.
| |
Collapse
|
42
|
Schoenfeld JD, Sibenaller ZA, Mapuskar KA, Bradley MD, Wagner BA, Buettner GR, Monga V, Milhem M, Spitz DR, Allen BG. Redox active metals and H 2O 2 mediate the increased efficacy of pharmacological ascorbate in combination with gemcitabine or radiation in pre-clinical sarcoma models. Redox Biol 2017; 14:417-422. [PMID: 29069637 PMCID: PMC5651553 DOI: 10.1016/j.redox.2017.09.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 08/21/2017] [Accepted: 09/18/2017] [Indexed: 11/15/2022] Open
Abstract
Soft tissue sarcomas are a histologically heterogeneous group of rare mesenchymal cancers for which treatment options leading to increased overall survival have not improved in over two decades. The current study shows that pharmacological ascorbate (systemic high dose vitamin C achieving ≥ 20 mM plasma levels) is a potentially efficacious and easily integrable addition to current standard of care treatment strategies in preclinical models of fibrosarcoma and liposarcoma both in vitro and in vivo. Furthermore, enhanced ascorbate-mediated toxicity and DNA damage in these sarcoma models were found to be dependent upon H2O2 and intracellular labile iron. Together, these data support the hypothesis that pharmacological ascorbate may represent an easily implementable and non-toxic addition to conventional sarcoma therapies based on taking advantage of fundamental differences in cancer cell oxidative metabolism. Ascorbate sensitizes sarcoma cells to radiation- or chemo-therapy by a mechanism involving redox active metal ions and H2O2. Pharmacological ascorbate in combination with radio-chemo-therapy decreases sarcoma disease burden in murine xenografts. Ascorbate-mediated DNA damage is dependent on intracellular redox active iron.
Collapse
Affiliation(s)
- Joshua D Schoenfeld
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Zita A Sibenaller
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Kranti A Mapuskar
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Megan D Bradley
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Brett A Wagner
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Garry R Buettner
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Varun Monga
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Mohammed Milhem
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Bryan G Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States.
| |
Collapse
|
43
|
Mastrangelo D, Pelosi E, Castelli G, Lo-Coco F, Testa U. Mechanisms of anti-cancer effects of ascorbate: Cytotoxic activity and epigenetic modulation. Blood Cells Mol Dis 2017; 69:57-64. [PMID: 28954710 DOI: 10.1016/j.bcmd.2017.09.005] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 09/20/2017] [Indexed: 12/16/2022]
Abstract
Vitamin C (Vit C or Ascorbate) is essential for many fundamental biochemical processes. Vit C is an essential nutrient with redox functions at normal physiologic concentrations. The main physiologic function of this vitamin is related to its capacity to act as a co-factor for a large family of enzymes, collectively known as Fe and 2-oxoglutarate-dependent dioxygenases. It also modulates epigenetic gene expression through the control of TET enzymes activity. Vit C also has several biological properties allowing to restore the deregulated epigenetic response observed in many tumors. High-dose Vit C has been investigated as a treatment for cancer patients since the 1969. Pharmacologic ascorbate acts as a pro-drug for hydrogen peroxide formation (H2O2) and, through this mechanism, kills cancer cells. To achieve high in vivo concentrations, Ascorbate must be injected by i.v. route. Initial clinical studies of Ascorbate cancer treatment have provided encouraging results, not confirmed in subsequent studies. Recent clinical studies using i.v. injection of high-dose Ascorbate have renewed the interest in the field, showing that significant anti-tumor activity. Pre-clinical studies have led to identify tumors sensitive to Ascorbate that could potentially benefit from this treatment either through an epigenetic modulator effect or through tumor killing by oxidative stress.
Collapse
Affiliation(s)
- Domenico Mastrangelo
- Department of Medical, Surgical and Neurological Sciences, University of Siena, Polo Scientifico San Miniato, Siena, Italy
| | - Elvira Pelosi
- Department of Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Germana Castelli
- Department of Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Francesco Lo-Coco
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy; Santa Lucia Foundation, I.R.C.C.S., Via del Fosso di Fiorano, Rome, Italy
| | - Ugo Testa
- Department of Oncology, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| |
Collapse
|
44
|
Brandt KE, Falls KC, Schoenfeld JD, Rodman SN, Gu Z, Zhan F, Cullen JJ, Wagner BA, Buettner GR, Allen BG, Berg DJ, Spitz DR, Fath MA. Augmentation of intracellular iron using iron sucrose enhances the toxicity of pharmacological ascorbate in colon cancer cells. Redox Biol 2017; 14:82-87. [PMID: 28886484 PMCID: PMC5591450 DOI: 10.1016/j.redox.2017.08.017] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 08/17/2017] [Accepted: 08/21/2017] [Indexed: 01/20/2023] Open
Abstract
Pharmacological doses (> 1 mM) of ascorbate (a.k.a., vitamin C) have been shown to selectively kill cancer cells through a mechanism that is dependent on the generation of H2O2 at doses that are safely achievable in humans using intravenous administration. The process by which ascorbate oxidizes to form H2O2 is thought to be mediated catalytically by redox active metal ions such as iron (Fe). Because intravenous iron sucrose is often administered to colon cancer patients to help mitigate anemia, the current study assessed the ability of pharmacological ascorbate to kill colon cancer cells in the presence and absence of iron sucrose. In vitro survival assays showed that 10 mM ascorbate exposure (2 h) clonogenically inactivated 40–80% of exponentially growing colon cancer cell lines (HCT116 and HT29). When the H2O2 scavenging enzyme, catalase, was added to the media, or conditionally over-expressed using a doxycycline inducible vector, the toxicity of pharmacological ascorbate was significantly blunted. When colon cancer cells were treated in the presence or absence of 250 µM iron sucrose, then rinsed, and treated with 10 mM ascorbate, the cells demonstrated increased levels of labile iron that resulted in significantly increased clonogenic cell killing, compared to pharmacological ascorbate alone. Interestingly, when colon cancer cells were treated with iron sucrose for 1 h and then 10 mM ascorbate was added to the media in the continued presence of iron sucrose, there was no enhancement of toxicity despite similar increases in intracellular labile iron. The combination of iron chelators, deferoxamine and diethylenetriaminepentaacetic acid, significantly inhibited the toxicity of either ascorbate alone or ascorbate following iron sucrose. These observations support the hypothesis that increasing intracellular labile iron pools, using iron sucrose, can be used to increase the toxicity of pharmacological ascorbate in human colon cancer cells by a mechanism involving increased generation of H2O2.
Collapse
Affiliation(s)
- Kristin E Brandt
- Free Radical and Radiation Biology Program, Departments of Radiation Oncology, Carver College of Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Kelly C Falls
- Free Radical and Radiation Biology Program, Departments of Radiation Oncology, Carver College of Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Joshua D Schoenfeld
- Free Radical and Radiation Biology Program, Departments of Radiation Oncology, Carver College of Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Samuel N Rodman
- Free Radical and Radiation Biology Program, Departments of Radiation Oncology, Carver College of Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Zhimin Gu
- Department of Internal Medicine, Carver College of Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Fenghuang Zhan
- Department of Internal Medicine, Carver College of Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Joseph J Cullen
- Free Radical and Radiation Biology Program, Departments of Radiation Oncology, Carver College of Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States; Department of Surgery, Carver College of Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Brett A Wagner
- Free Radical and Radiation Biology Program, Departments of Radiation Oncology, Carver College of Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Garry R Buettner
- Free Radical and Radiation Biology Program, Departments of Radiation Oncology, Carver College of Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Bryan G Allen
- Free Radical and Radiation Biology Program, Departments of Radiation Oncology, Carver College of Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Daniel J Berg
- Department of Internal Medicine, Carver College of Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Departments of Radiation Oncology, Carver College of Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States.
| | - Melissa A Fath
- Free Radical and Radiation Biology Program, Departments of Radiation Oncology, Carver College of Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, United States.
| |
Collapse
|
45
|
Wang K, Xiao T, Yue Q, Wu F, Yu P, Mao L. Selective Amperometric Recording of Endogenous Ascorbate Secretion from a Single Rat Adrenal Chromaffin Cell with Pretreated Carbon Fiber Microelectrodes. Anal Chem 2017; 89:9502-9507. [DOI: 10.1021/acs.analchem.7b02508] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Kai Wang
- Beijing
National Laboratory for Molecular Sciences, Key Laboratory of Analytical
Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (CAS), Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tongfang Xiao
- Beijing
National Laboratory for Molecular Sciences, Key Laboratory of Analytical
Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (CAS), Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingwei Yue
- Beijing
National Laboratory for Molecular Sciences, Key Laboratory of Analytical
Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (CAS), Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fei Wu
- Beijing
National Laboratory for Molecular Sciences, Key Laboratory of Analytical
Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (CAS), Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ping Yu
- Beijing
National Laboratory for Molecular Sciences, Key Laboratory of Analytical
Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (CAS), Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lanqun Mao
- Beijing
National Laboratory for Molecular Sciences, Key Laboratory of Analytical
Chemistry for Living Biosystems, Institute of Chemistry, Chinese Academy of Sciences (CAS), Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
46
|
Schoenfeld JD, Sibenaller ZA, Mapuskar KA, Wagner BA, Cramer-Morales KL, Furqan M, Sandhu S, Carlisle TL, Smith MC, Abu Hejleh T, Berg DJ, Zhang J, Keech J, Parekh KR, Bhatia S, Monga V, Bodeker KL, Ahmann L, Vollstedt S, Brown H, Kauffman EPS, Schall ME, Hohl RJ, Clamon GH, Greenlee JD, Howard MA, Schultz MK, Smith BJ, Riley DP, Domann FE, Cullen JJ, Buettner GR, Buatti JM, Spitz DR, Allen BG. O 2⋅- and H 2O 2-Mediated Disruption of Fe Metabolism Causes the Differential Susceptibility of NSCLC and GBM Cancer Cells to Pharmacological Ascorbate. Cancer Cell 2017; 32:268. [PMID: 28810149 DOI: 10.1016/j.ccell.2017.07.008] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
47
|
Schoenfeld JD, Sibenaller ZA, Mapuskar KA, Wagner BA, Cramer-Morales KL, Furqan M, Sandhu S, Carlisle TL, Smith MC, Abu Hejleh T, Berg DJ, Zhang J, Keech J, Parekh KR, Bhatia S, Monga V, Bodeker KL, Ahmann L, Vollstedt S, Brown H, Shanahan Kauffman EP, Schall ME, Hohl RJ, Clamon GH, Greenlee JD, Howard MA, Schultz MK, Smith BJ, Riley DP, Domann FE, Cullen JJ, Buettner GR, Buatti JM, Spitz DR, Allen BG. O 2⋅- and H 2O 2-Mediated Disruption of Fe Metabolism Causes the Differential Susceptibility of NSCLC and GBM Cancer Cells to Pharmacological Ascorbate. Cancer Cell 2017; 31:487-500.e8. [PMID: 28366679 PMCID: PMC5497844 DOI: 10.1016/j.ccell.2017.02.018] [Citation(s) in RCA: 280] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 12/13/2016] [Accepted: 02/27/2017] [Indexed: 01/10/2023]
Abstract
Pharmacological ascorbate has been proposed as a potential anti-cancer agent when combined with radiation and chemotherapy. The anti-cancer effects of ascorbate are hypothesized to involve the autoxidation of ascorbate leading to increased steady-state levels of H2O2; however, the mechanism(s) for cancer cell-selective toxicity remain unknown. The current study shows that alterations in cancer cell mitochondrial oxidative metabolism resulting in increased levels of O2⋅- and H2O2 are capable of disrupting intracellular iron metabolism, thereby selectively sensitizing non-small-cell lung cancer (NSCLC) and glioblastoma (GBM) cells to ascorbate through pro-oxidant chemistry involving redox-active labile iron and H2O2. In addition, preclinical studies and clinical trials demonstrate the feasibility, selective toxicity, tolerability, and potential efficacy of pharmacological ascorbate in GBM and NSCLC therapy.
Collapse
Affiliation(s)
- Joshua D Schoenfeld
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Zita A Sibenaller
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Kranti A Mapuskar
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Brett A Wagner
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Kimberly L Cramer-Morales
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Muhammad Furqan
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Sonia Sandhu
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Thomas L Carlisle
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Mark C Smith
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Taher Abu Hejleh
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Daniel J Berg
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Jun Zhang
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - John Keech
- Department of Surgery, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Kalpaj R Parekh
- Department of Surgery, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Sudershan Bhatia
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Varun Monga
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Kellie L Bodeker
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Logan Ahmann
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Sandy Vollstedt
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Heather Brown
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Erin P Shanahan Kauffman
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Mary E Schall
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Ray J Hohl
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Gerald H Clamon
- Division of Hematology, Oncology, and Blood & Marrow Transplantation, Department of Internal Medicine, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Jeremy D Greenlee
- Department of Neurosurgery, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Matthew A Howard
- Department of Neurosurgery, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Michael K Schultz
- Department of Radiology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Brian J Smith
- Departmet of Biostatistics, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | | | - Frederick E Domann
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Joseph J Cullen
- Department of Surgery, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Garry R Buettner
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - John M Buatti
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA
| | - Douglas R Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA.
| | - Bryan G Allen
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, The University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
48
|
Erudaitius D, Huang A, Kazmi S, Buettner GR, Rodgers VGJ. Peroxiporin Expression Is an Important Factor for Cancer Cell Susceptibility to Therapeutic H2O2: Implications for Pharmacological Ascorbate Therapy. PLoS One 2017; 12:e0170442. [PMID: 28107421 PMCID: PMC5249139 DOI: 10.1371/journal.pone.0170442] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 01/04/2017] [Indexed: 11/19/2022] Open
Abstract
Cancer cell toxicity to therapeutic H2O2 varies widely depending on cell type. Interestingly, it has been observed that different cancer cell types have varying peroxiporin expression. We hypothesize that variation in peroxiporin expression can alter cell susceptibility to therapeutic H2O2 concentrations. Here, we silence peroxiporin aquaporin-3 (AQP3) on the pancreatic cancer cell line MIA PaCa-2 and compare clonogenic survival response to the wild-type. The results showed a significantly higher surviving fraction in the clonogenic response for siAQP3 MIA PaCa-2 cells at therapeutic H2O2 doses (P < 0.05). These results suggest that peroxiporin expression is significant in modulating the susceptibility of cancer cells to ascorbate therapy.
Collapse
Affiliation(s)
- Dieanira Erudaitius
- Department of Bioengineering, University of California, Riverside, Riverside, California, United States of America
| | - Andrew Huang
- Department of Neuroscience, University of California, Riverside, Riverside, California, United States of America
| | - Sarah Kazmi
- Department of Bioengineering, University of California, Riverside, Riverside, California, United States of America
| | - Garry R. Buettner
- Free Radical & Radiation Biology, Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, IA, United States of America
| | - Victor G. J. Rodgers
- Department of Bioengineering, University of California, Riverside, Riverside, California, United States of America
| |
Collapse
|
49
|
Doskey CM, Buranasudja V, Wagner BA, Wilkes JG, Du J, Cullen JJ, Buettner GR. Tumor cells have decreased ability to metabolize H 2O 2: Implications for pharmacological ascorbate in cancer therapy. Redox Biol 2016; 10:274-284. [PMID: 27833040 PMCID: PMC5106370 DOI: 10.1016/j.redox.2016.10.010] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 10/22/2016] [Indexed: 12/15/2022] Open
Abstract
Ascorbate (AscH−) functions as a versatile reducing agent. At pharmacological doses (P-AscH−; [plasma AscH−] ≥≈20 mM), achievable through intravenous delivery, oxidation of P-AscH− can produce a high flux of H2O2 in tumors. Catalase is the major enzyme for detoxifying high concentrations of H2O2. We hypothesize that sensitivity of tumor cells to P-AscH− compared to normal cells is due to their lower capacity to metabolize H2O2. Rate constants for removal of H2O2 (kcell) and catalase activities were determined for 15 tumor and 10 normal cell lines of various tissue types. A differential in the capacity of cells to remove H2O2 was revealed, with the average kcell for normal cells being twice that of tumor cells. The ED50 (50% clonogenic survival) of P-AscH− correlated directly with kcell and catalase activity. Catalase activity could present a promising indicator of which tumors may respond to P-AscH−. Ascorbate oxidizes in cell culture medium to generate a flux of H2O2. The rate constants for removal of extracellular H2O2 are on average 2-fold higher in normal cells than in cancer cells. The ED50 of high-dose ascorbate correlated with the ability of tumor cells to remove extracellular H2O2. The response to pharmacological ascorbate in murine-models of pancreatic cancer paralleled the in vitro results.
Collapse
Affiliation(s)
- Claire M Doskey
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA 52242, USA
| | - Visarut Buranasudja
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA 52242, USA
| | - Brett A Wagner
- Free Radical & Radiation Biology Program in the Department of Radiation Oncology, The University of Iowa, Iowa City, IA 52242, USA
| | - Justin G Wilkes
- Department of Surgery, The University of Iowa, Iowa City, IA 52242, USA
| | - Juan Du
- Department of Surgery, The University of Iowa, Iowa City, IA 52242, USA
| | - Joseph J Cullen
- Free Radical & Radiation Biology Program in the Department of Radiation Oncology, The University of Iowa, Iowa City, IA 52242, USA; Department of Surgery, The University of Iowa, Iowa City, IA 52242, USA; Veterans Affairs Medical Center, Veterans Affairs Medical Center, Iowa City, IA 52246, USA
| | - Garry R Buettner
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, IA 52242, USA; Free Radical & Radiation Biology Program in the Department of Radiation Oncology, The University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
50
|
Lu N, Ding Y, Tian R, Yang Z, Chen J, Peng YY. Effects of pharmacological ascorbate on hemoglobin-induced cancer cell proliferation. Int J Biol Macromol 2016; 92:1215-1219. [PMID: 27527692 DOI: 10.1016/j.ijbiomac.2016.08.036] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Revised: 08/08/2016] [Accepted: 08/11/2016] [Indexed: 01/03/2023]
Abstract
The high heme content in red meat is associated with an increased risk of developing cancer. Pharmacologic concentrations of ascorbate can specifically kill a wide range of cancer cells. In this study, the impact of ascorbate at pharmacologic concentrations on hemoglobin (Hb)-modulated human hepatoma HepG2 cell survival was investigated. It was found that HepG2 cells were proliferated by Hb (5-25μM), but killed by high pharmacologic concentrations of ascorbate (2-10mM). Although ascorbate at the low pharmacologic concentration (0.5mM) alone exhibited insignificant effect on cell viability, it effectively inhibited Hb (10μM)-induced cancer cell proliferation. The mechanism of this cytotoxicity was based on the production of extracellular H2O2 and involved transition iron. The influence of ascorbate on Hb-dependent redox reactions (i.e. the oxidative stability of Hb and its cytotoxic ferryl intermediate) was further investigated to illustrate the reaction mechanism of ascorbate toxicity, where H2O2 was generated in the reaction of ascorbate with Hb. Furthermore, circular dichroism demonstrated no significant change in the secondary structure of Hb after ascorbate addition and molecular docking revealed binding modes of ascorbate with Hb. These results demonstrated that ascorbate could possess anti-cancer activity through interfering in Hb-dependent redox reactions.
Collapse
Affiliation(s)
- Naihao Lu
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education; Key Laboratory of Green Chemistry, Jiangxi Province and College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, China.
| | - Yun Ding
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education; Key Laboratory of Green Chemistry, Jiangxi Province and College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, China
| | - Rong Tian
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education; Key Laboratory of Green Chemistry, Jiangxi Province and College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, China
| | - Zhen Yang
- Department of Physics, University of Houston, Houston, TX, USA
| | - Jianfa Chen
- Department of Physics, University of Houston, Houston, TX, USA
| | - Yi-Yuan Peng
- Key Laboratory of Functional Small Organic Molecule, Ministry of Education; Key Laboratory of Green Chemistry, Jiangxi Province and College of Chemistry and Chemical Engineering, Jiangxi Normal University, Nanchang, China.
| |
Collapse
|