601
|
SVCT-2 determines the sensitivity to ascorbate-induced cell death in cholangiocarcinoma cell lines and patient derived xenografts. Cancer Lett 2017; 398:1-11. [PMID: 28385602 DOI: 10.1016/j.canlet.2017.03.039] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/28/2017] [Accepted: 03/29/2017] [Indexed: 12/21/2022]
Abstract
Cholangiocarcinoma (CC) is a devastating malignancy with late diagnosis and poor response to conventional chemotherapy. Recent studies have revealed anti-cancer effect of vitamin C (l-ascorbic acid, ascorbate) in several types of cancer. However, the effect of l-ascorbic acid (AA) in CC remains elusive. Herein, we demonstrated that AA induced cytotoxicity in CC cells by generating intracellular reactive oxygen species (ROS), and subsequently DNA damage, ATP depletion, mTOR pathway inhibition. Moreover, AA worked synergistically with chemotherapeutic agent cisplatin to impair CC cells growth both in vitro and in vivo. Intriguingly, sodium-dependent vitamin C transporter 2 (SVCT-2) expression was inversely correlated with IC50 values of AA. Knockdown of SVCT-2 dramatically alleviated DNA damage, ATP depletion, and inhibition of mTOR pathway induced by AA. Furthermore, SVCT-2 knockdown endowed CC cells with the resistance to AA treatment. Finally, the inhibitory effects of AA were further confirmed in patient-derived CC xenograft models. Thus, our results unravel therapeutic potential of AA alone or in combination with cisplatin for CC. SVCT2 expression level may serve as a positive outcome predictor for AA treatment in CC.
Collapse
|
602
|
Shah MA. Future Directions in Improving Outcomes for Patients with Gastric and Esophageal Cancer. Hematol Oncol Clin North Am 2017; 31:545-552. [PMID: 28501093 DOI: 10.1016/j.hoc.2017.01.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
"This issue of Hematology/Oncology Clinics of North America provides an update to the current understanding of the physiology of gastric and esophageal cancers and the state-of-the-art management of disease. Over the past 10 years, we have witnessed dramatic changes in both our understanding of the disease and its management. We have 2 new biological agents approved to treat advanced disease, with several more prospects under development. In this article, the author looks to the future, attempting to answer the question of which advancements will play the biggest role in improving patient outcomes in this still-devastating disease.
Collapse
Affiliation(s)
- Manish A Shah
- Weill Cornell Medicine/New York-Presbyterian Hospital, Division of Hematology and Medical Oncology, 1305 York Avenue, New York, NY 10021, USA.
| |
Collapse
|
603
|
Gunnink LK, Busscher BM, Wodarek JA, Rosette KA, Strohbehn LE, Looyenga BD, Louters LL. Caffeine inhibition of GLUT1 is dependent on the activation state of the transporter. Biochimie 2017; 137:99-105. [PMID: 28322926 DOI: 10.1016/j.biochi.2017.03.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 03/15/2017] [Indexed: 10/19/2022]
Abstract
Caffeine has been shown to be a robust uncompetitive inhibitor of glucose uptake in erythrocytes. It preferentially binds to the nucleotide-binding site on GLUT1 in its tetrameric form and mimics the inhibitory action of ATP. Here we demonstrate that caffeine is also a dose-dependent, uncompetitive inhibitor of 2-deoxyglucose (2DG) uptake in L929 fibroblasts. The inhibitory effect on 2DG uptake in these cells was reversible with a rapid onset and was additive to the competitive inhibitory effects of glucose itself, confirming that caffeine does not interfere with glucose binding. We also report for the first time that caffeine inhibition was additive to inhibition by curcumin, suggesting distinct binding sites for curcumin and caffeine. In contrast, caffeine inhibition was not additive to that of cytochalasin B, consistent with previous data that reported that these two inhibitors have overlapping binding sites. More importantly, we show that the magnitude of maximal caffeine inhibition in L929 cells is much lower than in erythrocytes (35% compared to 90%). Two epithelial cell lines, HCLE and HK2, have both higher concentrations of GLUT1 and increased basal 2DG uptake (3-4 fold) compared to L929 cells, and subsequently display greater maximal inhibition by caffeine (66-70%). Interestingly, activation of 2DG uptake (3-fold) in L929 cells by glucose deprivation shifted the responsiveness of these cells to caffeine inhibition (35%-70%) without a change in total GLUT1 concentration. These data indicate that the inhibition of caffeine is dependent on the activity state of GLUT1, not merely on the concentration.
Collapse
Affiliation(s)
- Leesha K Gunnink
- Department of Chemistry and Biochemistry, Calvin College, Grand Rapids, MI 49546, USA
| | - Brianna M Busscher
- Department of Chemistry and Biochemistry, Calvin College, Grand Rapids, MI 49546, USA
| | - Jeremy A Wodarek
- Department of Chemistry and Biochemistry, Calvin College, Grand Rapids, MI 49546, USA
| | - Kylee A Rosette
- Department of Chemistry and Biochemistry, Calvin College, Grand Rapids, MI 49546, USA
| | - Lauren E Strohbehn
- Department of Chemistry and Biochemistry, Calvin College, Grand Rapids, MI 49546, USA
| | - Brendan D Looyenga
- Department of Chemistry and Biochemistry, Calvin College, Grand Rapids, MI 49546, USA
| | - Larry L Louters
- Department of Chemistry and Biochemistry, Calvin College, Grand Rapids, MI 49546, USA.
| |
Collapse
|
604
|
Murray HC, Dun MD, Verrills NM. Harnessing the power of proteomics for identification of oncogenic, druggable signalling pathways in cancer. Expert Opin Drug Discov 2017; 12:431-447. [PMID: 28286965 DOI: 10.1080/17460441.2017.1304377] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Genomic and transcriptomic profiling of tumours has revolutionised our understanding of cancer. However, the majority of tumours possess multiple mutations, and determining which oncogene, or even which pathway, to target is difficult. Proteomics is emerging as a powerful approach to identify the functionally important pathways driving these cancers, and how they can be targeted therapeutically. Areas covered: The authors provide a technical overview of mass spectrometry based approaches for proteomic profiling, and review the current and emerging strategies available for the identification of dysregulated networks, pathways, and drug targets in cancer cells, with a key focus on the ability to profile cancer kinomes. The potential applications of mass spectrometry in the clinic are also highlighted. Expert opinion: The addition of proteomic information to genomic platforms - 'proteogenomics' - is providing unparalleled insight in cancer cell biology. Application of improved mass spectrometry technology and methodology, in particular the ability to analyse post-translational modifications (the PTMome), is providing a more complete picture of the dysregulated networks in cancer, and uncovering novel therapeutic targets. While the application of proteomics to discovery research will continue to rise, improved workflow standardisation and reproducibility is required before mass spectrometry can enter routine clinical use.
Collapse
Affiliation(s)
- Heather C Murray
- a School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, Priority Research Centre for Cancer Research, Innovation and Translation , University of Newcastle , Callaghan , NSW , Australia.,b Cancer Research Program , Hunter Medical Research Institute , Newcastle , NSW , Australia
| | - Matthew D Dun
- a School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, Priority Research Centre for Cancer Research, Innovation and Translation , University of Newcastle , Callaghan , NSW , Australia.,b Cancer Research Program , Hunter Medical Research Institute , Newcastle , NSW , Australia
| | - Nicole M Verrills
- a School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, Priority Research Centre for Cancer Research, Innovation and Translation , University of Newcastle , Callaghan , NSW , Australia.,b Cancer Research Program , Hunter Medical Research Institute , Newcastle , NSW , Australia
| |
Collapse
|
605
|
Galadari S, Rahman A, Pallichankandy S, Thayyullathil F. Reactive oxygen species and cancer paradox: To promote or to suppress? Free Radic Biol Med 2017; 104:144-164. [PMID: 28088622 DOI: 10.1016/j.freeradbiomed.2017.01.004] [Citation(s) in RCA: 635] [Impact Index Per Article: 90.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 12/16/2016] [Accepted: 01/03/2017] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS), a group of highly reactive ions and molecules, are increasingly being appreciated as powerful signaling molecules involved in the regulation of a variety of biological processes. Indeed, their role is continuously being delineated in a variety of pathophysiological conditions. For instance, cancer cells are shown to have increased ROS levels in comparison to their normal counterparts. This is partly due to an enhanced metabolism and mitochondrial dysfunction in cancer cells. The escalated ROS generation in cancer cells contributes to the biochemical and molecular changes necessary for the tumor initiation, promotion and progression, as well as, tumor resistance to chemotherapy. Therefore, increased ROS in cancer cells may provide a unique opportunity to eliminate cancer cells via elevating ROS to highly toxic levels intracellularly, thereby, activating various ROS-induced cell death pathways, or inhibiting cancer cell resistance to chemotherapy. Such results can be achieved by using agents that either increase ROS generation, or inhibit antioxidant defense, or even a combination of both. In fact, a large variety of anticancer drugs, and some of those currently under clinical trials, effectively kill cancer cells and overcome drug resistance via enhancing ROS generation and/or impeding the antioxidant defense mechanism. This review focuses on our current understanding of the tumor promoting (tumorigenesis, angiogenesis, invasion and metastasis, and chemoresistance) and the tumor suppressive (apoptosis, autophagy, and necroptosis) functions of ROS, and highlights the potential mechanism(s) involved. It also sheds light on a very novel and an actively growing field of ROS-dependent cell death mechanism referred to as ferroptosis.
Collapse
Affiliation(s)
- Sehamuddin Galadari
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE; Al Jalila Foundation Research Centre, P.O. Box 300100, Dubai, UAE.
| | - Anees Rahman
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE.
| | - Siraj Pallichankandy
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE.
| | - Faisal Thayyullathil
- Cell Signaling Laboratory, Department of Biochemistry, College of Medicine and Health Sciences, UAE University, P.O. Box 17666, Al Ain, Abu Dhabi, UAE.
| |
Collapse
|
606
|
Xia J, Xu H, Zhang X, Allamargot C, Coleman KL, Nessler R, Frech I, Tricot G, Zhan F. Multiple Myeloma Tumor Cells are Selectively Killed by Pharmacologically-dosed Ascorbic Acid. EBioMedicine 2017; 18:41-49. [PMID: 28229908 PMCID: PMC5405162 DOI: 10.1016/j.ebiom.2017.02.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 02/13/2017] [Accepted: 02/13/2017] [Indexed: 12/15/2022] Open
Abstract
High-dose chemotherapies to treat multiple myeloma (MM) can be life-threatening due to toxicities to normal cells and there is a need to target only tumor cells and/or lower standard drug dosage without losing efficacy. We show that pharmacologically-dosed ascorbic acid (PAA), in the presence of iron, leads to the formation of highly reactive oxygen species (ROS) resulting in cell death. PAA selectively kills CD138+ MM tumor cells derived from MM and smoldering MM (SMM) but not from monoclonal gammopathy undetermined significance (MGUS) patients. PAA alone or in combination with melphalan inhibits tumor formation in MM xenograft mice. This study shows PAA efficacy on primary cancer cells and cell lines in vitro and in vivo. Pharmacologically-dosed ascorbic acid kills Multiple Myeloma cells. Pharmacologically-dosed ascorbic leads to apoptosis-inducing factor 1 cleavage. Pharmacologically-dosed ascorbic lowers melphalan dosage.
Multiple myeloma (MM) remains a difficult to cure disease in the majority of cases. Several preclinical and clinical studies have shown that ascorbic acid in pharmacologic doses (PAA) selectively kills cancer cells, while sparing normal cells. This article reveals the biological mechanism by which PAA exerts its anti-cancer effects and should lead to the development of an innovative therapy in MM.
Collapse
Affiliation(s)
- Jiliang Xia
- Department of Medicine, Division of Hematology, Oncology and Blood and Marrow Transplantation, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States; Institute of Cancer Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Hongwei Xu
- Department of Medicine, Division of Hematology, Oncology and Blood and Marrow Transplantation, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| | - Xiaoyan Zhang
- Department of Medicine, Division of Hematology, Oncology and Blood and Marrow Transplantation, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States; East China University of Science and Technology, Shanghai, China
| | - Chantal Allamargot
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, United States
| | - Kristen L Coleman
- Department of Medicine, Division of Hematology, Oncology and Blood and Marrow Transplantation, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| | - Randy Nessler
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, United States
| | - Ivana Frech
- Department of Medicine, Division of Hematology, Oncology and Blood and Marrow Transplantation, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States
| | - Guido Tricot
- Department of Medicine, Division of Hematology, Oncology and Blood and Marrow Transplantation, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States.
| | - Fenghuang Zhan
- Department of Medicine, Division of Hematology, Oncology and Blood and Marrow Transplantation, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, United States.
| |
Collapse
|
607
|
Fernandes G, Barone AW, Dziak R. The effect of ascorbic acid on bone cancer cells in vitro. ACTA ACUST UNITED AC 2017. [DOI: 10.1080/23312025.2017.1288335] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Affiliation(s)
- Gabriela Fernandes
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, State University of New York, 3435 Main Street, Buffalo, NY 14201, USA
| | - Andrew W. Barone
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, State University of New York, 3435 Main Street, Buffalo, NY 14201, USA
| | - Rosemary Dziak
- Department of Oral Biology, School of Dental Medicine, University at Buffalo, State University of New York, 3435 Main Street, Buffalo, NY 14201, USA
| |
Collapse
|
608
|
Dharmaraja AT. Role of Reactive Oxygen Species (ROS) in Therapeutics and Drug Resistance in Cancer and Bacteria. J Med Chem 2017; 60:3221-3240. [DOI: 10.1021/acs.jmedchem.6b01243] [Citation(s) in RCA: 280] [Impact Index Per Article: 40.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Allimuthu T. Dharmaraja
- Department of Genetics and Genome Sciences and Comprehensive Cancer
Center, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, United States
| |
Collapse
|
609
|
Metabolomics: A Primer. Trends Biochem Sci 2017; 42:274-284. [PMID: 28196646 DOI: 10.1016/j.tibs.2017.01.004] [Citation(s) in RCA: 239] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 11/13/2016] [Accepted: 01/12/2017] [Indexed: 02/08/2023]
Abstract
Metabolomics generates a profile of small molecules that are derived from cellular metabolism and can directly reflect the outcome of complex networks of biochemical reactions, thus providing insights into multiple aspects of cellular physiology. Technological advances have enabled rapid and increasingly expansive data acquisition with samples as small as single cells; however, substantial challenges in the field remain. In this primer we provide an overview of metabolomics, especially mass spectrometry (MS)-based metabolomics, which uses liquid chromatography (LC) for separation, and discuss its utilities and limitations. We identify and discuss several areas at the frontier of metabolomics. Our goal is to give the reader a sense of what might be accomplished when conducting a metabolomics experiment, now and in the near future.
Collapse
|
610
|
Triethylenetetramine Synergizes with Pharmacologic Ascorbic Acid in Hydrogen Peroxide Mediated Selective Toxicity to Breast Cancer Cell. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:3481710. [PMID: 28280522 PMCID: PMC5320382 DOI: 10.1155/2017/3481710] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 01/05/2017] [Indexed: 12/29/2022]
Abstract
Breast cancer is characterized by overexpression of superoxide dismutase (SOD) and downregulation of catalase and more resistance to hydrogen peroxide (H2O2) than normal cells. Thus, relatively high H2O2 promotes breast cancer cell growth and proliferation. However, excessive intracellular H2O2 leads to death of breast cancer cells. In cancer cells, high level ascorbic acid (Asc) is able to be autoxidized and thus provides an electron to oxygen to generate H2O2. In the present study, we demonstrated that triethylenetetramine (TETA) enhances Asc autoxidation and thus elevates H2O2 production in MCF-7 cells. Furthermore, Asc/TETA combination significantly impaired cancer cell viability, while having much milder effects on normal cells, indicating Asc/TETA could be a promising therapy for breast cancer. Moreover, SOD1 and N-acetyl-L-cysteine failed to improve MCF-7 cells viability in the presence of Asc/TETA, while catalase significantly inhibited the cytotoxicity of Asc/TETA to breast cancer cells, strongly suggesting that the selective cytotoxicity of Asc/TETA to cancer cells is H2O2-dependent. In addition, Asc/TETA induces RAS/ERK downregulation in breast cancer cells. Animal studies confirmed that Asc/TETA effectively suppressed tumor growth in vivo. In conclusion, TETA synergizes pharmacologic Asc autoxidation and H2O2 overproduction in breast cancer cells, which suppresses RAS/ERK pathway and results in apoptosis.
Collapse
|
611
|
Dienstmann R, Vermeulen L, Guinney J, Kopetz S, Tejpar S, Tabernero J. Consensus molecular subtypes and the evolution of precision medicine in colorectal cancer. Nat Rev Cancer 2017; 17:79-92. [PMID: 28050011 DOI: 10.1038/nrc.2016.126] [Citation(s) in RCA: 553] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Critical driver genomic events in colorectal cancer have been shown to affect the response to targeted agents that were initially developed under the 'one gene, one drug' paradigm of precision medicine. Our current knowledge of the complexity of the cancer genome, clonal evolution patterns under treatment pressure and pharmacodynamic effects of target inhibition support the transition from a one gene, one drug approach to a 'multi-gene, multi-drug' model when making therapeutic decisions. Better characterization of the transcriptomic subtypes of colorectal cancer, encompassing tumour, stromal and immune components, has revealed convergent pathway dependencies that mandate a 'multi-molecular' perspective for the development of therapies to treat this disease.
Collapse
Affiliation(s)
- Rodrigo Dienstmann
- Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona 08035, Spain
- Sage Bionetworks, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, WA 98109, Seattle, USA
| | - Louis Vermeulen
- Laboratory for Experimental Oncology and Radiobiology (LEXOR), Center for Experimental Molecular Medicine (CEMM), Academic Medical Center (AMC), University of Amsterdam, 1012 WX Amsterdam, The Netherlands
| | - Justin Guinney
- Sage Bionetworks, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N, WA 98109, Seattle, USA
| | - Scott Kopetz
- The University of Texas, M.D. Anderson Cancer Center, 1515 Holcombe Blvd, Houston, Texas 77030, USA
| | - Sabine Tejpar
- Digestive Oncology Unit, University Hospital Gasthuisberg, Herestraat 49, 3000 Leuven, Belgium
| | - Josep Tabernero
- Vall d'Hebron Institute of Oncology (VHIO), Universitat Autònoma de Barcelona, Barcelona 08035, Spain
| |
Collapse
|
612
|
Zununi Vahed S, Salehi R, Davaran S, Sharifi S. Liposome-based drug co-delivery systems in cancer cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 71:1327-1341. [DOI: 10.1016/j.msec.2016.11.073] [Citation(s) in RCA: 190] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 11/10/2016] [Accepted: 11/21/2016] [Indexed: 02/07/2023]
|
613
|
Wei X, Xu Y, Xu FF, Chaiswing L, Schnell D, Noel T, Wang C, Chen J, St Clair DK, St Clair WH. RelB Expression Determines the Differential Effects of Ascorbic Acid in Normal and Cancer Cells. Cancer Res 2017; 77:1345-1356. [PMID: 28108513 DOI: 10.1158/0008-5472.can-16-0785] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 11/15/2016] [Accepted: 11/18/2016] [Indexed: 01/15/2023]
Abstract
Cancer cells typically experience higher oxidative stress than normal cells, such that elevating pro-oxidant levels can trigger cancer cell death. Although pre-exposure to mild oxidative agents will sensitize cancer cells to radiation, this pre-exposure may also activate the adaptive stress defense system in normal cells. Ascorbic acid is a prototype redox modulator that when infused intravenously appears to kill cancers without injury to normal tissues; however, the mechanisms involved remain elusive. In this study, we show how ascorbic acid kills cancer cells and sensitizes prostate cancer to radiation therapy while also conferring protection upon normal prostate epithelial cells against radiation-induced injury. We found that the NF-κB transcription factor RelB is a pivotal determinant in the differential radiosensitization effects of ascorbic acid in prostate cancer cells and normal prostate epithelial cells. Mechanistically, high reactive oxygen species concentrations suppress RelB in cancer cells. RelB suppression decreases expression of the sirtuin SIRT3 and the powerful antioxidant MnSOD, which in turn increases oxidative and metabolic stresses in prostate cancer cells. In contrast, ascorbic acid enhances RelB expression in normal cells, improving antioxidant and metabolic defenses against radiation injury. In addition to showing how RelB mediates the differential effects of ascorbic acid on cancer and normal tissue radiosensitivities, our work also provides a proof of concept for the existence of redox modulators that can improve the efficacy of radiotherapy while protecting against normal tissue injury in cancer settings. Cancer Res; 77(6); 1345-56. ©2017 AACR.
Collapse
Affiliation(s)
- Xiaowei Wei
- Department of Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, Kentucky.,Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Yong Xu
- Department of Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, Kentucky.,Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Fang Fang Xu
- Department of Radiation Medicine, Markey Cancer Center, University of Kentucky, Lexington, Kentucky
| | - Luksana Chaiswing
- Department of Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, Kentucky
| | - David Schnell
- Department of Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, Kentucky
| | - Teresa Noel
- Department of Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, Kentucky
| | - Chi Wang
- Biostatistics Core, Markey Cancer Center, University of Kentucky, Lexington, Kentucky
| | - Jinfei Chen
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| | - Daret K St Clair
- Department of Toxicology and Cancer Biology, Markey Cancer Center, University of Kentucky, Lexington, Kentucky.
| | - William H St Clair
- Department of Oncology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
614
|
Beaver LM, Kuintzle R, Buchanan A, Wiley MW, Glasser ST, Wong CP, Johnson GS, Chang JH, Löhr CV, Williams DE, Dashwood RH, Hendrix DA, Ho E. Long noncoding RNAs and sulforaphane: a target for chemoprevention and suppression of prostate cancer. J Nutr Biochem 2017; 42:72-83. [PMID: 28131897 DOI: 10.1016/j.jnutbio.2017.01.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Revised: 11/30/2016] [Accepted: 01/06/2017] [Indexed: 12/21/2022]
Abstract
Long noncoding RNAs (lncRNAs) have emerged as important in cancer development and progression. The impact of diet on lncRNA expression is largely unknown. Sulforaphane (SFN), obtained from vegetables like broccoli, can prevent and suppress cancer formation. Here we tested the hypothesis that SFN attenuates the expression of cancer-associated lncRNAs. We analyzed whole-genome RNA-sequencing data of normal human prostate epithelial cells and prostate cancer cells treated with 15 μM SFN or dimethylsulfoxide. SFN significantly altered expression of ~100 lncRNAs in each cell type and normalized the expression of some lncRNAs that were differentially expressed in cancer cells. SFN-mediated alterations in lncRNA expression correlated with genes that regulate cell cycle, signal transduction and metabolism. LINC01116 was functionally investigated because it was overexpressed in several cancers, and was transcriptionally repressed after SFN treatment. Knockdown of LINC01116 with siRNA decreased proliferation of prostate cancer cells and significantly up-regulated several genes including GAPDH (regulates glycolysis), MAP1LC3B2 (autophagy) and H2AFY (chromatin structure). A four-fold decrease in the ability of the cancer cells to form colonies was found when the LINC01116 gene was disrupted through a CRISPR/CAS9 method, further supporting an oncogenic function for LINC01116 in PC-3 cells. We identified a novel isoform of LINC01116 and bioinformatically investigated the possibility that LINC01116 could interact with target genes via ssRNA:dsDNA triplexes. Our data reveal that chemicals from the diet can influence the expression of functionally important lncRNAs, and suggest a novel mechanism by which SFN may prevent and suppress prostate cancer.
Collapse
Affiliation(s)
- Laura M Beaver
- Biological and Population Health Sciences, Oregon State University, 103 Milam Hall, Corvallis, OR 97331; Linus Pauling Institute, Oregon State University, 307 Linus Pauling Science Center, Corvallis, OR 97331.
| | - Rachael Kuintzle
- Department of Biochemistry and Biophysics, Oregon State University, 2011 Agriculture and Life Sciences Building, Corvallis, OR 97331.
| | - Alex Buchanan
- Biological and Population Health Sciences, Oregon State University, 103 Milam Hall, Corvallis, OR 97331; Department of Botany and Plant Pathology, Oregon State University, 2082 Cordley Hall, Corvallis, OR 97331.
| | - Michelle W Wiley
- Department of Biochemistry and Biophysics, Oregon State University, 2011 Agriculture and Life Sciences Building, Corvallis, OR 97331.
| | - Sarah T Glasser
- Biological and Population Health Sciences, Oregon State University, 103 Milam Hall, Corvallis, OR 97331.
| | - Carmen P Wong
- Biological and Population Health Sciences, Oregon State University, 103 Milam Hall, Corvallis, OR 97331; Linus Pauling Institute, Oregon State University, 307 Linus Pauling Science Center, Corvallis, OR 97331.
| | - Gavin S Johnson
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, Texas A&M Health Science Center, 2121 W. Holcombe Blvd., Mail Stop 1201, Houston, TX 77030-3303.
| | - Jeff H Chang
- Department of Botany and Plant Pathology, Oregon State University, 2082 Cordley Hall, Corvallis, OR 97331; Center for Genome Research and Biocomputing, Oregon State University, 3021 Agriculture and Life Sciences Building, Corvallis, OR 97331.
| | - Christiane V Löhr
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, 105 Magruder Hall, Corvallis, OR 97331.
| | - David E Williams
- Linus Pauling Institute, Oregon State University, 307 Linus Pauling Science Center, Corvallis, OR 97331; Environmental and Molecular Toxicology, Oregon State University, 1007 Agriculture & Life Sciences Building, Corvallis, OR 97331.
| | - Roderick H Dashwood
- Center for Epigenetics & Disease Prevention, Institute of Biosciences & Technology, Texas A&M Health Science Center, 2121 W. Holcombe Blvd., Mail Stop 1201, Houston, TX 77030-3303.
| | - David A Hendrix
- Department of Biochemistry and Biophysics, Oregon State University, 2011 Agriculture and Life Sciences Building, Corvallis, OR 97331; The School of Electrical Engineering and Computer Science, Oregon State University, 1148 Kelley Engineering Center, Corvallis, OR 97331.
| | - Emily Ho
- Biological and Population Health Sciences, Oregon State University, 103 Milam Hall, Corvallis, OR 97331; Linus Pauling Institute, Oregon State University, 307 Linus Pauling Science Center, Corvallis, OR 97331; Center for Genome Research and Biocomputing, Oregon State University, 3021 Agriculture and Life Sciences Building, Corvallis, OR 97331; Moore Family Center, Oregon State University, 103 Milam Hall, Corvallis, OR 97331.
| |
Collapse
|
615
|
Wu H, Song C, Zhang J, Zhao J, Fu B, Mao T, Zhang Y. Melatonin-mediated upregulation of GLUT1 blocks exit from pluripotency by increasing the uptake of oxidized vitamin C in mouse embryonic stem cells. FASEB J 2017; 31:1731-1743. [PMID: 28069827 DOI: 10.1096/fj.201601085r] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 01/03/2017] [Indexed: 12/21/2022]
Abstract
Melatonin and vitamin C are powerful antioxidants that improve the reprogramming efficiency of induced pluripotent stem cells (iPSCs). However, the effects of the combined treatment of vitamin C and melatonin on the differentiation of embryonic stem cells (ESCs) have not yet been examined. In this study, we showed that melatonin synergizes with vitamin C to derail exit from pluripotency of mouse ESCs. This effect is related to the increased uptake of dehydroascorbate, the oxidized form of vitamin C, through glucose transporter 1 (Glut1) transporter, which in turn, is upregulated by melatonin treatment. Analysis of the cell signaling pathway profiling systems and specific pathway inhibition indicated that melatonin enhances Glut1 expression by activating the PI3K/AKT and MAPK/ERK signaling pathways. Our findings provide a theoretical basis for application of melatonin in research on ESCs and iPSCs and for further investigation of the effect of combinatorial compounds on cell reprogramming.-Wu, H., Song, C., Zhang, J., Zhao, J., Fu, B., Mao, T., Zhang, Y. Melatonin-mediated upregulation of GLUT1 blocks exit from pluripotency by increasing the uptake of oxidized vitamin C in mouse embryonic stem cells.
Collapse
Affiliation(s)
- Haibo Wu
- College of Veterinary Medicine, Northwest Agriculture and Forestry (A&F) University, Yangling, China; and .,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, China
| | - Chao Song
- College of Veterinary Medicine, Northwest Agriculture and Forestry (A&F) University, Yangling, China; and.,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, China
| | - Jingcheng Zhang
- College of Veterinary Medicine, Northwest Agriculture and Forestry (A&F) University, Yangling, China; and.,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, China
| | - Jiamin Zhao
- College of Veterinary Medicine, Northwest Agriculture and Forestry (A&F) University, Yangling, China; and .,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, China
| | - Beibei Fu
- College of Veterinary Medicine, Northwest Agriculture and Forestry (A&F) University, Yangling, China; and.,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, China
| | - Tingchao Mao
- College of Veterinary Medicine, Northwest Agriculture and Forestry (A&F) University, Yangling, China; and.,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, China
| | - Yong Zhang
- College of Veterinary Medicine, Northwest Agriculture and Forestry (A&F) University, Yangling, China; and .,Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, China
| |
Collapse
|
616
|
Rezaei-Tavirani M, Rezaei-Tavirani S, Ahmadi N, Naderi N, Abdi S. Pancreatic adenocarcinoma protein-protein interaction network analysis. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2017; 10:S85-S92. [PMID: 29511477 PMCID: PMC5838186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
AIM Gene assessment of pancreatic adenocarcinoma disease via protein-protein interaction (PPI) Network Analysis. BACKGROUND Diagnosis, especially early detection of pancreatic adenocarcinoma as a lethal disease implies more investigation. PPI Network Analysis is a suitable tool to discover new aspects of molecular mechanism of diseases. METHODS In the present study the related genes to pancreatic adenocarcinoma are studied in the interactome unit and the key genes are highlighted. The significant clusters were introduced by Cluster-ONE application of Cytoscape software 3.4.0. The genes are retrieved from STRING date base and analyzed by Cytoscape software. The crucial genes based on analysis of central parameters were determined and enriched by ClueGO v2.3.5 via gene ontology. RESULTS The number of 24 key genes among 794 initial genes were highlighted as crucial nodes in relationship with pancreatic adenocarcinoma. All of the key genes were organized in a cluster including 216 nodes. The main related pathways and cancer diseases were determined. CONCLUSION It was concluded that the introduced 24 genes are possible biomarker panel of pancreatic adenocarcinoma.
Collapse
Affiliation(s)
| | - Sina Rezaei-Tavirani
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nayebali Ahmadi
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Nosratollah Naderi
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Abdi
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
617
|
Sun T, Wang Q, Bi Y, Chen X, Liu L, Ruan C, Zhao Z, Jiang C. Supramolecular amphiphiles based on cyclodextrin and hydrophobic drugs. J Mater Chem B 2017; 5:2644-2654. [DOI: 10.1039/c6tb03272a] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Herein we report a novel “supra-prodrug-type” superamphiphile design: via a redox-sensitive self-immolative linker, a hydrophobic drug molecule was labeled with an azobenzene moiety, which was designed to be capped by a hydrophilic cyclodextrin (CD) molecule.
Collapse
Affiliation(s)
- Tao Sun
- Key Laboratory of Smart Drug Delivery (Ministry of Education)
- State Key Laboratory of Medical Neurobiology
- Department of Pharmaceutics
- School of Pharmacy
- Fudan University
| | - Qingbing Wang
- Department of Chemistry
- Fudan University
- Shanghai 200433
- P. R. China
| | - Yunke Bi
- Department of Neurosurgery
- Shanghai First People's Hospital
- School of Medicine
- Shanghai Jiao Tong University
- Shanghai 201620
| | - Xinli Chen
- Key Laboratory of Smart Drug Delivery (Ministry of Education)
- State Key Laboratory of Medical Neurobiology
- Department of Pharmaceutics
- School of Pharmacy
- Fudan University
| | - Lisha Liu
- Key Laboratory of Smart Drug Delivery (Ministry of Education)
- State Key Laboratory of Medical Neurobiology
- Department of Pharmaceutics
- School of Pharmacy
- Fudan University
| | - Chunhui Ruan
- Key Laboratory of Smart Drug Delivery (Ministry of Education)
- State Key Laboratory of Medical Neurobiology
- Department of Pharmaceutics
- School of Pharmacy
- Fudan University
| | - Zhifeng Zhao
- Key Laboratory of Smart Drug Delivery (Ministry of Education)
- State Key Laboratory of Medical Neurobiology
- Department of Pharmaceutics
- School of Pharmacy
- Fudan University
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery (Ministry of Education)
- State Key Laboratory of Medical Neurobiology
- Department of Pharmaceutics
- School of Pharmacy
- Fudan University
| |
Collapse
|
618
|
Meščić A, Šalić A, Gregorić T, Zelić B, Raić-Malić S. Continuous flow-ultrasonic synergy in click reactions for the synthesis of novel 1,2,3-triazolyl appended 4,5-unsaturated l-ascorbic acid derivatives. RSC Adv 2017. [DOI: 10.1039/c6ra25244c] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
A combination of flow chemistry and batch-based synthetic procedures has been successfully applied to the assembly of novel 4,5-unsaturated l-ascorbic acid series 6a–6n with diverse C-6-substituted 1,2,3-triazole moiety.
Collapse
Affiliation(s)
- Andrijana Meščić
- University of Zagreb
- Faculty of Chemical Engineering and Technology
- Department of Organic Chemistry
- HR-10000 Zagreb
- Croatia
| | - Anita Šalić
- University of Zagreb
- Faculty of Chemical Engineering and Technology
- Department of Reaction Engineering and Catalysis
- HR-10000 Zagreb
- Croatia
| | - Tomislav Gregorić
- University of Zagreb
- Faculty of Chemical Engineering and Technology
- Department of Organic Chemistry
- HR-10000 Zagreb
- Croatia
| | - Bruno Zelić
- University of Zagreb
- Faculty of Chemical Engineering and Technology
- Department of Reaction Engineering and Catalysis
- HR-10000 Zagreb
- Croatia
| | - Silvana Raić-Malić
- University of Zagreb
- Faculty of Chemical Engineering and Technology
- Department of Organic Chemistry
- HR-10000 Zagreb
- Croatia
| |
Collapse
|
619
|
Zhang K, Sun W, Huang L, Zhu K, Pei F, Zhu L, Wang Q, Lu Y, Zhang H, Jin H, Zhang LH, Zhang L, Yue J. Identifying Glyceraldehyde 3-Phosphate Dehydrogenase as a Cyclic Adenosine Diphosphoribose Binding Protein by Photoaffinity Protein-Ligand Labeling Approach. J Am Chem Soc 2016; 139:156-170. [PMID: 27936653 DOI: 10.1021/jacs.6b08088] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Cyclic adenosine diphosphoribose (cADPR), an endogenous nucleotide derived from nicotinamide adenine dinucleotide (NAD+), mobilizes Ca2+ release from endoplasmic reticulum (ER) via ryanodine receptors (RyRs), yet the bridging protein(s) between cADPR and RyRs remain(s) unknown. Here we synthesized a novel photoaffinity labeling (PAL) cADPR agonist, PAL-cIDPRE, and subsequently applied it to purify its binding proteins in human Jurkat T cells. We identified glyceraldehyde 3-phosphate dehydrogenase (GAPDH) as one of the cADPR binding protein(s), characterized the binding affinity between cADPR and GAPDH in vitro by surface plasmon resonance (SPR) assay, and mapped cADPR's binding sites in GAPDH. We further demonstrated that cADPR induces the transient interaction between GAPDH and RyRs in vivo and that GAPDH knockdown abolished cADPR-induced Ca2+ release. However, GAPDH did not catalyze cADPR into any other known or novel compound(s). In summary, our data clearly indicate that GAPDH is the long-sought-after cADPR binding protein and is required for cADPR-mediated Ca2+ mobilization from ER via RyRs.
Collapse
Affiliation(s)
- Kehui Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University , Beijing 100191, China.,Department of Biomedical Sciences, City University of Hong Kong , Hong Kong, China
| | - Wei Sun
- Department of Biomedical Sciences, City University of Hong Kong , Hong Kong, China.,Department of Biology and Shenzhen Key Laboratory of Cell Microenvironment, South University of Science and Technology of China , Shenzhen 518052, China
| | - Lihong Huang
- Department of Biomedical Sciences, City University of Hong Kong , Hong Kong, China
| | - Kaiyuan Zhu
- Department of Biomedical Sciences, City University of Hong Kong , Hong Kong, China
| | - Fen Pei
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University , Beijing 100191, China
| | - Longchao Zhu
- Department of Biomedical Sciences, City University of Hong Kong , Hong Kong, China
| | - Qian Wang
- Department of Biomedical Sciences, City University of Hong Kong , Hong Kong, China
| | - Yingying Lu
- Department of Biomedical Sciences, City University of Hong Kong , Hong Kong, China
| | - Hongmin Zhang
- Department of Biology and Shenzhen Key Laboratory of Cell Microenvironment, South University of Science and Technology of China , Shenzhen 518052, China
| | - Hongwei Jin
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University , Beijing 100191, China
| | - Li-He Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University , Beijing 100191, China
| | - Liangren Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University , Beijing 100191, China
| | - Jianbo Yue
- Department of Biomedical Sciences, City University of Hong Kong , Hong Kong, China
| |
Collapse
|
620
|
Lei SF, Yang DH, Wang MW. A historic study that opened a new chapter in nutritional science. Acta Pharmacol Sin 2016; 37:1641-1644. [PMID: 27867188 DOI: 10.1038/aps.2016.131] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 10/26/2016] [Indexed: 12/15/2022] Open
|
621
|
Durand N, Storz P. Targeting reactive oxygen species in development and progression of pancreatic cancer. Expert Rev Anticancer Ther 2016; 17:19-31. [PMID: 27841037 DOI: 10.1080/14737140.2017.1261017] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
INTRODUCTION Pancreatic ductal adenocarcinoma (PDA) is characterized by expression of oncogenic KRas which drives all aspects of tumorigenesis. Oncogenic KRas induces the formation of reactive oxygen species (ROS) which have been implicated in initiation and progression of PDA. To facilitate tumor promoting levels and to avoid oncogene-induced senescence or cytotoxicity, ROS homeostasis in PDA cells is balanced by additional up-regulation of antioxidant systems. Areas covered: We examine the sources of ROS in PDA, the mechanisms by which ROS homeostasis is maintained, and the biological consequences of ROS in PDA. Additionally, we discuss the potential mechanisms for targeting ROS homoeostasis as a point of therapeutic intervention. An extensive review of the relevant literature as it relates to the topic was conducted using PubMed. Expert commentary: Even though oncogenic mutations in the KRAS gene have been detected in over 95% of human pancreatic adenocarcinoma, targeting its gene product, KRas, has been difficult. The dependency of PDA cells on balancing ROS homeostasis could be an angle for new prevention or treatment strategies. These include use of antioxidants to prevent formation or progression of precancerous lesions, or methods to increase ROS in tumor cells to toxic levels.
Collapse
Affiliation(s)
- Nisha Durand
- a Department of Cancer Biology , Mayo Clinic , Jacksonville , FL , USA
| | - Peter Storz
- a Department of Cancer Biology , Mayo Clinic , Jacksonville , FL , USA
| |
Collapse
|
622
|
Avci P, Freire F, Banvolgyi A, Mylonakis E, Wikonkal NM, Hamblin MR. Sodium ascorbate kills Candida albicans in vitro via iron-catalyzed Fenton reaction: importance of oxygenation and metabolism. Future Microbiol 2016; 11:1535-1547. [PMID: 27855492 DOI: 10.2217/fmb-2016-0063] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM Ascorbate can inhibit growth and even decrease viability of various microbial species including Candida albicans. However the optimum conditions and the mechanism of action are unclear. Materials/methodology: Candida albicans shaken for 90 min in a buffered solution of ascorbate (90 mM) gave a 5-log reduction of cell viability, while there was no killing without shaking, in growth media with different carbon sources or at 4°C. Killing was inhibited by the iron chelator 2,2'-bipyridyl. Hydroxyphenyl fluorescein probe showed the intracellular generation of hydroxyl radicals. RESULTS/CONCLUSION Ascorbate-mediated killing of C. albicans depends on oxygenation and metabolism, involves iron-catalyzed generation of hydroxyl radicals via Fenton reaction and depletion of intracellular NADH. Ascorbate could serve as a component of a topical antifungal therapy.
Collapse
Affiliation(s)
- Pinar Avci
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA.,Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA.,Department of Dermatology, Venerology & Dermato-Oncology, Semmelweis University, Budapest 1085, Hungary
| | - Fernanda Freire
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA.,Department of Biosciences & Oral Diagnosis, Institute of Science & Technology, Universidade Estadual Paulista (UNESP), São José dos Campos, São Paulo 12245-000, Brazil
| | - Andras Banvolgyi
- Department of Dermatology, Venerology & Dermato-Oncology, Semmelweis University, Budapest 1085, Hungary
| | - Eleftherios Mylonakis
- Infectious Diseases Division, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, RI 02912, USA
| | - Norbert M Wikonkal
- Department of Dermatology, Venerology & Dermato-Oncology, Semmelweis University, Budapest 1085, Hungary
| | - Michael R Hamblin
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, MA 02114, USA.,Department of Dermatology, Harvard Medical School, Boston, MA 02115, USA.,Harvard-MIT Division of Health Sciences & Technology, Cambridge, MA 02139, USA
| |
Collapse
|
623
|
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: 200] [Impact Index Per Article: 25.0] [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
|
624
|
Mambetsariev N, Lin WW, Wallis AM, Stunz LL, Bishop GA. TRAF3 deficiency promotes metabolic reprogramming in B cells. Sci Rep 2016; 6:35349. [PMID: 27752131 PMCID: PMC5082756 DOI: 10.1038/srep35349] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 09/27/2016] [Indexed: 12/23/2022] Open
Abstract
The adaptor protein TNF receptor-associated factor 3 (TRAF3) is a critical regulator of B lymphocyte survival. B cell-specific TRAF3 deficiency results in enhanced viability and is associated with development of lymphoma and multiple myeloma. We show that TRAF3 deficiency led to induction of two proteins important for glucose metabolism, Glut1 and Hexokinase 2 (HXK2). This was associated with increased glucose uptake. In the absence of TRAF3, anaerobic glycolysis and oxidative phosphorylation were increased in B cells without changes in mitochondrial mass or reactive oxygen species. Chemical inhibition of glucose metabolism or glucose deprivation substantially attenuated the enhanced survival of TRAF3-deficient B cells, with a decrease in the pro-survival protein Mcl-1. Changes in Glut1 and Mcl-1 levels, glucose uptake and B cell number in the absence of TRAF3 were all dependent upon NF-κB inducing kinase (NIK). These results indicate that TRAF3 deficiency suffices to metabolically reprogram B cells, a finding that improves our understanding of the role of TRAF3 as a tumor suppressor, and suggests potential therapeutic strategies.
Collapse
Affiliation(s)
- Nurbek Mambetsariev
- Dept. of Microbiology, The University of Iowa, 3-403 Bowen Science Building, 51 Newton Road, Iowa City, IA 52242, USA
- Medical Scientist Training Program, The University of Iowa, Carver College of Medicine, 2206 MERF, Iowa City, IA 52242-2600, USA
- Immunology Graduate Program, 357 Medical Research Center, Iowa City, IA 52242-1182, USA
| | - Wai W. Lin
- Dept. of Microbiology, The University of Iowa, 3-403 Bowen Science Building, 51 Newton Road, Iowa City, IA 52242, USA
- Immunology Graduate Program, 357 Medical Research Center, Iowa City, IA 52242-1182, USA
| | - Alicia M. Wallis
- Dept. of Microbiology, The University of Iowa, 3-403 Bowen Science Building, 51 Newton Road, Iowa City, IA 52242, USA
- Immunology Graduate Program, 357 Medical Research Center, Iowa City, IA 52242-1182, USA
| | - Laura L. Stunz
- Dept. of Microbiology, The University of Iowa, 3-403 Bowen Science Building, 51 Newton Road, Iowa City, IA 52242, USA
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Gail A. Bishop
- Dept. of Microbiology, The University of Iowa, 3-403 Bowen Science Building, 51 Newton Road, Iowa City, IA 52242, USA
- Medical Scientist Training Program, The University of Iowa, Carver College of Medicine, 2206 MERF, Iowa City, IA 52242-2600, USA
- Immunology Graduate Program, 357 Medical Research Center, Iowa City, IA 52242-1182, USA
- Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, 200 Hawkins Drive, Iowa City, IA 52242, USA
- University of Iowa and DVA Medical Center, 601 Highway 6 West, Iowa City, IA 52246, USA
- Internal Medicine, 200 Hawkins Drive, Iowa City, Iowa 52242, USA
| |
Collapse
|
625
|
Toda K, Kawada K, Iwamoto M, Inamoto S, Sasazuki T, Shirasawa S, Hasegawa S, Sakai Y. Metabolic Alterations Caused by KRAS Mutations in Colorectal Cancer Contribute to Cell Adaptation to Glutamine Depletion by Upregulation of Asparagine Synthetase. Neoplasia 2016; 18:654-665. [PMID: 27764698 PMCID: PMC5071549 DOI: 10.1016/j.neo.2016.09.004] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Revised: 09/23/2016] [Accepted: 09/26/2016] [Indexed: 12/28/2022] Open
Abstract
A number of clinical trials have shown that KRAS mutations of colorectal cancer (CRC) can predict a lack of responses to anti-epidermal growth factor receptor-based therapy. Recently, there have been several studies to elucidate metabolism reprogramming in cancer. However, it remains to be investigated how mutated KRAS can coordinate the metabolic shift to sustain CRC tumor growth. In this study, we found that KRAS mutation in CRC caused alteration in amino acid metabolism. KRAS mutation causes a marked decrease in aspartate level and an increase in asparagine level in CRC. Using several human CRC cell lines and clinical specimens of primary CRC, we demonstrated that the expression of asparagine synthetase (ASNS), an enzyme that synthesizes asparagine from aspartate, was upregulated by mutated KRAS and that ASNS expression was induced by KRAS-activated signaling pathway, in particular PI3K-AKT-mTOR pathway. Importantly, we demonstrated that KRAS-mutant CRC cells could become adaptive to glutamine depletion through asparagine biosynthesis by ASNS and that asparagine addition could rescue the inhibited growth and viability of cells grown under the glutamine-free condition in vitro. Notably, a pronounced growth suppression of KRAS-mutant CRC was observed upon ASNS knockdown in vivo. Furthermore, combination of L-asparaginase plus rapamycin markedly suppressed the growth of KRAS-mutant CRC xenografts in vivo, whereas either L-asparaginase or rapamycin alone was not effective. These results indicate ASNS might be a novel therapeutic target against CRCs with mutated KRAS.
Collapse
Affiliation(s)
- Kosuke Toda
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Kenji Kawada
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan.
| | - Masayoshi Iwamoto
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | - Susumu Inamoto
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| | | | - Senji Shirasawa
- Departments of Cell Biology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Suguru Hasegawa
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan; Gastroenterological Surgery, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Yoshiharu Sakai
- Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
626
|
Antioxidant Activity during Tumor Progression: A Necessity for the Survival of Cancer Cells? Cancers (Basel) 2016; 8:cancers8100092. [PMID: 27754368 PMCID: PMC5082382 DOI: 10.3390/cancers8100092] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 09/30/2016] [Accepted: 10/07/2016] [Indexed: 01/22/2023] Open
Abstract
Antioxidant defenses encompass a variety of distinct compounds and enzymes that are linked together through their capacity to neutralize and scavenge reactive oxygen species (ROS). While the relationship between ROS and tumorigenesis is clearly complex and context dependent, a number of recent studies have suggested that neutralizing ROS can facilitate tumor progression and metastasis in multiple cancer types through distinct mechanisms. These studies therefore infer that antioxidant activity may be necessary to support the viability and/or the invasive capacity of cancer cells during tumor progression and metastasis. Here, we discuss some of the accumulating evidence suggesting a role for antioxidant activity in facilitating tumor progression.
Collapse
|
627
|
Campbell EJ, Vissers MCM, Wohlrab C, Hicks KO, Strother RM, Bozonet SM, Robinson BA, Dachs GU. Pharmacokinetic and anti-cancer properties of high dose ascorbate in solid tumours of ascorbate-dependent mice. Free Radic Biol Med 2016; 99:451-462. [PMID: 27567539 DOI: 10.1016/j.freeradbiomed.2016.08.027] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 08/23/2016] [Accepted: 08/23/2016] [Indexed: 12/18/2022]
Abstract
Despite recent evidence for an anti-tumour role for high-dose ascorbate, potential mechanisms of action are still unclear. At mM concentrations that are achieved with high-dose intravenous administration, autoxidation of ascorbate can generate cytotoxic levels of H2O2. Ascorbate is also a required co-factor for the hydroxylases that suppress the transcription factor hypoxia-inducible factor (HIF-1). HIF-1 supports an aggressive tumour phenotype and is associated with poor prognosis, and previous studies have shown that optimizing intracellular ascorbate levels down-regulates HIF-1 activation. In this study we have simultaneously measured ascorbate concentrations and the HIF-1 pathway activity in tumour tissue following high dose ascorbate administration, and have studied tumour growth and physiology. Gulo-/- mice, a model of the human ascorbate dependency condition, were implanted with syngeneic Lewis lung tumours, 1g/kg ascorbate was administered into the peritoneum, and ascorbate concentrations were monitored in plasma, liver and tumours. Ascorbate levels peaked within 30min, and although plasma and liver ascorbate returned to baseline within 16h, tumour levels remained elevated for 48h, possibly reflecting increased stability in the hypoxic tumour environment. The expression of HIF-1 and its target proteins was down-regulated with tumour ascorbate uptake. Elevated tumour ascorbate levels could be maintained with daily administration, and HIF-1 and vascular endothelial growth factor protein levels were reduced in these conditions. Increased tumour ascorbate was associated with slowed tumour growth, reduced tumour microvessel density and decreased hypoxia. Alternate day administration of ascorbate resulted in lower tumour levels and did not consistently decrease HIF-1 pathway activity. Levels of sodium-dependent vitamin C transporters 1 and 2 were not clearly associated with ascorbate accumulation by murine tumour cells in vitro or in vivo. Our results support the suppression of the hypoxic response by ascorbate as a plausible mechanism of action of its anti-tumour activity, and this may be useful in a clinical setting.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/blood
- Antineoplastic Agents/pharmacokinetics
- Antineoplastic Agents/pharmacology
- Antioxidants/metabolism
- Antioxidants/pharmacokinetics
- Antioxidants/pharmacology
- Ascorbic Acid/blood
- Ascorbic Acid/pharmacokinetics
- Ascorbic Acid/pharmacology
- Carcinoma, Lewis Lung/drug therapy
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/metabolism
- Carcinoma, Lewis Lung/pathology
- Drug Administration Schedule
- Female
- Gene Expression Regulation, Neoplastic
- Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors
- Hypoxia-Inducible Factor 1, alpha Subunit/genetics
- Hypoxia-Inducible Factor 1, alpha Subunit/metabolism
- Injections, Intraperitoneal
- Isoenzymes/genetics
- Isoenzymes/metabolism
- Mice
- Mice, Knockout
- Signal Transduction
- Sodium-Coupled Vitamin C Transporters/genetics
- Sodium-Coupled Vitamin C Transporters/metabolism
- Vascular Endothelial Growth Factor A/antagonists & inhibitors
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
Collapse
Affiliation(s)
- Elizabeth J Campbell
- Mackenzie Cancer Research Group, Department of Pathology, University of Otago, Christchurch 8011, New Zealand
| | - Margreet C M Vissers
- Centre for Free Radical Research, Department of Pathology, University of Otago, Christchurch 8011, New Zealand
| | - Christina Wohlrab
- Mackenzie Cancer Research Group, Department of Pathology, University of Otago, Christchurch 8011, New Zealand
| | - Kevin O Hicks
- Auckland Cancer Society Research Centre, University of Auckland, Auckland 1142, New Zealand
| | - R Matthew Strother
- Canterbury Regional Cancer and Haematology Service, Canterbury District Health Board, Christchurch 8011, New Zealand
| | - Stephanie M Bozonet
- Centre for Free Radical Research, Department of Pathology, University of Otago, Christchurch 8011, New Zealand
| | - Bridget A Robinson
- Mackenzie Cancer Research Group, Department of Pathology, University of Otago, Christchurch 8011, New Zealand; Canterbury Regional Cancer and Haematology Service, Canterbury District Health Board, Christchurch 8011, New Zealand; Department of Medicine, University of Otago, Christchurch 8011, New Zealand
| | - Gabi U Dachs
- Mackenzie Cancer Research Group, Department of Pathology, University of Otago, Christchurch 8011, New Zealand.
| |
Collapse
|
628
|
Abstract
In recent years there has been a growing interest among cancer biologists in cancer metabolism. This Review summarizes past and recent advances in our understanding of the reprogramming of glucose metabolism in cancer cells, which is mediated by oncogenic drivers and by the undifferentiated character of cancer cells. The reprogrammed glucose metabolism in cancer cells is required to fulfil anabolic demands. This Review discusses the possibility of exploiting the reprogrammed glucose metabolism for therapeutic approaches that selectively target cancer cells.
Collapse
Affiliation(s)
- Nissim Hay
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois 60607 and Research and Development Section, Jesse Brown VA Medical Center, Chicago, Illinois 60612, USA
| |
Collapse
|
629
|
Zhang ZZ, Lee EE, Sudderth J, Yue Y, Zia A, Glass D, Deberardinis RJ, Wang RC. Glutathione Depletion, Pentose Phosphate Pathway Activation, and Hemolysis in Erythrocytes Protecting Cancer Cells from Vitamin C-induced Oxidative Stress. J Biol Chem 2016; 291:22861-22867. [PMID: 27660392 DOI: 10.1074/jbc.c116.748848] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Indexed: 01/02/2023] Open
Abstract
The discovery that oxidized vitamin C, dehydroascorbate (DHA), can induce oxidative stress and cell death in cancer cells has rekindled interest in the use of high dose vitamin C (VC) as a cancer therapy. However, high dose VC has shown limited efficacy in clinical trials, possibly due to the decreased bioavailability of oral VC. Because human erythrocytes express high levels of Glut1, take up DHA, and reduce it to VC, we tested how erythrocytes might impact high dose VC therapies. Cancer cells are protected from VC-mediated cell death when co-cultured with physiologically relevant numbers of erythrocytes. Pharmacological doses of VC induce oxidative stress, GSH depletion, and increased glucose flux through the oxidative pentose phosphate pathway (PPP) in erythrocytes. Incubation of erythrocytes with VC induced hemolysis, which was exacerbated in erythrocytes from glucose-6-phosphate dehydrogenase (G6PD) patients and rescued by antioxidants. Thus, erythrocytes protect cancer cells from VC-induced oxidative stress and undergo hemolysis in vitro, despite activation of the PPP. These results have implications on the use of high dose VC in ongoing clinical trials and highlight the importance of the PPP in the response to oxidative stress.
Collapse
Affiliation(s)
| | | | - Jessica Sudderth
- the Children's Medical Center Research Institute, the University of Texas Southwestern Medical Center, Dallas, Texas 75390, and
| | | | - Ayesha Zia
- Pediatrics, the University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Donald Glass
- From the Departments of Dermatology and.,the Eugene McDermott Center for Human Growth and Development, the University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Ralph J Deberardinis
- the Children's Medical Center Research Institute, the University of Texas Southwestern Medical Center, Dallas, Texas 75390, and.,Pediatrics, the University of Texas Southwestern Medical Center, Dallas, Texas 75390.,the Eugene McDermott Center for Human Growth and Development, the University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | | |
Collapse
|
630
|
Vitamin C in synergism with cisplatin induces cell death in cervical cancer cells through altered redox cycling and p53 upregulation. J Cancer Res Clin Oncol 2016; 142:2503-2514. [DOI: 10.1007/s00432-016-2235-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 08/30/2016] [Indexed: 12/16/2022]
|
631
|
Parkin and mitophagy in cancer. Oncogene 2016; 36:1315-1327. [PMID: 27593930 DOI: 10.1038/onc.2016.302] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Revised: 07/04/2016] [Accepted: 07/04/2016] [Indexed: 02/07/2023]
Abstract
Mitophagy, the selective engulfment and clearance of mitochondria, is essential for the homeostasis of a healthy network of functioning mitochondria and prevents excessive production of cytotoxic reactive oxygen species from damaged mitochondria. The mitochondrially targeted PTEN-induced kinase-1 (PINK1) and the E3 ubiquitin ligase Parkin are well-established synergistic mediators of the mitophagy of dysfunctional mitochondria. This pathway relies on the ubiquitination of a number of mitochondrial outer membrane substrates and subsequent docking of autophagy receptor proteins to selectively clear mitochondria. There are also alternate Parkin-independent mitophagy pathways mediated by BCL2/adenovirus E1B 19 kDa protein-interacting protein 3 and Nip-3 like protein X as well as other effectors. There is increasing evidence that ablation of mitophagy accelerates a number of pathologies. Familial Parkinsonism is associated with loss-of-function mutations in PINK1 and Parkin. A growing number of studies have observed a correlation between impaired Parkin activity and enhanced cancer development, leading to the emerging concept that Parkin activity, or mitophagy in general, is a tumour suppression mechanism. This review examines the molecular mechanisms of mitophagy and highlights the potential links between Parkin and the hallmarks of cancer that may influence tumour development and progression.
Collapse
|
632
|
Padayatty SJ, Levine M. Vitamin C: the known and the unknown and Goldilocks. Oral Dis 2016; 22:463-93. [PMID: 26808119 PMCID: PMC4959991 DOI: 10.1111/odi.12446] [Citation(s) in RCA: 403] [Impact Index Per Article: 50.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 01/08/2016] [Indexed: 12/11/2022]
Abstract
Vitamin C (Ascorbic Acid), the antiscorbutic vitamin, cannot be synthesized by humans and other primates, and has to be obtained from diet. Ascorbic acid is an electron donor and acts as a cofactor for fifteen mammalian enzymes. Two sodium-dependent transporters are specific for ascorbic acid, and its oxidation product dehydroascorbic acid is transported by glucose transporters. Ascorbic acid is differentially accumulated by most tissues and body fluids. Plasma and tissue vitamin C concentrations are dependent on amount consumed, bioavailability, renal excretion, and utilization. To be biologically meaningful or to be clinically relevant, in vitro and in vivo studies of vitamin C actions have to take into account physiologic concentrations of the vitamin. In this paper, we review vitamin C physiology; the many phenomena involving vitamin C where new knowledge has accrued or where understanding remains limited; raise questions about the vitamin that remain to be answered; and explore lines of investigations that are likely to be fruitful.
Collapse
Affiliation(s)
- S J Padayatty
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - M Levine
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
633
|
Abstract
Vitamin C deficiency is found in patients with cancer and might complicate various therapy paradigms. Here we show how this deficiency may influence the use of DNA methyltransferase inhibitors (DNMTis) for treatment of hematological neoplasias. In vitro, when vitamin C is added at physiological levels to low doses of the DNMTi 5-aza-2'-deoxycytidine (5-aza-CdR), there is a synergistic inhibition of cancer-cell proliferation and increased apoptosis. These effects are associated with enhanced immune signals including increased expression of bidirectionally transcribed endogenous retrovirus (ERV) transcripts, increased cytosolic dsRNA, and activation of an IFN-inducing cellular response. This synergistic effect is likely the result of both passive DNA demethylation by DNMTi and active conversion of 5-methylcytosine (5mC) to 5-hydroxymethylcytosine (5hmC) by ten-eleven translocation (TET) enzymes at LTR regions of ERVs, because vitamin C acts as a cofactor for TET proteins. In addition, TET2 knockout reduces the synergy between the two compounds. Furthermore, we show that many patients with hematological neoplasia are markedly vitamin C deficient. Thus, our data suggest that correction of vitamin C deficiency in patients with hematological and other cancers may improve responses to epigenetic therapy with DNMTis.
Collapse
|
634
|
Li Q, Chen P, Fan Y, Wang X, Xu K, Li L, Tang B. Multicolor Fluorescence Detection-Based Microfluidic Device for Single-Cell Metabolomics: Simultaneous Quantitation of Multiple Small Molecules in Primary Liver Cells. Anal Chem 2016; 88:8610-6. [DOI: 10.1021/acs.analchem.6b01775] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Qingling Li
- College of Chemistry, Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry of Education,
Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, P.R. China
| | - Peilin Chen
- College of Chemistry, Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry of Education,
Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, P.R. China
| | - Yuanyuan Fan
- College of Chemistry, Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry of Education,
Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, P.R. China
| | - Xu Wang
- College of Chemistry, Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry of Education,
Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, P.R. China
| | - Kehua Xu
- College of Chemistry, Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry of Education,
Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, P.R. China
| | - Lu Li
- College of Chemistry, Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry of Education,
Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, P.R. China
| | - Bo Tang
- College of Chemistry, Chemical
Engineering and Materials Science, Collaborative Innovation Center
of Functionalized Probes for Chemical Imaging in Universities of Shandong,
Key Laboratory of Molecular and Nano Probes, Ministry of Education,
Shandong Provincial Key Laboratory of Clean Production of Fine Chemicals, Shandong Normal University, Jinan, 250014, P.R. China
| |
Collapse
|
635
|
Armitage EG, Southam AD. Monitoring cancer prognosis, diagnosis and treatment efficacy using metabolomics and lipidomics. Metabolomics 2016; 12:146. [PMID: 27616976 PMCID: PMC4987388 DOI: 10.1007/s11306-016-1093-7] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Accepted: 08/02/2016] [Indexed: 02/06/2023]
Abstract
INTRODUCTION Cellular metabolism is altered during cancer initiation and progression, which allows cancer cells to increase anabolic synthesis, avoid apoptosis and adapt to low nutrient and oxygen availability. The metabolic nature of cancer enables patient cancer status to be monitored by metabolomics and lipidomics. Additionally, monitoring metabolic status of patients or biological models can be used to greater understand the action of anticancer therapeutics. OBJECTIVES Discuss how metabolomics and lipidomics can be used to (i) identify metabolic biomarkers of cancer and (ii) understand the mechanism-of-action of anticancer therapies. Discuss considerations that can maximize the clinical value of metabolic cancer biomarkers including case-control, prognostic and longitudinal study designs. METHODS A literature search of the current relevant primary research was performed. RESULTS Metabolomics and lipidomics can identify metabolic signatures that associate with cancer diagnosis, prognosis and disease progression. Discriminatory metabolites were most commonly linked to lipid or energy metabolism. Case-control studies outnumbered prognostic and longitudinal approaches. Prognostic studies were able to correlate metabolic features with future cancer risk, whereas longitudinal studies were most effective for studying cancer progression. Metabolomics and lipidomics can help to understand the mechanism-of-action of anticancer therapeutics and mechanisms of drug resistance. CONCLUSION Metabolomics and lipidomics can be used to identify biomarkers associated with cancer and to better understand anticancer therapies.
Collapse
Affiliation(s)
- Emily G. Armitage
- Centre for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, Universidad CEU San Pablo, Campus Monteprincipe, Boadilla del Monte, 28668 Madrid, Spain
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA UK
- Glasgow Polyomics, Wolfson Wohl Cancer Research Centre, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, G61 1QH UK
| | - Andrew D. Southam
- School of Biosciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT UK
| |
Collapse
|
636
|
Herbel C, Patsoukis N, Bardhan K, Seth P, Weaver JD, Boussiotis VA. Clinical significance of T cell metabolic reprogramming in cancer. Clin Transl Med 2016; 5:29. [PMID: 27510264 PMCID: PMC4980327 DOI: 10.1186/s40169-016-0110-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 07/15/2016] [Indexed: 02/06/2023] Open
Abstract
Conversion of normal cells to cancer is accompanied with changes in their metabolism. During this conversion, cell metabolism undergoes a shift from oxidative phosphorylation to aerobic glycolysis, also known as Warburg effect, which is a hallmark for cancer cell metabolism. In cancer cells, glycolysis functions in parallel with the TCA cycle and other metabolic pathways to enhance biosynthetic processes and thus support proliferation and growth. Similar metabolic features are observed in T cells during activation but, in contrast to cancer, metabolic transitions in T cells are part of a physiological process. Currently, there is intense interest in understanding the cause and effect relationship between metabolic reprogramming and T cell differentiation. After the recent success of cancer immunotherapy, the crosstalk between immune system and cancer has come to the forefront of clinical and basic research. One of the key goals is to delineate how metabolic alterations of cancer influence metabolism-regulated function and differentiation of tumor resident T cells and how such effects might be altered by immunotherapy. Here, we review the unique metabolic features of cancer, the implications of cancer metabolism on T cell metabolic reprogramming during antigen encounters, and the translational prospective of harnessing metabolism in cancer and T cells for cancer therapy.
Collapse
Affiliation(s)
- Christoph Herbel
- Division of Hematology-Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Nikolaos Patsoukis
- Division of Hematology-Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Kankana Bardhan
- Division of Hematology-Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Pankaj Seth
- Division of Hematology-Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA.,Beth Israel Deaconess Cancer Center, Harvard Medical School, 330 Brookline Avenue, Dana 513, Boston, MA, 02215, USA.,Division of Interdisciplinary Medicine and Biotechnology, Beth Israel Deaconess Medical Center, Boston, USA
| | - Jessica D Weaver
- Division of Hematology-Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Vassiliki A Boussiotis
- Division of Hematology-Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA. .,Beth Israel Deaconess Cancer Center, Harvard Medical School, 330 Brookline Avenue, Dana 513, Boston, MA, 02215, USA.
| |
Collapse
|
637
|
Chio IIC, Jafarnejad SM, Ponz-Sarvise M, Park Y, Rivera K, Palm W, Wilson J, Sangar V, Hao Y, Öhlund D, Wright K, Filippini D, Lee EJ, Da Silva B, Schoepfer C, Wilkinson JE, Buscaglia JM, DeNicola GM, Tiriac H, Hammell M, Crawford HC, Schmidt EE, Thompson CB, Pappin DJ, Sonenberg N, Tuveson DA. NRF2 Promotes Tumor Maintenance by Modulating mRNA Translation in Pancreatic Cancer. Cell 2016; 166:963-976. [PMID: 27477511 DOI: 10.1016/j.cell.2016.06.056] [Citation(s) in RCA: 288] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2015] [Revised: 05/05/2016] [Accepted: 06/29/2016] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer is a deadly malignancy that lacks effective therapeutics. We previously reported that oncogenic Kras induced the redox master regulator Nfe2l2/Nrf2 to stimulate pancreatic and lung cancer initiation. Here, we show that NRF2 is necessary to maintain pancreatic cancer proliferation by regulating mRNA translation. Specifically, loss of NRF2 led to defects in autocrine epidermal growth factor receptor (EGFR) signaling and oxidation of specific translational regulatory proteins, resulting in impaired cap-dependent and cap-independent mRNA translation in pancreatic cancer cells. Combined targeting of the EGFR effector AKT and the glutathione antioxidant pathway mimicked Nrf2 ablation to potently inhibit pancreatic cancer ex vivo and in vivo, representing a promising synthetic lethal strategy for treating the disease.
Collapse
Affiliation(s)
- Iok In Christine Chio
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Seyed Mehdi Jafarnejad
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| | - Mariano Ponz-Sarvise
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Youngkyu Park
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Keith Rivera
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Wilhelm Palm
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - John Wilson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Vineet Sangar
- Institute of Systems Biology, 401 Terry Avenue N, Seattle, WA 98109, USA
| | - Yuan Hao
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Daniel Öhlund
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Kevin Wright
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Dea Filippini
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Eun Jung Lee
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Brandon Da Silva
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Christina Schoepfer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY 11724, USA
| | - John Erby Wilkinson
- Departments of Molecular & Integrative Physiology and Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jonathan M Buscaglia
- Division of Gastroenterology, Department of Medicine, Stony Brook University School of Medicine, Stony Brook, NY 11794, USA
| | - Gina M DeNicola
- Meyer Cancer Center, Weill Cornell Medical College, New York, NY 10021, USA
| | - Herve Tiriac
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Molly Hammell
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Howard C Crawford
- Departments of Molecular & Integrative Physiology and Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Edward E Schmidt
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59718, USA
| | - Craig B Thompson
- Cancer Biology and Genetics Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Darryl J Pappin
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | - Nahum Sonenberg
- Department of Biochemistry and Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA; Lustgarten Foundation Pancreatic Cancer Research Laboratory, Cold Spring Harbor, NY 11724, USA.
| |
Collapse
|
638
|
Abstract
Decades ago, Otto Warburg observed that cancers ferment glucose in the presence of oxygen, suggesting that defects in mitochondrial respiration may be the underlying cause of cancer. We now know that the genetic events that drive aberrant cancer cell proliferation also alter biochemical metabolism, including promoting aerobic glycolysis, but do not typically impair mitochondrial function. Mitochondria supply energy; provide building blocks for new cells; and control redox homeostasis, oncogenic signaling, innate immunity, and apoptosis. Indeed, mitochondrial biogenesis and quality control are often upregulated in cancers. While some cancers have mutations in nuclear-encoded mitochondrial tricarboxylic acid (TCA) cycle enzymes that produce oncogenic metabolites, there is negative selection for pathogenic mitochondrial genome mutations. Eliminating mtDNA limits tumorigenesis, and rare human tumors with mutant mitochondrial genomes are relatively benign. Thus, mitochondria play a central and multifunctional role in malignant tumor progression, and targeting mitochondria provides therapeutic opportunities.
Collapse
Affiliation(s)
- Wei-Xing Zong
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers University, 164 Frelinghuysen Road, Piscataway, NJ 08854, USA; Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA
| | - Joshua D Rabinowitz
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA; Lewis-Sigler Institute for Integrative Genomics and Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Eileen White
- Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08903, USA; Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854, USA.
| |
Collapse
|
639
|
Topically applied ascorbic acid solution for the treatment of basal cell carcinoma (BCC). J Am Acad Dermatol 2016; 75:212-3. [DOI: 10.1016/j.jaad.2016.04.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 03/31/2016] [Accepted: 04/03/2016] [Indexed: 11/21/2022]
|
640
|
Looyenga B, VanOpstall C, Lee Z, Bell J, Lodge E, Wrobel K, Arnoys E, Louters L. Determination of GLUT1 Oligomerization Parameters using Bioluminescent Förster Resonance Energy Transfer. Sci Rep 2016; 6:29130. [PMID: 27357903 PMCID: PMC4928127 DOI: 10.1038/srep29130] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 06/15/2016] [Indexed: 12/24/2022] Open
Abstract
The facilitated glucose transporter GLUT1 (SLC2A1) is an important mediator of glucose homeostasis in humans. Though it is found in most cell types to some extent, the level of GLUT1 expression across different cell types can vary dramatically. Prior studies in erythrocytes-which express particularly high levels of GLUT1-have suggested that GLUT1 is able to form tetrameric complexes with enhanced transport activity. Whether dynamic aggregation of GLUT1 also occurs in cell types with more modest expression of GLUT1, however, is unclear. To address this question, we developed a genetically encoded bioluminescent Förster resonance energy transfer (BRET) assay using the luminescent donor Nanoluciferase and fluorescent acceptor mCherry. By tethering these proteins to the N-terminus of GLUT1 and performing saturation BRET analysis, we were able to demonstrate the formation of multimeric complexes in live cells. Parallel use of flow cytometry and immunoblotting further enabled us to estimate the density of GLUT1 proteins required for spontaneous oligomerization. These data provide new insights into the physiological relevance of GLUT1 multimerization as well as a new variant of BRET assay that is useful for measuring the interactions among other cell membrane proteins in live cells.
Collapse
Affiliation(s)
- Brendan Looyenga
- Calvin College, Department of Chemistry &Biochemistry, 3201 Burton St SE, Grand Rapids, MI, 49546, USA
| | - Calvin VanOpstall
- Calvin College, Department of Chemistry &Biochemistry, 3201 Burton St SE, Grand Rapids, MI, 49546, USA
| | - Zion Lee
- Calvin College, Department of Chemistry &Biochemistry, 3201 Burton St SE, Grand Rapids, MI, 49546, USA
| | - Jed Bell
- Calvin College, Department of Chemistry &Biochemistry, 3201 Burton St SE, Grand Rapids, MI, 49546, USA
| | - Evans Lodge
- Calvin College, Department of Chemistry &Biochemistry, 3201 Burton St SE, Grand Rapids, MI, 49546, USA
| | - Katherine Wrobel
- Calvin College, Department of Chemistry &Biochemistry, 3201 Burton St SE, Grand Rapids, MI, 49546, USA
| | - Eric Arnoys
- Calvin College, Department of Chemistry &Biochemistry, 3201 Burton St SE, Grand Rapids, MI, 49546, USA
| | - Larry Louters
- Calvin College, Department of Chemistry &Biochemistry, 3201 Burton St SE, Grand Rapids, MI, 49546, USA
| |
Collapse
|
641
|
Banerjee N, Kim H, Talcott ST, Turner ND, Byrne DH, Mertens-Talcott SU. Plum polyphenols inhibit colorectal aberrant crypt foci formation in rats: potential role of the miR-143/protein kinase B/mammalian target of rapamycin axis. Nutr Res 2016; 36:1105-1113. [PMID: 27865352 DOI: 10.1016/j.nutres.2016.06.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 05/18/2016] [Accepted: 06/10/2016] [Indexed: 12/25/2022]
Abstract
The nutritional prevention of aberrant crypt foci by polyphenols may be a crucial step to dietary cancer prevention. The objective of this study was to determine the underlying mechanisms that contribute to the anti-inflammatory and antitumorigenic properties of plum (Prunus salicina L.) polyphenols, including chlorogenic acid and neochlorogenic acid, in azoxymethane (AOM)-treated rats. The hypothesis was that plum polyphenolics suppress AOM-induced aberrant crypt foci formation through alterations in the protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway and relative micro-RNA expressions. Sprague-Dawley rats (n=10/group) received plum beverage (1346mg gallic acid equivalents/L) or a control beverage ad libitum for 10 weeks with subcutaneous injections of AOM (15mg/kg) at weeks 2 and 3. Results show that the consumption of the plum beverage decreased the number of dysplastic aberrant crypt foci by 48% (P<.05) and lowered proliferation of mucosal cells by 24% (P<.05). The plum beverage decreased the activity of glutathione peroxidase, superoxide dismutase, and catalase in mucosal scrapings, as well as the superoxide dismutase activity in serum. The results were accompanied by a down-regulation of proinflammatory enzymes nuclear factor κB, nitric oxide synthase, cyclooxygenase-2, and vascular cell adhesion molecule 1 messenger RNA. Plum inhibited the expression of AKT and mTOR messenger RNA, phosphorylated AKT, mTOR, and hypoxia-inducible factor-1α protein levels, and the ratio of the phosphorylated/total protein expression of mTOR. Also, the plum beverage increased the expression of miR-143, which is involved in the regulation of AKT. These results suggest that plum polyphenols may exhibit a chemopreventive potential against colon carcinogenesis by impacting the AKT/mTOR pathway and miR-143.
Collapse
Affiliation(s)
- Nivedita Banerjee
- Interdisciplinary Program of Toxicology, Texas A&M University, College Station, TX 77843, USA
| | - Hyemee Kim
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Stephen T Talcott
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - Nancy D Turner
- Interdisciplinary Program of Toxicology, Texas A&M University, College Station, TX 77843, USA; Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA
| | - David H Byrne
- Department of Horticulture, Texas A&M University, College Station, TX 77843, USA
| | - Susanne U Mertens-Talcott
- Department of Nutrition and Food Science, Texas A&M University, College Station, TX 77843, USA; Department of Veterinary Physiology & Pharmacology, Texas A&M University, College Station, TX 77843, USA.
| |
Collapse
|
642
|
Ratnikov BI, Scott DA, Osterman AL, Smith JW, Ronai ZA. Metabolic rewiring in melanoma. Oncogene 2016; 36:147-157. [PMID: 27270434 PMCID: PMC5140782 DOI: 10.1038/onc.2016.198] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 04/07/2016] [Accepted: 04/07/2016] [Indexed: 02/07/2023]
Abstract
Oncogene-driven metabolic rewiring is an adaptation to low nutrient and oxygen conditions in the tumor microenvironment that enables cancer cells of diverse origin to hyperproliferate. Aerobic glycolysis and enhanced reliance on glutamine utilization are prime examples of such rewiring. However, tissue of origin as well as specific genetic and epigenetic changes determines gene expression profiles underlying these metabolic alterations in specific cancers. In melanoma, activation of the mitogen-activated protein kinase (MAPK) pathway driven by mutant BRAF or NRAS is a primary cause of malignant transformation. Activity of the MAPK pathway, as well as other factors, such as HIF1α, Myc and MITF, are among those that control the balance between non-oxidative and oxidative branches of central carbon metabolism. Here, we discuss the nature of metabolic alterations that underlie melanoma development and affect its response to therapy.
Collapse
Affiliation(s)
- B I Ratnikov
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla CA, USA
| | - D A Scott
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla CA, USA
| | - A L Osterman
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla CA, USA
| | - J W Smith
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla CA, USA
| | - Z A Ronai
- Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla CA, USA
| |
Collapse
|
643
|
Wang X, Hai C. Novel insights into redox system and the mechanism of redox regulation. Mol Biol Rep 2016; 43:607-28. [DOI: 10.1007/s11033-016-4022-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 05/26/2016] [Indexed: 12/20/2022]
|
644
|
Rouleau L, Antony AN, Bisetto S, Newberg A, Doria C, Levine M, Monti DA, Hoek JB. Synergistic effects of ascorbate and sorafenib in hepatocellular carcinoma: New insights into ascorbate cytotoxicity. Free Radic Biol Med 2016; 95:308-322. [PMID: 27036367 PMCID: PMC4867251 DOI: 10.1016/j.freeradbiomed.2016.03.031] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 03/24/2016] [Accepted: 03/28/2016] [Indexed: 12/15/2022]
Abstract
We investigated the mechanism of selective ascorbate-induced cytotoxicity in tumor cells, including Hep G2 cells, compared to primary hepatocytes. H2O2 formation was required for ascorbate cytotoxicity, as extracellular catalase treatment protected tumor cells. H2O2 generated by glucose oxidase treatment also caused cell killing, but treatment with a pharmacologic dose (5-20mM) of ascorbate was significantly more cytotoxic at comparable rates of H2O2 production, suggesting that ascorbate enhanced H2O2 cytotoxicity. This was further supported by the finding that ascorbate at a non-cytotoxic dose (1mM) enhanced cell killing caused by glucose oxidase. Consistent with this conclusion, ascorbate treatment caused deregulation of cellular calcium homeostasis, resulting in massive mitochondrial calcium accumulation. Ascorbate acted synergistically with the chemotherapeutic sorafenib in killing Hep G2 cells, but not primary hepatocytes, suggesting adjuvant ascorbate treatment can broaden sorafenib's therapeutic range. Sorafenib caused mitochondrial depolarization and prevented mitochondrial calcium sequestration. Subsequent ascorbate addition further deregulated cellular calcium homeostasis promoting cell death. Additionally, we present the case of a patient with hepatocellular carcinoma (HCC) who had prolonged regression of a rib metastasis upon combination treatment with ascorbate and sorafenib, indicating that these studies have direct clinical relevance.
Collapse
Affiliation(s)
- Lauren Rouleau
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Anil Noronha Antony
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Sara Bisetto
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Andrew Newberg
- Jefferson-Myrna Brind Center of Integrative Medicine, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Cataldo Doria
- Division of Transplantation, Liver Tumor Center, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Mark Levine
- Molecular and Clinical Nutrition Section, Digestive Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Daniel A Monti
- Jefferson-Myrna Brind Center of Integrative Medicine, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Jan B Hoek
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
645
|
Abstract
New data show that as dermal fibroblasts grow old, they increase their secretion of the WNT antagonist sFRP2 to drive melanoma cell metastasis. sFRP2 suppresses β-catenin and MITF signaling in melanoma cells, downregulating the redox regulator APE1, making melanoma cells more sensitive to oxidative stress and driving resistance to BRAF inhibitors. Thus, the aging microenvironment in elderly patient skin activates a signaling pathway that drives more aggressive melanoma cell behavior. Cancer Discov; 6(6); 581-3. ©2016 AACR.
Collapse
Affiliation(s)
- Amaya Virós
- Molecular Oncology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom.
| | - Maria Romina Girotti
- Molecular Oncology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Richard Marais
- Molecular Oncology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
646
|
Jung SA, Lee DH, Moon JH, Hong SW, Shin JS, Hwang IY, Shin YJ, Kim JH, Gong EY, Kim SM, Lee EY, Lee S, Kim JE, Kim KP, Hong YS, Lee JS, Jin DH, Kim T, Lee WJ. L-Ascorbic acid can abrogate SVCT-2-dependent cetuximab resistance mediated by mutant KRAS in human colon cancer cells. Free Radic Biol Med 2016; 95:200-8. [PMID: 27012422 DOI: 10.1016/j.freeradbiomed.2016.03.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2015] [Revised: 03/03/2016] [Accepted: 03/09/2016] [Indexed: 01/05/2023]
Abstract
Colon cancer patients with mutant KRAS are resistant to cetuximab, an antibody directed against the epidermal growth factor receptor, which is an effective clinical therapy for patients with wild-type KRAS. Numerous combinatorial therapies have been tested to overcome the resistance to cetuximab. However, no combinations have been found that can be used as effective therapeutic strategies. In this study, we demonstrate that L-ascorbic acid partners with cetuximab to induce killing effects, which are influenced by sodium-dependent vitamin C transporter 2 (SVCT-2) in human colon cancer cells with a mutant KRAS. L-Ascorbic acid treatment of human colon cancer cells that express a mutant KRAS differentially and synergistically induced cell death with cetuximab in a SVCT-2-dependent manner. The ectopic expression of SVCT-2 induced sensitivity to L-ascorbic acid treatment in human colon cancer cells that do not express SVCT-2, whereas the knockdown of endogenous SVCT-2 induced resistance to L-ascorbic acid treatment in SVCT-2-positive cells. Moreover, tumor regression via the administration of L-ascorbic acid and cetuximab in mice bearing tumor cell xenografts corresponded to SVCT-2 protein levels. Interestingly, cell death induced by the combination of L-ascorbic acid and cetuximab resulted in both apoptotic and necrotic cell death. These cell death mechanisms were related to a disruption of the ERK pathway and were represented by the impaired activation of RAFs and the activation of the ASK-1-p38 pathway. Taken together, these results suggest that resistance to cetuximab in human colon cancer patients with a mutant KRAS can be bypassed by L-ascorbic acid in an SVCT-2-dependent manner. Furthermore, SVCT-2 in mutant KRAS colon cancer may act as a potent marker for potentiating L-ascorbic acid co-treatment with cetuximab.
Collapse
Affiliation(s)
- Soo-A Jung
- Innovative Cancer Research, Asan Institute for Life Science, Republic of Korea; Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, Republic of Korea
| | - Dae-Hee Lee
- Innovative Cancer Research, Asan Institute for Life Science, Republic of Korea; Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, Republic of Korea
| | - Jai-Hee Moon
- Innovative Cancer Research, Asan Institute for Life Science, Republic of Korea; Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, Republic of Korea
| | - Seung-Woo Hong
- Innovative Cancer Research, Asan Institute for Life Science, Republic of Korea; Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, Republic of Korea
| | - Jae-Sik Shin
- Innovative Cancer Research, Asan Institute for Life Science, Republic of Korea; Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, Republic of Korea
| | - Ih Yeon Hwang
- Innovative Cancer Research, Asan Institute for Life Science, Republic of Korea; Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, Republic of Korea
| | - Yu Jin Shin
- Innovative Cancer Research, Asan Institute for Life Science, Republic of Korea; Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, Republic of Korea
| | - Jeong Hee Kim
- Innovative Cancer Research, Asan Institute for Life Science, Republic of Korea; Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, Republic of Korea
| | - Eun-Yeung Gong
- Innovative Cancer Research, Asan Institute for Life Science, Republic of Korea; Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, Republic of Korea
| | - Seung-Mi Kim
- Innovative Cancer Research, Asan Institute for Life Science, Republic of Korea; Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, Republic of Korea
| | - Eun Young Lee
- Innovative Cancer Research, Asan Institute for Life Science, Republic of Korea; Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, Republic of Korea
| | - Seul Lee
- Innovative Cancer Research, Asan Institute for Life Science, Republic of Korea; Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, Republic of Korea
| | - Jeong Eun Kim
- Innovative Cancer Research, Asan Institute for Life Science, Republic of Korea; Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, Republic of Korea
| | - Kyu-Pyo Kim
- Innovative Cancer Research, Asan Institute for Life Science, Republic of Korea; Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, Republic of Korea
| | - Yong Sang Hong
- Innovative Cancer Research, Asan Institute for Life Science, Republic of Korea; Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, Republic of Korea
| | - Jung Shin Lee
- Innovative Cancer Research, Asan Institute for Life Science, Republic of Korea; Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, Republic of Korea
| | - Dong-Hoon Jin
- Innovative Cancer Research, Asan Institute for Life Science, Republic of Korea; Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, Republic of Korea; Department of Convergence Medicine, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, Republic of Korea.
| | - TaeWon Kim
- Innovative Cancer Research, Asan Institute for Life Science, Republic of Korea; Department of Oncology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympicro-43gil, Songpa-gu, Seoul, Republic of Korea.
| | - Wang Jae Lee
- Department of Anatomy and Tumor Immunity Medical Research Center, Seoul National University College of Medicine, Seoul 110-744, Republic of Korea
| |
Collapse
|
647
|
Abstract
Awareness that the metabolic phenotype of cells within tumours is heterogeneous - and distinct from that of their normal counterparts - is growing. In general, tumour cells metabolize glucose, lactate, pyruvate, hydroxybutyrate, acetate, glutamine, and fatty acids at much higher rates than their nontumour equivalents; however, the metabolic ecology of tumours is complex because they contain multiple metabolic compartments, which are linked by the transfer of these catabolites. This metabolic variability and flexibility enables tumour cells to generate ATP as an energy source, while maintaining the reduction-oxidation (redox) balance and committing resources to biosynthesis - processes that are essential for cell survival, growth, and proliferation. Importantly, experimental evidence indicates that metabolic coupling between cell populations with different, complementary metabolic profiles can induce cancer progression. Thus, targeting the metabolic differences between tumour and normal cells holds promise as a novel anticancer strategy. In this Review, we discuss how cancer cells reprogramme their metabolism and that of other cells within the tumour microenvironment in order to survive and propagate, thus driving disease progression; in particular, we highlight potential metabolic vulnerabilities that might be targeted therapeutically.
Collapse
|
648
|
Benhar M, Shytaj IL, Stamler JS, Savarino A. Dual targeting of the thioredoxin and glutathione systems in cancer and HIV. J Clin Invest 2016; 126:1630-9. [PMID: 27135880 PMCID: PMC4855928 DOI: 10.1172/jci85339] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Although the use of antioxidants for the treatment of cancer and HIV/AIDS has been proposed for decades, new insights gained from redox research have suggested a very different scenario. These new data show that the major cellular antioxidant systems, the thioredoxin (Trx) and glutathione (GSH) systems, actually promote cancer growth and HIV infection, while suppressing an effective immune response. Mechanistically, these systems control both the redox- and NO-based pathways (nitroso-redox homeostasis), which subserve innate and cellular immune defenses. Dual inhibition of the Trx and GSH systems synergistically kills neoplastic cells in vitro and in mice and decreases resistance to anticancer therapy. Similarly, the population of HIV reservoir cells that constitutes the major barrier to a cure for AIDS is exquisitely redox sensitive and could be selectively targeted by Trx and GSH inhibitors. Trx and GSH inhibition may lead to a reprogramming of the immune response, tilting the balance between the immune system and cancer or HIV in favor of the former, allowing elimination of diseased cells. Thus, therapies based on silencing of the Trx and GSH pathways represent a promising approach for the cure of both cancer and AIDS and warrant further investigation.
Collapse
Affiliation(s)
- Moran Benhar
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion – Israel Institute of Technology, Haifa, Israel
| | | | - Jonathan S. Stamler
- Institute for Transformative Molecular Medicine, Department of Medicine, and Harrington Discovery Institute, University Hospitals Case Medical Center, Cleveland, Ohio, USA
| | | |
Collapse
|
649
|
DeBerardinis RJ, Chandel NS. Fundamentals of cancer metabolism. SCIENCE ADVANCES 2016; 2:e1600200. [PMID: 27386546 PMCID: PMC4928883 DOI: 10.1126/sciadv.1600200] [Citation(s) in RCA: 1903] [Impact Index Per Article: 237.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 04/29/2016] [Indexed: 04/14/2023]
Abstract
Tumors reprogram pathways of nutrient acquisition and metabolism to meet the bioenergetic, biosynthetic, and redox demands of malignant cells. These reprogrammed activities are now recognized as hallmarks of cancer, and recent work has uncovered remarkable flexibility in the specific pathways activated by tumor cells to support these key functions. In this perspective, we provide a conceptual framework to understand how and why metabolic reprogramming occurs in tumor cells, and the mechanisms linking altered metabolism to tumorigenesis and metastasis. Understanding these concepts will progressively support the development of new strategies to treat human cancer.
Collapse
Affiliation(s)
- Ralph J. DeBerardinis
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Corresponding author. (R.J.D.); (N.S.C.)
| | - Navdeep S. Chandel
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
- Corresponding author. (R.J.D.); (N.S.C.)
| |
Collapse
|
650
|
sFRP2 in the aged microenvironment drives melanoma metastasis and therapy resistance. Nature 2016; 532:250-4. [PMID: 27042933 PMCID: PMC4833579 DOI: 10.1038/nature17392] [Citation(s) in RCA: 291] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 02/02/2016] [Indexed: 12/14/2022]
Abstract
Cancer is a disease of ageing. Clinically, aged cancer patients tend to have a poorer prognosis than young. This may be due to accumulated cellular damage, decreases in adaptive immunity, and chronic inflammation. However, the effects of the aged microenvironment on tumour progression have been largely unexplored. Since dermal fibroblasts can have profound impacts on melanoma progression, we examined whether age-related changes in dermal fibroblasts could drive melanoma metastasis and response to targeted therapy. Here we find that aged fibroblasts secrete a Wnt antagonist, sFRP2, which activates a multi-step signalling cascade in melanoma cells that results in a decrease in β-catenin and microphthalmia-associated transcription factor (MITF), and ultimately the loss of a key redox effector, APE1. Loss of APE1 attenuates the response of melanoma cells to DNA damage induced by reactive oxygen species, rendering the cells more resistant to targeted therapy (vemurafenib). Age-related increases in sFRP2 also augment both angiogenesis and metastasis of melanoma cells. These data provide an integrated view of how fibroblasts in the aged microenvironment contribute to tumour progression, offering new possibilities for the design of therapy for the elderly.
Collapse
|