551
|
Roh JL, Jang H, Kim EH, Shin D. Targeting of the Glutathione, Thioredoxin, and Nrf2 Antioxidant Systems in Head and Neck Cancer. Antioxid Redox Signal 2017; 27:106-114. [PMID: 27733046 DOI: 10.1089/ars.2016.6841] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
AIMS The glutathione (GSH), thioredoxin (Trx), and Nrf2 systems represent a major defense against reactive oxygen species (ROS), the cellular imbalance of which in cancer promotes growth and therapeutic resistance. This study investigated whether targeting the GSH, Trx, and Nrf2 antioxidant systems effectively eliminated head and neck cancer (HNC). RESULTS At high concentrations, auranofin, but not buthionine sulfoximine (BSO) alone, decreased the viability of HNC, whereas even at low concentrations, auranofin plus BSO synergized to kill HNC cells. Dual silencing of the genes for GCLM and TrxR1 induced GSH depletion, Trx activity inhibition, and ROS accumulation, synergistically killing HNC cells. Inhibition of the GSH and Trx systems resulted in activation of the Nrf2-antioxidant response element (ARE) pathway, which may result in suboptimal GSH and Trx inhibition where HNC is resistant. Genetic inhibition of Nrf2 and/or HO-1 or trigonelline enhanced growth suppression, ROS accumulation, and cell death from GSH and Trx inhibition. The in vivo effects of GSH, Trx, and Nrf2 system inhibition were confirmed in a mouse HNC xenograft model by achieving growth inhibition >60% compared with those of control. Innovations: This study is the first to show that triple inhibition of GSH, Trx, and Nrf2 pathways could be an effective method to overcome the resistance of HNC. CONCLUSIONS Inhibition of the Nrf2-ARE pathway in addition to dual inhibition of the GSH and Trx antioxidant systems can effectively eliminate resistant HNC. Antioxid. Redox Signal. 27, 106-114.
Collapse
Affiliation(s)
- Jong-Lyel Roh
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Republic of Korea
| | - Hyejin Jang
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Republic of Korea
| | - Eun Hye Kim
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Republic of Korea
| | - Daiha Shin
- Department of Otolaryngology, Asan Medical Center, University of Ulsan College of Medicine , Seoul, Republic of Korea
| |
Collapse
|
552
|
Oxidative Stress Gene Expression Profile Correlates with Cancer Patient Poor Prognosis: Identification of Crucial Pathways Might Select Novel Therapeutic Approaches. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:2597581. [PMID: 28770020 PMCID: PMC5523271 DOI: 10.1155/2017/2597581] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/30/2017] [Indexed: 12/17/2022]
Abstract
The role of altered redox status and high reactive oxygen species (ROS) is still controversial in cancer development and progression. Intracellular levels of ROS are elevated in cancer cells suggesting a role in cancer initiation and progression; on the contrary, ROS elevated levels may induce programmed cell death and have been associated with cancer suppression. Thus, it is crucial to consider the double-face of ROS, for novel therapeutic strategies targeting redox regulatory mechanisms. In this review, in order to derive cancer-type specific oxidative stress genes' profile and their potential prognostic role, we integrated a publicly available oxidative stress gene signature with patient survival data from the Cancer Genome Atlas database. Overall, we found several genes statistically significant associated with poor prognosis in the examined six tumor types. Among them, FoxM1 and thioredoxin reductase1 expression showed the same pattern in four out of six cancers, suggesting their specific critical role in cancer-related oxidative stress adaptation. Our analysis also unveiled an enriched cellular network, highlighting specific pathways, in which many genes are strictly correlated. Finally, we discussed novel findings on the correlation between oxidative stress and cancer stem cells in order to define those pathways to be prioritized in drug development.
Collapse
|
553
|
Biancur DE, Paulo JA, Małachowska B, Del Rey MQ, Sousa CM, Wang X, Sohn ASW, Chu GC, Gygi SP, Harper JW, Fendler W, Mancias JD, Kimmelman AC. Compensatory metabolic networks in pancreatic cancers upon perturbation of glutamine metabolism. Nat Commun 2017; 8:15965. [PMID: 28671190 PMCID: PMC5500878 DOI: 10.1038/ncomms15965] [Citation(s) in RCA: 210] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 05/16/2017] [Indexed: 12/22/2022] Open
Abstract
Pancreatic ductal adenocarcinoma is a notoriously difficult-to-treat cancer and patients are in need of novel therapies. We have shown previously that these tumours have altered metabolic requirements, making them highly reliant on a number of adaptations including a non-canonical glutamine (Gln) metabolic pathway and that inhibition of downstream components of Gln metabolism leads to a decrease in tumour growth. Here we test whether recently developed inhibitors of glutaminase (GLS), which mediates an early step in Gln metabolism, represent a viable therapeutic strategy. We show that despite marked early effects on in vitro proliferation caused by GLS inhibition, pancreatic cancer cells have adaptive metabolic networks that sustain proliferation in vitro and in vivo. We use an integrated metabolomic and proteomic platform to understand this adaptive response and thereby design rational combinatorial approaches. We demonstrate that pancreatic cancer metabolism is adaptive and that targeting Gln metabolism in combination with these adaptive responses may yield clinical benefits for patients.
Collapse
Affiliation(s)
- Douglas E. Biancur
- Division of Genomic Stability and DNA Repair, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Perlmutter Cancer Center, Department of Radiation Oncology, NYU Medical School, New York, New York 10016, USA
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Beata Małachowska
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz 91-738, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw 02-091, Poland
| | - Maria Quiles Del Rey
- Division of Genomic Stability and DNA Repair, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Cristovão M. Sousa
- Division of Genomic Stability and DNA Repair, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Xiaoxu Wang
- Division of Genomic Stability and DNA Repair, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
| | - Albert S. W. Sohn
- Perlmutter Cancer Center, Department of Radiation Oncology, NYU Medical School, New York, New York 10016, USA
| | - Gerald C. Chu
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - J. Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Wojciech Fendler
- Division of Genomic Stability and DNA Repair, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Lodz 91-738, Poland
| | - Joseph D. Mancias
- Division of Genomic Stability and DNA Repair, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Alec C. Kimmelman
- Division of Genomic Stability and DNA Repair, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215, USA
- Perlmutter Cancer Center, Department of Radiation Oncology, NYU Medical School, New York, New York 10016, USA
| |
Collapse
|
554
|
Toyokuni S, Ito F, Yamashita K, Okazaki Y, Akatsuka S. Iron and thiol redox signaling in cancer: An exquisite balance to escape ferroptosis. Free Radic Biol Med 2017; 108:610-626. [PMID: 28433662 DOI: 10.1016/j.freeradbiomed.2017.04.024] [Citation(s) in RCA: 161] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 04/11/2017] [Accepted: 04/18/2017] [Indexed: 02/06/2023]
Abstract
Epidemiological data indicate a constant worldwide increase in cancer mortality, although the age of onset is increasing. Recent accumulation of genomic data on human cancer via next-generation sequencing confirmed that cancer is a disease of genome alteration. In many cancers, the Nrf2 transcription system is activated via mutations either in Nrf2 or Keap1 ubiquitin ligase, leading to persistent activation of the genes with antioxidative functions. Furthermore, deep sequencing of passenger mutations is clarifying responsible cancer causative agent(s) in each case, including aging, APOBEC activation, smoking and UV. Therefore, it is most likely that oxidative stress is the principal initiating factor in carcinogenesis, with the involvement of two essential molecules for life, iron and oxygen. There is evidence based on epidemiological and animal studies that excess iron is a major risk for carcinogenesis, suggesting the importance of ferroptosis-resistance. Microscopic visualization of catalytic Fe(II) has recently become available. Although catalytic Fe(II) is largely present in lysosomes, proliferating cells harbor catalytic Fe(II) also in the cytosol and mitochondria. Oxidative stress catalyzed by Fe(II) is counteracted by thiol systems at different functional levels. Nitric oxide, carbon monoxide and hydrogen (per)sulfide modulate these reactions. Mitochondria generate not only energy but also heme/iron sulfur cluster cofactors and remain mostly dysfunctional in cancer cells, leading to Warburg effects. Cancer cells are under persistent oxidative stress with a delicate balance between catalytic iron and thiols, thereby escaping ferroptosis. Thus, high-dose L-ascorbate and non-thermal plasma as well as glucose/glutamine deprivation may provide additional benefits as cancer therapies over preexisting therapeutics.
Collapse
Affiliation(s)
- Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan; Sydney Medical School, The University of Sydney, NSW 2006, Australia.
| | - Fumiya Ito
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Kyoko Yamashita
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Yasumasa Okazaki
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Shinya Akatsuka
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| |
Collapse
|
555
|
Mvunta DH, Miyamoto T, Asaka R, Yamada Y, Ando H, Higuchi S, Ida K, Kashima H, Shiozawa T. SIRT1 Regulates the Chemoresistance and Invasiveness of Ovarian Carcinoma Cells. Transl Oncol 2017; 10:621-631. [PMID: 28667895 PMCID: PMC5491457 DOI: 10.1016/j.tranon.2017.05.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 05/15/2017] [Accepted: 05/16/2017] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND SIRT1 is a longevity gene that forestalls aging and age-related diseases including cancer, and has recently attracted widespread attention due to its overexpression in some cancers. We previously identified the overexpression of SIRT1 in ovarian carcinoma (OvCa) as a poor prognostic factor. However, mechanistic insights into the function of SIRT1 in OvCa have yet to be elucidated. METHODS Quantitative real-time reverse PCR (qRT-PCR) and Western blotting were employed to examine the expression of SIRT1 in a panel of human OvCa cell lines. si-RNA or sh-RNA and cDNA technologies were utilized to knockdown or overexpress SIRT1, respectively. The effects of SIRT1 on proliferation and chemoresistance were examined using a WST-1 assay, and the underlying mechanisms were confirmed using an apoptotic assay, and the quantification of glutathione (GSH), and reactive oxygen species (ROS). The aggressiveness of SIRT1 was analyzed using in vitro invasion and migration assays. RESULTS SIRT1 was more strongly expressed in OvCa cell lines than in the immortalized ovarian epithelium at the gene and protein levels. Stress up-regulated the expression of SIRT1 in dose- and time-dependent manners. SIRT1 significantly enhanced the proliferation (P<.05), chemoresistance (P<.05), and aggressiveness of OvCa cells by up-regulating multiple antioxidant pathways to inhibit oxidative stress. Further study into the overexpression of SIRT1 demonstrated the up-regulation of several stemness-associated genes and enrichment of CD44v9 via an as-yet-unidentified pathway. CONCLUSIONS Our results suggest that SIRT1 plays a role in the acquisition of aggressiveness and chemoresistance by OvCa, and has potential as a therapeutic target for OvCa.
Collapse
Affiliation(s)
- David Hamisi Mvunta
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Tsutomu Miyamoto
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan.
| | - Ryoichi Asaka
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Yasushi Yamada
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Hirofumi Ando
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Shotaro Higuchi
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Koichi Ida
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Hiroyasu Kashima
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| | - Tanri Shiozawa
- Department of Obstetrics and Gynecology, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto 390-8621, Japan
| |
Collapse
|
556
|
Abstract
Thioredoxin (Trx) and thioredoxin reductase (TrxR) are essential components of the Trx system which plays pivotal roles in regulating multiple cellular redox signaling pathways. In recent years TrxR/Trx have been increasingly recognized as an important modulator of tumor development, and hence targeting TrxR/Trx is a promising strategy for cancer treatment. In this review we first discuss the structural details of TrxR, the functions of the Trx system, and the rational of targeting TrxR/Trx for cancer treatment. We also highlight small-molecule TrxR/Trx inhibitors that have potential anticancer activity and review their mechanisms of action. Finally, we examine the challenges of developing TrxR/Trx inhibitors as anticancer agents and perspectives for selectively targeting TrxR/Trx.
Collapse
Affiliation(s)
- Junmin Zhang
- State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China; School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Xinming Li
- State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Xiao Han
- State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China
| | - Ruijuan Liu
- State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China; School of Pharmacy, Lanzhou University, Lanzhou, 730000, China
| | - Jianguo Fang
- State Key Laboratory of Applied Organic Chemistry, and College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, China.
| |
Collapse
|
557
|
Abstract
Background. The polysaccharide fucoidan is widely investigated as an anti-cancer agent. Here, we tested the effect of fucoidan on uveal melanoma cell lines. Methods. The effect of 100 µM fucoidan was investigated on five cell lines (92.1, Mel270 OMM1, OMM2.3, OMM2.5) and of 1 µg/mL–1 mg/mL fucoidan in two cell lines (OMM1, OMM2.3). Cell proliferation and viability were investigated with a WST-1 assay, migration in a wound healing (scratch) assay. Vascular Endothelial Growth Factor (VEGF) was measured in ELISA. Angiogenesis was evaluated in co-cultures with endothelial cells. Cell toxicity was induced by hydrogen-peroxide. Protein expression (Akt, ERK1/2, Bcl-2, Bax) was investigated in Western blot. Results. Fucoidan increased proliferation in two and reduced it in one cell line. Migration was reduced in three cell lines. The effect of fucoidan on VEGF was cell type and concentration dependent. In endothelial co-culture with 92.1, fucoidan significantly increased tubular structures. Moreover, fucoidan significantly protected all tested uveal melanoma cell lines from hydrogen-peroxide induced cell death. Under oxidative stress, fucoidan did not alter the expression of Bcl-2, Bax or ERK1/2, while inducing Akt expression in 92.1 cells but not in any other cell line. Conclusion. Fucoidan did not show anti-tumorigenic effects but displayed protective and pro-angiogenic properties, rendering fucoidan unsuitable as a potential new drug for the treatment of uveal melanoma.
Collapse
|
558
|
Huang Y, Pu F, Ren J, Qu X. Artificial Enzyme-based Logic Operations to Mimic an Intracellular Enzyme-participated Redox Balance System. Chemistry 2017; 23:9156-9161. [DOI: 10.1002/chem.201701353] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Yanyan Huang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences, Changchun; Jilin 130022 P. R. China
- University of Chinese Academy of Sciences; Beijing 100039 P. R. China
| | - Fang Pu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences, Changchun; Jilin 130022 P. R. China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences, Changchun; Jilin 130022 P. R. China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization; Changchun Institute of Applied Chemistry; Chinese Academy of Sciences, Changchun; Jilin 130022 P. R. China
| |
Collapse
|
559
|
Mak TW, Grusdat M, Duncan GS, Dostert C, Nonnenmacher Y, Cox M, Binsfeld C, Hao Z, Brüstle A, Itsumi M, Jäger C, Chen Y, Pinkenburg O, Camara B, Ollert M, Bindslev-Jensen C, Vasiliou V, Gorrini C, Lang PA, Lohoff M, Harris IS, Hiller K, Brenner D. Glutathione Primes T Cell Metabolism for Inflammation. Immunity 2017; 46:675-689. [PMID: 28423341 DOI: 10.1016/j.immuni.2017.03.019] [Citation(s) in RCA: 276] [Impact Index Per Article: 39.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 01/31/2017] [Accepted: 03/29/2017] [Indexed: 01/19/2023]
Abstract
Activated T cells produce reactive oxygen species (ROS), which trigger the antioxidative glutathione (GSH) response necessary to buffer rising ROS and prevent cellular damage. We report that GSH is essential for T cell effector functions through its regulation of metabolic activity. Conditional gene targeting of the catalytic subunit of glutamate cysteine ligase (Gclc) blocked GSH production specifically in murine T cells. Gclc-deficient T cells initially underwent normal activation but could not meet their increased energy and biosynthetic requirements. GSH deficiency compromised the activation of mammalian target of rapamycin-1 (mTOR) and expression of NFAT and Myc transcription factors, abrogating the energy utilization and Myc-dependent metabolic reprogramming that allows activated T cells to switch to glycolysis and glutaminolysis. In vivo, T-cell-specific ablation of murine Gclc prevented autoimmune disease but blocked antiviral defense. The antioxidative GSH pathway thus plays an unexpected role in metabolic integration and reprogramming during inflammatory T cell responses.
Collapse
Affiliation(s)
- Tak W Mak
- The Campbell Family Cancer Research Institute and University Health Network, Toronto, ON M5G 2C1, Canada; Departments of Medical Biophysics and Immunology, University of Toronto, Toronto, ON M5G 2M9, Canada.
| | - Melanie Grusdat
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, L-4354, Luxembourg
| | - Gordon S Duncan
- The Campbell Family Cancer Research Institute and University Health Network, Toronto, ON M5G 2C1, Canada
| | - Catherine Dostert
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, L-4354, Luxembourg
| | - Yannic Nonnenmacher
- Technische Universität Braunschweig, Braunschweig Integrated Center of Systems Biology, Braunschweig D-38106, Germany; Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, L-4367, Luxembourg
| | - Maureen Cox
- The Campbell Family Cancer Research Institute and University Health Network, Toronto, ON M5G 2C1, Canada
| | - Carole Binsfeld
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, L-4354, Luxembourg
| | - Zhenyue Hao
- The Campbell Family Cancer Research Institute and University Health Network, Toronto, ON M5G 2C1, Canada; The Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, ON M5S3E1 Canada
| | - Anne Brüstle
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia
| | - Momoe Itsumi
- Department of Molecular Virology, Tokyo Medical and Dental University, Tokyo, 113-8510, Japan
| | - Christian Jäger
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, L-4367, Luxembourg
| | - Ying Chen
- Department of Environmental Health Sciences, Yale School of Public Health, CT 06520, New Haven, USA
| | - Olaf Pinkenburg
- Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, D-35032 Germany
| | - Bärbel Camara
- Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, D-35032 Germany
| | - Markus Ollert
- Department of Infection and Immunity, Allergy and Clinical Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, L-4354, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, DK-5000, Denmark
| | - Carsten Bindslev-Jensen
- Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, DK-5000, Denmark
| | - Vasilis Vasiliou
- Department of Environmental Health Sciences, Yale School of Public Health, CT 06520, New Haven, USA
| | - Chiara Gorrini
- The Campbell Family Cancer Research Institute and University Health Network, Toronto, ON M5G 2C1, Canada
| | - Philipp A Lang
- Department of Molecular Medicine II, Medical Faculty, University of Düsseldorf, Düsseldorf, D-40225, Germany
| | - Michael Lohoff
- Institute for Medical Microbiology and Hospital Hygiene, University of Marburg, Marburg, D-35032 Germany
| | - Isaac S Harris
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Karsten Hiller
- Technische Universität Braunschweig, Braunschweig Integrated Center of Systems Biology, Braunschweig D-38106, Germany; Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Belvaux, L-4367, Luxembourg; Computational Biology of Infection Research, Helmholtz Centre for Infection Research, Braunschweig, D-38124, Germany
| | - Dirk Brenner
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, L-4354, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, DK-5000, Denmark.
| |
Collapse
|
560
|
Vander Heiden MG, DeBerardinis RJ. Understanding the Intersections between Metabolism and Cancer Biology. Cell 2017; 168:657-669. [PMID: 28187287 DOI: 10.1016/j.cell.2016.12.039] [Citation(s) in RCA: 1431] [Impact Index Per Article: 204.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/23/2016] [Accepted: 12/27/2016] [Indexed: 12/12/2022]
Abstract
Transformed cells adapt metabolism to support tumor initiation and progression. Specific metabolic activities can participate directly in the process of transformation or support the biological processes that enable tumor growth. Exploiting cancer metabolism for clinical benefit requires defining the pathways that are limiting for cancer progression and understanding the context specificity of metabolic preferences and liabilities in malignant cells. Progress toward answering these questions is providing new insight into cancer biology and can guide the more effective targeting of metabolism to help patients.
Collapse
Affiliation(s)
- Matthew G Vander Heiden
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Dana-Farber Cancer Institute, Boston, MA 02115, USA.
| | - Ralph J DeBerardinis
- Children's Medical Center Research Institute, Department of Pediatrics, and Eugene McDermott Center for Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, TX 75390.
| |
Collapse
|
561
|
Chio IIC, Tuveson DA. ROS in Cancer: The Burning Question. Trends Mol Med 2017; 23:411-429. [PMID: 28427863 PMCID: PMC5462452 DOI: 10.1016/j.molmed.2017.03.004] [Citation(s) in RCA: 340] [Impact Index Per Article: 48.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/16/2017] [Accepted: 03/16/2017] [Indexed: 02/07/2023]
Abstract
An unanswered question in human health is whether antioxidation prevents or promotes cancer. Antioxidation has historically been viewed as chemopreventive, but emerging evidence suggests that antioxidants may be supportive of neoplasia. We posit this contention to be rooted in the fact that ROS do not operate as one single biochemical entity, but as diverse secondary messengers in cancer cells. This cautions against therapeutic strategies to increase ROS at a global level. To leverage redox alterations towards the development of effective therapies necessitates the application of biophysical and biochemical approaches to define redox dynamics and to functionally elucidate specific oxidative modifications in cancer versus normal cells. An improved understanding of the sophisticated workings of redox biology is imperative to defeating cancer.
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.
| | - 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
|
562
|
Lee D, Xu IMJ, Chiu DKC, Lai RKH, Tse APW, Lan Li L, Law CT, Tsang FHC, Wei LL, Chan CYK, Wong CM, Ng IOL, Wong CCL. Folate cycle enzyme MTHFD1L confers metabolic advantages in hepatocellular carcinoma. J Clin Invest 2017; 127:1856-1872. [PMID: 28394261 PMCID: PMC5409797 DOI: 10.1172/jci90253] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 02/16/2017] [Indexed: 01/02/2023] Open
Abstract
Cancer cells preferentially utilize glucose and glutamine, which provide macromolecules and antioxidants that sustain rapid cell division. Metabolic reprogramming in cancer drives an increased glycolytic rate that supports maximal production of these nutrients. The folate cycle, through transfer of a carbon unit between tetrahydrofolate and its derivatives in the cytoplasmic and mitochondrial compartments, produces other metabolites that are essential for cell growth, including nucleotides, methionine, and the antioxidant NADPH. Here, using hepatocellular carcinoma (HCC) as a cancer model, we have observed a reduction in growth rate upon withdrawal of folate. We found that an enzyme in the folate cycle, methylenetetrahydrofolate dehydrogenase 1-like (MTHFD1L), plays an essential role in support of cancer growth. We determined that MTHFD1L is transcriptionally activated by NRF2, a master regulator of redox homeostasis. Our observations further suggest that MTHFD1L contributes to the production and accumulation of NADPH to levels that are sufficient to combat oxidative stress in cancer cells. The elevation of oxidative stress through MTHFD1L knockdown or the use of methotrexate, an antifolate drug, sensitizes cancer cells to sorafenib, a targeted therapy for HCC. Taken together, our study identifies MTHFD1L in the folate cycle as an important metabolic pathway in cancer cells with the potential for therapeutic targeting.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Chun-Ming Wong
- Department of Pathology and
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| | - Irene Oi-Lin Ng
- Department of Pathology and
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| | - Carmen Chak-Lui Wong
- Department of Pathology and
- State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
563
|
Cairns RA, Mak TW. Fire and water: Tumor cell adaptation to metabolic conditions. Exp Cell Res 2017; 356:204-208. [PMID: 28457987 DOI: 10.1016/j.yexcr.2017.04.029] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 04/25/2017] [Indexed: 12/23/2022]
Abstract
Lorenz Poellinger was a leader in understanding the effects of altered microenvironmental conditions in tumor biology and in normal physiology. His work examining the effects of hypoxia and the HIF transcription factors has expanded our understanding of the role of the microenvironment in affecting the behaviour of both normal and malignant cells. Furthermore, his work provides a model for understanding the adaptive responses to other metabolic stress conditions. By investigating the molecular mechanisms responsible for the adaptive responses to metabolic stress in normal physiological situations, across pathological conditions, and in different model organisms, his work shows the power of combining data from different model systems and physiological contexts. In cancers, it has become clear that in order to evolve to become an aggressive malignant disease, tumor cells must acquire the capacity to tolerate a host of abnormal and stressful metabolic conditions. This metabolic stress can be thought of as a fire that tumor cells must douse with enough water to survive, and may offer opportunities to exploit smoldering vulnerabilities in order to eradicate malignant cells.
Collapse
Affiliation(s)
- Rob A Cairns
- The Campbell Family Institute for Breast Cancer Research at Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada M5G 2C1.
| | - Tak W Mak
- The Campbell Family Institute for Breast Cancer Research at Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada M5G 2C1
| |
Collapse
|
564
|
Abstract
Oxidative stress is two sided: Whereas excessive oxidant challenge causes damage to biomolecules, maintenance of a physiological level of oxidant challenge, termed oxidative eustress, is essential for governing life processes through redox signaling. Recent interest has focused on the intricate ways by which redox signaling integrates these converse properties. Redox balance is maintained by prevention, interception, and repair, and concomitantly the regulatory potential of molecular thiol-driven master switches such as Nrf2/Keap1 or NF-κB/IκB is used for system-wide oxidative stress response. Nonradical species such as hydrogen peroxide (H2O2) or singlet molecular oxygen, rather than free-radical species, perform major second messenger functions. Chemokine-controlled NADPH oxidases and metabolically controlled mitochondrial sources of H2O2 as well as glutathione- and thioredoxin-related pathways, with powerful enzymatic back-up systems, are responsible for fine-tuning physiological redox signaling. This makes for a rich research field spanning from biochemistry and cell biology into nutritional sciences, environmental medicine, and molecular knowledge-based redox medicine.
Collapse
Affiliation(s)
- Helmut Sies
- Institute of Biochemistry and Molecular Biology I, Heinrich Heine University, Düsseldorf, University, D-40225, Düsseldorf, Germany; .,Leibniz Research Institute for Environmental Medicine, Heinrich Heine University, D-40225, Düsseldorf, Germany
| | - Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich Heine University, D-40225, Düsseldorf, Germany;
| | - Dean P Jones
- Department of Medicine, Emory University, Atlanta, Georgia 30322;
| |
Collapse
|
565
|
Lou M, Liu Q, Ren G, Zeng J, Xiang X, Ding Y, Lin Q, Zhong T, Liu X, Zhu L, Qi H, Shen J, Li H, Shao J. Physical interaction between human ribonucleotide reductase large subunit and thioredoxin increases colorectal cancer malignancy. J Biol Chem 2017; 292:9136-9149. [PMID: 28411237 DOI: 10.1074/jbc.m117.783365] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 04/11/2017] [Indexed: 11/06/2022] Open
Abstract
Ribonucleotide reductase (RR) is the rate-limiting enzyme in DNA synthesis, catalyzing the reduction of ribonucleotides to deoxyribonucleotides. During each enzymatic turnover, reduction of the active site disulfide in the catalytic large subunit is performed by a pair of shuttle cysteine residues in its C-terminal tail. Thioredoxin (Trx) and glutaredoxin (Grx) are ubiquitous redox proteins, catalyzing thiol-disulfide exchange reactions. Here, immunohistochemical examination of clinical colorectal cancer (CRC) specimens revealed that human thioredoxin1 (hTrx1), but not human glutaredoxin1 (hGrx1), was up-regulated along with human RR large subunit (RRM1) in cancer tissues, and the expression levels of both proteins were correlated with cancer malignancy stage. Ectopically expressed hTrx1 significantly increased RR activity, DNA synthesis, and cell proliferation and migration. Importantly, inhibition of both hTrx1 and RRM1 produced a synergistic anticancer effect in CRC cells and xenograft mice. Furthermore, hTrx1 rather than hGrx1 was the efficient reductase for RRM1 regeneration. We also observed a direct protein-protein interaction between RRM1 and hTrx1 in CRC cells. Interestingly, besides the known two conserved cysteines, a third cysteine (Cys779) in the RRM1 C terminus was essential for RRM1 regeneration and binding to hTrx1, whereas both Cys32 and Cys35 in hTrx1 played a counterpart role. Our findings suggest that the up-regulated RRM1 and hTrx1 in CRC directly interact with each other and promote RR activity, resulting in enhanced DNA synthesis and cancer malignancy. We propose that the RRM1-hTrx1 interaction might be a novel potential therapeutic target for cancer treatment.
Collapse
Affiliation(s)
- Meng Lou
- From the Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, Research Center for Air Pollution and Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Qian Liu
- From the Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, Research Center for Air Pollution and Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | | | | | - Xueping Xiang
- the Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China, and
| | | | - Qinghui Lin
- From the Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, Research Center for Air Pollution and Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Tingting Zhong
- From the Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, Research Center for Air Pollution and Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xia Liu
- From the Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, Research Center for Air Pollution and Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Lijun Zhu
- Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Hongyan Qi
- From the Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, Research Center for Air Pollution and Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jing Shen
- From the Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, Research Center for Air Pollution and Health, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Haoran Li
- Takeda Pharmaceuticals International Company, Cambridge, Massachusetts 02139
| | - Jimin Shao
- From the Department of Pathology and Pathophysiology, Key Laboratory of Disease Proteomics of Zhejiang Province, Research Center for Air Pollution and Health, Zhejiang University School of Medicine, Hangzhou 310058, China,
| |
Collapse
|
566
|
Zhao Y, Hu X, Liu Y, Dong S, Wen Z, He W, Zhang S, Huang Q, Shi M. ROS signaling under metabolic stress: cross-talk between AMPK and AKT pathway. Mol Cancer 2017. [PMID: 28407774 DOI: 10.1186/s12943-017-0648-1.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Cancer cells are frequently confronted with metabolic stress in tumor microenvironments due to their rapid growth and limited nutrient supply. Metabolic stress induces cell death through ROS-induced apoptosis. However, cancer cells can adapt to it by altering the metabolic pathways. AMPK and AKT are two primary effectors in response to metabolic stress: AMPK acts as an energy-sensing factor which rewires metabolism and maintains redox balance. AKT broadly promotes energy production in the nutrient abundance milieu, but the role of AKT under metabolic stress is in dispute. Recent studies show that AMPK and AKT display antagonistic roles under metabolic stress. Metabolic stress-induced ROS signaling lies in the hub between metabolic reprogramming and redox homeostasis. Here, we highlight the cross-talk between AMPK and AKT and their regulation on ROS production and elimination, which summarizes the mechanism of cancer cell adaptability under ROS stress and suggests potential options for cancer therapeutics.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xingbin Hu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yajing Liu
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shumin Dong
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Zhaowei Wen
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wanming He
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Shuyi Zhang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qiong Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
567
|
ROS signaling under metabolic stress: cross-talk between AMPK and AKT pathway. Mol Cancer 2017; 16:79. [PMID: 28407774 PMCID: PMC5390360 DOI: 10.1186/s12943-017-0648-1] [Citation(s) in RCA: 446] [Impact Index Per Article: 63.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Accepted: 04/05/2017] [Indexed: 02/06/2023] Open
Abstract
Cancer cells are frequently confronted with metabolic stress in tumor microenvironments due to their rapid growth and limited nutrient supply. Metabolic stress induces cell death through ROS-induced apoptosis. However, cancer cells can adapt to it by altering the metabolic pathways. AMPK and AKT are two primary effectors in response to metabolic stress: AMPK acts as an energy-sensing factor which rewires metabolism and maintains redox balance. AKT broadly promotes energy production in the nutrient abundance milieu, but the role of AKT under metabolic stress is in dispute. Recent studies show that AMPK and AKT display antagonistic roles under metabolic stress. Metabolic stress-induced ROS signaling lies in the hub between metabolic reprogramming and redox homeostasis. Here, we highlight the cross-talk between AMPK and AKT and their regulation on ROS production and elimination, which summarizes the mechanism of cancer cell adaptability under ROS stress and suggests potential options for cancer therapeutics.
Collapse
|
568
|
Schrier MS, Trivedi MS, Deth RC. Redox-Related Epigenetic Mechanisms in Glioblastoma: Nuclear Factor (Erythroid-Derived 2)-Like 2, Cobalamin, and Dopamine Receptor Subtype 4. Front Oncol 2017; 7:46. [PMID: 28424758 PMCID: PMC5371596 DOI: 10.3389/fonc.2017.00046] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 03/06/2017] [Indexed: 12/16/2022] Open
Abstract
Glioblastoma is an exceptionally difficult cancer to treat. Cancer is universally marked by epigenetic changes, which play key roles in sustaining a malignant phenotype, in addition to disease progression and patient survival. Studies have shown strong links between the cellular redox state and epigenetics. Nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a redox-sensitive transcription factor that upregulates endogenous antioxidant production, and is aberrantly expressed in many cancers, including glioblastoma. Methylation of DNA and histones provides a mode of epigenetic regulation, and cobalamin-dependent reactions link the redox state to methylation. Antagonists of dopamine receptor subtype 4 (D4 receptor) were recently shown to restrict glioblastoma stem cell growth by downregulating trophic signaling, resulting in inhibition of functional autophagy. In addition to stimulating glioblastoma stem cell growth, D4 receptors have the unique ability to catalyze cobalamin-dependent phospholipid methylation. Therefore, D4 receptors represent an important node in a molecular reflex pathway involving Nrf2 and cobalamin, operating in conjunction with redox status and methyl group donor availability. In this article, we describe the redox-related effects of Nrf2, cobalamin metabolism, and the D4 receptor on the regulation of the epigenetic state in glioblastoma.
Collapse
Affiliation(s)
- Matthew Scott Schrier
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Malav Suchin Trivedi
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Richard Carlton Deth
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL, USA
| |
Collapse
|
569
|
Reczek CR, Chandel NS. The Two Faces of Reactive Oxygen Species in Cancer. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2017. [DOI: 10.1146/annurev-cancerbio-041916-065808] [Citation(s) in RCA: 281] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Colleen R. Reczek
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| | - Navdeep S. Chandel
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611
| |
Collapse
|
570
|
Han E, Kwon B, Yoo D, Kang C, Khang G, Lee D. Dual Stimuli-Activatable Oxidative Stress Amplifying Agent as a Hybrid Anticancer Prodrug. Bioconjug Chem 2017; 28:968-978. [DOI: 10.1021/acs.bioconjchem.6b00683] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Eunji Han
- Department of BIN Convergence Technology and ‡Department of Polymer·Nano Science
and Technology, Chonbuk National University, Baekjedaero 567, Jeonju, Chonbuk, 567-756, Republic of Korea
| | - Byeongsu Kwon
- Department of BIN Convergence Technology and ‡Department of Polymer·Nano Science
and Technology, Chonbuk National University, Baekjedaero 567, Jeonju, Chonbuk, 567-756, Republic of Korea
| | - Donghyuck Yoo
- Department of BIN Convergence Technology and ‡Department of Polymer·Nano Science
and Technology, Chonbuk National University, Baekjedaero 567, Jeonju, Chonbuk, 567-756, Republic of Korea
| | - Changsun Kang
- Department of BIN Convergence Technology and ‡Department of Polymer·Nano Science
and Technology, Chonbuk National University, Baekjedaero 567, Jeonju, Chonbuk, 567-756, Republic of Korea
| | - Gilson Khang
- Department of BIN Convergence Technology and ‡Department of Polymer·Nano Science
and Technology, Chonbuk National University, Baekjedaero 567, Jeonju, Chonbuk, 567-756, Republic of Korea
| | - Dongwon Lee
- Department of BIN Convergence Technology and ‡Department of Polymer·Nano Science
and Technology, Chonbuk National University, Baekjedaero 567, Jeonju, Chonbuk, 567-756, Republic of Korea
| |
Collapse
|
571
|
Zou P, Xia Y, Chen W, Chen X, Ying S, Feng Z, Chen T, Ye Q, Wang Z, Qiu C, Yang S, Liang G. EF24 induces ROS-mediated apoptosis via targeting thioredoxin reductase 1 in gastric cancer cells. Oncotarget 2017; 7:18050-64. [PMID: 26919110 PMCID: PMC4951270 DOI: 10.18632/oncotarget.7633] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 02/14/2016] [Indexed: 12/22/2022] Open
Abstract
Gastric cancer (GC) is one of the leading causes of cancer mortality in the world, and finding novel agents for the treatment of advanced gastric cancer is of urgent need. Diphenyl difluoroketone (EF24), a molecule having structural similarity to curcumin, exhibits potent anti-tumor activities by arresting cell cycle and inducing apoptosis. Although EF24 demonstrates potent anticancer efficacy in numerous types of human cancer cells, the cellular targets of EF24 have not been fully defined. We report here that EF24 may interact with the thioredoxin reductase 1 (TrxR1), an important selenocysteine (Sec)-containing antioxidant enzyme, to induce reactive oxygen species (ROS)-mediated apoptosis in human gastric cancer cells. By inhibiting TrxR1 activity and increasing intracellular ROS levels, EF24 induces a lethal endoplasmic reticulum stress in human gastric cancer cells. Importantly, knockdown of TrxR1 sensitizes cells to EF24 treatment. In vivo, EF24 treatment markedly reduces the TrxR1 activity and tumor cell burden, and displays synergistic lethality with 5-FU against gastric cancer cells. Targeting TrxR1 with EF24 thus discloses a previously unrecognized mechanism underlying the biological activity of EF24, and reveals that TrxR1 is a good target for gastric cancer therapy.
Collapse
Affiliation(s)
- Peng Zou
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China.,Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yiqun Xia
- Department of Digestive Diseases, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Weiqian Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xi Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shilong Ying
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhiguo Feng
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Tongke Chen
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Qingqing Ye
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhe Wang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chenyu Qiu
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shulin Yang
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing, Jiangsu, China
| | - Guang Liang
- Chemical Biology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
572
|
Loss of Merlin induces metabolomic adaptation that engages dependence on Hedgehog signaling. Sci Rep 2017; 7:40773. [PMID: 28112165 PMCID: PMC5256100 DOI: 10.1038/srep40773] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Accepted: 12/09/2016] [Indexed: 02/06/2023] Open
Abstract
The tumor suppressor protein Merlin is proteasomally degraded in breast cancer. We undertook an untargeted metabolomics approach to discern the global metabolomics profile impacted by Merlin in breast cancer cells. We discerned specific changes in glutathione metabolites that uncovered novel facets of Merlin in impacting the cancer cell metabolome. Concordantly, Merlin loss increased oxidative stress causing aberrant activation of Hedgehog signaling. Abrogation of GLI-mediated transcription activity compromised the aggressive phenotype of Merlin-deficient cells indicating a clear dependence of cells on Hedgehog signaling. In breast tumor tissues, GLI1 expression enhanced tissue identification and discriminatory power of Merlin, cumulatively presenting a powerful substantiation of the relationship between these two proteins. We have uncovered, for the first time, details of the tumor cell metabolomic portrait modulated by Merlin, leading to activation of Hedgehog signaling. Importantly, inhibition of Hedgehog signaling offers an avenue to target the vulnerability of tumor cells with loss of Merlin.
Collapse
|
573
|
Hempel N, Trebak M. Crosstalk between calcium and reactive oxygen species signaling in cancer. Cell Calcium 2017; 63:70-96. [PMID: 28143649 DOI: 10.1016/j.ceca.2017.01.007] [Citation(s) in RCA: 149] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/13/2017] [Accepted: 01/14/2017] [Indexed: 02/07/2023]
Abstract
The interplay between Ca2+ and reactive oxygen species (ROS) signaling pathways is well established, with reciprocal regulation occurring at a number of subcellular locations. Many Ca2+ channels at the cell surface and intracellular organelles, including the endoplasmic reticulum and mitochondria are regulated by redox modifications. In turn, Ca2+ signaling can influence the cellular generation of ROS, from sources such as NADPH oxidases and mitochondria. This relationship has been explored in great depth during the process of apoptosis, where surges of Ca2+ and ROS are important mediators of cell death. More recently, coordinated and localized Ca2+ and ROS transients appear to play a major role in a vast variety of pro-survival signaling pathways that may be crucial for both physiological and pathophysiological functions. While much work is required to firmly establish this Ca2+-ROS relationship in cancer, existing evidence from other disease models suggests this crosstalk is likely of significant importance in tumorigenesis. In this review, we describe the regulation of Ca2+ channels and transporters by oxidants and discuss the potential consequences of the ROS-Ca2+ interplay in tumor cells.
Collapse
Affiliation(s)
- Nadine Hempel
- Department of Pharmacology, Penn State College of Medicine, Hershey PA 17033, United States; Penn State Hershey Cancer Institute, Penn State College of Medicine, Hershey PA 17033, United States.
| | - Mohamed Trebak
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey PA 17033, United States; Penn State Hershey Cancer Institute, Penn State College of Medicine, Hershey PA 17033, United States.
| |
Collapse
|
574
|
Krause M, Dubrovska A, Linge A, Baumann M. Cancer stem cells: Radioresistance, prediction of radiotherapy outcome and specific targets for combined treatments. Adv Drug Deliv Rev 2017; 109:63-73. [PMID: 26877102 DOI: 10.1016/j.addr.2016.02.002] [Citation(s) in RCA: 206] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 01/05/2016] [Accepted: 02/03/2016] [Indexed: 12/26/2022]
Abstract
Inactivation of cancer stem cells (CSCs) is of utmost importance for tumor cure after radiotherapy. An increasing body of evidence complies with a higher radioresistance of CSCs compared to the mass of tumor cells, supporting the use of CSC related biomarkers for prediction of radiotherapy outcome. Treatment individualization strategies for patient groups with vastly different risk of recurrence will most likely require application of more than one biomarker. Specifically, inclusion of established biomarkers like tumor size for primary radio(chemo)therapy or human papilloma virus (HPV) infection status in head and neck squamous cell carcinoma seems to be of very high relevance. The high heterogeneity of CSC subclones along with changes of the functional behavior of individual tumors under treatment underlines the importance of the selection of the optimal timepoint(s) of biomarker evaluation, but also provides a potential therapeutic target for combined treatment approaches with irradiation.
Collapse
Affiliation(s)
- Mechthild Krause
- German Cancer Consortium (DKTK) Dresden, Germany; Dept. of Radiation Oncology, Technische Universität Dresden, Germany; OncoRay, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf, Germany; German Cancer Research Center (DKFZ) Heidelberg, Germany.
| | - Anna Dubrovska
- German Cancer Consortium (DKTK) Dresden, Germany; OncoRay, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Research Center (DKFZ) Heidelberg, Germany
| | - Annett Linge
- German Cancer Consortium (DKTK) Dresden, Germany; Dept. of Radiation Oncology, Technische Universität Dresden, Germany; OncoRay, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; German Cancer Research Center (DKFZ) Heidelberg, Germany
| | - Michael Baumann
- German Cancer Consortium (DKTK) Dresden, Germany; Dept. of Radiation Oncology, Technische Universität Dresden, Germany; OncoRay, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf, Germany; German Cancer Research Center (DKFZ) Heidelberg, Germany
| |
Collapse
|
575
|
Gill JG, Piskounova E, Morrison SJ. Cancer, Oxidative Stress, and Metastasis. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2017; 81:163-175. [PMID: 28082378 DOI: 10.1101/sqb.2016.81.030791] [Citation(s) in RCA: 170] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) are highly reactive molecules that arise from a number of cellular sources, including oxidative metabolism in mitochondria. At low levels they can be advantageous to cells, activating signaling pathways that promote proliferation or survival. At higher levels, ROS can damage or kill cells by oxidizing proteins, lipids, and nucleic acids. It was hypothesized that antioxidants might benefit high-risk patients by reducing the rate of ROS-induced mutations and delaying cancer initiation. However, dietary supplementation with antioxidants has generally proven ineffective or detrimental in clinical trials. High ROS levels limit cancer cell survival during certain windows of cancer initiation and progression. During these periods, dietary supplementation with antioxidants may promote cancer cell survival and cancer progression. This raises the possibility that rather than treating cancer patients with antioxidants, they should be treated with pro-oxidants that exacerbate oxidative stress or block metabolic adaptations that confer oxidative stress resistance.
Collapse
Affiliation(s)
- Jennifer G Gill
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, Texas 75390.,Department of Pediatrics, Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Elena Piskounova
- Department of Pediatrics, Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Sean J Morrison
- Department of Pediatrics, Children's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390.,Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| |
Collapse
|
576
|
Zhao M, Liu Q, Gong Y, Xu X, Zhang C, Liu X, Zhang C, Guo H, Zhang X, Gong Y, Shao C. GSH-dependent antioxidant defense contributes to the acclimation of colon cancer cells to acidic microenvironment. Cell Cycle 2017; 15:1125-33. [PMID: 26950675 PMCID: PMC4889284 DOI: 10.1080/15384101.2016.1158374] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Due to increased glycolysis and poor local perfusion, solid tumors are usually immersed in an acidic microenvironment. While extracellular acidosis is cytotoxic, cancer cells eventually become acclimated to it. While previous studies have addressed the acute effect of acidosis on cancer cells, little is known about how cancer cells survive chronic acidosis. In this study we exposed colorectal cancer (CRC) cells (HCT15, HCT116 and LoVo) to acidic pH (pH 6.5) continuously for over three months and obtained CRC cells that become acclimated to acidic pH, designated as CRC-acidosis-acclimated or CRC-AA. We unexpectedly found that while acute exposure to low pH resulted in an increase in the level of intracellular reactive oxygen species (ROS), CRC-AA cells exhibited a significantly reduced level of ROS when compared to ancestor cells. CRC-AA cells were found to maintain a higher level of reduced glutathione, via the upregulation of CD44 and glutathione reductase (GSR), among others, than their ancestor cells. Importantly, CRC-AA cells were more sensitive to agents that deplete GSH. Moreover, downregulation of GSR by RNA interference was more deleterious to CRC-AA cells than to control cells. Together, our results demonstrate a critical role of glutathione-dependent antioxidant defense in acclimation of CRC cells to acidic extracellular pH.
Collapse
Affiliation(s)
- Minnan Zhao
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Qiao Liu
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Yanchao Gong
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Xiuhua Xu
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Chen Zhang
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Xiaojie Liu
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Caibo Zhang
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Haiyang Guo
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Xiyu Zhang
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Yaoqin Gong
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China
| | - Changshun Shao
- a Ministry of Education Key Laboratory of Experimental Teratology and Department of Molecular Medicine and Genetics, Shandong University School of Medicine , Jinan , China.,b Department of Genetics/Human Genetics Institute of New Jersey , Rutgers University , Piscataway , NJ , USA
| |
Collapse
|
577
|
Berger T, Saunders ME, Mak TW. Beyond the Oncogene Revolution: Four New Ways to Combat Cancer. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2017; 81:85-92. [PMID: 28057846 DOI: 10.1101/sqb.2016.81.031161] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
It has become clear that tumorigenesis results from much more than just the activation of an oncogene and/or the inactivation of a tumor-suppressor gene, and that the cancer cell genome contains many more alterations than can be specifically targeted at once. This observation has led our group to a search for alternative ways to kill cancer cells (while sparing normal cells) by focusing on properties unique to the former. We have identified four approaches with the potential to generate new anticancer therapies: combatting the tactics by which cancers evade antitumor immune responses, targeting metabolic adaptations that tumor cells use to survive conditions that would kill normal cells, manipulating a cancer cell's response to excessive oxidative stress, and exploiting aneuploidy. This review describes our progress to date on these fronts.
Collapse
Affiliation(s)
- Thorsten Berger
- The Campbell Family Institute for Breast Cancer Research and Ontario Cancer Institute, University Health Network, Toronto, Ontario M5G 2M9, Canada
| | - Mary E Saunders
- The Campbell Family Institute for Breast Cancer Research and Ontario Cancer Institute, University Health Network, Toronto, Ontario M5G 2M9, Canada
| | - Tak W Mak
- The Campbell Family Institute for Breast Cancer Research and Ontario Cancer Institute, University Health Network, Toronto, Ontario M5G 2M9, Canada
| |
Collapse
|
578
|
Cramer SL, Saha A, Liu J, Tadi S, Tiziani S, Yan W, Triplett K, Lamb C, Alters SE, Rowlinson S, Zhang YJ, Keating MJ, Huang P, DiGiovanni J, Georgiou G, Stone E. Systemic depletion of L-cyst(e)ine with cyst(e)inase increases reactive oxygen species and suppresses tumor growth. Nat Med 2017; 23:120-127. [PMID: 27869804 PMCID: PMC5218918 DOI: 10.1038/nm.4232] [Citation(s) in RCA: 381] [Impact Index Per Article: 54.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 10/14/2016] [Indexed: 02/07/2023]
Abstract
Cancer cells experience higher oxidative stress from reactive oxygen species (ROS) than do non-malignant cells because of genetic alterations and abnormal growth; as a result, maintenance of the antioxidant glutathione (GSH) is essential for their survival and proliferation. Under conditions of elevated ROS, endogenous L-cysteine (L-Cys) production is insufficient for GSH synthesis. This necessitates uptake of L-Cys that is predominantly in its disulfide form, L-cystine (CSSC), via the xCT(-) transporter. We show that administration of an engineered and pharmacologically optimized human cyst(e)inase enzyme mediates sustained depletion of the extracellular L-Cys and CSSC pool in mice and non-human primates. Treatment with this enzyme selectively causes cell cycle arrest and death in cancer cells due to depletion of intracellular GSH and ensuing elevated ROS; yet this treatment results in no apparent toxicities in mice even after months of continuous treatment. Cyst(e)inase suppressed the growth of prostate carcinoma allografts, reduced tumor growth in both prostate and breast cancer xenografts and doubled the median survival time of TCL1-Tg:p53-/- mice, which develop disease resembling human chronic lymphocytic leukemia. It was observed that enzyme-mediated depletion of the serum L-Cys and CSSC pool suppresses the growth of multiple tumors, yet is very well tolerated for prolonged periods, suggesting that cyst(e)inase represents a safe and effective therapeutic modality for inactivating antioxidant cellular responses in a wide range of malignancies.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Breast Neoplasms/metabolism
- Cell Line, Tumor
- Cystathionine gamma-Lyase/pharmacology
- Cysteine/drug effects
- Cysteine/metabolism
- Cystine/drug effects
- Cystine/metabolism
- Female
- Glutathione/metabolism
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Macaca fascicularis
- Male
- Mice
- Mice, Knockout
- Mice, Transgenic
- Neoplasm Transplantation
- Oxidative Stress
- Polyethylene Glycols/pharmacology
- Prostatic Neoplasms/metabolism
- Reactive Oxygen Species/metabolism
- Tumor Suppressor Protein p53/genetics
Collapse
Affiliation(s)
- Shira L. Cramer
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, United States
| | - Achinto Saha
- Division of Pharmacology and Toxicology and Dell Pediatric Research Institute, The University of Texas at Austin, Austin, TX 78712, United States
| | - Jinyun Liu
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Surendar Tadi
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX 78712, United States
| | - Stefano Tiziani
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, TX 78712, United States
| | - Wupeng Yan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, United States
| | - Kendra Triplett
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, United States
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, United States
| | - Candice Lamb
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, United States
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, United States
| | | | | | - Yan Jessie Zhang
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, United States
| | - Michael J. Keating
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Peng Huang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - John DiGiovanni
- Division of Pharmacology and Toxicology and Dell Pediatric Research Institute, The University of Texas at Austin, Austin, TX 78712, United States
| | - George Georgiou
- Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78712, United States
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, United States
| | - Everett Stone
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX 78712, United States
| |
Collapse
|
579
|
Zhong H, Xiao M, Zarkovic K, Zhu M, Sa R, Lu J, Tao Y, Chen Q, Xia L, Cheng S, Waeg G, Zarkovic N, Yin H. Mitochondrial control of apoptosis through modulation of cardiolipin oxidation in hepatocellular carcinoma: A novel link between oxidative stress and cancer. Free Radic Biol Med 2017; 102:67-76. [PMID: 27838437 DOI: 10.1016/j.freeradbiomed.2016.10.494] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 10/21/2016] [Accepted: 10/21/2016] [Indexed: 02/08/2023]
Abstract
Altered redox status in cancer cells has been linked to lipid peroxidation induced by reactive oxygen species (ROS) and subsequent formation of reactive lipid electrophiles, especially 4-hydroxy-nonenal (4-HNE). Emerging evidence suggests that cancer cells manipulate redox status to acquire anti-apoptotic phenotype but the underlying mechanisms are poorly understood. Cardiolipin (CL), a mitochondria-specific inner membrane phospholipid, is critical for maintaining mitochondrial function. Paradoxically, liver tissues contain tetralinoleoyl cardiolipin (TLCL) as the major CL in mitochondria yet emerging evidence suggests that ROS generated in mitochondria may lead to CL peroxidation and activation of intrinsic apoptosis. It remains unclear how CL oxidation leads to apoptosis and its relevance to the pathogenesis of hepatocellular carcinoma (HCC). We employed a mass spectrometry-based lipidomic approach to profile lipids in human tissues of HCC and found that CL was gradually decreased in tumor comparing to peripheral non-cancerous tissues, accompanied by a concomitant decrease of oxidized CL and its oxidation product, 4-HNE. Incubation of liver cancer cells with TLCL significantly restored apoptotic sensitivity accompanied by an increase of CL and its oxidation products when treated with staurosporine (STS) or Sorafenib (the standard treatment for late stage HCC patients). Our studies uncovered a novel mechanism by which cancer cells adopt to evade apoptosis, highlighting the importance of mitochondrial control of apoptosis through modulation of CL oxidation and subsequent 4-HNE formation in HCC. Thus manipulation of mitochondrial CL oxidation and lipid electrophile formation may have potential therapeutic value for diseases linked to oxidative stress and mitochondrial dysfunctions.
Collapse
Affiliation(s)
- Huiqin Zhong
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences (INS) Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China; University of the Chinese Academy of Sciences, CAS, Beijing, China
| | - Mengqing Xiao
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences (INS) Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China; University of the Chinese Academy of Sciences, CAS, Beijing, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Kamelija Zarkovic
- Division of Pathology, Clinical Hospital Centre & Medical Faculty, University of Zagreb, Croatia
| | - Mingjiang Zhu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences (INS) Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China
| | - Rina Sa
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences (INS) Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China; University of the Chinese Academy of Sciences, CAS, Beijing, China
| | - Jianhong Lu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences (INS) Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China; University of the Chinese Academy of Sciences, CAS, Beijing, China
| | - Yongzhen Tao
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences (INS) Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China
| | - Qun Chen
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences (INS) Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China; University of the Chinese Academy of Sciences, CAS, Beijing, China
| | - Lin Xia
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences (INS) Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China
| | - Shuqun Cheng
- The Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Georg Waeg
- Institute of Molecular Biosciences, Karl Franz University of Graz, Austria
| | - Neven Zarkovic
- Rudjer Boskovic Institute, Laboratory for Oxidative Stress, Zagreb, Croatia
| | - Huiyong Yin
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences (INS) Shanghai Institutes for Biological Sciences (SIBS), Chinese Academy of Sciences (CAS), Shanghai 200031, China; University of the Chinese Academy of Sciences, CAS, Beijing, China; Key Laboratory of Food Safety Risk Assessment, Ministry of Health, Beijing, China; School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
580
|
Semenza GL. Hypoxia-inducible factors: coupling glucose metabolism and redox regulation with induction of the breast cancer stem cell phenotype. EMBO J 2016; 36:252-259. [PMID: 28007895 DOI: 10.15252/embj.201695204] [Citation(s) in RCA: 266] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 09/19/2016] [Accepted: 09/22/2016] [Indexed: 12/12/2022] Open
Abstract
Reduced oxygen availability (hypoxia) leads to increased production of reactive oxygen species (ROS) by the electron transport chain. Here, I review recent work delineating mechanisms by which hypoxia-inducible factor 1 (HIF-1) mediates adaptive metabolic responses to hypoxia, including increased flux through the glycolytic pathway and decreased flux through the tricarboxylic acid cycle, in order to decrease mitochondrial ROS production. HIF-1 also mediates increased flux through the serine synthesis pathway and mitochondrial one-carbon (folate cycle) metabolism to increase mitochondrial antioxidant production (NADPH and glutathione). Dynamic maintenance of ROS homeostasis is required for induction of the breast cancer stem cell phenotype in response to hypoxia or cytotoxic chemotherapy. Consistently, inhibition of phosphoglycerate dehydrogenase, the first enzyme of the serine synthesis pathway, in breast cancer cells impairs tumor initiation, metastasis, and response to cytotoxic chemotherapy. I discuss how these findings have important implications for understanding the logic of the tumor microenvironment and for improving therapeutic responses in women with breast cancer.
Collapse
Affiliation(s)
- Gregg L Semenza
- Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry, McKusick-Nathans Institute of Genetic Medicine, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
581
|
Vyas S, Zaganjor E, Haigis MC. Mitochondria and Cancer. Cell 2016; 166:555-566. [PMID: 27471965 DOI: 10.1016/j.cell.2016.07.002] [Citation(s) in RCA: 1132] [Impact Index Per Article: 141.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 06/17/2016] [Accepted: 06/29/2016] [Indexed: 01/17/2023]
Abstract
Mitochondria are bioenergetic, biosynthetic, and signaling organelles that are integral in stress sensing to allow for cellular adaptation to the environment. Therefore, it is not surprising that mitochondria are important mediators of tumorigenesis, as this process requires flexibility to adapt to cellular and environmental alterations in addition to cancer treatments. Multiple aspects of mitochondrial biology beyond bioenergetics support transformation, including mitochondrial biogenesis and turnover, fission and fusion dynamics, cell death susceptibility, oxidative stress regulation, metabolism, and signaling. Thus, understanding mechanisms of mitochondrial function during tumorigenesis will be critical for the next generation of cancer therapeutics.
Collapse
Affiliation(s)
- Sejal Vyas
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School, Boston, MA 02115, USA
| | - Elma Zaganjor
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School, Boston, MA 02115, USA
| | - Marcia C Haigis
- Department of Cell Biology, Ludwig Center at Harvard, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
582
|
Trzeciecka A, Klossowski S, Bajor M, Zagozdzon R, Gaj P, Muchowicz A, Malinowska A, Czerwoniec A, Barankiewicz J, Domagala A, Chlebowska J, Prochorec-Sobieszek M, Winiarska M, Ostaszewski R, Gwizdalska I, Golab J, Nowis D, Firczuk M. Dimeric peroxiredoxins are druggable targets in human Burkitt lymphoma. Oncotarget 2016; 7:1717-31. [PMID: 26636537 PMCID: PMC4811492 DOI: 10.18632/oncotarget.6435] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 11/16/2015] [Indexed: 11/25/2022] Open
Abstract
Burkitt lymphoma is a fast-growing tumor derived from germinal center B cells. It is mainly treated with aggressive chemotherapy, therefore novel therapeutic approaches are needed due to treatment toxicity and developing resistance. Disturbance of red-ox homeostasis has recently emerged as an efficient antitumor strategy. Peroxiredoxins (PRDXs) are thioredoxin-family antioxidant enzymes that scavenge cellular peroxides and contribute to red-ox homeostasis. PRDXs are robustly expressed in various malignancies and critically involved in cell proliferation, differentiation and apoptosis. To elucidate potential role of PRDXs in lymphoma, we studied their expression level in B cell-derived primary lymphoma cells as well as in cell lines. We found that PRDX1 and PRDX2 are upregulated in tumor B cells as compared with normal counterparts. Concomitant knockdown of PRDX1 and PRDX2 significantly attenuated the growth rate of lymphoma cells. Furthermore, in human Burkitt lymphoma cell lines, we isolated dimeric 2-cysteine peroxiredoxins as targets for SK053, a novel thiol-specific small-molecule peptidomimetic with antitumor activity. We observed that treatment of lymphoma cells with SK053 triggers formation of covalent PRDX dimers, accumulation of intracellular reactive oxygen species, phosphorylation of ERK1/2 and AKT and leads to cell cycle arrest and apoptosis. Based on site-directed mutagenesis and modeling studies, we propose a mechanism of SK053-mediated PRDX crosslinking, involving double thioalkylation of active site cysteine residues. Altogether, our results suggest that peroxiredoxins are novel therapeutic targets in Burkitt lymphoma and provide the basis for new approaches to the treatment of this disease.
Collapse
Affiliation(s)
- Anna Trzeciecka
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Szymon Klossowski
- Institute of Organic Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Malgorzata Bajor
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Radoslaw Zagozdzon
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland.,Department of Bioinformatics, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Pawel Gaj
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | | | - Agata Malinowska
- Laboratory of Mass Spectrometry, Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Anna Czerwoniec
- Bioinformatics Laboratory, Institute of Molecular Biology and Biotechnology, Adam Mickiewicz University, Poznan, Poland
| | - Joanna Barankiewicz
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland.,Department of Hematology and Transfusion Medicine, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Antoni Domagala
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Justyna Chlebowska
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland.,Laboratory of Experimental Medicine, Center of New Technologies, University of Warsaw, Warsaw, Poland
| | - Monika Prochorec-Sobieszek
- Department of Diagnostic Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland.,Department of Pathology, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | | | | | | | - Jakub Golab
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Dominika Nowis
- Laboratory of Experimental Medicine, Center of New Technologies, University of Warsaw, Warsaw, Poland.,Genomic Medicine, Medical University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
583
|
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
|
584
|
Dunnill CJ, Ibraheem K, Mohamed A, Southgate J, Georgopoulos NT. A redox state-dictated signalling pathway deciphers the malignant cell specificity of CD40-mediated apoptosis. Oncogene 2016; 36:2515-2528. [PMID: 27869172 PMCID: PMC5422712 DOI: 10.1038/onc.2016.401] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Revised: 09/08/2016] [Accepted: 09/16/2016] [Indexed: 12/19/2022]
Abstract
CD40, a member of the tumour necrosis factor receptor (TNFR) superfamily, has the capacity to cause extensive apoptosis in carcinoma cells, while sparing normal epithelial cells. Yet, apoptosis is only achieved by membrane-presented CD40 ligand (mCD40L), as soluble receptor agonists are but weakly pro-apoptotic. Here, for the first time we have identified the precise signalling cascade underpinning mCD40L-mediated death as involving sequential TRAF3 stabilisation, ASK1 phosphorylation, MKK4 (but not MKK7) activation and JNK/AP-1 induction, leading to a Bak- and Bax-dependent mitochondrial apoptosis pathway. TRAF3 is central in the activation of the NADPH oxidase (Nox)-2 component p40phox and the elevation of reactive oxygen species (ROS) is essential in apoptosis. Strikingly, CD40 activation resulted in down-regulation of Thioredoxin (Trx)-1 to permit ASK1 activation and apoptosis. Although soluble receptor agonist alone could not induce death, combinatorial treatment incorporating soluble CD40 agonist and pharmacological inhibition of Trx-1 was functionally equivalent to the signal triggered by mCD40L. Finally, we demonstrate using normal, ‘para-malignant' and tumour-derived cells that progression to malignant transformation is associated with increase in oxidative stress in epithelial cells, which coincides with increased susceptibility to CD40 killing, while in normal cells CD40 signalling is cytoprotective. Our studies have revealed the molecular nature of the tumour specificity of CD40 signalling and explained the differences in pro-apoptotic potential between soluble and membrane-bound CD40 agonists. Equally importantly, by exploiting a unique epithelial culture system that allowed us to monitor alterations in the redox-state of epithelial cells at different stages of malignant transformation, our study reveals how pro-apoptotic signals can elevate ROS past a previously hypothesised ‘lethal pro-apoptotic threshold' to induce death; an observation that is both of fundamental importance and carries implications for cancer therapy.
Collapse
Affiliation(s)
- C J Dunnill
- Department of Biological Sciences, School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | - K Ibraheem
- Department of Biological Sciences, School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | - A Mohamed
- Department of Biological Sciences, School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| | - J Southgate
- Jack Birch Unit of Molecular Carcinogenesis, Department of Biology, University of York, York, UK
| | - N T Georgopoulos
- Department of Biological Sciences, School of Applied Sciences, University of Huddersfield, Huddersfield, UK
| |
Collapse
|
585
|
Role of thioredoxin reductase 1 in dysplastic transformation of human breast epithelial cells triggered by chronic oxidative stress. Sci Rep 2016; 6:36860. [PMID: 27845427 PMCID: PMC5109291 DOI: 10.1038/srep36860] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 10/20/2016] [Indexed: 12/21/2022] Open
Abstract
Thioredoxin reductase 1 (TrxR1) is a pivotal intracellular redox sensor and antioxidant enzyme. On the other hand, overexpression of TrxR1 is closely correlated with the initiation of various tumors including breast cancer, though the detailed mechanism remains unclear. Here we investigated the role of TrxR1 in dysplastic transformation of human breast epithelial cell line MCF-10A induced by chronic oxidative stress. Not surprisingly, sustained exposure to H2O2 significantly augmented the expression and activity of TrxR1 in MCF-10A cells. The dysplastically transformed MCF-10A (MCF-10AT) cells undergoing 8-week H2O2 treatment exhibited a certain degree of malignancy in tumorigenicity evaluation. Moreover, TrxR1 inhibitor ethaselen (BBSKE) could partially reverse some malignant phenotypes including epithelial to mesenchymal transition (EMT) of MCF-10AT as well as MCF-7 cells. Collectively, our results supported the considerable involvement of TrxR1 in the onset of breast cancer and BBSKE may be a promising agent against breast cancer.
Collapse
|
586
|
Sullivan LB, Gui DY, Vander Heiden MG. Altered metabolite levels in cancer: implications for tumour biology and cancer therapy. Nat Rev Cancer 2016; 16:680-693. [PMID: 27658530 DOI: 10.1038/nrc.2016.85] [Citation(s) in RCA: 275] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Altered cell metabolism is a characteristic feature of many cancers. Aside from well-described changes in nutrient consumption and waste excretion, altered cancer cell metabolism also results in changes to intracellular metabolite concentrations. Increased levels of metabolites that result directly from genetic mutations and cancer-associated modifications in protein expression can promote cancer initiation and progression. Changes in the levels of specific metabolites, such as 2-hydroxyglutarate, fumarate, succinate, aspartate and reactive oxygen species, can result in altered cell signalling, enzyme activity and/or metabolic flux. In this Review, we discuss the mechanisms that lead to changes in metabolite concentrations in cancer cells, the consequences of these changes for the cells and how they might be exploited to improve cancer therapy.
Collapse
Affiliation(s)
- Lucas B Sullivan
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Dan Y Gui
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Matthew G Vander Heiden
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts 02115, USA
| |
Collapse
|
587
|
Chen X, Lv Q, Hong Y, Chen X, Cheng B, Wu T. IL-1β maintains the redox balance by regulating glutaredoxin 1 expression during oral carcinogenesis. J Oral Pathol Med 2016; 46:332-339. [PMID: 27658048 DOI: 10.1111/jop.12502] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/15/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Xijuan Chen
- Department of Oral Medicine; Hospital of Stomatology; Guangzhou Guangdong China
- Guangdong Provincial Key Laboratory of Stomatology; Guanghua School of Stomatology; Sun Yat-sen University; Guangzhou Guangdong China
| | - Qianshu Lv
- Department of Oral Medicine; Affiliated Zhongshan Hospital; Sun Yat-sen University; Zhongshan Guangdong China
| | - Yun Hong
- Department of Oral Medicine; Hospital of Stomatology; Guangzhou Guangdong China
- Guangdong Provincial Key Laboratory of Stomatology; Guanghua School of Stomatology; Sun Yat-sen University; Guangzhou Guangdong China
| | - Xiaobing Chen
- Department of Oral Medicine; Hospital of Stomatology; Guangzhou Guangdong China
- Guangdong Provincial Key Laboratory of Stomatology; Guanghua School of Stomatology; Sun Yat-sen University; Guangzhou Guangdong China
| | - Bin Cheng
- Department of Oral Medicine; Hospital of Stomatology; Guangzhou Guangdong China
- Guangdong Provincial Key Laboratory of Stomatology; Guanghua School of Stomatology; Sun Yat-sen University; Guangzhou Guangdong China
| | - Tong Wu
- Department of Oral Medicine; Hospital of Stomatology; Guangzhou Guangdong China
- Guangdong Provincial Key Laboratory of Stomatology; Guanghua School of Stomatology; Sun Yat-sen University; Guangzhou Guangdong China
| |
Collapse
|
588
|
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
|
589
|
Abstract
Reactive oxygen species (ROS), which are both a natural byproduct of oxidative metabolism and an undesirable byproduct of many environmental stressors, can damage all classes of cellular macromolecules and promote diseases from cancer to neurodegeneration. The actions of ROS are mitigated by the transcription factor NRF2, which regulates expression of antioxidant genes via its interaction with cis-regulatory antioxidant response elements (AREs). However, despite the seemingly straightforward relationship between the opposing forces of ROS and NRF2, regulatory precision in the NRF2 network is essential. Genetic variants that alter NRF2 stability or alter ARE sequences have been linked to a range of diseases. NRF2 hyperactivating mutations are associated with tumorigenesis. On the subtler end of the spectrum, single nucleotide variants (SNVs) that alter individual ARE sequences have been linked to neurodegenerative disorders including progressive supranuclear palsy and Parkinson’s disease, as well as other diseases. Although the human health implications of NRF2 dysregulation have been recognized for some time, a systems level view of this regulatory network is beginning to highlight key NRF2-targeted AREs consistently associated with disease.
Collapse
|
590
|
Kim DK, Beaven MA, Kulinski JM, Desai A, Bandara G, Bai Y, Prussin C, Schwartz LB, Komarow H, Metcalfe DD, Olivera A. Regulation of Reactive Oxygen Species and the Antioxidant Protein DJ-1 in Mastocytosis. PLoS One 2016; 11:e0162831. [PMID: 27611333 PMCID: PMC5017616 DOI: 10.1371/journal.pone.0162831] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Accepted: 08/29/2016] [Indexed: 12/20/2022] Open
Abstract
Neoplastic accumulation of mast cells in systemic mastocytosis (SM) associates with activating mutations in the receptor tyrosine kinase KIT. Constitutive activation of tyrosine kinase oncogenes has been linked to imbalances in oxidant/antioxidant mechanisms in other myeloproliferative disorders. However, the impact of KIT mutations on the redox status in SM and the potential therapeutic implications are not well understood. Here, we examined the regulation of reactive oxygen species (ROS) and of the antioxidant protein DJ-1 (PARK-7), which increases with cancer progression and acts to lessen oxidative damage to malignant cells, in relationship with SM severity. ROS levels were increased in both indolent (ISM) and aggressive variants of the disease (ASM). However, while DJ-1 levels were reduced in ISM with lower mast cell burden, they rose in ISM with higher mast cell burden and were significantly elevated in patients with ASM. Studies on mast cell lines revealed that activating KIT mutations induced constant ROS production and consequent DJ-1 oxidation and degradation that could explain the reduced levels of DJ-1 in the ISM population, while IL-6, a cytokine that increases with disease severity, caused a counteracting transcriptional induction of DJ-1 which would protect malignant mast cells from oxidative damage. A mouse model of mastocytosis recapitulated the biphasic changes in DJ-1 and the escalating IL-6, ROS and DJ-1 levels as mast cells accumulate, findings which were reversed with anti-IL-6 receptor blocking antibody. Our findings provide evidence of increased ROS and a biphasic regulation of the antioxidant DJ-1 in variants of SM and implicate IL-6 in DJ-1 induction and expansion of mast cells with KIT mutations. We propose consideration of IL-6 blockade as a potential adjunctive therapy in the treatment of patients with advanced mastocytosis, as it would reduce DJ-1 levels making mutation-positive mast cells vulnerable to oxidative damage.
Collapse
Affiliation(s)
- Do-Kyun Kim
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Michael A. Beaven
- Laboratory of Molecular Immunology, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Joseph M. Kulinski
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Avanti Desai
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Geethani Bandara
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Yun Bai
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Calman Prussin
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lawrence B. Schwartz
- Department of Internal Medicine, Virginia Commonwealth University, Richmond, Virginia, United States of America
| | - Hirsh Komarow
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Dean D. Metcalfe
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ana Olivera
- Mast Cell Biology Section, Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
591
|
Tanaka G, Inoue KI, Shimizu T, Akimoto K, Kubota K. Dual pharmacological inhibition of glutathione and thioredoxin systems synergizes to kill colorectal carcinoma stem cells. Cancer Med 2016; 5:2544-57. [PMID: 27485632 PMCID: PMC5055185 DOI: 10.1002/cam4.844] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 07/06/2016] [Accepted: 07/07/2016] [Indexed: 12/14/2022] Open
Abstract
NRF2 stabilizes redox potential through genes for glutathione and thioredoxin antioxidant systems. Whether blockade of glutathione and thioredoxin is useful in eliminating cancer stem cells remain unknown. We used xenografts derived from colorectal carcinoma patients to investigate the pharmacological inhibition of glutathione and thioredoxin systems. Higher expression of five glutathione S‐transferase isoforms (GSTA1, A2, M4, O2, and P1) was observed in xenograft‐derived spheroids than in fibroblasts. Piperlongumine (2.5–10 μmol/L) and auranofin (0.25–4 μmol/L) were used to inhibit glutathione S‐transferase π and thioredoxin reductase, respectively. Piperlongumine or auranofin alone up‐regulated the expression of NRF2 target genes, but not TP53 targets. While piperlongumine showed modest cancer‐specific cell killing (IC50 difference between cancer spheroids and fibroblasts: P = 0.052), auranofin appeared more toxic to fibroblasts (IC50 difference between cancer spheroids and fibroblasts: P = 0.002). The synergism of dual inhibition was evaluated by determining the Combination Index, based on the number of surviving cells with combination treatments. Molar ratios indicated synergism in cancer spheroids, but not in fibroblasts: (auranofin:piperlongumine) = 2:5, 1:5, 1:10, and 1:20. Cancer‐specific cell killing was achieved at the following drug concentrations (auranofin:piperlongumine): 0.25:2.5 μmol/L, 0.5:2.5 μmol/L, or 0.25:5 μmol/L. The dual inhibition successfully decreased CD44v9 surface presentation and delayed tumor emergence in nude mouse. However, a small subpopulation persistently survived and accumulated phosphorylated histone H2A. Such “persisters” still retained lesser but significant tumorigenicity. Thus, dual inhibition of glutathione S‐transferase π and thioredoxin reductase could be a feasible option for decreasing the tumor mass and CD44v9‐positive fraction by disrupting redox regulation.
Collapse
Affiliation(s)
- Genki Tanaka
- Department of Second Surgery, Dokkyo Medical University, Shimotsuga-gun, Tochigi-ken, 321-0293, Japan
| | - Ken-Ichi Inoue
- Center for Research Support, Dokkyo Medial University, Shimotsuga-gun, Tochigi-ken, 321-0293, Japan
| | - Takayuki Shimizu
- Department of Second Surgery, Dokkyo Medical University, Shimotsuga-gun, Tochigi-ken, 321-0293, Japan
| | - Kazumi Akimoto
- Center for Research Support, Dokkyo Medial University, Shimotsuga-gun, Tochigi-ken, 321-0293, Japan
| | - Keiichi Kubota
- Department of Second Surgery, Dokkyo Medical University, Shimotsuga-gun, Tochigi-ken, 321-0293, Japan.
| |
Collapse
|
592
|
Kruspig B, Valter K, Skender B, Zhivotovsky B, Gogvadze V. Targeting succinate:ubiquinone reductase potentiates the efficacy of anticancer therapy. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:2065-71. [DOI: 10.1016/j.bbamcr.2016.04.026] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 04/12/2016] [Accepted: 04/28/2016] [Indexed: 10/21/2022]
|
593
|
Samanta D, Park Y, Andrabi SA, Shelton LM, Gilkes DM, Semenza GL. RETRACTED: PHGDH Expression Is Required for Mitochondrial Redox Homeostasis, Breast Cancer Stem Cell Maintenance, and Lung Metastasis. Cancer Res 2016; 76:4430-42. [PMID: 27280394 DOI: 10.1158/0008-5472.can-16-0530] [Citation(s) in RCA: 165] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 05/31/2016] [Indexed: 11/16/2022]
Abstract
Intratumoral hypoxia stimulates enrichment of breast cancer stem cells (BCSC), which are critical for metastasis and patient mortality. Here we report a metabolic adaptation that is required for hypoxia-induced BCSC enrichment and metastasis. Hypoxia-inducible factors coordinately regulate expression of genes encoding phosphoglycerate dehydrogenase (PHGDH) and five downstream enzymes in the serine synthesis pathway and mitochondrial one-carbon (folate) cycle. RNAi-mediated silencing of PHGDH expression in both estrogen receptor-positive and negative breast cancer cells led to decreased NADPH levels, disturbed mitochondrial redox homeostasis, and increased apoptosis, which abrogated BCSC enrichment under hypoxic conditions. PHGDH-deficient cells exhibited increased oxidant levels and apoptosis, as well as loss of BCSC enrichment, in response to treatment with carboplatin or doxorubicin. PHGDH-deficient cells were relatively weakly tumorigenic and tumors that did form were deficient in BCSCs, abolishing metastatic capacity. Our findings highlight a role for PHGDH in the formation of secondary (recurrent or metastatic) tumors, with potential implications for therapeutic targeting of advanced cancers. Cancer Res; 76(15); 4430-42. ©2016 AACR.
Collapse
Affiliation(s)
- Debangshu Samanta
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland. McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Youngrok Park
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Shaida A Andrabi
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland. Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Laura M Shelton
- Human Metabolome Technologies America, Inc., Boston, Massachusetts
| | - Daniele M Gilkes
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland. McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland. Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Gregg L Semenza
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland. McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland. Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland. Departments of Pediatrics, Medicine, Radiation Oncology, and Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, Maryland.
| |
Collapse
|
594
|
Engelman R, Ziv T, Arnér ESJ, Benhar M. Inhibitory nitrosylation of mammalian thioredoxin reductase 1: Molecular characterization and evidence for its functional role in cellular nitroso-redox imbalance. Free Radic Biol Med 2016; 97:375-385. [PMID: 27377780 DOI: 10.1016/j.freeradbiomed.2016.06.032] [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: 05/02/2016] [Revised: 06/28/2016] [Accepted: 06/30/2016] [Indexed: 12/18/2022]
Abstract
Mammalian thioredoxin 1 (Trx1) and the selenoprotein Trx reductase 1 (TrxR1) are key cellular enzymes that function coordinately in thiol-based redox regulation and signaling. Recent studies have revealed that the Trx1/TrxR1 system has an S-nitrosothiol reductase (denitrosylase) activity through which it can regulate nitric oxide-related cellular processes. In this study we revealed that TrxR1 is itself susceptible to nitrosylation, characterized the underlying mechanism, and explored its functional significance. We found that nitrosothiol or nitric oxide donating agents rapidly and effectively inhibited the activity of recombinant or endogenous TrxR1. In particular, the NADPH-reduced TrxR1 was partially and reversibly inhibited upon exposure to low concentrations (<10μM) of S-nitrosocysteine (CysNO) and markedly and continuously inhibited at higher doses. Concurrently, TrxR1 very efficiently reduced low, but not high, levels of CysNO. Biochemical and mass spectrometric analyses indicated that its active site selenocysteine residue renders TrxR1 highly susceptible to nitrosylation-mediated inhibition, and revealed both thiol and selenol modifications at the two redox active centers of the enzyme. Studies in HeLa cancer cells demonstrated that endogenous TrxR1 is sensitive to nitrosylation-dependent inactivation and pointed to an important role for glutathione in reversing or preventing this process. Notably, depletion of cellular glutathione with l-buthionine-sulfoximine synergized with nitrosating agents in promoting sustained nitrosylation and inactivation of TrxR1, events that were accompanied by significant oxidation of Trx1 and extensive cell death. Collectively, these findings expand our knowledge of the role and regulation of the mammalian Trx system in relation to cellular nitroso-redox imbalance. The observations raise the possibility of exploiting the nitrosylation susceptibility of TrxR1 for killing tumor cells.
Collapse
Affiliation(s)
- Rotem Engelman
- Department of Biochemistry, Rappaport Institute for Research in the Medical Sciences, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Tamar Ziv
- Smoler Proteomics Center and Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Elias S J Arnér
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Moran Benhar
- Department of Biochemistry, Rappaport Institute for Research in the Medical Sciences, Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
595
|
Chen X, Yang M, Hao W, Han J, Ma J, Wang C, Sun S, Zheng Q. Differentiation-inducing and anti-proliferative activities of isoliquiritigenin and all-trans-retinoic acid on B16F0 melanoma cells: Mechanisms profiling by RNA-seq. Gene 2016; 592:86-98. [PMID: 27461947 DOI: 10.1016/j.gene.2016.07.052] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 07/14/2016] [Accepted: 07/22/2016] [Indexed: 12/11/2022]
Abstract
Melanoma is a cancer that arises from melanocytes, specialized pigmented cells that are found predominantly in the skin. The incidence of malignant melanoma has significantly increased over the last decade. With the development of therapy, the survival rate of some kind of cancer has been improved greatly. But the treatment of melanoma remains unsatisfactory. Much of melanoma's resistance to traditional chemotherapy is believed to arise intrinsically, by virtue of potent growth and cell survival-promoting genetic alteration. Therefore, significant attention has recently been focused on differentiation therapy, as well as differentiation inducer compounds. In previous study, we found isoliquiritigenin (ISL), a natural product extracted from licorice, could induce B16F0 melanoma cell differentiation. Here we investigated the transcriptional response of melanoma differentiation process induced by ISL and all-trans-retinoic acid (RA). Results showed that 390 genes involves in 201 biochemical pathways were differentially expressed in ISL treatment and 304 genes in 193 pathways in RA treatment. Differential expressed genes (DGEs, fold-change (FC)≥10) with the function of anti-proliferative and differentiation inducing indicated a loss of grade malignancy characteristic. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis indicated glutathione metabolism, glycolysis/gluconeogenesis and pentose phosphate pathway were the top three relative pathway perturbed by ISL, and mitogen-activated protein kinase (MAPK) signaling pathway was the most important pathway in RA treatment. In the analysis of hierarchical clustering of DEGs, we discovered 72 DEGs involved in the process of drug action. We thought Cited1, Tgm2, Xaf1, Cd59a, Fbxo2, Adh7 may have critical role in the differentiation of melanoma. The evidence displayed herein confirms the critical role of reactive oxygen species (ROS) in melanoma pathobiology and provides evidence for future targets in the development of next-generation biomarkers and therapeutics.
Collapse
Affiliation(s)
- Xiaoyu Chen
- College of Science, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Ming Yang
- BGI-Tech, BGI-Shenzhen, Shenzhen, Guangdong 518083, China
| | - Wenjin Hao
- Binzhou Medical University, Yantai, Shandong 264003, China
| | - Jichun Han
- Binzhou Medical University, Yantai, Shandong 264003, China
| | - Jun Ma
- Binzhou Medical University, Yantai, Shandong 264003, China
| | - Caixia Wang
- Key Laboratory of Coastal Biology and Bioresource Utilization, Yantai Institute of Coastal Zone Research, Chinese Academy of Sciences, Yantai 264003, China
| | - Shiguo Sun
- College of Science, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Qiusheng Zheng
- Binzhou Medical University, Yantai, Shandong 264003, China.
| |
Collapse
|
596
|
Starheim KK, Holien T, Misund K, Johansson I, Baranowska KA, Sponaas AM, Hella H, Buene G, Waage A, Sundan A, Bjørkøy G. Intracellular glutathione determines bortezomib cytotoxicity in multiple myeloma cells. Blood Cancer J 2016; 6:e446. [PMID: 27421095 PMCID: PMC5141348 DOI: 10.1038/bcj.2016.56] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 05/03/2016] [Indexed: 12/19/2022] Open
Abstract
Multiple myeloma (myeloma in short) is an incurable cancer of antibody-producing plasma cells that comprise 13% of all hematological malignancies. The proteasome inhibitor bortezomib has improved treatment significantly, but inherent and acquired resistance to the drug remains a problem. We here show that bortezomib-induced cytotoxicity was completely dampened when cells were supplemented with cysteine or its derivative, glutathione (GSH) in ANBL-6 and INA-6 myeloma cell lines. GSH is a major component of the antioxidative defense in eukaryotic cells. Increasing intracellular GSH levels fully abolished bortezomib-induced cytotoxicity and transcriptional changes. Elevated intracellular GSH levels blocked bortezomib-induced nuclear factor erythroid 2-related factor 2 (NFE2L2, NRF2)-associated stress responses, including upregulation of the xCT subunit of the Xc- cystine-glutamate antiporter. INA-6 cells conditioned to increasing bortezomib doses displayed reduced bortezomib sensitivity and elevated xCT levels. Inhibiting Xc- activity potentiated bortezomib-induced cytotoxicity in myeloma cell lines and primary cells, and re-established sensitivity to bortezomib in bortezomib-conditioned cells. We propose that intracellular GSH level is the main determinant of bortezomib-induced cytotoxicity in a subset of myeloma cells, and that combined targeting of the proteasome and the Xc- cystine-glutamate antiporter can circumvent bortezomib resistance.
Collapse
Affiliation(s)
- K K Starheim
- Department of Cancer Research and Molecular Medicine, K.G. Jebsen Center for Myeloma Research, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Cancer Research and Molecular Medicine, Center of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - T Holien
- Department of Cancer Research and Molecular Medicine, K.G. Jebsen Center for Myeloma Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - K Misund
- Department of Cancer Research and Molecular Medicine, K.G. Jebsen Center for Myeloma Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - I Johansson
- Department of Cancer Research and Molecular Medicine, Center of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Laboratory Medicine, Children's and Women's Health, Faculty of Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - K A Baranowska
- Department of Cancer Research and Molecular Medicine, K.G. Jebsen Center for Myeloma Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - A-M Sponaas
- Department of Cancer Research and Molecular Medicine, K.G. Jebsen Center for Myeloma Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - H Hella
- Department of Cancer Research and Molecular Medicine, K.G. Jebsen Center for Myeloma Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - G Buene
- Department of Cancer Research and Molecular Medicine, K.G. Jebsen Center for Myeloma Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - A Waage
- Department of Cancer Research and Molecular Medicine, K.G. Jebsen Center for Myeloma Research, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Hematology, St. Olavs University Hospital, Trondheim, Norway
| | - A Sundan
- Department of Cancer Research and Molecular Medicine, K.G. Jebsen Center for Myeloma Research, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Cancer Research and Molecular Medicine, Center of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway
| | - G Bjørkøy
- Department of Cancer Research and Molecular Medicine, K.G. Jebsen Center for Myeloma Research, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Cancer Research and Molecular Medicine, Center of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim, Norway.,Department of Medical Laboratory Technology, Faculty of Technology, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
597
|
Jin L, Alesi GN, Kang S. Glutaminolysis as a target for cancer therapy. Oncogene 2016; 35:3619-25. [PMID: 26592449 PMCID: PMC5225500 DOI: 10.1038/onc.2015.447] [Citation(s) in RCA: 290] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 10/15/2015] [Accepted: 10/22/2015] [Indexed: 02/06/2023]
Abstract
Cancer cells display an altered metabolic circuitry that is directly regulated by oncogenic mutations and loss of tumor suppressors. Mounting evidence indicates that altered glutamine metabolism in cancer cells has critical roles in supporting macromolecule biosynthesis, regulating signaling pathways, and maintaining redox homeostasis, all of which contribute to cancer cell proliferation and survival. Thus, intervention in these metabolic processes could provide novel approaches to improve cancer treatment. This review summarizes current findings on the role of glutaminolytic enzymes in human cancers and provides an update on the development of small molecule inhibitors to target glutaminolysis for cancer therapy.
Collapse
Affiliation(s)
- L Jin
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - G N Alesi
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| | - S Kang
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
598
|
Sun CW, Willmon C, Wu LC, Knopick P, Thoerner J, Vile R, Townes TM, Terman DS. Sickle Cells Abolish Melanoma Tumorigenesis in Hemoglobin SS Knockin Mice and Augment the Tumoricidal Effect of Oncolytic Virus In Vivo. Front Oncol 2016; 6:166. [PMID: 27458571 PMCID: PMC4937018 DOI: 10.3389/fonc.2016.00166] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2016] [Accepted: 06/20/2016] [Indexed: 01/19/2023] Open
Abstract
Insights from the study of cancer resistance in animals have led to the discovery of novel anticancer pathways and opened new venues for cancer prevention and treatment. Sickle cells (SSRBCs) from subjects with homozygous sickle cell anemia (SCA) have been shown to target hypoxic tumor niches, induce diffuse vaso-occlusion, and potentiate a tumoricidal response in a heme- and oxidant-dependent manner. These findings spawned the hypothesis that SSRBCs and the vasculopathic microenvironment of subjects with SCA might be inimical to tumor outgrowth and thereby constitute a natural antitumor defense. We therefore implanted the B16F10 melanoma into humanized hemoglobin SS knockin mice which exhibit the hematologic and vasculopathic sequelae of human SCA. Over the 31-day observation period, hemoglobin SS mice showed no significant melanoma outgrowth. By contrast, 68-100% of melanomas implanted in background and hemoglobin AA knockin control mice reached the tumor growth end point (p < 0.0001). SS knockin mice also exhibited established markers of underlying vasculopathy, e.g., chronic hemolysis (anemia, reticulocytosis) and vascular inflammation (leukocytosis) that differed significantly from all control groups. Genetic differences or normal AA gene knockin do not explain the impaired tumor outgrowth in SS knockin mice. These data point instead to the chronic pro-oxidative vasculopathic network in these mice as the predominant cause. In related studies, we demonstrate the ability of the sickle cell component of this system to function as a therapeutic vehicle in potentiating the oncolytic/vasculopathic effect of RNA reovirus. Sickle cells were shown to efficiently adsorb and transfer the virus to melanoma cells where it induced apoptosis even in the presence of anti-reovirus neutralizing antibodies. In vivo, SSRBCs along with their viral cargo rapidly targeted the tumor and initiated a tumoricidal response exceeding that of free virus and similarly loaded normal RBCs without toxicity. Collectively, these data unveil two hitherto unrecognized findings: hemoglobin SS knockin mice appear to present a natural barrier to melanoma tumorigenesis while SSRBCs demonstrate therapeutic function as a vehicle for enhancing the oncolytic effect of free reovirus against established melanoma.
Collapse
Affiliation(s)
- Chiang Wang Sun
- Department of Biochemistry and Molecular Genetics, University of Alabama Medical School at Birmingham, Birmingham, AL, USA
| | - Candice Willmon
- Department of Molecular Medicine, Mayo Clinic Foundation, Rochester, MN, USA
| | - Li-Chen Wu
- Department of Biochemistry and Molecular Genetics, University of Alabama Medical School at Birmingham, Birmingham, AL, USA
| | - Peter Knopick
- Department of Immunology, University of North Dakota Medical School, Grand Forks, ND, USA
| | - Jutta Thoerner
- Hisotpathology Section, Hospital of the Monterey Peninsula, Monterey, CA, USA
| | - Richard Vile
- Department of Molecular Medicine, Mayo Clinic Foundation, Rochester, MN, USA
| | - Tim M. Townes
- Department of Biochemistry and Molecular Genetics, University of Alabama Medical School at Birmingham, Birmingham, AL, USA
| | - David S. Terman
- Department of Biochemistry and Molecular Genetics, University of Alabama Medical School at Birmingham, Birmingham, AL, USA
| |
Collapse
|
599
|
Shi H, Hu L, Chen S, Bao W, Yang S, Zhao X, Sun C. Metabolomics analysis of urine from rats administered with long-term, low-dose acrylamide by ultra-performance liquid chromatography-mass spectrometry. Xenobiotica 2016; 47:439-449. [DOI: 10.1080/00498254.2016.1196509] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Affiliation(s)
- Haidan Shi
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Heilongjiang, China
| | - Liyan Hu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Heilongjiang, China
| | - Shuai Chen
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Heilongjiang, China
| | - Wei Bao
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Heilongjiang, China
| | - Shuang Yang
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiujuan Zhao
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Heilongjiang, China
| | - Changhao Sun
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
600
|
Redox Homeostasis and Cellular Antioxidant Systems: Crucial Players in Cancer Growth and Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:6235641. [PMID: 27418953 PMCID: PMC4932173 DOI: 10.1155/2016/6235641] [Citation(s) in RCA: 188] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/18/2016] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) and their products are components of cell signaling pathways and play important roles in cellular physiology and pathophysiology. Under physiological conditions, cells control ROS levels by the use of scavenging systems such as superoxide dismutases, peroxiredoxins, and glutathione that balance ROS generation and elimination. Under oxidative stress conditions, excessive ROS can damage cellular proteins, lipids, and DNA, leading to cell damage that may contribute to carcinogenesis. Several studies have shown that cancer cells display an adaptive response to oxidative stress by increasing expression of antioxidant enzymes and molecules. As a double-edged sword, ROS influence signaling pathways determining beneficial or detrimental outcomes in cancer therapy. In this review, we address the role of redox homeostasis in cancer growth and therapy and examine the current literature regarding the redox regulatory systems that become upregulated in cancer and their role in promoting tumor progression and resistance to chemotherapy.
Collapse
|