501
|
Yang YF, Wu CC, Chen WP, Chen YL, Su MJ. Prazosin induces p53-mediated autophagic cell death in H9C2 cells. Naunyn Schmiedebergs Arch Pharmacol 2011; 384:209-16. [PMID: 21614555 DOI: 10.1007/s00210-011-0657-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2011] [Accepted: 05/11/2011] [Indexed: 12/22/2022]
Abstract
Prazosin, a quinazoline-based α(1)-adrenoceptor antagonist, is known to induce cell death, and this effect is independent of its α-blockade activity. However, the detailed molecular mechanisms involved are still not fully understood. In this study, we found that prazosin, but not doxazosin, could induce patterns of autophagy in H9C2 cells, including intracellular vacuole formation, microtubule-associated protein 1 light chain 3 (LC3) conversion, and acidic vesicular organelle (AVO) augmentation. Western blot analysis of phosphorylated proteins showed that exposure to prazosin increased the levels of phospho-p53 and phospho-adenosine monophosphate-activated protein kinase (AMPK) but dramatically decreased the levels of phospho-mammalian target of rapamycin (mTOR), phospho-protein kinase B (Akt), and phospho-ribosomal protein S6 kinase (p70S6K). Furthermore, although pretreatments with the pharmacological autophagy inhibitor 3-methyladenine and the p53 inhibitor pifithrin-α suppressed prazosin-induced AVO formation, they did not reverse prazosin-induced decline in cell viability but enhanced prazosin-induced caspase-3 activation. From these results we suggest that prazosin induces autophagic cell death via a p53-mediated mechanism. When the autophagy pathway was inhibited, prazosin still induced programmed cell death, at least in part through apoptotic caspase-3 cascade enhancement. Thus, our results indicate a potential new target in prazosin-induced cell death.
Collapse
Affiliation(s)
- Yi-Fan Yang
- Institute of Pharmacology, College of Medicine, National Taiwan University, No. 1 Sec. 1 Jen-Ai Road, Taipei, Taiwan
| | | | | | | | | |
Collapse
|
502
|
Sermeus A, Michiels C. Reciprocal influence of the p53 and the hypoxic pathways. Cell Death Dis 2011; 2:e164. [PMID: 21614094 PMCID: PMC3122125 DOI: 10.1038/cddis.2011.48] [Citation(s) in RCA: 212] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Accepted: 04/19/2011] [Indexed: 12/17/2022]
Abstract
When cells sense a decrease in oxygen availability (hypoxia), they develop adaptive responses in order to sustain this condition and survive. If hypoxia lasts too long or is too severe, the cells eventually die. Hypoxia is also known to modulate the p53 pathway, in a manner dependent or not of HIF-1 (hypoxia-inducible factor-1), the main transcription factor activated by hypoxia. The p53 protein is a transcription factor, which is rapidly stabilised by cellular stresses and which has a major role in the cell responses to these stresses. The aim of this review is to compile what has been reported until now about the interconnection between these two important pathways. Indeed, according to the cell line, the severity and the duration of hypoxia, oxygen deficiency influences very differently p53 protein level and activity. Conversely, p53 is also described to affect HIF-1α stability, one of the two subunits of HIF-1, and HIF-1 activity. The direct and indirect interactions between HIF-1α and p53 are described as well as the involvement in this complex network of their respective ubiquitin ligases von Hippel Lindau protein and murine double minute 2. Finally, the synergistic or antagonistic effects of p53 and HIF-1 on some important cellular pathways are discussed.
Collapse
Affiliation(s)
- A Sermeus
- Laboratory of Biochemistry and Cellular Biology (URBC), NARILIS, University of Namur–FUNDP, Namur, Belgium
| | - C Michiels
- Laboratory of Biochemistry and Cellular Biology (URBC), NARILIS, University of Namur–FUNDP, Namur, Belgium
| |
Collapse
|
503
|
Patel J, Pathak RR, Mujtaba S. The biology of lysine acetylation integrates transcriptional programming and metabolism. Nutr Metab (Lond) 2011; 8:12. [PMID: 21371315 PMCID: PMC3060110 DOI: 10.1186/1743-7075-8-12] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Accepted: 03/03/2011] [Indexed: 11/18/2022] Open
Abstract
The biochemical landscape of lysine acetylation has expanded from a small number of proteins in the nucleus to a multitude of proteins in the cytoplasm. Since the first report confirming acetylation of the tumor suppressor protein p53 by a lysine acetyltransferase (KAT), there has been a surge in the identification of new, non-histone targets of KATs. Added to the known substrates of KATs are metabolic enzymes, cytoskeletal proteins, molecular chaperones, ribosomal proteins and nuclear import factors. Emerging studies demonstrate that no fewer than 2000 proteins in any particular cell type may undergo lysine acetylation. As described in this review, our analyses of cellular acetylated proteins using DAVID 6.7 bioinformatics resources have facilitated organization of acetylated proteins into functional clusters integral to cell signaling, the stress response, proteolysis, apoptosis, metabolism, and neuronal development. In addition, these clusters also depict association of acetylated proteins with human diseases. These findings not only support lysine acetylation as a widespread cellular phenomenon, but also impel questions to clarify the underlying molecular and cellular mechanisms governing target selectivity by KATs. Present challenges are to understand the molecular basis for the overlapping roles of KAT-containing co-activators, to differentiate between global versus dynamic acetylation marks, and to elucidate the physiological roles of acetylated proteins in biochemical pathways. In addition to discussing the cellular 'acetylome', a focus of this work is to present the widespread and dynamic nature of lysine acetylation and highlight the nexus that exists between epigenetic-directed transcriptional regulation and metabolism.
Collapse
Affiliation(s)
- Jigneshkumar Patel
- Department of Structural and Chemical Biology, Mount Sinai School of Medicine New York, NY 10029 USA.
| | | | | |
Collapse
|
504
|
Sinthupibulyakit C, Ittarat W, St Clair WH, St Clair DK. p53 Protects lung cancer cells against metabolic stress. Int J Oncol 2011; 37:1575-81. [PMID: 21042727 DOI: 10.3892/ijo_00000811] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The preferential use of aerobic glycolysis for energy production by cancer cells, a phenomenon known as the 'Warburg effect', is well recognized and is being considered for therapeutic applications. However, whether inhibition of glycolysis will be effective in all types of cancer is unclear. The current study shows that a glycolytic inhibitor, 2-deoxy-D-glucose (2DG), exhibits the cytotoxic effect on non-small cell lung cancer in a p53-dependent manner. 2DG significantly inhibits ATP production in p53-deficient lung cancer cells (H358) but not in p53-wt cells (A549). In contrast to p53-wt cells, p53-defective cells are unable to compensate for their need of energy via oxidative phosphorylation (OXPHOS) when glycolysis is inhibited. In the presence of p53, increased ROS from OXPHOS increases the expression of p53 target genes known to modulate metabolism, including synthesis of cytochrome c oxidase 2 (SCO2) and TP53-induced glycolysis and apoptosis regulator (TIGAR). Importantly, 2DG selectively induces the expression of the antioxidant enzymes manganese superoxide dismutase (MnSOD) and glutathione peroxidase 1 (GPx1) in a p53-dependent manner. The results demonstrate that the killing of cancer cells by the inhibitor of glycolysis is more efficient in cancer cells without functional p53 and that p53 protects against metabolic stress by up-regulation of oxidative phosphorylation and modulation of antioxidants.
Collapse
|
505
|
Maddocks ODK, Vousden KH. Metabolic regulation by p53. J Mol Med (Berl) 2011; 89:237-45. [PMID: 21340684 PMCID: PMC3043245 DOI: 10.1007/s00109-011-0735-5] [Citation(s) in RCA: 214] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2011] [Accepted: 01/21/2011] [Indexed: 01/24/2023]
Abstract
We are increasingly aware that cellular metabolism plays a vital role in diseases such as cancer, and that p53 is an important regulator of metabolic pathways. By transcriptional activation and other means, p53 is able to contribute to the regulation of glycolysis, oxidative phosphorylation, glutaminolysis, insulin sensitivity, nucleotide biosynthesis, mitochondrial integrity, fatty acid oxidation, antioxidant response, autophagy and mTOR signalling. The ability to positively and negatively regulate many of these pathways, combined with feedback signalling from these pathways to p53, demonstrates the reciprocal and flexible nature of the regulation, facilitating a diverse range of responses to metabolic stress. Intriguingly, metabolic stress triggers primarily an adaptive (rather than pro-apoptotic) p53 response, and p53 is emerging as an important regulator of metabolic homeostasis. A better understanding of how p53 coordinates metabolic adaptation will facilitate the identification of novel therapeutic targets and will also illuminate the wider role of p53 in human biology.
Collapse
|
506
|
|
507
|
Zheltukhin AO, Chumakov PM. Constitutive and induced functions of the p53 gene. BIOCHEMISTRY (MOSCOW) 2011; 75:1692-721. [DOI: 10.1134/s0006297910130110] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
508
|
Zhao Y, Liu H, Riker AI, Fodstad O, Ledoux SP, Wilson GL, Tan M. Emerging metabolic targets in cancer therapy. Front Biosci (Landmark Ed) 2011; 16:1844-60. [PMID: 21196269 DOI: 10.2741/3826] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cancer cells are different from normal cells in their metabolic properties. Normal cells mostly rely on mitochondrial oxidative phosphorylation to produce energy. In contrast, cancer cells depend mostly on glycolysis, the aerobic breakdown of glucose into ATP. This altered energy dependency is known as the "Warburg effect" and is a hallmark of cancer cells. In recent years, investigating the metabolic changes within cancer cells has been a rapidly growing area. Emerging evidence shows that oncogenes that drive the cancer-promoting signals also drive the altered metabolism. Although the exact mechanisms underlying the Warburg effect are unclear, the existing evidence suggests that increased glycolysis plays an important role in support malignant behavior of cancer cells. A thorough understanding of the unique metabolism of cancer cells will help to design of more effective drugs targeting metabolic pathways, which will greatly impact the capacity to effectively treat cancer patients. Here we provide an overview of the current understanding of the Warburg effect upon tumor cell growth and survival, and discussion on the potential metabolic targets for cancer therapy.
Collapse
Affiliation(s)
- Yuhua Zhao
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL 36604, USA
| | | | | | | | | | | | | |
Collapse
|
509
|
Abstract
Cellular energy metabolism is one of the main processes affected during the transition from normal to cancer cells, and it is a crucial determinant of cell proliferation or cell death. As a support for rapid proliferation, cancer cells choose to use glycolysis even in the presence of oxygen (Warburg effect) to fuel macromolecules for the synthesis of nucleotides, fatty acids, and amino acids for the accelerated mitosis, rather than fuel the tricarboxylic acid cycle and oxidative phosphorylation. Mitochondria biogenesis is also reprogrammed in cancer cells, and the destiny of those cells is determined by the balance between energy and macromolecule supplies, and the efficiency of buffering of the cumulative radical oxygen species. In glioblastoma, the most frequent and malignant adult brain tumor, a metabolic shift toward aerobic glycolysis is observed, with regulation by well known genes as integrants of oncogenic pathways such as phosphoinositide 3-kinase/protein kinase, MYC, and hypoxia regulated gene as hypoxia induced factor 1. The expression profile of a set of genes coding for glycolysis and the tricarboxylic acid cycle in glioblastoma cases confirms this metabolic switch. An understanding of how the main metabolic pathways are modified by cancer cells and the interactions between oncogenes and tumor suppressor genes with these pathways may enlighten new strategies in cancer therapy. In the present review, the main metabolic pathways are compared in normal and cancer cells, and key regulations by the main oncogenes and tumor suppressor genes are discussed. Potential therapeutic targets of the cancer energetic metabolism are enumerated, highlighting the astrocytomas, the most common brain cancer.
Collapse
|
510
|
Reconstruction of protein-protein interaction network of insulin signaling in Homo sapiens. J Biomed Biotechnol 2010; 2010:690925. [PMID: 21197403 PMCID: PMC3010689 DOI: 10.1155/2010/690925] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2010] [Revised: 07/22/2010] [Accepted: 10/27/2010] [Indexed: 01/23/2023] Open
Abstract
Diabetes is one of the most prevalent diseases in the world. Type 1 diabetes is characterized by the failure of synthesizing and secreting of insulin because of destroyed pancreatic β-cells. Type 2 diabetes, on the other hand, is described by the decreased synthesis and secretion of insulin because of the defect in pancreatic β-cells as well as by the failure of responding to insulin because of malfunctioning of insulin signaling. In order to understand the signaling mechanisms of responding to insulin, it is necessary to identify all components in the insulin signaling network. Here, an interaction network consisting of proteins that have statistically high probability of being biologically related to insulin signaling in Homo sapiens was reconstructed by integrating Gene Ontology (GO) annotations and interactome data. Furthermore, within this reconstructed network, interacting proteins which mediate the signal from insulin hormone to glucose transportation were identified using linear paths. The identification of key components functioning in insulin action on glucose metabolism is crucial for the efforts of preventing and treating type 2 diabetes mellitus.
Collapse
|
511
|
Bodily JM, Mehta KPM, Laimins LA. Human papillomavirus E7 enhances hypoxia-inducible factor 1-mediated transcription by inhibiting binding of histone deacetylases. Cancer Res 2010; 71:1187-95. [PMID: 21148070 DOI: 10.1158/0008-5472.can-10-2626] [Citation(s) in RCA: 113] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Infection by human papillomaviruses (HPV) leads to the formation of benign lesions, warts, and in some cases, cervical cancer. The formation of these lesions is dependent upon increased expression of proangiogenic factors. Angiogenesis is linked to tissue hypoxia through the activity of the oxygen-sensitive hypoxia-inducible factor 1α (HIF-1α). Our studies indicate that the HPV E7 protein enhances HIF-1 transcriptional activity whereas E6 functions to counteract the repressive effects of p53. Both high- and low-risk HPV E7 proteins were found to bind to HIF-1α through a domain located in the N-terminus. Importantly, the ability of E7 to enhance HIF-1 activity mapped to the C-terminus and correlated with the displacement of the histone deacetylases HDAC1, HDAC4, and HDAC7 from HIF-1α by E7. Our findings describe a novel role of the E7 oncoprotein in activating the function of a key transcription factor mediating hypoxic responses by blocking the binding of HDACs.
Collapse
Affiliation(s)
- Jason M Bodily
- Department of Microbiology-Immunology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois 60611, USA
| | | | | |
Collapse
|
512
|
Groheux D, Giacchetti S, Moretti JL, Porcher R, Espié M, Lehmann-Che J, de Roquancourt A, Hamy AS, Cuvier C, Vercellino L, Hindié E. Correlation of high 18F-FDG uptake to clinical, pathological and biological prognostic factors in breast cancer. Eur J Nucl Med Mol Imaging 2010; 38:426-35. [PMID: 21057787 DOI: 10.1007/s00259-010-1640-9] [Citation(s) in RCA: 294] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2010] [Accepted: 10/04/2010] [Indexed: 12/13/2022]
Abstract
PURPOSE The aim of this study was to determine the impact of the main clinicopathological and biological prognostic factors of breast cancer on (18)F-fluorodeoxyglucose (FDG) uptake. Only women with tumours larger than 20 mm (T2-T4) were included in order to minimize bias of partial volume effect. METHODS In this prospective study, 132 consecutive women received FDG PET/CT imaging before starting neoadjuvant chemotherapy. Maximum standardized uptake values (SUV(max)) were compared to tumour characteristics as assessed on core biopsy. RESULTS There was no influence of T and N stage on SUV. Invasive ductal carcinoma showed higher SUV than lobular carcinoma. However, the highest uptake was found for metaplastic tumours, representing 5% of patients in this series. Several biological features usually considered as bad prognostic factors were associated with an increase in FDG uptake: the median of SUV(max) was 9.7 for grade 3 tumours vs 4.8 for the lower grades (p < 0.0001); negativity for oestrogen receptors (ER) was associated with higher SUV (ER+ SUV = 5.5; ER- SUV = 7.6; p = 0.003); triple-negative tumours (oestrogen and progesterone receptor negative, no overexpression of c-erbB-2) had an SUV of 9.2 vs 5.8 for all others (p = 0005); p53 mutated tumours also had significantly higher SUV (7.8 vs 5.0; p < 0.0001). Overexpression of c-erbB-2 had no effect on the SUV value. CONCLUSION Knowledge of the factors influencing uptake is important when interpreting FDG PET/CT scans. Also, findings that FDG uptake is highest in those patients with poor prognostic features (high grade, hormone receptor negativity, triple negativity, metaplastic tumours) is helpful to determine who are the best candidates for baseline staging.
Collapse
Affiliation(s)
- David Groheux
- Department of Nuclear Medicine, Saint-Louis Hospital, Assistance publique Hôpitaux de Paris, Paris, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
513
|
Requirement of the ATM/p53 tumor suppressor pathway for glucose homeostasis. Mol Cell Biol 2010; 30:5787-94. [PMID: 20956556 DOI: 10.1128/mcb.00347-10] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Ataxia telangiectasia (A-T) patients can develop multiple clinical pathologies, including neuronal degeneration, an elevated risk of cancer, telangiectasias, and growth retardation. Patients with A-T can also exhibit an increased risk of insulin resistance and type 2 diabetes. The ATM protein kinase, the product of the gene mutated in A-T patients (Atm), has been implicated in metabolic disease, which is characterized by insulin resistance and increased cholesterol and lipid levels, blood pressure, and atherosclerosis. ATM phosphorylates the p53 tumor suppressor on a site (Ser15) that regulates transcription activity. To test whether the ATM pathway that regulates insulin resistance is mediated by p53 phosphorylation, we examined insulin sensitivity in mice with a germ line mutation that replaces the p53 phosphorylation site with alanine. The loss of p53 Ser18 (murine Ser15) led to increased metabolic stress, including severe defects in glucose homeostasis. The mice developed glucose intolerance and insulin resistance. The insulin resistance correlated with the loss of antioxidant gene expression and decreased insulin signaling. N-Acetyl cysteine (NAC) treatment restored insulin signaling in late-passage primary fibroblasts. The addition of an antioxidant in the diet rendered the p53 Ser18-deficient mice glucose tolerant. This analysis demonstrates that p53 phosphorylation on an ATM site is an important mechanism in the physiological regulation of glucose homeostasis.
Collapse
|
514
|
Schneider G, Krämer OH. NFκB/p53 crosstalk-a promising new therapeutic target. Biochim Biophys Acta Rev Cancer 2010; 1815:90-103. [PMID: 20951769 DOI: 10.1016/j.bbcan.2010.10.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Revised: 10/02/2010] [Accepted: 10/07/2010] [Indexed: 12/12/2022]
Abstract
The transcription factors p53 and NFκB determine cellular fate and are involved in the pathogenesis of most-if not all-cancers. The crosstalk between these transcription factors becomes increasingly appreciated as an important mechanism operative during all stages of tumorigenesis, metastasis, and immunological surveillance. In this review, we summarize molecular mechanisms regulating cross-signaling between p53 and NFκB proteins and how dysregulated interactions between p53 and NFκB family members contribute to oncogenesis. We furthermore analyze how such signaling modules represent targets for the design of novel intervention strategies using established compounds and powerful combination therapies.
Collapse
Affiliation(s)
- Günter Schneider
- Technische Universität München, Klinikum rechts der Isar, II. Medizinische Klinik, Ismaninger Str. 22, D-81675 München, Germany
| | | |
Collapse
|
515
|
Watanabe M, Naraba H, Sakyo T, Kitagawa T. DNA damage-induced modulation of GLUT3 expression is mediated through p53-independent extracellular signal-regulated kinase signaling in HeLa cells. Mol Cancer Res 2010; 8:1547-57. [PMID: 20870738 DOI: 10.1158/1541-7786.mcr-10-0011] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Many cancer cells exhibit increased rates of uptake and metabolism of glucose compared with normal cells. Glucose uptake in mammalian cells is mediated by the glucose transporter (GLUT) family. Here, we report that DNA-damaging anticancer agents such as Adriamycin and etoposide suppressed the expression of GLUT3, but not GLUT1, in HeLa cells and a tumorigenic HeLa cell hybrid. Suppression of GLUT3 expression determined by the real-time PCR was also evident with another DNA-damaging agent, camptothecin, which reduced the promoter's activity as determined with a luciferase-linked assay. The suppression by these agents seemed to be induced independently of p53, and it was evident when wild-type p53 was overproduced in these cells. In contrast, the mitogen-activated protein kinase/extracellular signal regulated kinase (MAPK/ERK) kinase (MEK) inhibitor U0126 (but not the phosphoinositide 3-kinase inhibitor LY294002) prevented the drug-induced suppression as determined by reverse transcription-PCR and promoter assays. Furthermore, overexpression of GLUT3 in HeLa cell hybrids increased resistance to these drugs, whereas depletion of the gene by small interfering RNA rendered the cells more sensitive to the drugs, decreasing glucose consumption. The results suggest that DNA-damaging agents reduce GLUT3 expression in cancer cells through activation of the MEK-ERK pathway independently of p53, leading to cell death or apoptosis. The findings may contribute to the development of new chemotherapeutic drugs based on the GLUT3-dependent metabolism of glucose.
Collapse
Affiliation(s)
- Masaru Watanabe
- Department of Cell Biology and Molecular Pathology, Iwate Medical University, School of Pharmacy, 2-1-1 Nishitokuta, Yahaba, Shiwa, Iwate 028-3694, Japan
| | | | | | | |
Collapse
|
516
|
Fan J, Zhou JQ, Yu GR, Lu DD. Glucose transporter protein 1-targeted RNA interference inhibits growth and invasion of the osteosarcoma cell line MG63 in vitro. Cancer Biother Radiopharm 2010; 25:521-7. [PMID: 20854211 DOI: 10.1089/cbr.2010.0784] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Malignant cells show increased glucose uptake, which is thought to be mediated by glucose transporters. Glucose transporter protein 1 (Glut-1) is critical for growth, proliferation, and migration of tumor cells and Glut-1 overexpression is associated with poor overall survival in osteosarcoma patients. The present study was designed to determine the role of Glut-1 in the growth and invasion of the osteosarcoma cell line MG63, using RNA interference technology in vitro. shRNA-expressing lentiviral vectors targeting the Glut-1 gene were constructed, which were stably expressed in MG63 cells. The level of Glut-1 mRNA was investigated using real-time reverse transcription-polymerase chain reaction, and the protein expression of Glut-1 mRNA was observed using western blotting. MG63 cellular glucose uptake, proliferation, and migration were detected by methyl thiazole tetrazolium assay and flow cytometry. A Glut-1-specific shRNA-expressing lentiviral vector was obtained, which could efficiently inhibit the mRNA and protein expression of Glut-1 to ∼82%-85% in MG63 cells. Downregulation of Glut-1 inhibited the cellular glucose uptake, growth, and invasion of MG63 cells in vitro. These results indicate that RNA interference targeting of Glut-1 could be an effective strategy for the treatment of osteoscarcoma patients.
Collapse
Affiliation(s)
- Jian Fan
- Department of Orthopedics, Tongji University, Tongji Hospital, Shanghai, People's Republic of China
| | | | | | | |
Collapse
|
517
|
The metabolic switch and its regulation in cancer cells. SCIENCE CHINA-LIFE SCIENCES 2010; 53:942-58. [PMID: 20821293 DOI: 10.1007/s11427-010-4041-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2010] [Accepted: 05/07/2010] [Indexed: 02/05/2023]
Abstract
The primary features of cancer are maintained via intrinsically modified metabolic activity, which is characterized by enhanced nutrient supply, energy production, and biosynthetic activity to synthesize a variety of macromolecular components during each passage through the cell cycle. This metabolic shift in transformed cells, as compared with non-proliferating cells, involves aberrant activation of aerobic glycolysis, de novo lipid biosynthesis and glutamine-dependent anaplerosis to fuel robust cell growth and proliferation. Here, we discuss the unique metabolic characteristics of cancer, the constitutive regulation of metabolism through a variety of signal transduction pathways and/or enzymes involved in metabolic reprogramming in cancer cells, and their implications in cancer diagnosis and therapy.
Collapse
|
518
|
Abstract
The p53 tumor suppressor gene has recently been shown to mediate metabolic changes in cells under physiological and pathological conditions. It has been revealed that p53 regulates energy metabolism, oxidative stress, and amino acid metabolism through balancing glycolysis and oxidative phosphorylation (OXPHOS) as well as the autophagy pathway. p53 is activated by metabolic stress through AMP-activated protein kinase (AMPK) and the mammalian target of rapamycin (mTOR) signaling pathways. p53 regulates OXPHOS through the transcriptional regulation of fructose-2,6-bisphosophatase, TP53-induced glycolysis regulator (TIGAR) and synthesis of cytochrome c oxidase (SCO2) subunit of complex IV of the electron transport chain. p53 also indirectly influences the energy metabolism through regulating glucose transporter (GLUT) expression, glutaminase 2 (GLS2) and fatty acid synthase (FAS). In addition, p53 regulates autophagy to provide cell metabolites for surviving through damage regulated autophagy modulator (DRAM1). Here we review the recent findings to elucidate the important role of p53 in cell metabolism.
Collapse
Affiliation(s)
- Xing-ding Zhang
- Department of Pharmacology and Aging and Nervous Diseases, Soochow University School of Medicine, Suzhou 215123, China
| | - Zheng-hong Qin
- Department of Pharmacology and Aging and Nervous Diseases, Soochow University School of Medicine, Suzhou 215123, China
| | - Jin Wang
- Department of Pharmacology and Aging and Nervous Diseases, Soochow University School of Medicine, Suzhou 215123, China
| |
Collapse
|
519
|
Aung W, Hasegawa S, Koshikawa-Yano M, Tsuji AB, Sogawa C, Sudo H, Sugyo A, Koizumi M, Furukawa T, Saga T. Noninvasive assessment of regulable transferred-p53 gene expression and evaluation of therapeutic response with FDG-PET in tumor model. Gene Ther 2010; 17:1142-1151. [PMID: 20445579 DOI: 10.1038/gt.2010.70] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2009] [Revised: 02/23/2010] [Accepted: 03/18/2010] [Indexed: 01/18/2023]
Abstract
The use of tumor-suppressor gene p53 as an anticancer therapeutic has been vigorously investigated. However, progress has met with limited success to date. Some major drawbacks are the difficulty in achieving controllable and efficient gene transfer as well as in analyzing the transferred gene expression in real time and the treatment response in a timely manner. Thus, development of novel gene transfer vector with a regulative gene expression system coupled with the reporter gene, by which transgene can be monitored simultaneously, is critical. Moreover, noninvasive imaging-based assessment of the therapeutic response to exogenous wild-type p53 gene transfer is crucial for refining treatment protocols. In this study, as a simple preclinical model, we constructed a doxycycline-regulated bidirectional vector harboring a reporter gene encoding red fluorescence protein and p53. Then, we determined the controllable and simultaneously coordinated expression of both proteins and the p53-mediated anticancer effects in vitro and in vivo. Next, we observed that cells or tumors with induced p53 overexpression exhibited decreased uptake of [(14)C]FDG in cellular assay and [(18)F]FDG in positron emission tomography (PET) imaging. Thus, by coupling with bidirectional vector, controllable p53 transfer was achieved and the capability of fluoro-2-deoxy-D-glucose (FDG)-PET to assess the therapeutic response to p53 gene therapy was evidently confirmed, which may have an impact on the improvement of p53 gene therapy.
Collapse
Affiliation(s)
- W Aung
- Diagnostic Imaging Group, Molecular Imaging Center, National Institute of Radiological Sciences, Chiba, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
520
|
The regulation of energy metabolism and the IGF-1/mTOR pathways by the p53 protein. Trends Cell Biol 2010; 20:427-34. [PMID: 20399660 DOI: 10.1016/j.tcb.2010.03.004] [Citation(s) in RCA: 291] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 03/19/2010] [Accepted: 03/24/2010] [Indexed: 01/09/2023]
Abstract
In response to stress, p53 initiates the transcriptional regulation of selected target genes and various cellular responses, including cell cycle arrest, apoptosis and senescence. Recent studies revealed two additional functions of p53 in the regulation of IGF-1/AKT/mTOR pathways and energy metabolism, contributing to p53's role as a tumor suppressor. Oncogenic processes give rise to metabolic pathways focused upon the use of aerobic glycolysis (the Warburg effect) and the pentose shunt, providing higher levels of reducing activities. p53 shuts down these pathways and refocuses cells to utilize mitochondrial oxidative phosphorylation, thereby maximizing efficient ATP production and minimizing the synthesis of substrates for cell division. The use of these alternative metabolic pathways is an integral part of both normal and oncogenic phenotypes.
Collapse
|
521
|
Chen G, Wang F, Trachootham D, Huang P. Preferential killing of cancer cells with mitochondrial dysfunction by natural compounds. Mitochondrion 2010; 10:614-25. [PMID: 20713185 DOI: 10.1016/j.mito.2010.08.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Revised: 08/03/2010] [Accepted: 08/04/2010] [Indexed: 01/06/2023]
Abstract
Mitochondria play essential roles in cellular metabolism, redox homeostasis, and regulation of cell death. Emerging evidences suggest that cancer cells exhibit various degrees of mitochondrial dysfunctions and metabolic alterations, which may serve as a basis to develop therapeutic strategies to preferentially kill the malignant cells. Mitochondria as a therapeutic target for cancer treatment is gaining much attention in the recent years, and agents that impact mitochondria with anticancer activity have been identified and tested in vitro and in vivo using various experimental systems. Anticancer agents that directly target mitochondria or indirectly affect mitochondrial functions are collectively classified as mitocans. This review article focuses on several natural compounds that preferentially kill cancer cells with mitochondrial dysfunction, and discusses the possible underlying mechanisms and their therapeutic implications in cancer treatment. Mitocans that have been comprehensively reviewed recently are not included in this article. Important issues such as therapeutic selectivity and the relevant biochemical basis are discussed in the context of future perspectives.
Collapse
Affiliation(s)
- Gang Chen
- Department of Molecular Pathology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | | | | | | |
Collapse
|
522
|
Mulukutla BC, Khan S, Lange A, Hu WS. Glucose metabolism in mammalian cell culture: new insights for tweaking vintage pathways. Trends Biotechnol 2010; 28:476-84. [PMID: 20691487 DOI: 10.1016/j.tibtech.2010.06.005] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Revised: 06/10/2010] [Accepted: 06/16/2010] [Indexed: 10/19/2022]
Abstract
Cultured mammalian cells are major vehicles for producing therapeutic proteins, and energy metabolism in those cells profoundly affects process productivity. The characteristic high glucose consumption and lactate production of industrial cell lines as well as their adverse effects on productivity have been the target of both cell line and process improvement for several decades. Recent research advances have shed new light on regulation of glucose metabolism and its links to cell proliferation. This review highlights our current understanding in this area of crucial importance in bioprocessing and further discusses strategies for harnessing new findings toward process enhancement through the manipulation of cellular energy metabolism.
Collapse
Affiliation(s)
- Bhanu Chandra Mulukutla
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, MN, USA
| | | | | | | |
Collapse
|
523
|
Ai B, Fu X, Liao Y. [The relationship of GLUT1 and lung cancer and its PET imaging]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2010; 13:165-7. [PMID: 20673512 PMCID: PMC6000514 DOI: 10.3779/j.issn.1009-3419.2010.02.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Bo Ai
- Department of Thoracic Surgery, Tongji Hospital, Huazhong University of Science and Technology, Wuhan 430030, China.
| | | | | |
Collapse
|
524
|
Ak P, Levine AJ. p53 and NF-κB: different strategies for responding to stress lead to a functional antagonism. FASEB J 2010; 24:3643-52. [PMID: 20530750 DOI: 10.1096/fj.10-160549] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The p53 transcription factor responds to a variety of intrinsic stresses, such as DNA damage, hypoxia, and even oncogene activation. NF-κB responds to a large number of extrinsic stresses such as cytokine activation and infectious diseases. The p53 tumor suppressor limits the consequences of stress by initiating cell death, senescence, or cell cycle arrest and promotes metabolic patterns in the cell to favor oxidative phosphorylation. NF-κB, the oncogene, promotes cell division, which initiates the innate and adaptive immune responses utilizing large amounts of glucose in aerobic glycolysis, resulting in the synthesis of substrates for cell division. Thus these two transcription factors, both of which have evolved to respond to different types of stress, have adopted opposite strategies and cannot function in the same cell at the same time. On activation of one of these transcription factors, the other is inactivated. This is achieved at several places in the p53 and NF-κB pathways where regulatory proteins act on both p53 and NF-κB with opposite functional consequences. These internodal sites create core regulatory circuits essential for integrating two central pathways in cells.
Collapse
Affiliation(s)
- Prashanth Ak
- Institute for Advanced Study, Princeton, NJ 08540, USA
| | | |
Collapse
|
525
|
Fritz V, Fajas L. Metabolism and proliferation share common regulatory pathways in cancer cells. Oncogene 2010; 29:4369-77. [PMID: 20514019 DOI: 10.1038/onc.2010.182] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cancer development involves major alterations in cells' metabolism. Enhanced glycolysis and de novo fatty acids synthesis are indeed characteristic features of cancer. Cell proliferation and metabolism are tightly linked cellular processes. Others and we have previously shown a close relationship between metabolic responses and proliferative stimuli. In addition to trigger proliferative and survival signaling pathways, most oncoproteins also trigger metabolic changes to transform the cell. We present herein the view that participation of cell-cycle regulators and oncogenic proteins to cancer development extend beyond the control of cell proliferation, and discuss how these new functions may be implicated in metabolic alterations concomitant to the pathogenesis of human cancers.
Collapse
Affiliation(s)
- V Fritz
- Institut de Recherche en Cancérologie de Montpellier, Montpellier, France
| | | |
Collapse
|
526
|
The role of p53 in glucose metabolism. Curr Opin Cell Biol 2010; 22:186-91. [DOI: 10.1016/j.ceb.2009.12.006] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Revised: 12/08/2009] [Accepted: 12/10/2009] [Indexed: 02/04/2023]
|
527
|
Phosphate-activated glutaminase (GLS2), a p53-inducible regulator of glutamine metabolism and reactive oxygen species. Proc Natl Acad Sci U S A 2010; 107:7461-6. [PMID: 20351271 DOI: 10.1073/pnas.1002459107] [Citation(s) in RCA: 512] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
We identified a p53 target gene, phosphate-activated mitochondrial glutaminase (GLS2), a key enzyme in conversion of glutamine to glutamate, and thereby a regulator of glutathione (GSH) synthesis and energy production. GLS2 expression is induced in response to DNA damage or oxidative stress in a p53-dependent manner, and p53 associates with the GLS2 promoter. Elevated GLS2 facilitates glutamine metabolism and lowers intracellular reactive oxygen species (ROS) levels, resulting in an overall decrease in DNA oxidation as determined by measurement of 8-OH-dG content in both normal and stressed cells. Further, siRNA down-regulation of either GLS2 or p53 compromises the GSH-dependent antioxidant system and increases intracellular ROS levels. High ROS levels following GLS2 knockdown also coincide with stimulation of p53-induced cell death. We propose that GLS2 control of intracellular ROS levels and the apoptotic response facilitates the ability of p53 to protect cells from accumulation of genomic damage and allows cells to survive after mild and repairable genotoxic stress. Indeed, overexpression of GLS2 reduces the growth of tumor cells and colony formation. Further, compared with normal tissue, GLS2 expression is reduced in liver tumors. Thus, our results provide evidence for a unique metabolic role for p53, linking glutamine metabolism, energy, and ROS homeostasis, which may contribute to p53 tumor suppressor function.
Collapse
|
528
|
Airley R, Evans A, Mobasheri A, Hewitt SM. Glucose transporter Glut-1 is detectable in peri-necrotic regions in many human tumor types but not normal tissues: Study using tissue microarrays. Ann Anat 2010; 192:133-8. [PMID: 20395120 DOI: 10.1016/j.aanat.2010.03.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Revised: 02/11/2010] [Accepted: 03/01/2010] [Indexed: 12/16/2022]
Abstract
The hypoxic tumor microenvironment is associated with malignant progression and poor treatment response. The glucose transporter Glut-1 is a prognostic factor and putative hypoxia marker. So far, studies of Glut-1 in cancer have utilized conventional immunohistochemical analysis in a series of individual biopsy or surgical specimens. Tissue microarrays, however, provide a rapid, inexpensive means of profiling biomarker expression. To evaluate hypoxia markers, tissue cores must show the architectural features of hypoxia; i.e. viable tissue surrounding necrotic regions. Glut-1 may be a useful biomarker to validate tissue microarrays for use in studies of hypoxia-regulated genes in cancer. In this study, we carried out immunohistochemical detection of Glut-1 protein in many tumor and normal tissue types in a range of tissue microarrays. Glut-1 was frequently found in peri-necrotic regions, occurring in 9/34 lymphomas, 6/12 melanomas, and 5/16 glioblastomas; and in 43/54 lung, 22/84 colon, and 23/60 ovarian tumors. Expression was rare in breast (6/40) and prostate (1/57) tumors, and in normal tissue, was restricted to spleen, tongue, and CNS endothelium. In conclusion, tissue microarrays enable the observation of Glut-1 expression in peri-necrotic regions, which may be linked to hypoxia, and reflect previous studies showing differential Glut-1 expression across tumor types and non-malignant tissue.
Collapse
Affiliation(s)
- Rachel Airley
- Division of Pharmacy and Pharmaceutical Sciences, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield, West Yorkshire, UK.
| | | | | | | |
Collapse
|
529
|
Calvo MB, Figueroa A, Pulido EG, Campelo RG, Aparicio LA. Potential role of sugar transporters in cancer and their relationship with anticancer therapy. Int J Endocrinol 2010; 2010:205357. [PMID: 20706540 PMCID: PMC2913528 DOI: 10.1155/2010/205357] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2010] [Accepted: 06/20/2010] [Indexed: 12/18/2022] Open
Abstract
Sugars, primarily glucose and fructose, are the main energy source of cells. Because of their hydrophilic nature, cells use a number of transporter proteins to introduce sugars through their plasma membrane. Cancer cells are well known to display an enhanced sugar uptake and consumption. In fact, sugar transporters are deregulated in cancer cells so they incorporate higher amounts of sugar than normal cells. In this paper, we compile the most significant data available about biochemical and biological properties of sugar transporters in normal tissues and we review the available information about sugar carrier expression in different types of cancer. Moreover, we describe the possible pharmacological interactions between drugs currently used in anticancer therapy and the expression or function of facilitative sugar transporters. Finally, we also go into the insights about the future design of drugs targeted against sugar utilization in cancer cells.
Collapse
Affiliation(s)
- Moisés Blanco Calvo
- Biomedical Research Institute, A Coruña University Hospital, As Xubias 84, 15006 A Coruña, Spain
| | - Angélica Figueroa
- Biomedical Research Institute, A Coruña University Hospital, As Xubias 84, 15006 A Coruña, Spain
| | - Enrique Grande Pulido
- Clinical Oncology Department, Ramón y Cajal University Hospital, Ctra. de Colmenar Viejo Km. 9,100, 28034 Madrid, Spain
| | - Rosario García Campelo
- Clinical Oncology Department, A Coruña University Hospital, As Xubias 84, 15006 A Coruña, Spain
| | - Luís Antón Aparicio
- Clinical Oncology Department, A Coruña University Hospital, As Xubias 84, 15006 A Coruña, Spain
- Medicine Department, University of A Coruña, Oza s/n, 15006 A Coruña, Spain
- *Luís Antón Aparicio:
| |
Collapse
|
530
|
Abstract
During the course of tumorigenesis, cells acquire a number of alterations that contribute to the acquisition of the malignant phenotype, allowing them to survive and flourish in increasingly hostile environments. Cancer cells can be characterized by perturbations in the control of cell proliferation and growth, resistance to death, and alterations in their interactions with the microenvironment. Underpinning many of these changes are shifts in metabolism that allow cancer cells to use alternative pathways for energy production and building the macromolecules necessary for growth, as well as regulating the generation of signaling molecules such as reactive oxygen species (ROS). In the past few years, it became clear that p53, the most studied, if not most important, tumor suppressor protein, can also directly control metabolic traits of cells.
Collapse
Affiliation(s)
- Eyal Gottlieb
- The Beatson Institute for Cancer Research, Bearsden, Glasgow, United Kingdom
| | | |
Collapse
|
531
|
Abstract
Historically, it has been assumed that glycolytic metabolism, not mitochondrial metabolism, is essential for tumor cell proliferation. However, most tumor cells have functional mitochondria, and recent studies suggest that the citric acid cycle (TCA) cycle intermediates are precursors for synthesis of nucleotides, lipids, and amino acids. Here we review the accumulating evidence that mitochondrial metabolism plays an essential role in tumor cell proliferation.
Collapse
Affiliation(s)
- Frank Weinberg
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, Northwestern University Medical School, Chicago, IL 60611-2010, USA
| | | |
Collapse
|
532
|
Liu X, Wang X, Zhang J, Lam EKY, Shin VY, Cheng ASL, Yu J, Chan FKL, Sung JJY, Jin HC. Warburg effect revisited: an epigenetic link between glycolysis and gastric carcinogenesis. Oncogene 2009; 29:442-50. [PMID: 19881551 DOI: 10.1038/onc.2009.332] [Citation(s) in RCA: 140] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In cancer cells, glucose is often converted into lactic acid, which is known as the 'Warburg effect'. The reason that cancer cells have a higher rate of aerobic glycolysis, but not oxidative phosphorylation, remains largely unclear. Herein, we proposed an epigenetic mechanism of the Warburg effect. Fructose-1,6-bisphosphatase-1 (FBP1), which functions to antagonize glycolysis was downregulated through NF-kappaB pathway in Ras-transformed NIH3T3 cells. Restoration of FBP1 expression suppressed anchorage-independent growth, indicating the relevance of FBP1 downregulation in carcinogenesis. Indeed, FBP1 was downregulated in gastric carcinomas (P<0.01, n=22) and gastric cancer cell lines (57%, 4/7). Restoration of FBP1 expression reduced growth and glycolysis in gastric cancer cells. Moreover, FBP1 downregulation was reversed by pharmacological demethylation. Its promoter was hypermethylated in gastric cancer cell lines (57%, 4/7) and gastric carcinomas (33%, 33/101). Inhibition of NF-kappaB restored FBP1 expression, partially through demethylation of FBP1 promoter. Notably, Cox regression analysis revealed FBP1 promoter methylation as an independent prognosis predicator for gastric cancer (hazard ratio: 3.60, P=0.010). In summary, we found that NF-kappaB functions downstream of Ras to promote epigenetic downregulation of FBP1. Promoter methylation of FBP1 can be used as a new biomarker for prognosis prediction of gastric cancer. Such an important epigenetic link between glycolysis and carcinogenesis partly explains the Warburg effect.
Collapse
Affiliation(s)
- X Liu
- Institute of Digestive Disease, The Chinese University of Hong Kong, Hong Kong, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
533
|
Sung JY, Kim GY, Lim SJ, Park YK, Kim YW. Expression of the GLUT1 glucose transporter and p53 in carcinomas of the pancreatobiliary tract. Pathol Res Pract 2009; 206:24-9. [PMID: 19819644 DOI: 10.1016/j.prp.2009.07.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2009] [Revised: 07/10/2009] [Accepted: 07/30/2009] [Indexed: 01/12/2023]
Abstract
Only few studies have evaluated the usefulness of the GLUT1 and p53 status of pancreatobiliary tract carcinomas in revealing tumorigenesis. We studied GLUT1 and p53 immunoexpression in a total of 355 cases of the pancreatobiliary carcinoma to determine the biological significance of GLUT1 and p53 expression. Positive expression of GLUT1 was identified in 38 out of 67 (57.7%) ampulla of Vater (AV) carcinomas, in 27 out of 52 (51.8%) pancreatic (PA) carcinomas, in 38 out of 121 (31.4%) extrahepatic bile duct (EBD) carcinomas, and in 53 out of 115 (46.5%) gallbladder (GB) carcinomas. GLUT1 expression in pancreatobiliary carcinomas showed some positive correlation with histological grade, T stage, N stage, TNM stage, and lymphatic invasion. However, p53 expression showed no correlation with any prognostic factors. In the Kaplan-Meier test, positive GLUT1 expression was a poor prognostic factor in the pancreatobiliary tract cancers; however, only GB cancers were statistically significant (P=0.002). Our results suggest that GLUT1 expression in the AV, EBD, and GB carcinomas is associated with some prognostic factors, and GLUT1 expression is associated with a worse prognosis in the patients with GB carcinomas.
Collapse
Affiliation(s)
- Ji-Youn Sung
- Department of Pathology, School of Medicine, Kyung Hee University, #1 Hoiki-dong, Dongdaemun-gu, Seoul 130-701, Republic of Korea
| | | | | | | | | |
Collapse
|
534
|
Abstract
Although metabolic alterations have been observed in cancer for almost a century, only recently have the mechanisms underlying these changes been identified and the importance of metabolic transformation realized. p53 has been shown to respond to metabolic changes and to influence metabolic pathways through several mechanisms. The contributions of these activities to tumour suppression are complex and potentially rather surprising: some reflect the function of basal p53 levels that do not require overt activation and others might even promote, rather than inhibit, tumour progression.
Collapse
Affiliation(s)
- Karen H Vousden
- The Beatson Institute for Cancer Research, Garscube Estate, Glasgow G61 1BD, UK.
| | | |
Collapse
|
535
|
Lee SM, Kim JH, Cho EJ, Youn HD. A nucleocytoplasmic malate dehydrogenase regulates p53 transcriptional activity in response to metabolic stress. Cell Death Differ 2009; 16:738-48. [DOI: 10.1038/cdd.2009.5] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
536
|
Ma G, Shimada H, Hiroshima K, Tada Y, Suzuki N, Tagawa M. Gene medicine for cancer treatment: commercially available medicine and accumulated clinical data in China. DRUG DESIGN DEVELOPMENT AND THERAPY 2009; 2:115-22. [PMID: 19920899 PMCID: PMC2761194 DOI: 10.2147/dddt.s3535] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Loss of p53 function compromises genetic homeostasis, which induces deregulated DNA replication, damages DNA, and subsequently results in increased resistance to anticancer agents. Pharmacological approaches using recombinant adenoviruses (Ad) have been developed to restore the p53 functions. Another approach for gene medicine is to modify Ad replication in a tumor-specific manner, which induces tumor cell death without damaging normal tissues in the vicinity. The Ad-derived gene medicines, Ad expressing the wild-type p53 gene and replication-competent Ad defective of the E1B-55kDa gene, have been tested for their clinical feasibility and became commercially available in China. These agents demonstrated their antitumor activities as a monotherapy and in combination with conventional chemotherapeutic agents. In this article, we summarize the outcomes of clinical trials in China, most of which have been published in domestic Chinese journals, and discuss potential directions of cancer gene therapy with these agents.
Collapse
Affiliation(s)
- Guangyu Ma
- Division of Pathology, Chiba Cancer Center Research Institute, 666-1 Nitona, Chuo-ku, Chiba, Japan
| | | | | | | | | | | |
Collapse
|
537
|
Natalicchio A, De Stefano F, Perrini S, Laviola L, Cignarelli A, Caccioppoli C, Quagliara A, Melchiorre M, Leonardini A, Conserva A, Giorgino F. Involvement of the p66Shc protein in glucose transport regulation in skeletal muscle myoblasts. Am J Physiol Endocrinol Metab 2009; 296:E228-37. [PMID: 18957618 DOI: 10.1152/ajpendo.90347.2008] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The p66(Shc) protein isoform regulates MAP kinase activity and the actin cytoskeleton turnover, which are both required for normal glucose transport responses. To investigate the role of p66(Shc) in glucose transport regulation in skeletal muscle cells, L6 myoblasts with antisense-mediated reduction (L6/p66(Shc)as) or adenovirus-mediated overexpression (L6/p66(Shc)adv) of the p66(Shc) protein were examined. L6/(Shc)as myoblasts showed constitutive activation of ERK-1/2 and disruption of the actin network, associated with an 11-fold increase in basal glucose transport. GLUT1 and GLUT3 transporter proteins were sevenfold and fourfold more abundant, respectively, and were localized throughout the cytoplasm. Conversely, in L6 myoblasts overexpressing p66(Shc), basal glucose uptake rates were reduced by 30% in parallel with a approximately 50% reduction in total GLUT1 and GLUT3 transporter levels. Inhibition of the increased ERK-1/2 activity with PD98059 in L6/(Shc)as cells had a minimal effect on increased GLUT1 and GLUT3 protein levels, but restored the actin cytoskeleton, and reduced the abnormally high basal glucose uptake by 70%. In conclusion, p66(Shc) appears to regulate the glucose transport system in skeletal muscle myoblasts by controlling, via MAP kinase, the integrity of the actin cytoskeleton and by modulating cellular expression of GLUT1 and GLUT3 transporter proteins via ERK-independent pathways.
Collapse
Affiliation(s)
- Annalisa Natalicchio
- Dept. of Emergency and Organ Transplantation, Section on Internal Medicine, Endocrinology and Metabolic Diseases, Univ. of Bari, Piazza Giulio Cesare, 11, I-70124 Bari, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
538
|
Cuezva JM, Ortega AD, Willers I, Sánchez-Cenizo L, Aldea M, Sánchez-Aragó M. The tumor suppressor function of mitochondria: translation into the clinics. Biochim Biophys Acta Mol Basis Dis 2009; 1792:1145-58. [PMID: 19419707 DOI: 10.1016/j.bbadis.2009.01.006] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Revised: 12/15/2008] [Accepted: 01/16/2009] [Indexed: 01/30/2023]
Abstract
Recently, the inevitable metabolic reprogramming experienced by cancer cells as a result of the onset of cellular proliferation has been added to the list of hallmarks of the cancer cell phenotype. Proliferation is bound to the synchronous fluctuation of cycles of an increased glycolysis concurrent with a restrained oxidative phosphorylation. Mitochondria are key players in the metabolic cycling experienced during proliferation because of their essential roles in the transduction of biological energy and in defining the life-death fate of the cell. These two activities are molecularly and functionally integrated and are both targets of commonly altered cancer genes. Moreover, energetic metabolism of the cancer cell also affords a target to develop new therapies because the activity of mitochondria has an unquestionable tumor suppressor function. In this review, we summarize most of these findings paying special attention to the opportunity that translation of energetic metabolism into the clinics could afford for the management of cancer patients. More specifically, we emphasize the role that mitochondrial beta-F1-ATPase has as a marker for the prognosis of different cancer patients as well as in predicting the tumor response to therapy.
Collapse
Affiliation(s)
- José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, CSIC-UAM and CIBER de Enfermedades Raras (CIBERER), Universidad Autónoma de Madrid, 28049 Madrid, Spain.
| | | | | | | | | | | |
Collapse
|
539
|
Olovnikov IA, Kravchenko JE, Chumakov PM. Homeostatic functions of the p53 tumor suppressor: regulation of energy metabolism and antioxidant defense. Semin Cancer Biol 2008; 19:32-41. [PMID: 19101635 DOI: 10.1016/j.semcancer.2008.11.005] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Accepted: 11/25/2008] [Indexed: 12/19/2022]
Abstract
The p53 tumor suppressor plays pivotal role in the organism by supervising strict compliance of individual cells to needs of the whole organisms. It has been widely accepted that p53 acts in response to stresses and abnormalities in cell physiology by mobilizing the repair processes or by removing the diseased cells through initiating the cell death programs. Recent studies, however, indicate that even under normal physiological conditions certain activities of p53 participate in homeostatic regulation of metabolic processes and that these activities are important for prevention of cancer. These novel functions of p53 help to align metabolic processes with the proliferation and energy status, to maintain optimal mode of glucose metabolism and to boost the energy efficient mitochondrial respiration in response to ATP deficiency. Additional activities of p53 in non-stressed cells tune up the antioxidant defense mechanisms reducing the probability of mutations caused by DNA oxidation under conditions of daily stresses. The deficiency in the p53-mediated regulation of glycolysis and mitochondrial respiration greatly accounts for the deficient respiration of the predominance of aerobic glycolysis in cancer cells (the Warburg effect), while the deficiency in the p53-modulated antioxidant defense mechanisms contributes to mutagenesis and additionally boosts the carcinogenesis process.
Collapse
|
540
|
Calreticulin regulates insulin receptor expression and its downstream PI3 Kinase/Akt signalling pathway. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2008; 1783:2344-51. [DOI: 10.1016/j.bbamcr.2008.08.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2008] [Revised: 08/27/2008] [Accepted: 08/28/2008] [Indexed: 01/09/2023]
|
541
|
Fan TWM, Kucia M, Jankowski K, Higashi RM, Ratajczak J, Ratajczak MZ, Lane AN. Rhabdomyosarcoma cells show an energy producing anabolic metabolic phenotype compared with primary myocytes. Mol Cancer 2008; 7:79. [PMID: 18939998 PMCID: PMC2577687 DOI: 10.1186/1476-4598-7-79] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2008] [Accepted: 10/21/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The functional status of a cell is expressed in its metabolic activity. We have applied stable isotope tracing methods to determine the differences in metabolic pathways in proliferating Rhabdomysarcoma cells (Rh30) and human primary myocytes in culture. Uniformly 13C-labeled glucose was used as a source molecule to follow the incorporation of 13C into more than 40 marker metabolites using NMR and GC-MS. These include metabolites that report on the activity of glycolysis, Krebs' cycle, pentose phosphate pathway and pyrimidine biosynthesis. RESULTS The Rh30 cells proliferated faster than the myocytes. Major differences in flux through glycolysis were evident from incorporation of label into secreted lactate, which accounts for a substantial fraction of the glucose carbon utilized by the cells. Krebs' cycle activity as determined by 13C isotopomer distributions in glutamate, aspartate, malate and pyrimidine rings was considerably higher in the cancer cells than in the primary myocytes. Large differences were also evident in de novo biosynthesis of riboses in the free nucleotide pools, as well as entry of glucose carbon into the pyrimidine rings in the free nucleotide pool. Specific labeling patterns in these metabolites show the increased importance of anaplerotic reactions in the cancer cells to maintain the high demand for anabolic and energy metabolism compared with the slower growing primary myocytes. Serum-stimulated Rh30 cells showed higher degrees of labeling than serum starved cells, but they retained their characteristic anabolic metabolism profile. The myocytes showed evidence of de novo synthesis of glycogen, which was absent in the Rh30 cells. CONCLUSION The specific 13C isotopomer patterns showed that the major difference between the transformed and the primary cells is the shift from energy and maintenance metabolism in the myocytes toward increased energy and anabolic metabolism for proliferation in the Rh30 cells. The data further show that the mitochondria remain functional in Krebs' cycle activity and respiratory electron transfer that enables continued accelerated glycolysis. This may be a common adaptive strategy in cancer cells.
Collapse
Affiliation(s)
- Teresa W M Fan
- Department of Chemistry, University of Louisville, KY, USA.
| | | | | | | | | | | | | |
Collapse
|
542
|
Expression of glucose transporter 1 is associated with loss of heterozygosity of chromosome 1p in oligodendroglial tumors WHO grade II. J Mol Histol 2008; 39:553-60. [DOI: 10.1007/s10735-008-9191-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2008] [Accepted: 08/05/2008] [Indexed: 12/26/2022]
|
543
|
Gillies RJ, Robey I, Gatenby RA. Causes and consequences of increased glucose metabolism of cancers. J Nucl Med 2008; 49 Suppl 2:24S-42S. [PMID: 18523064 DOI: 10.2967/jnumed.107.047258] [Citation(s) in RCA: 453] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
In this review we examine the mechanisms (causes) underlying the increased glucose consumption observed in tumors within a teleological context (consequences). In other words, we will ask not only "How do cancers have high glycolysis?" but also, "Why?" We believe that the insights gained from answering the latter question support the conclusion that elevated glucose consumption is a necessary component of carcinogenesis. Specifically we propose that glycolysis is elevated because it produces acid, which provides an evolutionary advantage to cancer cells vis-à-vis normal parenchyma into which they invade.
Collapse
|
544
|
Karnieli E, Armoni M. Transcriptional regulation of the insulin-responsive glucose transporter GLUT4 gene: from physiology to pathology. Am J Physiol Endocrinol Metab 2008; 295:E38-45. [PMID: 18492767 DOI: 10.1152/ajpendo.90306.2008] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The insulin-responsive glucose transporter 4 (GLUT4) plays a key role in glucose uptake and metabolism in insulin target tissues. Being a rate-limiting step in glucose metabolism, the expression and function of the GLUT4 isoform has been extensively studied and found to be tightly regulated at both mRNA and protein levels. Adaptation to states of enhanced metabolic demand is associated with increased glucose metabolism and GLUT4 gene expression, whereas states of insulin resistance such as type 2 diabetes mellitus (DM2), obesity, and aging are associated with impaired regulation of GLUT4 gene expression and function. The present review focuses on the interplay among hormonal, nutritional, and transcription factors in the regulation of GLUT4 transcription in health and sickness.
Collapse
Affiliation(s)
- Eddy Karnieli
- Institute of Endocrinology, Diabetes and Metabolism, 12 Haliah St., PO Box 9602, Rambam Medical Center, Haifa 31096, Israel.
| | | |
Collapse
|
545
|
Kawauchi K, Araki K, Tobiume K, Tanaka N. p53 regulates glucose metabolism through an IKK-NF-κB pathway and inhibits cell transformation. Nat Cell Biol 2008; 10:611-8. [DOI: 10.1038/ncb1724] [Citation(s) in RCA: 473] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2007] [Accepted: 03/07/2008] [Indexed: 12/14/2022]
|
546
|
Kim SJ, Lee HW, Kim DC, Rha SH, Hong SH, Jeong JS. Significance of GLUT1 expression in adenocarcinoma and adenoma of the ampulla of Vater. Pathol Int 2008; 58:233-8. [PMID: 18324916 DOI: 10.1111/j.1440-1827.2008.02216.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
The expression of glucose transporter protein 1 (GLUT1) in malignant tumors is increased due to the higher metabolic needs of the proliferating cell populations. Aberrant GLUT1 expression is exhibited in a wide spectrum of epithelial malignancies and their precursors, which occur with low frequency and intensity; aberrant GLUT1 expression does not occur in normal epithelial cells. The expression of GLUT1 in tumors of the ampulla of Vater was evaluated on immunohistochemistry, and the relationships of GLUT1 expression to histological parameters and p53 expression were analyzed. Twenty-one (58.3%) of 36 adenocarcinomas and three (17.6%) of 17 adenomas had GLUT1 immunoreactivity. None of the regenerating or normal epithelia had any immunoreactivity. No significant relationships were found between GLUT1 expression and histological parameters or p53 expression. It was found that histological subtypes originated from different epithelium were strongly related to different macroscopic types. In the ampulla of Vater, GLUT1 expression was associated with malignant change, and might be a useful marker of malignancy.
Collapse
Affiliation(s)
- Su Jin Kim
- Department of Pathology, Dong-A University College of Medicine, Busan, Korea
| | | | | | | | | | | |
Collapse
|
547
|
Ma W, Sung HJ, Park JY, Matoba S, Hwang PM. A pivotal role for p53: balancing aerobic respiration and glycolysis. J Bioenerg Biomembr 2008; 39:243-6. [PMID: 17551815 DOI: 10.1007/s10863-007-9083-0] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
The genetic basis of increased glycolytic activity observed in cancer cells is likely to be the result of complex interactions of multiple regulatory pathways. Here we review the recent evidence of a simple genetic mechanism by which tumor suppressor p53 regulates mitochondrial respiration with secondary changes in glycolysis that are reminiscent of the Warburg effect. The biological significance of this regulation of the two major pathways of energy generation by p53 remains to be seen.
Collapse
Affiliation(s)
- Wenzhe Ma
- Cardiology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | |
Collapse
|
548
|
Alvarado-Vásquez N, Zapata E, Alcázar-Leyva S, Massó F, Montaño LF. Reduced NO synthesis and eNOS mRNA expression in endothelial cells from newborns with a strong family history of type 2 diabetes. Diabetes Metab Res Rev 2007; 23:559-66. [PMID: 17385193 DOI: 10.1002/dmrr.743] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND A deficient synthesis of nitric oxide (NO) may play a role in the early endothelial dysfunction of healthy humans with a strong family history of type 2 diabetes (DM2). In this study, we evaluate the intracellular synthesis of NO and the expression of eNOS transcripts in human umbilical vein endothelial cells (HUVECs), exposed to high glucose concentrations, of healthy newborns with (experimental) and without (control) a strong family history of DM2. METHODS HUVECs were incubated in M-199 culture media (containing a 5 mmol/L physiological glucose concentration) or supraphysiological glucose concentrations (15 or 30 mmol/L), for 48 h. Flow cytometry, reactive of Griess and RT-PCR were used to determine intracellular NO synthesis, presence of NO metabolites, and expression of eNOS, GLUT1 or p53 transcripts. RESULTS NO synthesis in experimental HUVECs showed a progressive reduction in the presence of increasing glucose concentration (11% for 5 mmol to 8% for 30 mmol; p < 0.01), whereas control HUVECs showed an increase in NO synthesis (3% for 5 mmol to 31% for 30 mmol; p < 0.001). In experimental HUVECs, we found a diminished expression of eNOS and p53, and also an enhanced expression of GLUT1 mRNA transcripts. Control HUVECs showed an increase in eNOS, and no modifications in p53 or GLUT1 mRNA transcripts. CONCLUSIONS Our results show how HUVECs, isolated from healthy newborns with a strong family history of DM2, have an abnormal intracellular synthesis of NO and an impaired expression of eNOS, GLUT1 and p53 genes, all associated with NO synthesis.
Collapse
Affiliation(s)
- Noé Alvarado-Vásquez
- Departamento de Bioquímica, Instituto Nacional de Enfermedades Respiratorias, México.
| | | | | | | | | |
Collapse
|
549
|
Chung HK, Cheon GJ, Choi CW, Kim MJ, Lee SJ, Lim SM. Comparison of Cellular Metabolic Responses of18F-FDG According to the Effect ofβ-Irradiation in p53 Wild and Deleted Cell Lines. Cancer Biother Radiopharm 2007; 22:636-43. [DOI: 10.1089/cbr.2007.331] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Affiliation(s)
- Hye-Kyung Chung
- Laboratory of Nuclear Medicine Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Gi Jeong Cheon
- Laboratory of Nuclear Medicine Research, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
- Department of Nuclear Medicine, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Chang Woon Choi
- Department of Nuclear Medicine, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Min-Jung Kim
- Laboratory of Radiation Experimental Therapeutics, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Su-Jae Lee
- Laboratory of Radiation Experimental Therapeutics, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Sang Moo Lim
- Department of Nuclear Medicine, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| |
Collapse
|
550
|
Huc L, Tekpli X, Holme JA, Rissel M, Solhaug A, Gardyn C, Le Moigne G, Gorria M, Dimanche-Boitrel MT, Lagadic-Gossmann D. c-Jun NH2-terminal kinase-related Na+/H+ exchanger isoform 1 activation controls hexokinase II expression in benzo(a)pyrene-induced apoptosis. Cancer Res 2007; 67:1696-705. [PMID: 17308111 DOI: 10.1158/0008-5472.can-06-2327] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Regulation of the balance between survival, proliferation, and apoptosis on carcinogenic polycyclic aromatic hydrocarbon (PAH) exposure is still poorly understood and more particularly the role of physiologic variables, including intracellular pH (pH(i)). Although the involvement of the ubiquitous pH(i) regulator Na(+)/H(+) exchanger isoform 1 (NHE1) in tumorigenesis is well documented, less is known about its role and regulation during apoptosis. Our previous works have shown the primordial role of NHE1 in carcinogenic PAH-induced apoptosis. This alkalinizing transporter was activated by an early CYP1-dependent H(2)O(2) production, subsequently promoting mitochondrial dysfunction leading to apoptosis. The aim of this study was to further elucidate how NHE1 was activated by benzo(a)pyrene (BaP) and what the downstream events were in the context of apoptosis. Our results indicate that the mitogen-activated protein kinase kinase 4/c-Jun NH(2)-terminal kinase (MKK4/JNK) pathway was a link between BaP-induced H(2)O(2) production and NHE1 activation. This activation, in combination with BaP-induced phosphorylated p53, promoted mitochondrial superoxide anion production, supporting the existence of a common target for NHE1 and p53. Furthermore, we showed that the mitochondrial expression of glycolytic enzyme hexokinase II (HKII) was decreased following a combined action of NHE1 and p53 pathways, thereby enhancing the BaP-induced apoptosis. Taken together, our findings suggest that, on BaP exposure, MKK4/JNK targets NHE1 with consequences on HKII protein, which might thus be a key protein during carcinogenic PAH apoptosis.
Collapse
Affiliation(s)
- Laurence Huc
- Institut National de la Santé et de la Recherche Médicale U620, Université Rennes 1, IFR 140, 2 Avenue du Pr. Léon Bernard, Rennes, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|