351
|
Wiley CD, Campisi J. From Ancient Pathways to Aging Cells-Connecting Metabolism and Cellular Senescence. Cell Metab 2016; 23:1013-1021. [PMID: 27304503 PMCID: PMC4911819 DOI: 10.1016/j.cmet.2016.05.010] [Citation(s) in RCA: 296] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2016] [Revised: 05/19/2016] [Accepted: 05/25/2016] [Indexed: 02/07/2023]
Abstract
Cellular senescence is a complex stress response that permanently arrests the proliferation of cells at risk for oncogenic transformation. However, senescent cells can also drive phenotypes associated with aging. Although the senescence-associated growth arrest prevents the development of cancer, and the metabolism of cancer cells has been studied in depth, the metabolic causes and consequences of cellular senescence were largely unexplored until recently. New findings reveal key roles for several aspects of cellular metabolism in the establishment and control of senescent phenotypes. These discoveries have important implications for both cancer and aging. In this review, we highlight some of the recent links between metabolism and phenotypes that are commonly associated with senescent cells.
Collapse
Affiliation(s)
- Christopher D Wiley
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA.
| | - Judith Campisi
- Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, CA 94945, USA; Lawrence Berkeley National Laboratory, 1 Cyclotron Road, Berkeley, CA 94720, USA.
| |
Collapse
|
352
|
Fekry B, Jeffries KA, Esmaeilniakooshkghazi A, Ogretmen B, Krupenko SA, Krupenko NI. CerS6 Is a Novel Transcriptional Target of p53 Protein Activated by Non-genotoxic Stress. J Biol Chem 2016; 291:16586-96. [PMID: 27302066 PMCID: PMC4974374 DOI: 10.1074/jbc.m116.716902] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Indexed: 12/19/2022] Open
Abstract
Our previous study suggested that ceramide synthase 6 (CerS6), an enzyme in sphingolipid biosynthesis, is regulated by p53: CerS6 was elevated in several cell lines in response to transient expression of p53 or in response to folate stress, which is known to activate p53. It was not clear, however, whether CerS6 gene is a direct transcriptional target of p53 or whether this was an indirect effect through additional regulatory factors. In the present study, we have shown that the CerS6 promoter is activated by p53 in luciferase assays, whereas transcriptionally inactive R175H p53 mutant failed to induce the luciferase expression from this promoter. In vitro immunoprecipitation assays and gel shift analyses have further demonstrated that purified p53 binds within the CerS6 promoter sequence spanning 91 bp upstream and 60 bp downstream of the transcription start site. The Promo 3.0.2 online tool for the prediction of transcription factor binding sites indicated the presence of numerous putative non-canonical p53 binding motifs in the CerS6 promoter. Luciferase assays and gel shift analysis have identified a single motif upstream of the transcription start as a key p53 response element. Treatment of cells with Nutlin-3 or low concentrations of actinomycin D resulted in a strong elevation of CerS6 mRNA and protein, thus demonstrating that CerS6 is a component of the non-genotoxic p53-dependent cellular stress response. This study has shown that by direct transcriptional activation of CerS6, p53 can regulate specific ceramide biosynthesis, which contributes to the pro-apoptotic cellular response.
Collapse
Affiliation(s)
- Baharan Fekry
- From the Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina 28081
| | - Kristen A Jeffries
- From the Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina 28081
| | - Amin Esmaeilniakooshkghazi
- From the Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina 28081
| | - Besim Ogretmen
- the Department of Biochemistry and Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina 29425, and
| | - Sergey A Krupenko
- From the Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina 28081, the Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| | - Natalia I Krupenko
- From the Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina 28081, the Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599
| |
Collapse
|
353
|
Piątkiewicz P, Bernat-Karpińska M, Miłek T, Rabijewski M, Rosiak E. NK cell count and glucotransporter 4 (GLUT4) expression in subjects with type 2 diabetes and colon cancer. Diabetol Metab Syndr 2016; 8:38. [PMID: 27303448 PMCID: PMC4906701 DOI: 10.1186/s13098-016-0152-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2015] [Accepted: 05/26/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) and colon cancer (CC) are numbered among the most common diseases in the world. The decreased activity of natural killer (NK) cells previously revealed in both mentioned pathological states may be correlated with impaired expression of GLUT4 as the major insulin-dependent glucose transporter in these cells. METHODS The aim of this study was to evaluate GLUT4 expression and NK cells number in subjects with T2D and/or CC in comparison with control group. We evaluated 78 individuals divided into four groups: (1) patients with CC and T2DM, (2) patients with CC, (3) patients with T2DM (4) healthy control. GLUT4 expression on the surface of NK cells was measured using flow cytometry and phenotyping of NK cell was performed by immunofluorescent method. RESULTS Subjects with diabetes had the highest GLUT4 expression (21.35 ± 7.2 %) in comparison with other groups (P < 0.01). The mean values of GLUT4 expression in group with CC and in patients with both T2D and CC were similar (1.4 ± 0.4 % vs 1.5 ± 1.0 %; respectively). These values were significantly lower than in control group (12.6 ± 2.9 %; P < 0.01). In patients with T2D and CC the number of NK cells (20.15 ± 6.6 %) was significantly higher than in other groups, i.e. in group with T2D (14.08 ± 5.7 %), in group with CC (9.21 ± 3.6 %) and in control group (9.48 ± 4.7 %), respectively (P < 0.01). CONCLUSIONS It seems that there is a need to pay more attention to the high incidence of colon cancer among patients with type 2 diabetes. Decreased GLUT4 expression observed on NK cells in patients with colon cancer may be responsible for dysfunction of these cells and the higher carcinogenic risk in type 2 diabetic subjects.
Collapse
Affiliation(s)
- Paweł Piątkiewicz
- />Department of Internal Diseases, Diabetology and Endocrinology, Warsaw Medical University, Ul. Kondratowicza 8, 03-242 Warsaw, Poland
| | - Małgorzata Bernat-Karpińska
- />Department of Internal Diseases, Diabetology and Endocrinology, Warsaw Medical University, Ul. Kondratowicza 8, 03-242 Warsaw, Poland
| | - Tomasz Miłek
- />Department of General and Vascular Surgery, Warsaw Medical University, Warsaw, Poland
| | - Michał Rabijewski
- />Department of Internal Diseases, Diabetology and Endocrinology, Warsaw Medical University, Ul. Kondratowicza 8, 03-242 Warsaw, Poland
| | - Elżbieta Rosiak
- />Department of Nuclear Medicine, Warsaw Medical University, Warsaw, Poland
| |
Collapse
|
354
|
Hjelmeland A, Zhang J. Metabolic, autophagic, and mitophagic activities in cancer initiation and progression. Biomed J 2016; 39:98-106. [PMID: 27372165 PMCID: PMC5514543 DOI: 10.1016/j.bj.2015.10.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 10/19/2015] [Indexed: 12/11/2022] Open
Abstract
Cancer is a complex disease marked by uncontrolled cell growth and invasion. These processes are driven by the accumulation of genetic and epigenetic alterations that promote cancer initiation and progression. Contributing to genome changes are the regulation of oxidative stress and reactive species-induced damage to molecules and organelles. Redox regulation, metabolic plasticity, autophagy, and mitophagy play important and interactive roles in cancer hallmarks including sustained proliferation, activated invasion, and replicative immortality. However, the impact of these processes can differ depending on the signaling pathways altered in cancer, tumor type, tumor stage, and/or the differentiation state. Here, we highlight some of the representative studies on the impact of oxidative and nitrosative activities, mitochondrial bioenergetics, metabolism, and autophagy and mitophagy in the context of tumorigenesis. We discuss the implications of these processes for cellular activities in cancer for anti-cancer-based therapeutics.
Collapse
Affiliation(s)
- Anita Hjelmeland
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA; Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jianhua Zhang
- Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Veterans Affairs, Birmingham VA Medical Center, Birmingham, AL, USA.
| |
Collapse
|
355
|
Pinweha P, Rattanapornsompong K, Charoensawan V, Jitrapakdee S. MicroRNAs and oncogenic transcriptional regulatory networks controlling metabolic reprogramming in cancers. Comput Struct Biotechnol J 2016; 14:223-33. [PMID: 27358718 PMCID: PMC4915959 DOI: 10.1016/j.csbj.2016.05.005] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Revised: 05/25/2016] [Accepted: 05/27/2016] [Indexed: 12/15/2022] Open
Abstract
Altered cellular metabolism is a fundamental adaptation of cancer during rapid proliferation as a result of growth factor overstimulation. We review different pathways involving metabolic alterations in cancers including aerobic glycolysis, pentose phosphate pathway, de novo fatty acid synthesis, and serine and glycine metabolism. Although oncoproteins, c-MYC, HIF1α and p53 are the major drivers of this metabolic reprogramming, post-transcriptional regulation by microRNAs (miR) also plays an important role in finely adjusting the requirement of the key metabolic enzymes underlying this metabolic reprogramming. We also combine the literature data on the miRNAs that potentially regulate 40 metabolic enzymes responsible for metabolic reprogramming in cancers, with additional miRs from computational prediction. Our analyses show that: (1) a metabolic enzyme is frequently regulated by multiple miRs, (2) confidence scores from prediction algorithms might be useful to help narrow down functional miR-mRNA interaction, which might be worth further experimental validation. By combining known and predicted interactions of oncogenic transcription factors (TFs) (c-MYC, HIF1α and p53), sterol regulatory element binding protein 1 (SREBP1), 40 metabolic enzymes, and regulatory miRs we have established one of the first reference maps for miRs and oncogenic TFs that regulate metabolic reprogramming in cancers. The combined network shows that glycolytic enzymes are linked to miRs via p53, c-MYC, HIF1α, whereas the genes in serine, glycine and one carbon metabolism are regulated via the c-MYC, as well as other regulatory organization that cannot be observed by investigating individual miRs, TFs, and target genes.
Collapse
Key Words
- 2-HG, 2-hydroxyglutarate
- ACC, acetyl-CoA carboxylase
- ACL, ATP-citrate lyase
- BRCA1, breast cancer type 1 susceptibility protein
- Cancer
- FAS, fatty acid synthase
- FH, fumarate hydratase
- G6PD, glucose-6-phosphate dehydrogenase
- GDH, glutamate dehydrogenase
- GLS, glutaminase
- GLUT, glucose transporter
- HIF1α, hypoxia inducible factor 1α
- HK, hexokinase
- IDH, isocitrate dehydrogenase
- MCT, monocarboxylic acid transporter
- ME, malic enzyme
- Metabolism
- MicroRNA
- Oncogene
- PC, pyruvate carboxylase
- PDH, pyruvate dehydrogenase
- PDK, pyruvate dehydrogenase kinase
- PEP, phosphoenolpyruvate
- PEPCK, phosphoenolpyruvate carboxykinase
- PFK, phosphofructokinase
- PGK, phosphoglycerate kinase (PGK)
- PHGDH, phosphoglycerate dehydrogenase
- PKM, muscle-pyruvate kinase
- PPP, pentose phosphate pathway
- PSAT, phosphoserine aminotransferase
- PSPH, phosphoserine phosphatase
- SDH, succinate dehydrogenase
- SHMT, serine hydroxymethyl transferase
- SREBP1, sterol regulatory element binding protein 1
- TCA, tricarboxylic acid
- TFs, transcription factors
- Transcriptional regulation network
- c-MYC, V-myc avian myelocytomatosis viral oncogene homolog
- miR/miRNA, LDH, lactate dehydrogenase micro RNA
- p53, tumor protein p53
Collapse
Affiliation(s)
- Pannapa Pinweha
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | | | - Varodom Charoensawan
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
- Integrative Computational BioScience (ICBS) Center, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Sarawut Jitrapakdee
- Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| |
Collapse
|
356
|
Abstract
Deregulated cellular energetics was one of the cancer hallmarks. Several underlying mechanisms of deregulated cellular energetics are associated with mitochondrial dysfunction caused by mitochondrial DNA mutations, mitochondrial enzyme defects, or altered oncogenes/tumor suppressors. In this review, we summarize the current understanding about the role of mitochondrial dysfunction in cancer progression. Point mutations and copy number changes are the two most common mitochondrial DNA alterations in cancers, and mitochondrial dysfunction induced by chemical depletion of mitochondrial DNA or impairment of mitochondrial respiratory chain in cancer cells promotes cancer progression to a chemoresistance or invasive phenotype. Moreover, defects in mitochondrial enzymes, such as succinate dehydrogenase, fumarate hydratase, and isocitrate dehydrogenase, are associated with both familial and sporadic forms of cancer. Deregulated mitochondrial deacetylase sirtuin 3 might modulate cancer progression by regulating cellular metabolism and oxidative stress. These mitochondrial defects during oncogenesis and tumor progression activate cytosolic signaling pathways that ultimately alter nuclear gene expression, a process called retrograde signaling. Changes in the intracellular level of reactive oxygen species, Ca(2+), or oncometabolites are important in the mitochondrial retrograde signaling for neoplastic transformation and cancer progression. In addition, altered oncogenes/tumor suppressors including hypoxia-inducible factor 1 and tumor suppressor p53 regulate mitochondrial respiration and cellular metabolism by modulating the expression of their target genes. We thus suggest that mitochondrial dysfunction plays a critical role in cancer progression and that targeting mitochondrial alterations and mitochondrial retrograde signaling might be a promising strategy for the development of selective anticancer therapy.
Collapse
Affiliation(s)
- Chia-Chi Hsu
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| | - Ling-Ming Tseng
- Department of Surgery, Taipei Veterans General Hospital, Taipei 112, Taiwan Department of Surgery, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan Taipei-Veterans General Hospital Comprehensive Breast Health Center, Taipei 112, Taiwan
| | - Hsin-Chen Lee
- Department and Institute of Pharmacology, School of Medicine, National Yang-Ming University, Taipei 112, Taiwan
| |
Collapse
|
357
|
Abstract
The fundamental biological importance of the Tp53 gene family is highlighted by its evolutionary conservation for more than one billion years dating back to the earliest multicellular organisms. The TP53 protein provides essential functions in the cellular response to diverse stresses and safeguards maintenance of genomic integrity, and this is manifest in its critical role in tumor suppression. The importance of Tp53 in tumor prevention is exemplified in human cancer where it is the most frequently detected genetic alteration. This is confirmed in animal models, in which a defective Tp53 gene leads inexorably to cancer development, whereas reinstatement of TP53 function results in regression of established tumors that had been initiated by loss of TP53. Remarkably, despite extensive investigation, the specific mechanisms by which TP53 acts as a tumor suppressor are yet to be fully defined. We review the history and current standing of efforts to understand these mechanisms and how they complement each other in tumor suppression.
Collapse
Affiliation(s)
- Brandon J Aubrey
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3050, Australia Department of Clinical Haematology and Bone Marrow Transplant Service, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3050, Australia
| | - Gemma L Kelly
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia Department of Medical Biology, University of Melbourne, Parkville, Victoria 3050, Australia
| |
Collapse
|
358
|
Lim DC, Brady DC, Soans R, Kim EY, Valverde L, Keenan BT, Guo X, Kim WY, Park MJ, Galante R, Shackleford JA, Pack AI. Different cyclical intermittent hypoxia severities have different effects on hippocampal microvasculature. J Appl Physiol (1985) 2016; 121:78-88. [PMID: 27125850 DOI: 10.1152/japplphysiol.01040.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 04/27/2016] [Indexed: 01/12/2023] Open
Abstract
Recent studies have shown an association between obstructive sleep apnea (OSA) and cognitive impairment. This study was done to investigate whether varied levels of cyclical intermittent hypoxia (CIH) differentially affect the microvasculature in the hippocampus, operating as a mechanistic link between OSA and cognitive impairment. We exposed C57BL/6 mice to sham [continuous air, arterial O2 saturation (SaO2 ) 97%], severe CIH to inspired O2 fraction (FiO2 ) = 0.10 (CIH10; SaO2 nadir of 61%), or very severe CIH to FiO2 = 0.05 (CIH5; SaO2 nadir of 37%) for 12 h/day for 2 wk. We quantified capillary length using neurostereology techniques in the dorsal hippocampus and utilized quantitative PCR methods to measure changes in sets of genes related to angiogenesis and to metabolism. Next, we employed immunohistochemistry semiquantification algorithms to quantitate GLUT1 protein on endothelial cells within hippocampal capillaries. Capillary length differed among CIH severity groups (P = 0.013) and demonstrated a linear relationship with CIH severity (P = 0.002). There was a strong association between CIH severity and changes in mRNA for VEGFA (P < 0.0001). Less strong, but nominally significant associations with CIH severity were also observed for ANGPT2 (PANOVA = 0.065, PTREND = 0.040), VEGFR2 (PANOVA = 0.032, PTREND = 0.429), and TIE-2 (PANOVA = 0.006, PTREND = 0.010). We found that the CIH5 group had increased GLUT1 protein relative to sham (P = 0.006) and CIH10 (P = 0.001). There was variation in GLUT1 protein along the microvasculature in different hippocampal subregions. An effect of CIH5 on GLUT1 mRNA was seen (PANOVA = 0.042, PTREND = 0.012). Thus CIH affects the microvasculature in the hippocampus, but consequences depend on CIH severity.
Collapse
Affiliation(s)
- Diane C Lim
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania;
| | - Daniel C Brady
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Rajath Soans
- Department of Electrical and Computer Engineering, Drexel University, Philadelphia, Pennsylvania
| | - Emily Y Kim
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Laise Valverde
- Superior School of Health Sciences, Brasilia, Brazil; and
| | - Brendan T Keenan
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Xiaofeng Guo
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | - Raymond Galante
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - James A Shackleford
- Department of Electrical and Computer Engineering, Drexel University, Philadelphia, Pennsylvania
| | - Allan I Pack
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
359
|
Schneider LS, Ulrich M, Lehr T, Menche D, Müller R, von Schwarzenberg K. MDM2 antagonist nutlin-3a sensitizes tumors to V-ATPase inhibition. Mol Oncol 2016; 10:1054-62. [PMID: 27157929 DOI: 10.1016/j.molonc.2016.04.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 04/19/2016] [Accepted: 04/19/2016] [Indexed: 01/08/2023] Open
Abstract
Treating cancer is one of the big challenges of this century and it has become evident that single chemotherapeutic treatment is rarely effective. As tumors often carry multiple mutations using combination therapy which addresses different targets seems therefore more beneficial. One of the most frequently mutated genes in tumors is the tumor suppressor p53. Significant work has been put in the development of p53 activators, which are now in clinical studies against diverse cancers. Recently, we could show that inhibition of V-ATPase, a multisubunit proton pump, by archazolid induces p53 protein levels in cancer cells. In this study, we provide evidence that the combination of archazolid with the p53 activator nutlin-3a is synergistically inducing cell death in different p53 wild type tumor cell lines. Mechanistically, this effect could presumably be attributed to reduction of glycolysis as TIGAR mRNA levels were increased and glucose uptake and Glut1 protein levels were reduced. In addition, combination treatment highly activated pro-apoptotic pathways including IGFBP3 and Bax inducing caspase-9 and PARP cleavage. Remarkably, combination of archazolid and nutlin-3a was more efficient in reducing tumor growth compared to single dose treatment in a U87MG mouse model in vivo. Hence, our findings suggest the combination of archazolid and nutlin-3a as a highly promising strategy for the treatment of p53 wild type tumors.
Collapse
Affiliation(s)
- Lina S Schneider
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University of Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Melanie Ulrich
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University of Munich, Butenandtstrasse 5-13, 81377 Munich, Germany
| | - Thorsten Lehr
- Clinical Pharmacy, Saarland University, Campus C2 2, 66123 Saarbrücken, Germany
| | - Dirk Menche
- Kekulé Institute of Organic Chemistry and Biochemistry, University of Bonn, Gerhard-Domagk-Str. 1, 53121 Bonn, Germany
| | - Rolf Müller
- Saarland University, Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research and Department of Pharmaceutical Biotechnology, PO 151150, 66042 Saarbrücken, Germany
| | - Karin von Schwarzenberg
- Department of Pharmacy, Pharmaceutical Biology, Ludwig-Maximilians-University of Munich, Butenandtstrasse 5-13, 81377 Munich, Germany.
| |
Collapse
|
360
|
Kaplon J, van Dam L, Peeper D. Two-way communication between the metabolic and cell cycle machineries: the molecular basis. Cell Cycle 2016; 14:2022-32. [PMID: 26038996 DOI: 10.1080/15384101.2015.1044172] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The relationship between cellular metabolism and the cell cycle machinery is by no means unidirectional. The ability of a cell to enter the cell cycle critically depends on the availability of metabolites. Conversely, the cell cycle machinery commits to regulating metabolic networks in order to support cell survival and proliferation. In this review, we will give an account of how the cell cycle machinery and metabolism are interconnected. Acquiring information on how communication takes place among metabolic signaling networks and the cell cycle controllers is crucial to increase our understanding of the deregulation thereof in disease, including cancer.
Collapse
Affiliation(s)
- Joanna Kaplon
- a Division of Molecular Oncology; The Netherlands Cancer Institute ; Amsterdam ; The Netherlands
| | | | | |
Collapse
|
361
|
Identification of novel GLUT inhibitors. Bioorg Med Chem Lett 2016; 26:1732-7. [DOI: 10.1016/j.bmcl.2016.02.050] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 02/15/2016] [Accepted: 02/18/2016] [Indexed: 01/01/2023]
|
362
|
TP53 mutation, mitochondria and cancer. Curr Opin Genet Dev 2016; 38:16-22. [PMID: 27003724 DOI: 10.1016/j.gde.2016.02.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/09/2016] [Accepted: 02/22/2016] [Indexed: 12/20/2022]
Abstract
Under normal conditions, basal levels of wild-type p53 promote mitochondrial function through multiple mechanisms. Remarkably, some missense mutations of p53, in contrast to the null state, can result in the retention of its metabolic activities. These effects are particularly prominent in the mitochondria and demonstrate a functional role for mutant p53 in cancer metabolism. This review summarizes accumulating data on the mechanisms by which p53 missense mutations can regulate mitochondrial metabolism and promote the viability and survival of both normal and cancer cells, thus acting as a double edged sword for the host. Greater understanding of these mechanisms may provide insights for developing new treatment or preventive strategies against cancer.
Collapse
|
363
|
Burkart AM, Tan K, Warren L, Iovino S, Hughes KJ, Kahn CR, Patti ME. Insulin Resistance in Human iPS Cells Reduces Mitochondrial Size and Function. Sci Rep 2016; 6:22788. [PMID: 26948272 PMCID: PMC4780029 DOI: 10.1038/srep22788] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 02/17/2016] [Indexed: 12/20/2022] Open
Abstract
Insulin resistance, a critical component of type 2 diabetes (T2D), precedes and predicts T2D onset. T2D is also associated with mitochondrial dysfunction. To define the cause-effect relationship between insulin resistance and mitochondrial dysfunction, we compared mitochondrial metabolism in induced pluripotent stem cells (iPSC) from 5 healthy individuals and 4 patients with genetic insulin resistance due to insulin receptor mutations. Insulin-resistant iPSC had increased mitochondrial number and decreased mitochondrial size. Mitochondrial oxidative function was impaired, with decreased citrate synthase activity and spare respiratory capacity. Simultaneously, expression of multiple glycolytic enzymes was decreased, while lactate production increased 80%. These perturbations were accompanied by an increase in ADP/ATP ratio and 3-fold increase in AMPK activity, indicating energetic stress. Insulin-resistant iPSC also showed reduced catalase activity and increased susceptibility to oxidative stress. Thus, insulin resistance can lead to mitochondrial dysfunction with reduced mitochondrial size, oxidative activity, and energy production.
Collapse
Affiliation(s)
- Alison M Burkart
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Kelly Tan
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Laura Warren
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Salvatore Iovino
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Katelyn J Hughes
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center and Harvard Medical School, Boston, MA, 02215, USA
| | - C Ronald Kahn
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center and Harvard Medical School, Boston, MA, 02215, USA
| | - Mary-Elizabeth Patti
- Integrative Physiology and Metabolism Research Division, Joslin Diabetes Center and Harvard Medical School, Boston, MA, 02215, USA
| |
Collapse
|
364
|
Leung CON, Wong CCL, Fan DNY, Kai AKL, Tung EKK, Xu IMJ, Ng IOL, Lo RCL. PIM1 regulates glycolysis and promotes tumor progression in hepatocellular carcinoma. Oncotarget 2016; 6:10880-92. [PMID: 25834102 PMCID: PMC4484426 DOI: 10.18632/oncotarget.3534] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 02/18/2015] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is characteristically one of the most rapidly proliferating tumors which outgrows functional blood supply and results in regional oxygen deprivation. Overexpression of PIM1, a serine/threonine kinase, has been identified recently in human cancers. Knowledge on PIM1 in HCC is however, scarce. By immunohistochemical analysis on 56 human primary HCC samples, we observed overexpression of PIM1 in 39% of the cases. In two independent cohorts of paired primary and extra-hepatic metastatic HCC tissues, PIM1 expression was higher (p=0.002) in the extra-hepatic metastatic HCC tissues as compared with the corresponding primary HCCs. PIM1 was markedly up-regulated in multiple HCC cell lines in hypoxic condition (1% O2) versus normoxia (20% O2). Silencing of PIM1 suppressed HCC cell invasion in vitro as compared to non-target control, and decreased HCC cell proliferation in vitro and tumor growth and metastatic potential in vivo. Knockdown of PIM1 significantly reduced glucose uptake by HCC cells and was associated with decreased levels of p-AKT and key molecules in the glycolytic pathway. Taken together, PIM1 is up-regulated by hypoxia in HCC and promotes tumor growth and metastasis through facilitating cancer cell glycolysis. Targeting PIM1 may have potential role in the management of HCC.
Collapse
Affiliation(s)
| | - Carmen Chak-lui Wong
- Department of Pathology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| | | | - Alan Ka-lun Kai
- Department of Pathology, The University of Hong Kong, Hong Kong, China
| | | | - Iris Ming-jing Xu
- Department of Pathology, The University of Hong Kong, Hong Kong, China
| | - Irene Oi-lin Ng
- Department of Pathology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| | - Regina Cheuk-lam Lo
- Department of Pathology, The University of Hong Kong, Hong Kong, China.,State Key Laboratory for Liver Research, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
365
|
Abstract
Cancer is a disease characterized by uncontrolled growth. Metabolic demands to sustain rapid proliferation must be compelling since aerobic glycolysis is the first as well as the most commonly shared characteristic of cancer. During the last decade, the significance of metabolic reprogramming of cancer has been at the center of attention. Nonetheless, despite all the knowledge gained on cancer biology, the field is not able to reach agreement on the issue of mitochondria: Are damaged mitochondria the cause for aerobic glycolysis in cancer? Warburg proposed the damaged mitochondria theory over 80 years ago; the field has been testing the theory equally long. In this review, we will discuss alterations in metabolic fluxes of cancer cells, and provide an opinion on the damaged mitochondria theory.
Collapse
Affiliation(s)
- Aekyong Kim
- School of Pharmacy, Catholic University of Daegu, Gyeongbuk, Korea
| |
Collapse
|
366
|
Lee YM, Chang WC, Ma WL. Hypothesis: solid tumours behave as systemic metabolic dictators. J Cell Mol Med 2016; 20:1076-85. [PMID: 26843513 PMCID: PMC4882994 DOI: 10.1111/jcmm.12794] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Accepted: 12/22/2015] [Indexed: 02/06/2023] Open
Abstract
Current knowledge regarding mechanisms of carcinogenesis in human beings centres around the accumulation of genetic instability, amplified cellular signalling, disturbed cellular energy metabolism and microenvironmental regulation governed by complicated cell-cell interactions. In this article, we provide an alternative view of cancer biology. We propose that cancer behaves as a systemic dictator that interacts with tissues throughout the body to control their metabolism and eventually homeostasis. The mechanism of development of this endocrine organ-like tumour (EOLT) tissue might be the driving force for cancer progression. Here, we review the literature that led to the development of this hypothesis. The EOLT phenotype can be defined as a tumour that alters systemic homeostasis. The literature indicates that the EOLT phenotype is present throughout cancer progression. The feedback mechanism that governs the interaction between tumours and various organs is unknown. We believe that investigating the mechanism of EOLT development may advance the current knowledge of regulation within the tumour macroenvironment and consequently lead to new diagnostic methods and therapy.
Collapse
Affiliation(s)
- Yang-Ming Lee
- Sex Hormone Research Center, Graduate Institution of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Department of Endocrinology and Metabolism, Changhua Christian Hospital, Changhua, Taiwan
| | - Wei-Chun Chang
- Sex Hormone Research Center, Department of Gynecology and Obstetric, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Lung Ma
- Sex Hormone Research Center, Graduate Institution of Clinical Medical Science, China Medical University, Taichung, Taiwan.,Sex Hormone Research Center, Department of Gynecology and Obstetric, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
367
|
Smith H, Board M, Pellagatti A, Turley H, Boultwood J, Callaghan R. The Effects of Severe Hypoxia on Glycolytic Flux and Enzyme Activity in a Model of Solid Tumors. J Cell Biochem 2016; 117:1890-901. [PMID: 26755257 DOI: 10.1002/jcb.25488] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Accepted: 01/06/2016] [Indexed: 12/14/2022]
Abstract
Solid tumors contend with, and adapt to, a hostile micro-environment that includes limited availability of nutrient fuels and oxygen. The presence of hypoxia (O2 <5%) stabilizes the transcription factor Hif1 and results in numerous cellular adaptations including increased flux of glucose through glycolysis. Increasingly, more sophisticated analysis of tumor oxygenation has revealed large gradients of oxygen tension and significant regions under severe hypoxia (O2 ∼0.1%). The present investigation has demonstrated a significant increase in the glycolytic flux rate when tumor spheroids were exposed to 0.1% O2 . The severe hypoxia was associated with uniform pimonidazole adduct formation and elevated levels of Hif1α and c-Myc. This resulted in elevated expression of GLUT and MCT transporters, in addition to increased activity of PFK1 in comparison to that observed in normoxia. However, the protein expression and enzymatic capacity of HK2, G6PDH, PK, and LDH were all reduced by severe hypoxia. Clearly, the effects of exposure to severe hypoxia lead to a significantly abridged Hif1 response, yet one still able to elevate glycolytic flux and prevent loss of intermediates to anabolism. J. Cell. Biochem. 117: 1890-1901, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hannah Smith
- Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, The University of Oxford, Headington, UK
| | - Mary Board
- Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, The University of Oxford, Headington, UK
| | - Andrea Pellagatti
- Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, The University of Oxford, Headington, UK.,NIHR Biomedical Research Centre, Oxford, UK
| | - Helen Turley
- Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, The University of Oxford, Headington, UK
| | - Jacqueline Boultwood
- Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, The University of Oxford, Headington, UK.,NIHR Biomedical Research Centre, Oxford, UK
| | - Richard Callaghan
- Nuffield Department of Clinical Laboratory Sciences, John Radcliffe Hospital, The University of Oxford, Headington, UK.,Division of Biomedical Science and Biochemistry, Research School of Biology, The Australian National University Canberra, ACT 0200, Australia
| |
Collapse
|
368
|
Luthra PM, Lal N. Prospective of curcumin, a pleiotropic signalling molecule from Curcuma longa in the treatment of Glioblastoma. Eur J Med Chem 2016; 109:23-35. [DOI: 10.1016/j.ejmech.2015.11.049] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 11/25/2015] [Accepted: 11/29/2015] [Indexed: 12/13/2022]
|
369
|
Barron CC, Bilan PJ, Tsakiridis T, Tsiani E. Facilitative glucose transporters: Implications for cancer detection, prognosis and treatment. Metabolism 2016; 65:124-39. [PMID: 26773935 DOI: 10.1016/j.metabol.2015.10.007] [Citation(s) in RCA: 294] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Revised: 09/22/2015] [Accepted: 10/01/2015] [Indexed: 12/11/2022]
Abstract
It is long recognized that cancer cells display increased glucose uptake and metabolism. In a rate-limiting step for glucose metabolism, the glucose transporter (GLUT) proteins facilitate glucose uptake across the plasma membrane. Fourteen members of the GLUT protein family have been identified in humans. This review describes the major characteristics of each member of the GLUT family and highlights evidence of abnormal expression in tumors and cancer cells. The regulation of GLUTs by key proliferation and pro-survival pathways including the phosphatidylinositol 3-kinase (PI3K)-Akt, hypoxia-inducible factor-1 (HIF-1), Ras, c-Myc and p53 pathways is discussed. The clinical utility of GLUT expression in cancer has been recognized and evidence regarding the use of GLUTs as prognostic or predictive biomarkers is presented. GLUTs represent attractive targets for cancer therapy and this review summarizes recent studies in which GLUT1, GLUT3, GLUT5 and others are inhibited to decrease cancer growth.
Collapse
Affiliation(s)
- Carly C Barron
- Department of Health Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Philip J Bilan
- Program in Cell Biology, Peter Gilgan Centre for Research and Learning, The Hospital for Sick Children, Toronto, ON, M5G 0A4, Canada
| | - Theodoros Tsakiridis
- Department of Oncology, and Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, L8S 4L8, Canada
| | - Evangelia Tsiani
- Department of Health Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada.
| |
Collapse
|
370
|
Haupt S, Raghu D, Haupt Y. Mutant p53 Drives Cancer by Subverting Multiple Tumor Suppression Pathways. Front Oncol 2016; 6:12. [PMID: 26858938 PMCID: PMC4728204 DOI: 10.3389/fonc.2016.00012] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 01/12/2016] [Indexed: 11/13/2022] Open
Abstract
The tumor suppressor p53 normally acts as a brake to halt damaged cells from perpetrating their genetic errors into future generations. If p53 is disrupted by mutation, it may not only lose these corrective powers, but counterproductively acquire new capacities that drive cancer. A newly emerging manner in which mutant p53 executes its cancer promoting functions is by harnessing key proteins, which normally partner with its wild type, tumor-inhibiting counterpart. In association with the subverted activities of these protein partners, mutant p53 is empowered to act across multiple fundamental cellular pathways (regulating cell division and metabolism) and corrupt them to become cancer promoting.
Collapse
Affiliation(s)
- Sue Haupt
- Tumour Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Department of Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - Dinesh Raghu
- Tumour Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Ygal Haupt
- Tumour Suppression Laboratory, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia; Department of Pathology, The University of Melbourne, Parkville, VIC, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
371
|
Li Z, Zhang H. Reprogramming of glucose, fatty acid and amino acid metabolism for cancer progression. Cell Mol Life Sci 2016; 73:377-92. [PMID: 26499846 PMCID: PMC11108301 DOI: 10.1007/s00018-015-2070-4] [Citation(s) in RCA: 520] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 10/08/2015] [Accepted: 10/13/2015] [Indexed: 02/08/2023]
Abstract
Metabolic reprogramming is widely observed during cancer development to confer cancer cells the ability to survive and proliferate, even under the stressed, such as nutrient-limiting, conditions. It is famously known that cancer cells favor the "Warburg effect", i.e., the enhanced glycolysis or aerobic glycolysis, even when the ambient oxygen supply is sufficient. In addition, deregulated anabolism/catabolism of fatty acids and amino acids, especially glutamine, serine and glycine, have been identified to function as metabolic regulators in supporting cancer cell growth. Furthermore, extensive crosstalks are being revealed between the deregulated metabolic network and cancer cell signaling. These exciting advancements have inspired new strategies for treating various malignancies by targeting cancer metabolism. Here we review recent findings related to the regulation of glucose, fatty acid and amino acid metabolism, their crosstalk, and relevant cancer therapy strategy.
Collapse
Affiliation(s)
- Zhaoyong Li
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, 230027, China.
| | - Huafeng Zhang
- Hefei National Laboratory for Physical Sciences at Microscale, The CAS Key Laboratory of Innate Immunity and Chronic Disease, Innovation Center for Cell Signaling Network, School of Life Sciences and Medical Center, University of Science and Technology of China, Hefei, 230027, China.
| |
Collapse
|
372
|
Du W, Amarachintha S, Wilson AF, Pang Q. SCO2 Mediates Oxidative Stress-Induced Glycolysis to Oxidative Phosphorylation Switch in Hematopoietic Stem Cells. Stem Cells 2015; 34:960-71. [PMID: 26676373 DOI: 10.1002/stem.2260] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 10/30/2015] [Accepted: 11/20/2015] [Indexed: 11/10/2022]
Abstract
Fanconi anemia (FA) is an inherited bone marrow (BM) failure syndrome, presumably resulting from defects in hematopoietic stem cells (HSCs). Normal HSCs depend more on glycolysis than on oxidative phosphorylation (OXPHOS) for energy production. Here, we show that FA HSCs are more sensitive to the respiration inhibitor NaN3 treatment than to glycolytic inhibitor 2-deoxy-d-glucose (2-DG), indicating more dependence on OXPHOS. FA HSCs undergo glycolysis-to-OXPHOS switch in response to oxidative stress through a p53-dependent mechanism. Metabolic stresses induce upregulation of p53 metabolic targets in FA HSCs. Inactivation of p53 in FA HSCs prevents glycolysis-to-OXPHOS switch. Furthermore, p53-deficient FA HSCs are more sensitive to 2-DG-mediated metabolic stress. Finally, oxidative stress-induced glycolysis-to-OXPHOS switch is mediated by synthesis of cytochrome c oxidase 2 (SCO2). These findings demonstrate p53-mediated OXPHOS function as a compensatory alteration in FA HSCs to ensure a functional but mildly impaired energy metabolism and suggest a cautious approach to manipulating p53 signaling in FA.
Collapse
Affiliation(s)
- Wei Du
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Divisions of Radiation Health, College of Pharmacy, UAMS, Little Rock, Arkansas, USA
| | - Surya Amarachintha
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Andrew F Wilson
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Qishen Pang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
373
|
Phelan JJ, Feighery R, Eldin OS, Meachair SÓ, Cannon A, Byrne R, MacCarthy F, O'Toole D, Reynolds JV, O'Sullivan J. Examining the connectivity between different cellular processes in the Barrett tissue microenvironment. Cancer Lett 2015; 371:334-46. [PMID: 26688097 DOI: 10.1016/j.canlet.2015.11.041] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 11/27/2015] [Accepted: 11/30/2015] [Indexed: 12/20/2022]
Abstract
In Barrett associated tumorigenesis, oxidative phosphorylation and glycolysis are reprogrammed early in the disease sequence and act mutually to promote disease progression. However, the link between energy metabolism and its connection with other central cellular processes within the Barrett microenvironment is unknown. The aim of this study was to examine the relationship between metabolism (ATP5B/GAPDH), hypoxia (HIF1α), inflammation (IL1β/SERPINA3), p53 and obesity status using in-vivo and ex-vivo models of Barrett oesophagus. At the protein level, ATP5B (r = 0.71, P < 0.0001) and p53 (r = 0.455, P = 0.015) were found to be strongly associated with hypoxia. In addition, levels of ATP5B (r = 0.53, P = 0.0031) and GAPDH (r = -0.39, P = 0.0357) were positively associated with p53 expression. Moreover, we demonstrate that ATP5B (r = 0.8, P < 0.0001) and GAPDH (r = 0.43, P = 0.022) were positively associated with IL1β expression. Interestingly, obesity was negatively associated with oxidative phosphorylation (r = -0.6016, P = 0.0177) but positively associated with glycolysis (r = 0.743, P = 0.0015). Comparable correlations were exhibited in the ex-vivo explant tissue between metabolism, p53, hypoxia, inflammation and angiogenesis (P < 0.05). We have shown that metabolism is closely linked with many cellular processes in the Barrett tissue microenvironment.
Collapse
Affiliation(s)
- J J Phelan
- Department of Surgery, Institute of Molecular Medicine, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - R Feighery
- Department of Surgery, Institute of Molecular Medicine, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - O S Eldin
- Department of Histopathology, St. James's Hospital, Dublin, Ireland
| | - S Ó Meachair
- Centre for Health Decision Science (CHeDS), School of Computer Science and Statistics, Trinity College Dublin, Dublin, Ireland
| | - A Cannon
- Department of Surgery, Institute of Molecular Medicine, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - R Byrne
- Department of Surgery, Institute of Molecular Medicine, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - F MacCarthy
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - D O'Toole
- Department of Clinical Medicine, Institute of Molecular Medicine, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - J V Reynolds
- Department of Surgery, Institute of Molecular Medicine, Trinity College Dublin, St. James's Hospital, Dublin, Ireland
| | - J O'Sullivan
- Department of Surgery, Institute of Molecular Medicine, Trinity College Dublin, St. James's Hospital, Dublin, Ireland.
| |
Collapse
|
374
|
Wu S, Yin X, Fang X, Zheng J, Li L, Liu X, Chu L. c-MYC responds to glucose deprivation in a cell-type-dependent manner. Cell Death Discov 2015; 1:15057. [PMID: 27551483 PMCID: PMC4979460 DOI: 10.1038/cddiscovery.2015.57] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Revised: 10/14/2015] [Accepted: 10/21/2015] [Indexed: 12/17/2022] Open
Abstract
Metabolic reprogramming supports cancer cells’ demands for rapid proliferation and growth. Previous work shows that oncogenes, such as MYC, hypoxia-inducible factor 1 (HIF1), have a central role in driving metabolic reprogramming. A lot of metabolic enzymes, which are deregulated in most cancer cells, are the targets of these oncogenes. However, whether metabolic change affects these oncogenes is still unclear. Here we show that glucose deprivation (GD) affects c-MYC protein levels in a cell-type-dependent manner regardless of P53 mutation status. GD dephosphorylates and then decreases c-MYC protein stability through PI3K signaling pathway in HeLa cells, but not in MDA-MB-231 cells. Role of c-MYC in sensitivity of GD also varies with cell types. c-MYC-mediated glutamine metabolism partially improves the sensitivity of GD in MDA-MB-231 cells. Our results reveal that the heterogeneity of cancer cells in response to metabolic stress should be considered in metabolic therapy for cancer.
Collapse
Affiliation(s)
- S Wu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , 320 Yue-Yang Road, Shanghai 200031, China
| | - X Yin
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , 320 Yue-Yang Road, Shanghai 200031, China
| | - X Fang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , 320 Yue-Yang Road, Shanghai 200031, China
| | - J Zheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, China
| | - L Li
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu 221002, China
| | - X Liu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences , 320 Yue-Yang Road, Shanghai 200031, China
| | - L Chu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical College, Xuzhou, Jiangsu 221002, China
| |
Collapse
|
375
|
Wang X, Simpson ER, Brown KA. p53: Protection against Tumor Growth beyond Effects on Cell Cycle and Apoptosis. Cancer Res 2015; 75:5001-7. [PMID: 26573797 DOI: 10.1158/0008-5472.can-15-0563] [Citation(s) in RCA: 204] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 07/20/2015] [Indexed: 01/10/2023]
Abstract
The tumor suppressor p53 has established functions in cancer. Specifically, it has been shown to cause cell-cycle arrest and apoptosis in response to DNA damage. It is also one of the most commonly mutated or silenced genes in cancer and for this reason has been extensively studied. Recently, the role of p53 has been shown to go beyond its effects on cell cycle and apoptosis, with effects on metabolism emerging as a key contributor to cancer growth in situations where p53 is lost. Beyond this, the role of p53 in the tumor microenvironment is poorly understood. The publication by Wang and colleagues demonstrates for the first time that p53 is a key negative regulator of aromatase and, hence, estrogen production in the breast tumor microenvironment. It goes further by demonstrating that an important regulator of aromatase, the obesity-associated and tumor-derived factor prostaglandin E2, inhibits p53 in the breast adipose stroma. This review presents these findings in the context of established and emerging roles of p53 and discusses possible implications for the treatment of breast cancer.
Collapse
Affiliation(s)
- Xuyi Wang
- Metabolism and Cancer Laboratory, Centres for Cancer Research and Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria, Australia. Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Evan R Simpson
- Metabolism and Cancer Laboratory, Centres for Cancer Research and Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria, Australia. Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Kristy A Brown
- Metabolism and Cancer Laboratory, Centres for Cancer Research and Endocrinology and Metabolism, Hudson Institute of Medical Research, Clayton, Victoria, Australia. Department of Physiology, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
376
|
Mehdad A, Campos NA, Arruda SF, Siqueira EMDA. Iron Deprivation May Enhance Insulin Receptor and Glut4 Transcription in Skeletal Muscle of Adult Rats. J Nutr Health Aging 2015; 19:846-54. [PMID: 26412289 DOI: 10.1007/s12603-015-0541-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
OBJECTIVES Considering that phenotype related to iron overload associated with pathological conditions differs from that caused by dietary iron excess, our study set out to evaluate the impact of dietary iron restriction and dietary iron supplementation on oxidative stress and functional outcome in adult, healthy rats. METHODS adult rats were divided into the three groups and fed diets containing 10, 35 or 350 mg/kg iron (restricted-diet, control-diet and supplemented- diet groups, respectively) for 78 days. Hematological variables, fasting blood glucose, hepatic enzyme activity and C-reactive protein levels were analyzed. Iron and glycogen concentrations in liver and skeletal muscle were determined. The extent of tissue damage caused by either dietary iron restriction or iron supplementation was accessed by measuring malondialdehyde, carbonyl, NADPH oxidase, glutathione peroxidase, glutathione reductase and glutathione-s-transferase in various tissues. The mRNA expression levels of insulin receptor, glucose transporter 4 and p53 were also determined. RESULTS Fasting blood glucose values trended toward a decrease by dietary iron restriction, moreover, hepatic glycogen content decreased with concomitant increases in skeletal muscle. In addition, dietary iron restriction resulted in a twofold increase in mRNA expression of Insr and fourfold increase in Glut4 expression in skeletal muscle. Although the dietary iron restriction did not affect body iron status, it caused hepatic low oxidative damages. However, high liver NADPH oxidase activity and increased levels of protein oxidation in muscle were observed. Chronic feeding of high iron diet induces iron overload and resulted in elevated levels of stress markers in tissues. CONCLUSION Dietary iron deprivation may improve insulin receptor and glucose transporter transcription in muscle; however, our results show that dietary iron restriction can prevent and/or promote oxidative damage in a tissue-specific manner, emphasizing the importance of maintaining optimal iron intake.
Collapse
Affiliation(s)
- A Mehdad
- Azadeh Mehdad, Laboratory of Molecular biophysic, Instituto of Biology, Dep. Cellular Biology, University of Brasilia (UnB) Brazil,
| | | | | | | |
Collapse
|
377
|
Brito AF, Abrantes AM, Ribeiro M, Oliveira R, Casalta-Lopes J, Gonçalves AC, Sarmento-Ribeiro AB, Tralhão JG, Botelho MF. Fluorine-18 Fluorodeoxyglucose Uptake in Hepatocellular Carcinoma: Correlation with Glucose Transporters and p53 Expression. J Clin Exp Hepatol 2015; 5:183-9. [PMID: 26628835 PMCID: PMC4632095 DOI: 10.1016/j.jceh.2015.05.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 05/25/2015] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Hepatocellular Carcinoma (HCC) is one of most lethal cancers worldwide. The prognosis is very poor and therapeutic options are limited. The aim of this study was to determine the correlation of the [(18)F]FDG uptake profile of three HCC cell lines with p53 and glucose transporters (GLUTs) 1, 2, 3, 5 and 12 expression and with the glucose level present in the cell culture medium. METHODS Cell lines used are HepG2 (wp53), HuH7 (overexpress p53) and Hep3B2.1-7 (p53null). An immunocytochemical analysis was performed to evaluate p53 expression. Through uptake studies were analyzed the [(18)F]FDG uptake profiles of all cell lines under study. The expression of GLUTs were quantified by flow cytometry. The [(18)F]FDG uptake studies GLUTs expression analysis were performed on cells that grew in a high and low glucose medium in order to determine the effect of glucose concentration on GLUTs expression and on [(18)F]FDG uptake. RESULTS Immunocytochemical analysis confirmed the p53 expression profiles of all cell lines. It was found out that for all cell lines, [(18)F]FDG uptake is higher when cells grow in low glucose medium, however, the glucose level doesn't affect mostly the GLUTs expression. The Hep3B2.1-7 (p53null) is always the one that have higher [(18)F]FDG uptake. It was found that not always GLUT1 and GLUT3 are the most expressed by these cell lines. CONCLUSIONS Our results shown that the p53 expression influences [(18)F]FDG uptake. This suggests that [(18)F]FDG may be used in HCC diagnosis, and may even provide some information about the genetic profile of the tumor.
Collapse
Affiliation(s)
- Ana F. Brito
- Biophysics Unit, Faculty of Medicine of University of Coimbra, Coimbra, Portugal,Center of Investigation on Environmental, Genetics and Oncobiology (CIMAGO), Faculty of Medicine of University of Coimbra, Coimbra, Portugal,CNC.IBILI, University of Coimbra, Coimbra, Portugal,Address for correspondence: Ana F. Brito, Biophysics Unit, Faculty of Medicine of University of Coimbra, Pólo III – Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Celas. 3000-548 Coimbra, Portugal. Tel.: +351 239480200; fax: +351 239480217.
| | - Ana M. Abrantes
- Biophysics Unit, Faculty of Medicine of University of Coimbra, Coimbra, Portugal,Center of Investigation on Environmental, Genetics and Oncobiology (CIMAGO), Faculty of Medicine of University of Coimbra, Coimbra, Portugal
| | - Marina Ribeiro
- Biophysics Unit, Faculty of Medicine of University of Coimbra, Coimbra, Portugal,Faculty of Sciences and Technology of University of Coimbra, Coimbra, Portugal
| | - Rui Oliveira
- Biophysics Unit, Faculty of Medicine of University of Coimbra, Coimbra, Portugal,Center of Investigation on Environmental, Genetics and Oncobiology (CIMAGO), Faculty of Medicine of University of Coimbra, Coimbra, Portugal,Anatomical Pathology Department, CHUC, Coimbra, Portugal
| | - João Casalta-Lopes
- Biophysics Unit, Faculty of Medicine of University of Coimbra, Coimbra, Portugal,Center of Investigation on Environmental, Genetics and Oncobiology (CIMAGO), Faculty of Medicine of University of Coimbra, Coimbra, Portugal
| | - Ana C. Gonçalves
- Center of Investigation on Environmental, Genetics and Oncobiology (CIMAGO), Faculty of Medicine of University of Coimbra, Coimbra, Portugal,CNC.IBILI, University of Coimbra, Coimbra, Portugal,Applied Molecular Biology and Hematology Group, Faculty of Medicine of University of Coimbra, Coimbra, Portugal
| | - Ana B. Sarmento-Ribeiro
- Center of Investigation on Environmental, Genetics and Oncobiology (CIMAGO), Faculty of Medicine of University of Coimbra, Coimbra, Portugal,CNC.IBILI, University of Coimbra, Coimbra, Portugal,Applied Molecular Biology and Hematology Group, Faculty of Medicine of University of Coimbra, Coimbra, Portugal
| | - José G. Tralhão
- Biophysics Unit, Faculty of Medicine of University of Coimbra, Coimbra, Portugal,Center of Investigation on Environmental, Genetics and Oncobiology (CIMAGO), Faculty of Medicine of University of Coimbra, Coimbra, Portugal,Surgical Department A, CHUC, Coimbra, Portugal
| | - Maria F. Botelho
- Biophysics Unit, Faculty of Medicine of University of Coimbra, Coimbra, Portugal,Center of Investigation on Environmental, Genetics and Oncobiology (CIMAGO), Faculty of Medicine of University of Coimbra, Coimbra, Portugal,CNC.IBILI, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
378
|
Cohen R, Neuzillet C, Tijeras-Raballand A, Faivre S, de Gramont A, Raymond E. Targeting cancer cell metabolism in pancreatic adenocarcinoma. Oncotarget 2015; 6:16832-47. [PMID: 26164081 PMCID: PMC4627277 DOI: 10.18632/oncotarget.4160] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 05/29/2015] [Indexed: 12/13/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is expected to become the second leading cause of cancer death by 2030. Current therapeutic options are limited, warranting an urgent need to explore innovative treatment strategies. Due to specific microenvironment constraints including an extensive desmoplastic stroma reaction, PDAC faces major metabolic challenges, principally hypoxia and nutrient deprivation. Their connection with oncogenic alterations such as KRAS mutations has brought metabolic reprogramming to the forefront of PDAC therapeutic research. The Warburg effect, glutamine addiction, and autophagy stand as the most important adaptive metabolic mechanisms of cancer cells themselves, however metabolic reprogramming is also an important feature of the tumor microenvironment, having a major impact on epigenetic reprogramming and tumor cell interactions with its complex stroma. We present a comprehensive overview of the main metabolic adaptations contributing to PDAC development and progression. A review of current and future therapies targeting this range of metabolic pathways is provided.
Collapse
Affiliation(s)
- Romain Cohen
- INSERM U728, Beaujon University Hospital (AP-HP – PRES Paris 7 Diderot), Clichy La Garenne, France
| | - Cindy Neuzillet
- INSERM U728, Beaujon University Hospital (AP-HP – PRES Paris 7 Diderot), Clichy La Garenne, France
- Department of Medical Oncology, Henri Mondor University Hospital, Créteil, France
| | | | - Sandrine Faivre
- Medical Oncology, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Armand de Gramont
- New Drug Evaluation Laboratory, Centre of Experimental Therapeutics and Medical Oncology, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Eric Raymond
- Medical Oncology, Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| |
Collapse
|
379
|
Zhang C, Liu J, Wu R, Liang Y, Lin M, Liu J, Chan CS, Hu W, Feng Z. Tumor suppressor p53 negatively regulates glycolysis stimulated by hypoxia through its target RRAD. Oncotarget 2015; 5:5535-46. [PMID: 25114038 PMCID: PMC4170611 DOI: 10.18632/oncotarget.2137] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Cancer cells display enhanced glycolysis to meet their energetic and biosynthetic demands even under normal oxygen concentrations. Recent studies have revealed that tumor suppressor p53 represses glycolysis under normoxia as a novel mechanism for tumor suppression. As the common microenvironmental stress for tumors, hypoxia drives the metabolic switch from the oxidative phosphorylation to glycolysis, which is crucial for survival and proliferation of cancer cells under hypoxia. The p53's role and mechanism in regulating glycolysis under hypoxia is poorly understood. Here, we found that p53 represses hypoxia-stimulated glycolysis in cancer cells through RRAD, a newly-identified p53 target. RRAD expression is frequently decreased in lung cancer. Ectopic expression of RRAD greatly reduces glycolysis whereas knockdown of RRAD promotes glycolysis in lung cancer cells. Furthermore, RRAD represses glycolysis mainly through inhibition of GLUT1 translocation to the plasma membrane. Under hypoxic conditions, p53 induces RRAD, which in turn inhibits the translocation of GLUT1 and represses glycolysis in lung cancer cells. Blocking RRAD by siRNA greatly abolishes p53's function in repressing glycolysis under hypoxia. Taken together, our results revealed an important role and mechanism of p53 in antagonizing the stimulating effect of hypoxia on glycolysis, which contributes to p53's function in tumor suppression.
Collapse
Affiliation(s)
- Cen Zhang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, USA; These two authors contributed equally to this work
| | - Juan Liu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, USA; These two authors contributed equally to this work
| | - Rui Wu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, USA
| | - Yingjian Liang
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, USA
| | - Meihua Lin
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, USA
| | - Jia Liu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, USA
| | - Chang S Chan
- Department of Medicine, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, USA
| | - Wenwei Hu
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, USA
| | - Zhaohui Feng
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, Rutgers, State University of New Jersey, New Brunswick, USA
| |
Collapse
|
380
|
Iurlaro R, León-Annicchiarico CL, Muñoz-Pinedo C. Regulation of cancer metabolism by oncogenes and tumor suppressors. Methods Enzymol 2015; 542:59-80. [PMID: 24862260 DOI: 10.1016/b978-0-12-416618-9.00003-0] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cell proliferation requires the coordination of multiple signaling pathways as well as the provision of metabolic substrates. Nutrients are required to generate such building blocks and their form of utilization differs to significant extents between malignant tissues and their nontransformed counterparts. Thus, oncogenes and tumor suppressor genes regulate the proliferation of cancer cells also by controlling their metabolism. Here, we discuss the central anabolic functions of the signaling pathways emanating from mammalian target of rapamycin, MYC, and hypoxia-inducible factor-1. Moreover, we analyze how oncogenic proteins like phosphoinositide-3-kinase, AKT, and RAS, tumor suppressors such as phosphatase and tensin homolog, retinoblastoma, and p53, as well as other factors associated with the proliferation or survival of cancer cells, such as NF-κB, regulate cellular metabolism.
Collapse
Affiliation(s)
- Raffaella Iurlaro
- Cell Death Regulation Group, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | | | - Cristina Muñoz-Pinedo
- Cell Death Regulation Group, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain.
| |
Collapse
|
381
|
Molecular Connections between Cancer Cell Metabolism and the Tumor Microenvironment. Int J Mol Sci 2015; 16:11055-86. [PMID: 25988385 PMCID: PMC4463690 DOI: 10.3390/ijms160511055] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 04/30/2015] [Accepted: 05/08/2015] [Indexed: 12/13/2022] Open
Abstract
Cancer cells preferentially utilize glycolysis, instead of oxidative phosphorylation, for metabolism even in the presence of oxygen. This phenomenon of aerobic glycolysis, referred to as the “Warburg effect”, commonly exists in a variety of tumors. Recent studies further demonstrate that both genetic factors such as oncogenes and tumor suppressors and microenvironmental factors such as spatial hypoxia and acidosis can regulate the glycolytic metabolism of cancer cells. Reciprocally, altered cancer cell metabolism can modulate the tumor microenvironment which plays important roles in cancer cell somatic evolution, metastasis, and therapeutic response. In this article, we review the progression of current understandings on the molecular interaction between cancer cell metabolism and the tumor microenvironment. In addition, we discuss the implications of these interactions in cancer therapy and chemoprevention.
Collapse
|
382
|
Shi S, Ji S, Qin Y, Xu J, Zhang B, Xu W, Liu J, Long J, Liu C, Liu L, Ni Q, Yu X. Metabolic tumor burden is associated with major oncogenomic alterations and serum tumor markers in patients with resected pancreatic cancer. Cancer Lett 2015; 360:227-233. [PMID: 25687883 DOI: 10.1016/j.canlet.2015.02.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Revised: 02/08/2015] [Accepted: 02/09/2015] [Indexed: 12/27/2022]
Abstract
Pancreatic cancer is an aggressive and lethal disease with an overall 5-year survival rate of only 5%. Studies have demonstrated the ability of (18)F-fludrodeoxyglucose ((18)F-FDG) positron emission tomography/computed tomography (PET/CT) to measure the metabolic tumor burden in patients with various tumors, including pancreatic cancer. In a previous study, we investigated the predictive significance of the metabolic tumor burden in terms of the metabolic tumor volume (MTV) and total lesion glycolysis (TLG). In this study, we analyzed the correlation between metabolic tumor burden and the status of the KRAS, TP53, CDKN2A/p16, and SMAD4/DPC4 genes. Our results showed that the metabolic tumor burden was associated with oncogenomic alterations that reflected the abnormal expression of carbohydrate metabolic enzymes (GLUT1, ALDOA and FBP1). We also identified a linear correlation between serum tumor markers and the metabolic tumor burden. To estimate the metabolic tumor burden when (18)F-FDG PET/CT is not available, we used the linear regression models to establish equations for MTV and TLG using CA19-9 and CA125 as independent variables. Our results suggest that the metabolic tumor burden, as evaluated by (18)F-FDG PET/CT or estimated by serum tumor markers, may be suitable for monitoring treatment response and disease progression of pancreatic cancer. Further research is needed to better understand why pancreatic cancer patients with abnormal expressions of TP53, CDKN2A/p16, and SMAD4/DPC4 get high metabolic tumor burden.
Collapse
Affiliation(s)
- Si Shi
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College; and Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Shunrong Ji
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College; and Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Yi Qin
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College; and Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College; and Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Bo Zhang
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College; and Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Wenyan Xu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College; and Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jiang Liu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College; and Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jiang Long
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College; and Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Chen Liu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College; and Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Liang Liu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College; and Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Quanxing Ni
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College; and Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic and Hepatobiliary Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College; and Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| |
Collapse
|
383
|
Aird KM, Worth AJ, Snyder NW, Lee JV, Sivanand S, Liu Q, Blair IA, Wellen KE, Zhang R. ATM couples replication stress and metabolic reprogramming during cellular senescence. Cell Rep 2015; 11:893-901. [PMID: 25937285 DOI: 10.1016/j.celrep.2015.04.014] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 03/23/2015] [Accepted: 04/05/2015] [Indexed: 11/18/2022] Open
Abstract
Replication stress induced by nucleotide deficiency plays an important role in cancer initiation. Replication stress in primary cells typically activates the cellular senescence tumor-suppression mechanism. Senescence bypass correlates with development of cancer, a disease characterized by metabolic reprogramming. However, the role of metabolic reprogramming in the cellular response to replication stress has been little explored. Here, we report that ataxia telangiectasia mutated (ATM) plays a central role in regulating the cellular response to replication stress by shifting cellular metabolism. ATM inactivation bypasses senescence induced by replication stress triggered by nucleotide deficiency. This was due to restoration of deoxyribonucleotide triphosphate (dNTP) levels through both upregulation of the pentose phosphate pathway via increased glucose-6-phosphate dehydrogenase (G6PD) activity and enhanced glucose and glutamine consumption. These phenotypes were mediated by a coordinated suppression of p53 and upregulation of c-MYC downstream of ATM inactivation. Our data indicate that ATM status couples replication stress and metabolic reprogramming during senescence.
Collapse
Affiliation(s)
- Katherine M Aird
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Andrew J Worth
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nathaniel W Snyder
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joyce V Lee
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sharanya Sivanand
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Qin Liu
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Ian A Blair
- Department of Pharmacology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kathryn E Wellen
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rugang Zhang
- Gene Expression and Regulation Program, The Wistar Institute, Philadelphia, PA 19104, USA.
| |
Collapse
|
384
|
Park MS. Molecular Dynamics Simulations of the Human Glucose Transporter GLUT1. PLoS One 2015; 10:e0125361. [PMID: 25919356 PMCID: PMC4412407 DOI: 10.1371/journal.pone.0125361] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 03/12/2015] [Indexed: 01/31/2023] Open
Abstract
Glucose transporters (GLUTs) provide a pathway for glucose transport across membranes. Human GLUTs are implicated in devastating diseases such as heart disease, hyper- and hypo-glycemia, type 2 diabetes and cancer. The human GLUT1 has been recently crystalized in the inward-facing open conformation. However, there is no other structural information for other conformations. The X-ray structures of E. coli Xylose permease (XylE), a glucose transporter homolog, are available in multiple conformations with and without the substrates D-xylose and D-glucose. XylE has high sequence homology to human GLUT1 and key residues in the sugar-binding pocket are conserved. Here we construct a homology model for human GLUT1 based on the available XylE crystal structure in the partially occluded outward-facing conformation. A long unbiased all atom molecular dynamics simulation starting from the model can capture a new fully opened outward-facing conformation. Our investigation of molecular interactions at the interface between the transmembrane (TM) domains and the intracellular helices (ICH) domain in the outward- and inward-facing conformation supports that the ICH domain likely stabilizes the outward-facing conformation in GLUT1. Furthermore, inducing a conformational transition, our simulations manifest a global asymmetric rocker switch motion and detailed molecular interactions between the substrate and residues through the water-filled selective pore along a pathway from the extracellular to the intracellular side. The results presented here are consistent with previously published biochemical, mutagenesis and functional studies. Together, this study shed light on the structure and functional relationships of GLUT1 in multiple conformational states.
Collapse
Affiliation(s)
- Min-Sun Park
- Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, United States of America
- * E-mail:
| |
Collapse
|
385
|
Zhao K, Zhou Y, Qiao C, Ni T, Li Z, Wang X, Guo Q, Lu N, Wei L. Oroxylin A promotes PTEN-mediated negative regulation of MDM2 transcription via SIRT3-mediated deacetylation to stabilize p53 and inhibit glycolysis in wt-p53 cancer cells. J Hematol Oncol 2015; 8:41. [PMID: 25902914 PMCID: PMC4419472 DOI: 10.1186/s13045-015-0137-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Accepted: 04/11/2015] [Indexed: 02/06/2023] Open
Abstract
Introduction p53 plays important roles in regulating the metabolic reprogramming of cancer, such as aerobic glycolysis. Oroxylin A is a natural active flavonoid with strong anticancer effects both in vitro and in vivo. Methods wt-p53 (MCF-7 and HCT116 cells) cancer cells and p53-null H1299 cancer cells were used. The glucose uptake and lactate production were analyzed using Lactic Acid production Detection kit and the Amplex Red Glucose Assay Kit. Then, the protein levels and RNA levels of p53, mouse double minute 2 (MDM2), and p53-targeted glycolytic enzymes were quantified using Western blotting and quantitative polymerase chain reaction (PCR), respectively. Immunoprecipitation were performed to assess the binding between p53, MDM2, and sirtuin-3 (SIRT3), and the deacetylation of phosphatase and tensin homolog (PTEN). Reporter assays were performed to assess the transcriptional activity of PTEN. In vivo, effects of oroxylin A was investigated in nude mice xenograft tumor-inoculated MCF-7 or HCT116 cells. Results Here, we analyzed the underlying mechanisms that oroxylin A regulated p53 level and glycolytic metabolism in wt-p53 cancer cells, and found that oroxylin A inhibited glycolysis through upregulating p53 level. Oroxylin A did not directly affect the transcription of wt-p53, but suppressed the MDM2-mediated degradation of p53 via downregulating MDM2 transcription in wt-p53 cancer cells. In further studies, we found that oroxylin A induced a reduction in MDM2 transcription by promoting the lipid phosphatase activity of phosphatase and tensin homolog, which was upregulated via sirtuin3-mediated deacetylation. In vivo, oroxylin A inhibited the tumor growth of nude mice-inoculated MCF-7 or HCT116 cells. The expression of MDM2 protein in tumor tissue was downregulated by oroxylin A as well. Conclusions These results provide a p53-independent mechanism of MDM2 transcription and reveal the potential of oroxylin A on glycolytic regulation in both wt-p53 and mut-p53 cancer cells. The studies have important implications for the investigation on anticancer effects of oroxylin A, and provide the academic basis for the clinical trial of oroxylin A in cancer patients. Electronic supplementary material The online version of this article (doi:10.1186/s13045-015-0137-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kai Zhao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, The People's Republic of China.
| | - Yuxin Zhou
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, The People's Republic of China.
| | - Chen Qiao
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, The People's Republic of China.
| | - Ting Ni
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, The People's Republic of China.
| | - Zhiyu Li
- Department of Medicinal Chemistry, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, The People's Republic of China.
| | - Xiaotang Wang
- Department of Chemistry and Biochemistry, Florida International University, Miami, FL, 33199, USA.
| | - Qinglong Guo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, The People's Republic of China.
| | - Na Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, The People's Republic of China.
| | - Libin Wei
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Carcinogenesis and Intervention, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing, 210009, The People's Republic of China.
| |
Collapse
|
386
|
Diabetes and its link with cancer: providing the fuel and spark to launch an aggressive growth regime. BIOMED RESEARCH INTERNATIONAL 2015; 2015:390863. [PMID: 25961014 PMCID: PMC4413255 DOI: 10.1155/2015/390863] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 09/27/2014] [Indexed: 01/21/2023]
Abstract
Diabetes is a disease involving metabolic derangements in multiple organs. While the spectrum of diabetic complications has been known for years, recent evidence suggests that diabetes could also contribute to the initiation and propagation of certain cancers. The mechanism(s) underlying this relationship are not completely resolved but likely involve changes in hormone and nutrient levels, as well as activation of inflammatory and stress-related pathways. Interestingly, some of the drugs used clinically to treat diabetes also appear to have antitumour effects, further highlighting the interaction between these two conditions. In this contribution we review recent literature on this emerging relationship and explore the potential mechanisms that may promote cancer in diabetic patients.
Collapse
|
387
|
Briehl MM. Oxygen in human health from life to death--An approach to teaching redox biology and signaling to graduate and medical students. Redox Biol 2015; 5:124-139. [PMID: 25912168 PMCID: PMC4412967 DOI: 10.1016/j.redox.2015.04.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 04/08/2015] [Indexed: 02/07/2023] Open
Abstract
In the absence of oxygen human life is measured in minutes. In the presence of oxygen, normal metabolism generates reactive species (ROS) that have the potential to cause cell injury contributing to human aging and disease. Between these extremes, organisms have developed means for sensing oxygen and ROS and regulating their cellular processes in response. Redox signaling contributes to the control of cell proliferation and death. Aberrant redox signaling underlies many human diseases. The attributes acquired by altered redox homeostasis in cancer cells illustrate this particularly well. This teaching review and the accompanying illustrations provide an introduction to redox biology and signaling aimed at instructors of graduate and medical students. The ability to sense oxygen and respond to oxidative stress is ancient. Chemical and kinetic properties of ROS are key to understanding redox signaling. Redox signaling participates in normal control of cell proliferation and death. Aberrant redox signaling contributes to the hallmarks of cancer. Novel redox-based chemotherapeutics are being developed.
Collapse
Affiliation(s)
- Margaret M Briehl
- Department of Pathology, University of Arizona, PO Box 24-5043, Tucson, AZ 85724-5043, USA.
| |
Collapse
|
388
|
Docanto MM, Ham S, Corbould A, Brown KA. Obesity-Associated Inflammatory Cytokines and Prostaglandin E2 Stimulate Glucose Transporter mRNA Expression and Glucose Uptake in Primary Human Adipose Stromal Cells. J Interferon Cytokine Res 2015; 35:600-5. [PMID: 25839190 DOI: 10.1089/jir.2014.0194] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Obesity is associated with chronic low-grade inflammation. This occurs largely as a result of the infiltration of immune cells within the obese adipose, which produce a number of inflammatory factors, including interleukin-6 (IL-6), IL-1β, tumor necrosis factor-α (TNFα), and prostaglandin E(2) (PGE(2)). These factors have previously been shown to affect insulin-mediated glucose uptake in differentiated adipocytes. However, the insulin-independent effect of inflammation on adipocyte precursors, the adipose stromal cells, has not been explored. This study therefore aimed to examine the effect of obesity-associated inflammatory factors on the expression of insulin-independent glucose transporters (GLUT1 and GLUT3) and on the uptake of glucose within adipose stromal cells. Primary human subcutaneous adipose stromal cells were isolated from abdominoplasty, and the effect of inflammatory cytokines (IL-6, IL-1β, and TNFα) and PGE(2) on GLUT mRNA expression and glucose transport was assessed using real-time polymerase chain reaction and radiolabeled deoxyglucose uptake assays, respectively. Results demonstrate that all four inflammatory mediators caused a dose-dependent increase in GLUT1 mRNA expression and glucose uptake. GLUT3 mRNA expression was also upregulated by IL-6 (0.5 ng/mL), TNFα (0.1 and 10 ng/mL), and PGE(2) (0.1 μM). Overall, these results demonstrate that obesity-associated inflammation increases insulin-independent glucose transporter expression and glucose uptake in undifferentiated adipose stromal cells.
Collapse
Affiliation(s)
- Maria M Docanto
- 1 Metabolism & Cancer Laboratory, Centre for Cancer Research, MIMR-PHI Institute of Medical Research , Victoria, Australia
| | - Seungmin Ham
- 1 Metabolism & Cancer Laboratory, Centre for Cancer Research, MIMR-PHI Institute of Medical Research , Victoria, Australia
| | - Anne Corbould
- 1 Metabolism & Cancer Laboratory, Centre for Cancer Research, MIMR-PHI Institute of Medical Research , Victoria, Australia .,2 Department of Physiology, Monash University , Clayton, Victoria, Australia
| | - Kristy A Brown
- 1 Metabolism & Cancer Laboratory, Centre for Cancer Research, MIMR-PHI Institute of Medical Research , Victoria, Australia .,2 Department of Physiology, Monash University , Clayton, Victoria, Australia .,3 Department of Molecular and Translational Sciences, Monash University , Clayton, Victoria, Australia
| |
Collapse
|
389
|
Mayers JR, Vander Heiden MG. Famine versus feast: understanding the metabolism of tumors in vivo. Trends Biochem Sci 2015; 40:130-40. [PMID: 25639751 PMCID: PMC4340757 DOI: 10.1016/j.tibs.2015.01.004] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 01/07/2015] [Accepted: 01/08/2015] [Indexed: 12/20/2022]
Abstract
To fuel unregulated proliferation, cancer cells alter metabolism to support macromolecule biosynthesis. Cell culture studies have revealed how different oncogenic mutations and nutrients impact metabolism. Glucose and glutamine are the primary fuels used in vitro; however, recent studies have suggested that utilization of other amino acids as well as lipids and protein can also be important to cancer cells. Early investigations of tumor metabolism are translating these findings to the biology of whole tumors and suggest that additional complexity exists beyond nutrient availability alone in vivo. Whole-body metabolism and tumor heterogeneity also influence the metabolism of tumor cells, and successful targeting of metabolism for cancer therapy will require an understanding of tumor metabolism in vivo.
Collapse
Affiliation(s)
- Jared R Mayers
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA
| | - Matthew G Vander Heiden
- Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology (MIT), Cambridge, MA, USA; Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard University, Cambridge, MA, USA.
| |
Collapse
|
390
|
Analysis of p53 transactivation domain mutants reveals Acad11 as a metabolic target important for p53 pro-survival function. Cell Rep 2015; 10:1096-109. [PMID: 25704813 DOI: 10.1016/j.celrep.2015.01.043] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 12/10/2014] [Accepted: 01/16/2015] [Indexed: 11/22/2022] Open
Abstract
The p53 tumor suppressor plays a key role in maintaining cellular integrity. In response to diverse stress signals, p53 can trigger apoptosis to eliminate damaged cells or cell-cycle arrest to enable cells to cope with stress and survive. However, the transcriptional networks underlying p53 pro-survival function are incompletely understood. Here, we show that in oncogenic-Ras-expressing cells, p53 promotes oxidative phosphorylation (OXPHOS) and cell survival upon glucose starvation. Analysis of p53 transcriptional activation domain mutants reveals that these responses depend on p53 transactivation function. Using gene expression profiling and ChIP-seq analysis, we identify several p53-inducible fatty acid metabolism-related genes. One such gene, Acad11, encoding a protein involved in fatty acid oxidation, is required for efficient OXPHOS and cell survival upon glucose starvation. This study provides new mechanistic insight into the pro-survival function of p53 and suggests that targeting this pathway may provide a strategy for therapeutic intervention based on metabolic perturbation.
Collapse
|
391
|
Sakamoto A, Hino S, Nagaoka K, Anan K, Takase R, Matsumori H, Ojima H, Kanai Y, Arita K, Nakao M. Lysine Demethylase LSD1 Coordinates Glycolytic and Mitochondrial Metabolism in Hepatocellular Carcinoma Cells. Cancer Res 2015; 75:1445-56. [PMID: 25649769 DOI: 10.1158/0008-5472.can-14-1560] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 12/22/2014] [Indexed: 11/16/2022]
Abstract
The hallmark of most cancer cells is the metabolic shift from mitochondrial to glycolytic metabolism for adapting to the surrounding environment. Although epigenetic modification is intimately linked to cancer, the molecular mechanism, by which epigenetic factors regulate cancer metabolism, is poorly understood. Here, we show that lysine-specific demethylase-1 (LSD1, KDM1A) has an essential role in maintaining the metabolic shift in human hepatocellular carcinoma cells. Inhibition of LSD1 reduced glucose uptake and glycolytic activity, with a concurrent activation of mitochondrial respiration. These metabolic changes coexisted with the inactivation of the hypoxia-inducible factor HIF1α, resulting in a decreased expression of GLUT1 and glycolytic enzymes. In contrast, during LSD1 inhibition, a set of mitochondrial metabolism genes was activated with the concomitant increase of methylated histone H3 at lysine 4 in the promoter regions. Consistently, both LSD1 and GLUT1 were significantly overexpressed in carcinoma tissues. These findings demonstrate the epigenetic plasticity of cancer cell metabolism, which involves an LSD1-mediated mechanism.
Collapse
Affiliation(s)
- Akihisa Sakamoto
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan Department of Neurosurgery, Faculty of Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Shinjiro Hino
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Katsuya Nagaoka
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Kotaro Anan
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Ryuta Takase
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Haruka Matsumori
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Hidenori Ojima
- Division of Molecular Pathology, National Cancer Center Research Institute, Tokyo, Japan
| | - Yae Kanai
- Division of Molecular Pathology, National Cancer Center Research Institute, Tokyo, Japan
| | - Kazunori Arita
- Department of Neurosurgery, Faculty of Medicine, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Mitsuyoshi Nakao
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan Core Research for Evolutional Science and Technology (CREST), Japan Science and Technology Agency, Tokyo, Japan.
| |
Collapse
|
392
|
Chang CF, Diers AR, Hogg N. Cancer cell metabolism and the modulating effects of nitric oxide. Free Radic Biol Med 2015; 79:324-36. [PMID: 25464273 PMCID: PMC5275750 DOI: 10.1016/j.freeradbiomed.2014.11.012] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Revised: 10/31/2014] [Accepted: 11/08/2014] [Indexed: 12/18/2022]
Abstract
Altered metabolic phenotype has been recognized as a hallmark of tumor cells for many years, but this aspect of the cancer phenotype has come into greater focus in recent years. NOS2 (inducible nitric oxide synthase of iNOS) has been implicated as a component in many aggressive tumor phenotypes, including melanoma, glioblastoma, and breast cancer. Nitric oxide has been well established as a modulator of cellular bioenergetics pathways, in many ways similar to the alteration of cellular metabolism observed in aggressive tumors. In this review we attempt to bring these concepts together with the general hypothesis that one function of NOS2 and NO in cancer is to modulate metabolic processes to facilitate increased tumor aggression. There are many mechanisms by which NO can modulate tumor metabolism, including direct inhibition of respiration, alterations in mitochondrial mass, oxidative inhibition of bioenergetic enzymes, and the stimulation of secondary signaling pathways. Here we review metabolic alterations in the context of cancer cells and discuss the role of NO as a potential mediator of these changes.
Collapse
Affiliation(s)
- Ching-Fang Chang
- Department of Biophysics and Redox Biology Program, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Anne R Diers
- Department of Biophysics and Redox Biology Program, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Neil Hogg
- Department of Biophysics and Redox Biology Program, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA.
| |
Collapse
|
393
|
Wang X, Docanto MM, Sasano H, Lo C, Simpson ER, Brown KA. Prostaglandin E2 inhibits p53 in human breast adipose stromal cells: a novel mechanism for the regulation of aromatase in obesity and breast cancer. Cancer Res 2015; 75:645-55. [PMID: 25634217 DOI: 10.1158/0008-5472.can-14-2164] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Obesity is a risk factor for postmenopausal breast cancer and the majority of these cancers are estrogen dependent. Aromatase converts androgens into estrogens and its increased expression in breast adipose stromal cells (ASC) is a major driver of estrogen receptor-positive breast cancer. In particular, obesity-associated and tumor-derived factors, such as prostaglandin E2 (PGE2), have been shown to drive the expression of aromatase by stimulating the activity of the proximal promoter II (PII). The tumor-suppressor p53 is a key regulator of cell-cycle arrest and apoptosis and is frequently mutated in breast cancer. Mutations in p53 are rare in tumor-associated ASCs. Therefore, it was hypothesized that p53 is regulated by PGE2 and involved in the PGE2-mediated regulation of aromatase. Results demonstrate that PGE2 causes a significant decrease in p53 transcript and nuclear protein expression, as well as phosphorylation at Ser15 in primary human breast ASCs. Stabilization of p53 with RITA leads to a significant decrease in the PGE2-stimulated aromatase mRNA expression and activity, and PII activity. Interaction of p53 with PII was demonstrated and this interaction is decreased in the presence of PGE2. Moreover, mutation of the identified p53 response element leads to an increase in the basal activity of the promoter. Immunofluorescence on clinical samples demonstrates that p53 is decreased in tumor-associated ASCs compared with ASCs from normal breast tissue, and that there is a positive association between perinuclear (inactive) p53 and aromatase expression in these cells. Furthermore, aromatase expression is increased in breast ASCs from Li-Fraumeni patients (germline TP53 mutations) compared with non-Li-Fraumeni breast tissue. Overall, our results demonstrate that p53 is a negative regulator of aromatase in the breast and its inhibition by PGE2 provides a novel mechanism for aromatase regulation in obesity and breast cancer.
Collapse
Affiliation(s)
- Xuyi Wang
- Metabolism and Cancer Laboratory, Centre for Cancer Research, MIMR-PHI Institute of Medical Research, Clayton, Victoria, Australia. Department of Physiology, Monash University, Clayton, Victoria, Australia
| | - Maria M Docanto
- Metabolism and Cancer Laboratory, Centre for Cancer Research, MIMR-PHI Institute of Medical Research, Clayton, Victoria, Australia
| | - Hironobu Sasano
- Department of Pathology, Tohoku University School of Medicine, Sendai, Japan
| | - Camden Lo
- Monash Micro Imaging, Monash University, Clayton, Victoria, Australia
| | - Evan R Simpson
- Metabolism and Cancer Laboratory, Centre for Cancer Research, MIMR-PHI Institute of Medical Research, Clayton, Victoria, Australia. Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Kristy A Brown
- Metabolism and Cancer Laboratory, Centre for Cancer Research, MIMR-PHI Institute of Medical Research, Clayton, Victoria, Australia. Department of Physiology, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
394
|
Chandra V, Hong KM. Effects of deranged metabolism on epigenetic changes in cancer. Arch Pharm Res 2015; 38:321-37. [PMID: 25628247 DOI: 10.1007/s12272-015-0561-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 01/09/2015] [Indexed: 12/17/2022]
Abstract
The concept of epigenetics is now providing the mechanisms by which cells transfer their new environmental-change-induced phenotypes to their daughter cells. However, how extracellular or cytoplasmic environmental cues are connected to the nuclear epigenome remains incompletely understood. Recently emerging evidence suggests that epigenetic changes are correlated with metabolic changes via chromatin remodeling. As many human complex diseases including cancer harbor both epigenetic changes and metabolic dysregulation, understanding the molecular processes linking them has huge implications for disease pathogenesis and therapeutic intervention. In this review, the impacts of metabolic changes on cancer epigenetics are discussed, along with the current knowledge on cancer metabolism and epigenetics.
Collapse
Affiliation(s)
- Vishal Chandra
- Cancer Cell and Molecular Biology Branch, Research Institute, National Cancer Center, 323 Ilsan-ro, Ilsandong-gu, Goyang, 410-769, Korea
| | | |
Collapse
|
395
|
Tambe Y, Hasebe M, Kim CJ, Yamamoto A, Inoue H. The drs tumor suppressor regulates glucose metabolism via lactate dehydrogenase-B. Mol Carcinog 2015; 55:52-63. [PMID: 25620379 DOI: 10.1002/mc.22258] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 10/03/2014] [Accepted: 11/03/2014] [Indexed: 02/04/2023]
Abstract
Previously, we showed that drs contributes to suppression of malignant tumor formation in drs-knockout (KO) mice. In this study, we demonstrate the regulation of glucose metabolism by drs using comparisons of drs-KO and wild-type (WT) mouse embryonic fibroblasts (MEFs). Extracellular acidification, lactate concentration, and glucose consumption in drs-KO cells were significantly greater than those in WT cells. Metabolomic analyses also confirmed enhanced glycolysis in drs-KO cells. Among glycolysis-regulating proteins, expression of lactate dehydrogenase (LDH)-B was upregulated at the post-transcriptional level in drs-KO cells and increased LDH-B expression, LDH activity, and acidification of culture medium in drs-KO cells were suppressed by retroviral rescue of drs, indicating that LDH-B plays a critical role for glycolysis regulation mediated by drs. In WT cells transformed by activated K-ras, expression of endogenous drs mRNA was markedly suppressed and LDH-B expression was increased. In human cancer cell lines with low drs expression, LDH-B expression was increased. Database analyses also showed the correlation between downregulation of drs and upregulation of LDH-B in human colorectal cancer and lung adenocarcinoma tissues. Furthermore, an LDH inhibitor suppressed anchorage-independent growth of human cancer cells and MEF cells transformed by activated K-ras. These results indicate that drs regulates glucose metabolism via LDH-B. Downregulating drs may contribute to the Warburg effect, which is closely associated with malignant progression of cancer cells.
Collapse
Affiliation(s)
- Yukihiro Tambe
- Division of Microbiology and Infectious Diseases, Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Masahiro Hasebe
- Division of Microbiology and Infectious Diseases, Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan.,Department of Cell Biology and Bioscience, Nagahama Institute of Bioscience and Technology, Nagahama, Shiga, Japan
| | - Chul Jang Kim
- Department of Urology, Kohka Public Hospital, Kohka, Shiga, Japan
| | - Akitsugu Yamamoto
- Department of Cell Biology and Bioscience, Nagahama Institute of Bioscience and Technology, Nagahama, Shiga, Japan
| | - Hirokazu Inoue
- Division of Microbiology and Infectious Diseases, Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| |
Collapse
|
396
|
Kee HJ, Cheong JH. Tumor bioenergetics: an emerging avenue for cancer metabolism targeted therapy. BMB Rep 2014; 47:158-66. [PMID: 24499670 PMCID: PMC4163877 DOI: 10.5483/bmbrep.2014.47.3.273] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 12/28/2013] [Accepted: 01/28/2014] [Indexed: 12/28/2022] Open
Abstract
Cell proliferation is a delicately regulated process that couples growth signals and metabolic demands to produce daughter cells. Interestingly, the proliferation of tumor cells immensely depends on glycolysis, the Warburg effect, to ensure a sufficient amount of metabolic flux and bioenergetics for macromolecule synthesis and cell division. This unique metabolic derangement ould provide an opportunity for developing cancer therapeutic strategy, particularly when other diverse anti-cancer treatments have been proved ineffective in achieving durable response, largely due to the emergence of resistance. Recent advances in deeper understanding of cancer metabolism usher in new horizons of the next generation strategy for cancer therapy. Here, we discuss the focused review of cancer energy metabolism, and the therapeutic exploitation of glycolysis and OXPHOS as a novel anti-cancer strategy, with particular emphasis on the promise of this approach, among other cancer metabolism targeted therapies that reveal unexpected complexity and context-dependent metabolic adaptability, complicating the development of effective strategies. [BMB Reports 2014; 47(3): 158-166]
Collapse
Affiliation(s)
- Hyun Jung Kee
- Departments of Biomedical Science, Yonsei University College of Medicine, Seoul 120-752, Korea
| | - Jae-Ho Cheong
- Departments of Surgery and; Biochemistry & Molecular Biology, Yonsei University College of Medicine, Seoul 120-752, Korea
| |
Collapse
|
397
|
Abstract
Accumulation of DNA damage has been linked to the process of aging and to the onset of age-related diseases including diabetes. Studies on progeroid syndromes have suggested that the DNA damage response is involved in regulation of metabolic homeostasis. DNA damage could impair metabolic organ functions by causing cell death or senescence. DNA damage also could induce tissue inflammation that disturbs the homeostasis of systemic metabolism. Various roles of molecules related to DNA repair in cellular metabolism are being uncovered, and such molecules could also have an impact on systemic metabolism. This review explores mechanisms by which the DNA damage response could contribute to metabolic dysfunction.
Collapse
Affiliation(s)
- Ippei Shimizu
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; Department of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Yohko Yoshida
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; Department of Molecular Aging and Cell Biology, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Masayoshi Suda
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Tohru Minamino
- Department of Cardiovascular Biology and Medicine, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan; PRESTO, Japan Science and Technology Agency, 4-1-8 Honcho Kawaguchi, Saitama 332-0012, Japan.
| |
Collapse
|
398
|
Eleftheriadis T, Pissas G, Antoniadi G, Spanoulis A, Liakopoulos V, Stefanidis I. Indoleamine 2,3-dioxygenase increases p53 levels in alloreactive human T cells, and both indoleamine 2,3-dioxygenase and p53 suppress glucose uptake, glycolysis and proliferation. Int Immunol 2014; 26:673-684. [PMID: 25064493 DOI: 10.1093/intimm/dxu077] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Indoleamine 2,3-dioxygenase (IDO) suppresses adaptive immunity by inhibiting T-cell proliferation and altering glucose metabolism. The tumor suppressor p53 also alters these cellular processes with similar results. The effect of IDO on p53 and on glucose metabolism was evaluated in alloreactive T cells. Mixed-lymphocyte reactions (MLRs) were performed in the presence or not of the IDO inhibitor, 1-dl-methyl-tryptophan (1-MT) and/or the p53 inhibitor, pifithrin-α (PFT). Cell proliferation, glucose consumption and lactate production were assessed. 1-MT increased cell proliferation, glucose influx and lactate production, whereas PFT enhanced cell proliferation and glucose influx, leaving lactate production unaffected. In MLR-derived T cells, protein analysis revealed that IDO activated general control non-derepressible 2 kinase and induced p53, p-p53 (p53 phosphorylated at serine 15) and p21. In addition, both IDO and p53 decreased glucose transporter 1 and TP53-induced glycolysis and apoptosis regulator and increased synthesis of cytochrome c oxidase 2. IDO also reduced lactate dehydrogenase-A and glutaminase 2 levels, whereas p53 left them unaffected. Neither 1-MT nor PFT affected glucose-6-phosphate dehydrogenase. In conclusion, in alloreactive T cells, IDO increases p53 levels, and both IDO and p53 inhibit cell proliferation, glucose consumption and glycolysis. Lactate production and glutaminolysis are also suppressed by IDO, but not by p53.
Collapse
Affiliation(s)
- Theodoros Eleftheriadis
- Department of Nephrology, Medical School, University of Thessaly, Neo Ktirio, Mezourlo Hill, 41110 Larissa, Greece
| | - Georgios Pissas
- Department of Nephrology, Medical School, University of Thessaly, Neo Ktirio, Mezourlo Hill, 41110 Larissa, Greece
| | - Georgia Antoniadi
- Department of Nephrology, Medical School, University of Thessaly, Neo Ktirio, Mezourlo Hill, 41110 Larissa, Greece
| | - Aginor Spanoulis
- Department of Nephrology, Medical School, University of Thessaly, Neo Ktirio, Mezourlo Hill, 41110 Larissa, Greece
| | - Vassilios Liakopoulos
- Department of Nephrology, Medical School, University of Thessaly, Neo Ktirio, Mezourlo Hill, 41110 Larissa, Greece
| | - Ioannis Stefanidis
- Department of Nephrology, Medical School, University of Thessaly, Neo Ktirio, Mezourlo Hill, 41110 Larissa, Greece
| |
Collapse
|
399
|
Ou Y, Wang SJ, Jiang L, Zheng B, Gu W. p53 Protein-mediated regulation of phosphoglycerate dehydrogenase (PHGDH) is crucial for the apoptotic response upon serine starvation. J Biol Chem 2014; 290:457-66. [PMID: 25404730 DOI: 10.1074/jbc.m114.616359] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Although p53 is frequently mutated in human cancers, about 80% of human melanomas retain wild-type p53. Here we report that PHGDH, the key metabolic enzyme that catalyzes the rate-limiting step of the serine biosynthesis pathway, is a target of p53 in human melanoma cells. p53 suppresses PHGDH expression and inhibits de novo serine biosynthesis. Notably, upon serine starvation, p53-mediated cell death is enhanced dramatically in response to Nutlin-3 treatment. Moreover, PHGDH has been found recently to be amplified frequently in human melanomas. We found that PHGDH overexpression significantly suppresses the apoptotic response, whereas RNAi-mediated knockdown of endogenous PHGDH promotes apoptosis under the same treatment. These results demonstrate an important role of p53 in regulating the serine biosynthesis pathway through suppressing PHGDH expression and reveal serine deprivation as a novel approach to sensitize p53-mediated apoptotic responses in human melanoma cells.
Collapse
Affiliation(s)
- Yang Ou
- From the Institute for Cancer Genetics and Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York 10032 and
| | - Shang-Jui Wang
- From the Institute for Cancer Genetics and Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York 10032 and
| | - Le Jiang
- From the Institute for Cancer Genetics and Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York 10032 and
| | - Bin Zheng
- the Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129
| | - Wei Gu
- From the Institute for Cancer Genetics and Department of Pathology and Cell Biology, College of Physicians and Surgeons, Columbia University, New York, New York 10032 and
| |
Collapse
|
400
|
Siegl C, Prusty BK, Karunakaran K, Wischhusen J, Rudel T. Tumor suppressor p53 alters host cell metabolism to limit Chlamydia trachomatis infection. Cell Rep 2014; 9:918-29. [PMID: 25437549 DOI: 10.1016/j.celrep.2014.10.004] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Revised: 07/21/2014] [Accepted: 09/30/2014] [Indexed: 11/30/2022] Open
Abstract
Obligate intracellular bacteria depend entirely on nutrients from the host cell for their reproduction. Here, we show that obligate intracellular Chlamydia downregulate the central tumor suppressor p53 in human cells. This reduction of p53 levels is mediated by the PI3K-Akt signaling pathway, activation of HDM2, and subsequent proteasomal degradation of p53. The stabilization of p53 in human cells severely impaired chlamydial development and caused the loss of infectious particle formation. DNA-damage-induced p53 interfered with chlamydial development through downregulation of the pentose phosphate pathway (PPP). Increased expression of the PPP key enzyme glucose-6-phosphate dehydrogenase rescued the inhibition of chlamydial growth induced by DNA damage or stabilized p53. Thus, downregulation of p53 is a key event in the chlamydial life cycle that reprograms the host cell to create a metabolic environment supportive of chlamydial growth.
Collapse
Affiliation(s)
- Christine Siegl
- Department of Microbiology, University of Würzburg Biocenter, Am Hubland, 97074 Würzburg, Germany
| | - Bhupesh K Prusty
- Department of Microbiology, University of Würzburg Biocenter, Am Hubland, 97074 Würzburg, Germany
| | - Karthika Karunakaran
- Department of Microbiology, University of Würzburg Biocenter, Am Hubland, 97074 Würzburg, Germany
| | - Jörg Wischhusen
- Frauenklinik, University Clinic Würzburg, Josef-Schneider-Str. 4, 97080 Würzburg, Germany
| | - Thomas Rudel
- Department of Microbiology, University of Würzburg Biocenter, Am Hubland, 97074 Würzburg, Germany.
| |
Collapse
|