Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Mol Cell ; 81(18): 3749-3759, 2021 09 16.
Article in English | MEDLINE | ID: mdl-34469752

ABSTRACT

The expression of the urea cycle (UC) proteins is dysregulated in multiple cancers, providing metabolic benefits to tumor survival, proliferation, and growth. Here, we review the main changes described in the expression of UC enzymes and metabolites in different cancers at various stages and suggest that these changes are dynamic and should hence be viewed in a context-specific manner. Understanding the evolvability in the activity of the UC pathway in cancer has implications for cancer-immune cell interactions and for cancer diagnosis and therapy.


Subject(s)
Carcinogenesis/metabolism , Cell Transformation, Neoplastic/metabolism , Urea/metabolism , Ammonia/metabolism , Cell Line, Tumor , Cell Proliferation , Gene Expression/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/genetics , Humans , Urea Cycle Disorders, Inborn/metabolism , Urea Cycle Disorders, Inborn/physiopathology
2.
Semin Cell Dev Biol ; 98: 15-25, 2020 02.
Article in English | MEDLINE | ID: mdl-31085323

ABSTRACT

Cancer is now considered a multifactorial disorder with different aetiologies and outcomes. Yet, all cancers share some common molecular features. Among these, the reprogramming of cellular metabolism has emerged as a key player in tumour initiation and progression. The finding that metabolic enzymes such as fumarate hydratase (FH), succinate dehydrogenase (SDH) and isocitrate dehydrogenase (IDH), when mutated, cause cancer suggested that metabolic dysregulation is not only a consequence of oncogenic transformation but that it can act as cancer driver. However, the mechanisms underpinning the link between metabolic dysregulation and cancer remain only partially understood. In this review we discuss the role of FH loss in tumorigenesis, focusing on the role of fumarate as a key activator of a variety of oncogenic cascades. We also discuss how these alterations are integrated and converge towards common biological processes. This review highlights the complexity of the signals elicited by FH loss, describes that fumarate can act as a bona fide oncogenic event, and provides a compelling hypothesis of the stepwise neoplastic progression after FH loss.


Subject(s)
Fumarate Hydratase/metabolism , Neoplasms/enzymology , Fumarate Hydratase/genetics , Humans , Mutation , Neoplasms/metabolism , Neoplasms/pathology
3.
Mol Syst Biol ; 17(1): e9730, 2021 01.
Article in English | MEDLINE | ID: mdl-33502086

ABSTRACT

Multi-omics datasets can provide molecular insights beyond the sum of individual omics. Various tools have been recently developed to integrate such datasets, but there are limited strategies to systematically extract mechanistic hypotheses from them. Here, we present COSMOS (Causal Oriented Search of Multi-Omics Space), a method that integrates phosphoproteomics, transcriptomics, and metabolomics datasets. COSMOS combines extensive prior knowledge of signaling, metabolic, and gene regulatory networks with computational methods to estimate activities of transcription factors and kinases as well as network-level causal reasoning. COSMOS provides mechanistic hypotheses for experimental observations across multi-omics datasets. We applied COSMOS to a dataset comprising transcriptomics, phosphoproteomics, and metabolomics data from healthy and cancerous tissue from eleven clear cell renal cell carcinoma (ccRCC) patients. COSMOS was able to capture relevant crosstalks within and between multiple omics layers, such as known ccRCC drug targets. We expect that our freely available method will be broadly useful to extract mechanistic insights from multi-omics studies.


Subject(s)
Carcinoma, Renal Cell/genetics , Computational Biology/methods , Gene Regulatory Networks , Kidney Neoplasms/genetics , Carcinoma, Renal Cell/metabolism , Case-Control Studies , Gene Expression Profiling , Humans , Kidney Neoplasms/metabolism , Metabolomics , Phosphoproteins
4.
Nature ; 537(7621): 544-547, 2016 08 31.
Article in English | MEDLINE | ID: mdl-27580029

ABSTRACT

Mutations of the tricarboxylic acid cycle enzyme fumarate hydratase cause hereditary leiomyomatosis and renal cell cancer. Fumarate hydratase-deficient renal cancers are highly aggressive and metastasize even when small, leading to a very poor clinical outcome. Fumarate, a small molecule metabolite that accumulates in fumarate hydratase-deficient cells, plays a key role in cell transformation, making it a bona fide oncometabolite. Fumarate has been shown to inhibit α-ketoglutarate-dependent dioxygenases that are involved in DNA and histone demethylation. However, the link between fumarate accumulation, epigenetic changes, and tumorigenesis is unclear. Here we show that loss of fumarate hydratase and the subsequent accumulation of fumarate in mouse and human cells elicits an epithelial-to-mesenchymal-transition (EMT), a phenotypic switch associated with cancer initiation, invasion, and metastasis. We demonstrate that fumarate inhibits Tet-mediated demethylation of a regulatory region of the antimetastatic miRNA cluster mir-200ba429, leading to the expression of EMT-related transcription factors and enhanced migratory properties. These epigenetic and phenotypic changes are recapitulated by the incubation of fumarate hydratase-proficient cells with cell-permeable fumarate. Loss of fumarate hydratase is associated with suppression of miR-200 and the EMT signature in renal cancer and is associated with poor clinical outcome. These results imply that loss of fumarate hydratase and fumarate accumulation contribute to the aggressive features of fumarate hydratase-deficient tumours.


Subject(s)
Epigenesis, Genetic , Epithelial-Mesenchymal Transition , Fumarates/metabolism , Animals , Cell Movement , Cells, Cultured , Fumarate Hydratase/deficiency , Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , HEK293 Cells , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Mesoderm/metabolism , Mice , MicroRNAs/genetics , Transcription Factors/metabolism , Transcriptome
5.
PLoS Genet ; 13(3): e1006620, 2017 03.
Article in English | MEDLINE | ID: mdl-28267784

ABSTRACT

Tubulointerstitial kidney disease is an important cause of progressive renal failure whose aetiology is incompletely understood. We analysed a large pedigree with maternally inherited tubulointerstitial kidney disease and identified a homoplasmic substitution in the control region of the mitochondrial genome (m.547A>T). While mutations in mtDNA coding sequence are a well recognised cause of disease affecting multiple organs, mutations in the control region have never been shown to cause disease. Strikingly, our patients did not have classical features of mitochondrial disease. Patient fibroblasts showed reduced levels of mitochondrial tRNAPhe, tRNALeu1 and reduced mitochondrial protein translation and respiration. Mitochondrial transfer demonstrated mitochondrial transmission of the defect and in vitro assays showed reduced activity of the heavy strand promoter. We also identified further kindreds with the same phenotype carrying a homoplasmic mutation in mitochondrial tRNAPhe (m.616T>C). Thus mutations in mitochondrial DNA can cause maternally inherited renal disease, likely mediated through reduced function of mitochondrial tRNAPhe.


Subject(s)
DNA, Mitochondrial/genetics , Kidney Diseases/genetics , Kidney Tubules/pathology , Mutation , Acetylglucosaminidase/urine , Biopsy , Female , Fibroblasts/metabolism , Genetic Linkage , Humans , Leucine/chemistry , Male , Mitochondria/metabolism , Oxygen Consumption , Pedigree , Phenotype , Phenylalanine/chemistry , Polymorphism, Single Nucleotide , Promoter Regions, Genetic , Quadriceps Muscle/pathology , RNA, Transfer/genetics
7.
Metab Eng ; 45: 149-157, 2018 01.
Article in English | MEDLINE | ID: mdl-29191787

ABSTRACT

Deregulated signal transduction and energy metabolism are hallmarks of cancer and both play a fundamental role in tumorigenesis. While it is increasingly recognised that signalling and metabolism are highly interconnected, the underpinning mechanisms of their co-regulation are still largely unknown. Here we designed and acquired proteomics, phosphoproteomics, and metabolomics experiments in fumarate hydratase (FH) deficient cells and developed a computational modelling approach to identify putative regulatory phosphorylation-sites of metabolic enzymes. We identified previously reported functionally relevant phosphosites and potentially novel regulatory residues in enzymes of the central carbon metabolism. In particular, we showed that pyruvate dehydrogenase (PDHA1) enzymatic activity is inhibited by increased phosphorylation in FH-deficient cells, restricting carbon entry from glucose to the tricarboxylic acid cycle. Moreover, we confirmed PDHA1 phosphorylation in human FH-deficient tumours. Our work provides a novel approach to investigate how post-translational modifications of enzymes regulate metabolism and could have important implications for understanding the metabolic transformation of FH-deficient cancers with potential clinical applications.


Subject(s)
Fumarate Hydratase/deficiency , Neoplasm Proteins , Neoplasms , Protein Processing, Post-Translational , Pyruvate Dehydrogenase (Lipoamide) , Cell Line, Tumor , Fumarate Hydratase/metabolism , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Pyruvate Dehydrogenase (Lipoamide)/genetics , Pyruvate Dehydrogenase (Lipoamide)/metabolism
9.
Proc Natl Acad Sci U S A ; 107(2): 726-31, 2010 Jan 12.
Article in English | MEDLINE | ID: mdl-20080742

ABSTRACT

We studied human cancer cell models in which we detected constitutive activation of ERK. A fraction of active ERK was found to be located in mitochondria in RWPE-2 cells, obtained by v-Ki-Ras transformation of the epithelial prostate RWPE-1 cell line; in metastatic prostate cancer DU145 cells; and in osteosarcoma SAOS-2 cells. All these tumor cells displayed marked resistance to death caused by apoptotic stimuli like arachidonic acid and the BH3 mimetic EM20-25, which cause cell death through the mitochondrial permeability transition pore (PTP). PTP desensitization and the ensuing resistance to cell death induced by arachidonic acid or EM20-25 could be ablated by inhibiting ERK with the drug PD98059 or with a selective ERK activation inhibitor peptide. ERK inhibition enhanced glycogen synthase kinase-3 (GSK-3)-dependent phosphorylation of the pore regulator cyclophilin D, whereas GSK-3 inhibition protected from PTP opening. Neither active ERK in mitochondria nor pore desensitization was observed in non-transformed RWPE-1 cells. Thus, in tumor cells mitochondrial ERK activation desensitizes the PTP through a signaling axis that involves GSK-3 and cyclophilin D, a finding that provides a mechanistic basis for increased resistance to apoptosis of neoplastic cells.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Prostatic Neoplasms/pathology , Apoptosis , Blotting, Western , Cell Death , Cell Division/drug effects , Cell Line, Tumor , Enzyme Activation , Flow Cytometry , Humans , Male , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/metabolism , Mitochondria/pathology , Mitochondria/physiology , Mitochondrial Permeability Transition Pore , Prostatic Neoplasms/metabolism
10.
Cell Rep ; 42(7): 112751, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37405921

ABSTRACT

Hereditary leiomyomatosis and renal cell cancer (HLRCC) is a cancer syndrome caused by inactivating germline mutations in fumarate hydratase (FH) and subsequent accumulation of fumarate. Fumarate accumulation leads to profound epigenetic changes and the activation of an anti-oxidant response via nuclear translocation of the transcription factor NRF2. The extent to which chromatin remodeling shapes this anti-oxidant response is currently unknown. Here, we explored the effects of FH loss on the chromatin landscape to identify transcription factor networks involved in the remodeled chromatin landscape of FH-deficient cells. We identify FOXA2 as a key transcription factor that regulates anti-oxidant response genes and subsequent metabolic rewiring cooperating without direct interaction with the anti-oxidant regulator NRF2. The identification of FOXA2 as an anti-oxidant regulator provides additional insights into the molecular mechanisms behind cell responses to fumarate accumulation and potentially provides further avenues for therapeutic intervention for HLRCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Leiomyomatosis , Neoplastic Syndromes, Hereditary , Skin Neoplasms , Uterine Neoplasms , Female , Humans , Fumarate Hydratase/genetics , Antioxidants , NF-E2-Related Factor 2/genetics , Leiomyomatosis/genetics , Uterine Neoplasms/genetics , Skin Neoplasms/genetics , Neoplastic Syndromes, Hereditary/genetics , Chromatin , Kidney Neoplasms/genetics , Carcinoma, Renal Cell/genetics , Hepatocyte Nuclear Factor 3-beta/genetics
11.
Nat Commun ; 13(1): 7830, 2022 12 20.
Article in English | MEDLINE | ID: mdl-36539415

ABSTRACT

Metabolic reprogramming is critical for tumor initiation and progression. However, the exact impact of specific metabolic changes on cancer progression is poorly understood. Here, we integrate multimodal analyses of primary and metastatic clonally-related clear cell renal cancer cells (ccRCC) grown in physiological media to identify key stage-specific metabolic vulnerabilities. We show that a VHL loss-dependent reprogramming of branched-chain amino acid catabolism sustains the de novo biosynthesis of aspartate and arginine enabling tumor cells with the flexibility of partitioning the nitrogen of the amino acids depending on their needs. Importantly, we identify the epigenetic reactivation of argininosuccinate synthase (ASS1), a urea cycle enzyme suppressed in primary ccRCC, as a crucial event for metastatic renal cancer cells to acquire the capability to generate arginine, invade in vitro and metastasize in vivo. Overall, our study uncovers a mechanism of metabolic flexibility occurring during ccRCC progression, paving the way for the development of novel stage-specific therapies.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinoma, Renal Cell/genetics , Amino Acids, Branched-Chain , Nitrogen , Kidney Neoplasms/genetics , Arginine/metabolism , Cell Line, Tumor
12.
Cancers (Basel) ; 13(7)2021 Apr 06.
Article in English | MEDLINE | ID: mdl-33917317

ABSTRACT

Tumor cells display metabolic alterations when compared to non-transformed cells. These characteristics are crucial for tumor development, maintenance and survival providing energy supplies and molecular precursors. Anaplerosis is the property of replenishing the TCA cycle, the hub of carbon metabolism, participating in the biosynthesis of precursors for building blocks or signaling molecules. In advanced prostate cancer, an upshift of succinate-driven oxidative phosphorylation via mitochondrial Complex II was reported. Here, using untargeted metabolomics, we found succinate accumulation mainly in malignant cells and an anaplerotic effect contributing to biosynthesis, amino acid, and carbon metabolism. Succinate also stimulated oxygen consumption. Malignant prostate cells displayed higher mitochondrial affinity for succinate when compared to non-malignant prostate cells and the succinate-driven accumulation of metabolites induced expression of mitochondrial complex subunits and their activities. Moreover, extracellular succinate stimulated migration, invasion, and colony formation. Several enzymes linked to accumulated metabolites in the malignant cells were found upregulated in tumor tissue datasets, particularly NME1 and SHMT2 mRNA expression. High expression of the two genes was associated with shorter disease-free survival in prostate cancer cohorts. Moreover, in-vitro expression of both genes was enhanced in prostate cancer cells upon succinate stimulation. In conclusion, the data indicate that uptake of succinate from the tumor environment has an anaplerotic effect that enhances the malignant potential of prostate cancer cells.

13.
Biology (Basel) ; 10(5)2021 Apr 23.
Article in English | MEDLINE | ID: mdl-33922660

ABSTRACT

Cell proliferation and escape from apoptosis are important pathological features of hepatocellular carcinoma (HCC), one of the tumors with the highest mortality rate worldwide. The aim of the study was to evaluate the expression of the pro-apoptotic p66shc and the anti-apoptotic SerpinB3 in HCCs in relation to clinical outcome, cell fate and tumor growth. p66shc and SerpinB3 were evaluated in 67 HCC specimens and the results were correlated with overall survival. Proliferation and cell death markers were analyzed in hepatoma cells overexpressing SerpinB3, under different stress conditions. p66shc-/- mice and xenograft models were also used to assess the effects of p66shc and SerpinB3 on tumor growth. In patients with HCC, the best survival was observed in the subgroup with p66shc levels below median values and SerpinB3 levels above median values. Mice p66shc-/- showed high levels of SerpinB3, while in HepG2 cells overexpressing SerpinB3, p66shc expression was trivial. HepG2 overexpressing SerpinB3 cells were more prone to die after oxidizing treatments, such as diamide or high concentration H2O2. These cells injected in nude mice developed tumors five times smaller than those from control HepG2 cells. Tumors originating from HepG2 overexpressing SerpinB3 cells showed decreased activated Caspase-8, with concomitant increase of RIP3K and decreased levels of cleaved RIP3K, typical features of necroptosis. In conclusion, in patients affected by HCC, the pattern characterized by p66shc downregulation and elevated SerpinB3 levels was associated with markedly better survival. This pattern favored necroptosis in experimental high-stress conditions.

14.
Nat Commun ; 12(1): 4814, 2021 08 10.
Article in English | MEDLINE | ID: mdl-34376668

ABSTRACT

Glutamoptosis is the induction of apoptotic cell death as a consequence of the aberrant activation of glutaminolysis and mTORC1 signaling during nutritional imbalance in proliferating cells. The role of the bioenergetic sensor AMPK during glutamoptosis is not defined yet. Here, we show that AMPK reactivation blocks both the glutamine-dependent activation of mTORC1 and glutamoptosis in vitro and in vivo. We also show that glutamine is used for asparagine synthesis and the GABA shunt to produce ATP and to inhibit AMPK, independently of glutaminolysis. Overall, our results indicate that glutamine metabolism is connected with mTORC1 activation through two parallel pathways: an acute alpha-ketoglutarate-dependent pathway; and a secondary ATP/AMPK-dependent pathway. This dual metabolic connection between glutamine and mTORC1 must be considered for the future design of therapeutic strategies to prevent cell growth in diseases such as cancer.


Subject(s)
Apoptosis/physiology , Cell Proliferation/physiology , Glutamine/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Signal Transduction/physiology , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , HCT116 Cells , HEK293 Cells , Humans , Male , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Xenograft Model Antitumor Assays/methods
15.
FEBS J ; 284(19): 3132-3144, 2017 10.
Article in English | MEDLINE | ID: mdl-28444969

ABSTRACT

Several lines of evidence indicate that during transformation epithelial cancer cells can acquire mesenchymal features via a process called epithelial-to-mesenchymal transition (EMT). This process endows cancer cells with increased invasive and migratory capacity, enabling tumour dissemination and metastasis. EMT is associated with a complex metabolic reprogramming, orchestrated by EMT transcription factors, which support the energy requirements of increased motility and growth in harsh environmental conditions. The discovery that mutations in metabolic genes such as FH, SDH and IDH activate EMT provided further evidence that EMT and metabolism are intertwined. In this review, we discuss the role of EMT in cancer and the underpinning metabolic reprogramming. We also put forward the hypothesis that, by altering chromatin structure and function, metabolic pathways engaged by EMT are necessary for its full activation.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cellular Reprogramming/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Metabolic Networks and Pathways/genetics , Neoplasms/metabolism , Cell Movement , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Chromatin/chemistry , Chromatin/metabolism , Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mutation , Neoplasm Invasiveness , Neoplasms/genetics , Neoplasms/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Succinate Dehydrogenase/genetics , Succinate Dehydrogenase/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Cell Rep ; 21(4): 1036-1047, 2017 Oct 24.
Article in English | MEDLINE | ID: mdl-29069586

ABSTRACT

Fumarate hydratase (FH) is an enzyme of the tricarboxylic acid (TCA) cycle mutated in hereditary and sporadic cancers. Despite recent advances in understanding its role in tumorigenesis, the effects of FH loss on mitochondrial metabolism are still unclear. Here, we used mouse and human cell lines to assess mitochondrial function of FH-deficient cells. We found that human and mouse FH-deficient cells exhibit decreased respiration, accompanied by a varying degree of dysfunction of respiratory chain (RC) complex I and II. Moreover, we show that fumarate induces succination of key components of the iron-sulfur cluster biogenesis family of proteins, leading to defects in the biogenesis of iron-sulfur clusters that affect complex I function. We also demonstrate that suppression of complex II activity is caused by product inhibition due to fumarate accumulation. Overall, our work provides evidence that the loss of a single TCA cycle enzyme is sufficient to cause combined RC activity dysfunction.


Subject(s)
Electron Transport Complex II/metabolism , Electron Transport Complex I/metabolism , Fumarate Hydratase/metabolism , Animals , Cell Line, Tumor , Cell Respiration , Fumarate Hydratase/deficiency , Fumarate Hydratase/genetics , Fumarates/metabolism , Humans , Iron-Sulfur Proteins/metabolism , Mice
17.
Free Radic Biol Med ; 100: 175-181, 2016 11.
Article in English | MEDLINE | ID: mdl-27117029

ABSTRACT

Cancer is a complex and heterogeneous disease thought to be caused by multiple genetic lesions. The recent finding that enzymes of the tricarboxylic acid (TCA) cycle are mutated in cancer rekindled the hypothesis that altered metabolism might also have a role in cellular transformation. Attempts to link mitochondrial dysfunction to cancer uncovered the unexpected role of small molecule metabolites, now known as oncometabolites, in tumorigenesis. In this review, we describe how oncometabolites can contribute to tumorigenesis. We propose that lesions of oncogenes and tumour suppressors are only one of the possible routes to tumorigenesis, which include accumulation of oncometabolites triggered by environmental cues.


Subject(s)
Carcinogenesis , Citric Acid Cycle , Neoplasms/metabolism , Signal Transduction , Animals , Humans
18.
Nat Commun ; 6: 6001, 2015 Jan 23.
Article in English | MEDLINE | ID: mdl-25613188

ABSTRACT

Mutations in the tricarboxylic acid (TCA) cycle enzyme fumarate hydratase (FH) are associated with a highly malignant form of renal cancer. We combined analytical chemistry and metabolic computational modelling to investigate the metabolic implications of FH loss in immortalized and primary mouse kidney cells. Here, we show that the accumulation of fumarate caused by the inactivation of FH leads to oxidative stress that is mediated by the formation of succinicGSH, a covalent adduct between fumarate and glutathione. Chronic succination of GSH, caused by the loss of FH, or by exogenous fumarate, leads to persistent oxidative stress and cellular senescence in vitro and in vivo. Importantly, the ablation of p21, a key mediator of senescence, in Fh1-deficient mice resulted in the transformation of benign renal cysts into a hyperplastic lesion, suggesting that fumarate-induced senescence needs to be bypassed for the initiation of renal cancers.


Subject(s)
Fumarates/chemistry , Glutathione/metabolism , Animals , Cell Line , Cell Proliferation , Cell Transformation, Neoplastic , Cellular Senescence , Chromatography, Liquid , Computational Biology , Female , Fibroblasts/metabolism , Fumarate Hydratase/chemistry , Glutamine/chemistry , Immunohistochemistry , Kidney/metabolism , Magnetic Resonance Spectroscopy , Male , Mass Spectrometry , Metabolomics , Mice , Mice, Inbred C57BL , Mutation , Oxidation-Reduction , Oxidative Stress , Transcriptome
19.
Methods Enzymol ; 542: 1-23, 2014.
Article in English | MEDLINE | ID: mdl-24862258

ABSTRACT

Cancer cells exhibit profound metabolic alterations, allowing them to fulfill the metabolic needs that come with increased proliferation and additional facets of malignancy. Such a metabolic transformation is orchestrated by the genetic changes that drive tumorigenesis, that is, the activation of oncogenes and/or the loss of oncosuppressor genes, and further shaped by environmental cues, such as oxygen concentration and nutrient availability. Understanding this metabolic rewiring is essential to elucidate the fundamental mechanisms of tumorigenesis as well as to find novel, therapeutically exploitable liabilities of malignant cells. Here, we describe key features of the metabolic transformation of cancer cells, which frequently include the switch to aerobic glycolysis, a profound mitochondrial reprogramming, and the deregulation of lipid metabolism, highlighting the notion that these pathways are not independent but rather cooperate to sustain proliferation. Finally, we hypothesize that only those genetic defects that effectively support anabolism are selected in the course of tumor progression, implying that cancer-associated mutations may undergo a metabolically convergent evolution.


Subject(s)
Glycolysis , Mitochondria/metabolism , Neoplasms/metabolism , Cell Transformation, Neoplastic , Diphosphoglyceric Acids/metabolism , Fatty Acids/biosynthesis , Humans , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Lipid Metabolism , Mutation , Neoplasms/genetics , Oncogenes , Phosphofructokinase-1/metabolism , Phosphofructokinase-2/metabolism , Phosphoglycerate Dehydrogenase/metabolism , Phosphoglycerate Mutase/metabolism , Pyruvate Kinase/metabolism
20.
Oncotarget ; 5(23): 11897-908, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25564869

ABSTRACT

TRAP1 is a mitochondrial chaperone highly expressed in many tumor types; it inhibits respiratory complex II, down-modulating its succinate dehydrogenase (SDH) enzymatic activity. SDH inhibition in turn leads to a pseudohypoxic state caused by succinate-dependent HIF1α stabilization and promotes neoplastic growth. Here we report that TRAP1 inhibition of SDH also shields cells from oxidative insults and from the ensuing lethal opening of the mitochondrial permeability transition pore. This anti-oxidant activity of TRAP1 protects tumor cells from death in conditions of nutrient paucity that mimic those encountered in the neoplasm during the process of malignant accrual, and it is required for in vitro tumorigenic growth. Our findings demonstrate that SDH inhibition by TRAP1 is oncogenic not only by inducing pseudohypoxia, but also by protecting tumor cells from oxidative stress.


Subject(s)
Apoptosis/physiology , HSP90 Heat-Shock Proteins/metabolism , Neoplasms/metabolism , Oxidative Stress/physiology , Succinate Dehydrogenase/antagonists & inhibitors , Blotting, Western , Cell Line, Tumor , Cell Survival/physiology , Flow Cytometry , Humans , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore
SELECTION OF CITATIONS
SEARCH DETAIL