Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Cell Rep ; 20(13): 3135-3148, 2017 Sep 26.
Article in English | MEDLINE | ID: mdl-28954230

ABSTRACT

We explored the role of the Krebs cycle enzyme fumarate hydratase (FH) in glucose-stimulated insulin secretion (GSIS). Mice lacking Fh1 in pancreatic ß cells (Fh1ßKO mice) appear normal for 6-8 weeks but then develop progressive glucose intolerance and diabetes. Glucose tolerance is rescued by expression of mitochondrial or cytosolic FH but not by deletion of Hif1α or Nrf2. Progressive hyperglycemia in Fh1ßKO mice led to dysregulated metabolism in ß cells, a decrease in glucose-induced ATP production, electrical activity, cytoplasmic [Ca2+]i elevation, and GSIS. Fh1 loss resulted in elevated intracellular fumarate, promoting succination of critical cysteines in GAPDH, GMPR, and PARK 7/DJ-1 and cytoplasmic acidification. Intracellular fumarate levels were increased in islets exposed to high glucose and in islets from human donors with type 2 diabetes (T2D). The impaired GSIS in islets from diabetic Fh1ßKO mice was ameliorated after culture under normoglycemic conditions. These studies highlight the role of FH and dysregulated mitochondrial metabolism in T2D.


Subject(s)
Diabetes Mellitus, Type 2/genetics , Fumarate Hydratase/deficiency , Insulin-Secreting Cells/metabolism , Islets of Langerhans/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Humans , Mice
2.
J Exp Med ; 214(3): 719-735, 2017 03 06.
Article in English | MEDLINE | ID: mdl-28202494

ABSTRACT

Strict regulation of stem cell metabolism is essential for tissue functions and tumor suppression. In this study, we investigated the role of fumarate hydratase (Fh1), a key component of the mitochondrial tricarboxylic acid (TCA) cycle and cytosolic fumarate metabolism, in normal and leukemic hematopoiesis. Hematopoiesis-specific Fh1 deletion (resulting in endogenous fumarate accumulation and a genetic TCA cycle block reflected by decreased maximal mitochondrial respiration) caused lethal fetal liver hematopoietic defects and hematopoietic stem cell (HSC) failure. Reexpression of extramitochondrial Fh1 (which normalized fumarate levels but not maximal mitochondrial respiration) rescued these phenotypes, indicating the causal role of cellular fumarate accumulation. However, HSCs lacking mitochondrial Fh1 (which had normal fumarate levels but defective maximal mitochondrial respiration) failed to self-renew and displayed lymphoid differentiation defects. In contrast, leukemia-initiating cells lacking mitochondrial Fh1 efficiently propagated Meis1/Hoxa9-driven leukemia. Thus, we identify novel roles for fumarate metabolism in HSC maintenance and hematopoietic differentiation and reveal a differential requirement for mitochondrial Fh1 in normal hematopoiesis and leukemia propagation.


Subject(s)
Fumarate Hydratase/physiology , Hematopoietic Stem Cells/physiology , Animals , Female , Fumarates/metabolism , Hematopoiesis , Histones/metabolism , Leukemia, Myeloid, Acute/etiology , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , NF-E2-Related Factor 2/physiology , Oxygen Consumption
3.
Cancer Cell ; 30(4): 578-594, 2016 10 10.
Article in English | MEDLINE | ID: mdl-27693047

ABSTRACT

Isocitrate dehydrogenase 1 mutations drive human gliomagenesis, probably through neomorphic enzyme activity that produces D-2-hydroxyglutarate. To model this disease, we conditionally expressed Idh1R132H in the subventricular zone (SVZ) of the adult mouse brain. The mice developed hydrocephalus and grossly dilated lateral ventricles, with accumulation of 2-hydroxyglutarate and reduced α-ketoglutarate. Stem and transit amplifying/progenitor cell populations were expanded, and proliferation increased. Cells expressing SVZ markers infiltrated surrounding brain regions. SVZ cells also gave rise to proliferative subventricular nodules. DNA methylation was globally increased, while hydroxymethylation was decreased. Mutant SVZ cells overexpressed Wnt, cell-cycle and stem cell genes, and shared an expression signature with human gliomas. Idh1R132H mutation in the major adult neurogenic stem cell niche causes a phenotype resembling gliomagenesis.


Subject(s)
Brain Neoplasms/enzymology , Glioma/enzymology , Isocitrate Dehydrogenase/biosynthesis , Lateral Ventricles/enzymology , Neoplastic Stem Cells/enzymology , Stem Cell Niche , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , DNA Methylation , Glioma/genetics , Glioma/pathology , Isocitrate Dehydrogenase/genetics , Lateral Ventricles/pathology , Mice , Mice, Transgenic , Mutation , Neoplastic Stem Cells/pathology , Transcriptome
4.
Mol Cell ; 56(3): 343-344, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25514180

ABSTRACT

Metabolic reprogramming is a characteristic of cancer cells. Three studies published in this month's Molecular Cell provide novel insights into the role of mitochondrial pyruvate in tumor metabolism and describe how targeting pyruvate transport and metabolism may afford therapeutic benefit.


Subject(s)
Mitochondria/metabolism , Neoplasms/metabolism , Pyruvic Acid/metabolism , Biological Transport , Glycolysis , Humans , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/pathology
5.
Int J Gynecol Pathol ; 33(6): 560-7, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25272294

ABSTRACT

The contemporary oncologic pathology report conveys diagnostic, prognostic, predictive, and hereditary predisposition information. Each component may be premised on a morphologic feature or a biomarker. Clinical validity and reproducibility are paramount as is standardization of reporting and clinical response to ensure individualization of patient care. Regarding hereditary predisposition, morphology-based genetic referral systems in some instances have eclipsed genealogy-based systems, for example, cell type in ovarian cancer and BRCA screening. In other instances such as Lynch syndrome, morphology-based schemas supplement clinical schemas and there is an emerging standard of care for reflex biomarker testing. Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) syndrome predisposes patients to uterine and cutaneous leiomyomas (LMs) and renal cell carcinomas (RCCs). Several authors have emphasized the role pathologists may play in identifying this syndrome by recognizing the morphologic characteristics of syndromic uterine LMs and RCCs. Recently immunohistochemical overexpression of S-(2-succinyl) cysteine (2SC) has been demonstrated as a robust biomarker of mutation status in tumors from HLRCC patients. In this blinded control-cohort study we demonstrate that the proposed morphologic criteria used to identify uterine LMs in HLRCC syndrome are largely irreproducible among pathologists and lack sufficient robustness to serve as a trigger to triage cases for 2SC immunohistochemistry or patients for further family/personal history inquiry. Although refinement of morphologic criteria can be considered, in view of the availability of a clinically robust biomarker, consideration should be given to reflex testing of uterine LMs with an appropriate age cut off or in the setting of a suspicious family history.


Subject(s)
Leiomyomatosis/diagnosis , Pathology, Clinical/standards , Skin Neoplasms/diagnosis , Uterine Neoplasms/diagnosis , Adult , Aged , Cohort Studies , Female , Humans , Immunohistochemistry , Middle Aged , Neoplastic Syndromes, Hereditary , Reproducibility of Results , Sensitivity and Specificity
6.
Metabolites ; 4(3): 640-54, 2014 Aug 07.
Article in English | MEDLINE | ID: mdl-25105836

ABSTRACT

Inherited mutations in the Krebs cycle enzyme fumarate hydratase (FH) predispose to hereditary leiomyomatosis and renal cell cancer (HLRCC). Loss of FH activity in HLRCC tumours causes accumulation of the Krebs cycle intermediate fumarate to high levels, which may act as an oncometabolite through various, but not necessarily mutually exclusive, mechanisms. One such mechanism, succination, is an irreversible non-enzymatic modification of cysteine residues by fumarate, to form S-(2-succino)cysteine (2SC). Previous studies have demonstrated that succination of proteins including glyceraldehyde 3-phosphate dehydrogenase (GAPDH), kelch-like ECH-associated protein 1 (KEAP1) and mitochondrial aconitase (ACO2) can have profound effects on cellular metabolism. Furthermore, immunostaining for 2SC is a sensitive and specific biomarker for HLRCC tumours. Here, we performed a proteomic screen on an FH-mutant tumour and two HLRCC-derived cancer cell lines and identified 60 proteins where one or more cysteine residues were succinated; 10 of which were succinated at cysteine residues either predicted, or experimentally proven, to be functionally significant. Bioinformatic enrichment analyses identified most succinated targets to be involved in redox signaling. To our knowledge, this is the first proteomic-based succination screen performed in human tumours and cancer-derived cells and has identified novel 2SC targets that may be relevant to the pathogenesis of HLRCC.

7.
Hypoxia (Auckl) ; 2: 127-142, 2014.
Article in English | MEDLINE | ID: mdl-27774472

ABSTRACT

The hypoxia-inducible factor (HIF) prolyl hydroxylase domain enzymes (PHDs) regulate the stability of HIF protein by post-translational hydroxylation of two conserved prolyl residues in its α subunit in an oxygen-dependent manner. Trans-4-prolyl hydroxylation of HIFα under normal oxygen (O2) availability enables its association with the von Hippel-Lindau (VHL) tumor suppressor pVHL E3 ligase complex, leading to the degradation of HIFα via the ubiquitin-proteasome pathway. Due to the obligatory requirement of molecular O2 as a co-substrate, the activity of PHDs is inhibited under hypoxic conditions, resulting in stabilized HIFα, which dimerizes with HIFß and, together with transcriptional co-activators CBP/p300, activates the transcription of its target genes. As a key molecular regulator of adaptive response to hypoxia, HIF plays important roles in multiple cellular processes and its overexpression has been detected in various cancers. The HIF1α isoform in particular has a strong impact on cellular metabolism, most notably by promoting anaerobic, whilst inhibiting O2-dependent, metabolism of glucose. The PHD enzymes also seem to have HIF-independent functions and are subject to regulation by factors other than O2, such as by metabolic status, oxidative stress, and abnormal levels of endogenous metabolites (oncometabolites) that have been observed in some types of cancers. In this review, we aim to summarize current understandings of the function and regulation of PHDs in cancer with an emphasis on their roles in metabolism.

8.
Endocr Relat Cancer ; 21(4): 653-61, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24951106

ABSTRACT

Hotspot mutations in the promoter of the telomerase reverse transcriptase (TERT) gene have been recently reported in human cancers and proposed as a novel mechanism of telomerase activation. To explore TERT promoter mutations in tumors originating from the adrenal gland and extra-adrenal paraganglia, a set of 253 tumors (38 adrenocortical carcinomas (ACCs), 127 pheochromocytomas (PCCs), 18 extra-adrenal paragangliomas (ea PGLs), 37 head and neck PGLs (HN PGLs), and 33 peripheral neuroblastic tumors) was selected along with 16 human neuroblastoma (NBL) and two ACC cell lines to assess TERT promoter mutations by the Sanger sequencing method. All mutations detected were confirmed by a SNaPshot assay. Additionally, 36 gastrointestinal stromal tumors (GISTs) were added to explore an association between TERT promoter mutations and SDH deficiency. TERT promoter mutations were found in seven out of 289 tumors and in three out of 18 human cell lines; four C228T mutations in 38 ACCs (10.5%), two C228T mutations in 18 ea PGLs (11.1%), one C250T mutation in 36 GISTs (2.8%), and three C228T mutations in 16 human NBL cell lines (18.75%). No mutation was detected in PCCs, HN PGLs, neuroblastic tumors as well as ACC cell lines. TERT promoter mutations preferentially occurred in a SDH-deficient setting (P=0.01) being present in three out of 47 (6.4%) SDH-deficient tumors vs zero out of 171 (0%) SDH-intact tumors. We conclude that TERT promoter mutations occur in ACCs and ea PGLs. In addition, preliminary evidence indicates a potential association with the acquisition of TERT promoter mutations in SDH-deficient tumors.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenocortical Carcinoma/genetics , Paraganglioma/genetics , Promoter Regions, Genetic/genetics , Telomerase/genetics , Adult , Cell Line, Tumor , Female , Head and Neck Neoplasms/genetics , Humans , Male , Middle Aged , Mutation , Neuroblastoma/genetics , Pheochromocytoma/genetics
9.
Stem Cells ; 32(6): 1390-7, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24496882

ABSTRACT

Adult hematopoiesis depends on rare multipotent hematopoietic stem cells (HSCs) that self-renew and give rise to progenitor cells, which differentiate to all blood lineages. The strict regulation of the fine balance between self-renewal and differentiation is essential for normal hematopoiesis and suppression of leukemia development. HSCs and progenitor cells are commonly assumed to reside within the hypoxic BM microenvironment, however, there is no direct evidence supporting this notion. Nevertheless, HSCs and progenitors do exhibit a hypoxic profile and strongly express Hif-1α. Although hypoxia signaling pathways are thought to play important roles in adult HSC maintenance and leukemogenesis, the precise function of Hif-dependent signaling in HSCs remains to be uncovered. Here we discuss recent gain-of-function and loss-of-function studies that shed light on the complex roles of hypoxia-signaling pathways in HSCs and their niches in normal and malignant hematopoiesis. Importantly, we comment on the current and often contrasting interpretations of the role of Hif-dependent signaling in stem cell functions.


Subject(s)
Hematopoietic Stem Cells/metabolism , Leukemia/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction/genetics , Animals , Cell Hypoxia/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Leukemia/pathology
10.
Proc Natl Acad Sci U S A ; 111(11): 4031-6, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24550447

ABSTRACT

2-Oxoglutarate (2OG) and Fe(II)-dependent oxygenase domain-containing protein 1 (OGFOD1) is predicted to be a conserved 2OG oxygenase, the catalytic domain of which is related to hypoxia-inducible factor prolyl hydroxylases. OGFOD1 homologs in yeast are implicated in diverse cellular functions ranging from oxygen-dependent regulation of sterol response genes (Ofd1, Schizosaccharomyces pombe) to translation termination/mRNA polyadenylation (Tpa1p, Saccharomyces cerevisiae). However, neither the biochemical activity of OGFOD1 nor the identity of its substrate has been defined. Here we show that OGFOD1 is a prolyl hydroxylase that catalyzes the posttranslational hydroxylation of a highly conserved residue (Pro-62) in the small ribosomal protein S23 (RPS23). Unusually OGFOD1 retained a high affinity for, and forms a stable complex with, the hydroxylated RPS23 substrate. Knockdown or inactivation of OGFOD1 caused a cell type-dependent induction of stress granules, translational arrest, and growth impairment in a manner complemented by wild-type but not inactive OGFOD1. The work identifies a human prolyl hydroxylase with a role in translational regulation.


Subject(s)
Carrier Proteins/metabolism , Nuclear Proteins/metabolism , Prolyl Hydroxylases/metabolism , Protein Biosynthesis/physiology , Protein Processing, Post-Translational/physiology , Ribosomal Proteins/metabolism , Analysis of Variance , Carrier Proteins/genetics , Computational Biology , Fluorescent Antibody Technique , Gene Knockdown Techniques , Humans , Hydroxylation , Immunoblotting , Immunoprecipitation , Ketoglutaric Acids/metabolism , Luciferases , Nuclear Proteins/genetics , Proline/metabolism , Protein Biosynthesis/genetics , Yeasts
11.
Hum Mol Genet ; 23(9): 2440-6, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24334767

ABSTRACT

Malignant pheochromocytoma (PCC) and paraganglioma (PGL) are mostly caused by germline mutations of SDHB, encoding a subunit of succinate dehydrogenase. Using whole-exome sequencing, we recently identified a mutation in the FH gene encoding fumarate hydratase, in a PCC with an 'SDH-like' molecular phenotype. Here, we investigated the role of FH in PCC/PGL predisposition, by screening for germline FH mutations in a large international cohort of patients. We screened 598 patients with PCC/PGL without mutations in known PCC/PGL susceptibility genes. We searched for FH germline mutations and large deletions, by direct sequencing and multiplex ligation-dependent probe amplification methods. Global alterations in DNA methylation and protein succination were assessed by immunohistochemical staining for 5-hydroxymethylcytosine (5-hmC) and S-(2-succinyl) cysteine (2SC), respectively. We identified five pathogenic germline FH mutations (four missense and one splice mutation) in five patients. Somatic inactivation of the second allele, resulting in a loss of fumarate hydratase activity, was demonstrated in tumors with FH mutations. Low tumor levels of 5-hmC, resembling those in SDHB-deficient tumors, and positive 2SC staining were detected in tumors with FH mutations. Clinically, metastatic phenotype (P = 0.007) and multiple tumors (P = 0.02) were significantly more frequent in patients with FH mutations than those without such mutations. This study reveals a new role for FH in susceptibility to malignant and/or multiple PCC/PGL. Remarkably, FH-deficient PCC/PGLs display the same pattern of epigenetic deregulation as SDHB-mutated malignant PCC/PGL. Therefore, we propose that mutation screening for FH should be included in PCC/PGL genetic testing, at least for tumors with malignant behavior.


Subject(s)
Fumarate Hydratase/genetics , Germ-Line Mutation/genetics , Paraganglioma/genetics , Pheochromocytoma/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Child , Exons/genetics , Female , Genetic Predisposition to Disease/genetics , Humans , Male , Middle Aged , Young Adult
12.
J Clin Invest ; 123(9): 3652-8, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23999438

ABSTRACT

The discovery of cancer-associated mutations in genes encoding key metabolic enzymes has provided a direct link between altered metabolism and cancer. Advances in mass spectrometry and nuclear magnetic resonance technologies have facilitated high-resolution metabolite profiling of cells and tumors and identified the accumulation of metabolites associated with specific gene defects. Here we review the potential roles of such "oncometabolites" in tumor evolution and as clinical biomarkers for the detection of cancers characterized by metabolic dysregulation.


Subject(s)
Biomarkers, Tumor/metabolism , Carboxylic Acids/metabolism , Neoplasms/metabolism , Animals , Cell Transformation, Neoplastic/metabolism , Epigenesis, Genetic , Fumarate Hydratase/genetics , Humans , Metabolic Networks and Pathways/genetics , Mutation , Neoplasms/genetics , Neoplasms/pathology , Succinate Dehydrogenase/genetics
13.
Kidney Int ; 84(4): 667-81, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23823604

ABSTRACT

The heterogeneity of renal cell carcinoma (RCC) poses a challenge for designing clinically applicable diagnostic and screening investigations, predictive and prognostic biomarkers, and targeted molecular therapies. Hereditary RCC syndromes harbor specific driver gene mutations, and their discoveries have provided unequivocal insight into the pathogenomic landscape of RCCs. These observed genetic aberrations correspond to a diverse range of dysplastic metabolic processes, including mutations in genes encoding tricarboxylic acid (TCA) cycle enzymes, defects in hypoxic and antioxidant signaling, and abnormalities in nutrient-sensing phosphorylation cascades. Medical management of RCC focused on understanding and correcting these metabolic abnormalities may refine current RCC screening, diagnosis, and treatment. This review describes RCC subtypes associated with TCA and intermediary metabolic defects, outlining salient clinical features, genetic and molecular pathologies, medical management, and dynamic research areas that may affect future practice.


Subject(s)
Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/therapy , Disease Management , Kidney Neoplasms/metabolism , Kidney Neoplasms/therapy , Animals , Antioxidants/physiology , Carcinoma, Renal Cell/physiopathology , Citric Acid Cycle/physiology , Disease Models, Animal , Early Detection of Cancer , Humans , Hypoxia/physiopathology , Kidney Neoplasms/physiopathology , Mice , Mitochondria/physiology , Signal Transduction/physiology
14.
Cancer Cell ; 23(6): 709-11, 2013 Jun 10.
Article in English | MEDLINE | ID: mdl-23763995

ABSTRACT

Epigenetic reprogramming is a feature of many human cancers. In this issue of Cancer Cell, Letouzé and colleagues describe DNA hypermethylation in paragangliomas harboring mutations in succinate dehydrogenase genes. These tumors accumulate succinate, which inhibits 2-oxoglutarate-dependent histone and DNA demethylase enzymes, resulting in epigenetic silencing that affects neuroendocrine differentiation.


Subject(s)
DNA Methylation , Paraganglioma/pathology , Succinate Dehydrogenase/genetics , Animals , Female , Humans , Male
15.
Cell Rep ; 3(5): 1440-8, 2013 May 30.
Article in English | MEDLINE | ID: mdl-23643539

ABSTRACT

The identification of mutated metabolic enzymes in hereditary cancer syndromes has established a direct link between metabolic dysregulation and cancer. Mutations in the Krebs cycle enzyme, fumarate hydratase (FH), predispose affected individuals to leiomyomas, renal cysts, and cancers, though the respective pathogenic roles of mitochondrial and cytosolic FH isoforms remain undefined. On the basis of comprehensive metabolomic analyses, we demonstrate that FH1-deficient cells and tissues exhibit defects in the urea cycle/arginine metabolism. Remarkably, transgenic re-expression of cytosolic FH ameliorated both renal cyst development and urea cycle defects associated with renal-specific FH1 deletion in mice. Furthermore, acute arginine depletion significantly reduced the viability of FH1-deficient cells in comparison to controls. Our findings highlight the importance of extramitochondrial metabolic pathways in FH-associated oncogenesis and the urea cycle/arginine metabolism as a potential therapeutic target.


Subject(s)
Fumarate Hydratase/metabolism , Kidney Neoplasms/enzymology , Animals , Arginine/metabolism , Argininosuccinic Acid/metabolism , Cell Line , Citric Acid Cycle , Fumarate Hydratase/deficiency , Fumarate Hydratase/genetics , Fumarates/metabolism , Kidney/enzymology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Metabolome , Mice , Mice, Knockout , Mice, Transgenic , Mitochondria/metabolism , Mutation , Protein Isoforms/genetics , Protein Isoforms/metabolism , Urea/metabolism
16.
Cell Rep ; 3(3): 689-700, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23499446

ABSTRACT

The gene encoding the Krebs cycle enzyme fumarate hydratase (FH) is mutated in hereditary leiomyomatosis and renal cell cancer (HLRCC). Loss of FH activity causes accumulation of intracellular fumarate, which can directly modify cysteine residues to form 2-succinocysteine through succination. We undertook a proteomic-based screen in cells and renal cysts from Fh1 (murine FH)-deficient mice and identified 94 protein succination targets. Notably, we identified the succination of three cysteine residues in mitochondrial Aconitase2 (ACO2) crucial for iron-sulfur cluster binding. We show that fumarate exerts a dose-dependent inhibition of ACO2 activity, which correlates with increased succination as determined by mass spectrometry, possibly by interfering with iron chelation. Importantly, we show that aconitase activity is impaired in FH-deficient cells. Our data provide evidence that succination, resulting from FH deficiency, targets and potentially alters the function of multiple proteins and may contribute to the dysregulated metabolism observed in HLRCC.


Subject(s)
Aconitate Hydratase/metabolism , Fumarate Hydratase/deficiency , Fumarate Hydratase/metabolism , Kidney Neoplasms/metabolism , Leiomyomatosis/metabolism , Mitochondria/metabolism , Neoplastic Syndromes, Hereditary/metabolism , Succinic Acid/metabolism , Aconitate Hydratase/antagonists & inhibitors , Animals , Cell Line , Cysteine/metabolism , Fumarate Hydratase/genetics , Fumarates/metabolism , Humans , Iron/metabolism , Mice , Mice, Transgenic , Proteome/metabolism , Skin Neoplasms , Uterine Neoplasms
17.
Front Oncol ; 2: 85, 2012.
Article in English | MEDLINE | ID: mdl-22866264

ABSTRACT

The drive to understand how altered cellular metabolism and cancer are linked has caused a paradigm shift in the focus of cancer research. The discovery of a mutated metabolic enzyme, isocitrate dehydrogenase 1, that leads to accumulation of the oncometabolite 2-hydroxyglutarate, provided significant direct evidence that dysfunctional metabolism plays an important role in oncogenesis. Striking parallels exist with the Krebs cycle enzyme fumarate hydratase (FH), a tumor suppressor, whose mutation is associated with the development of leiomyomata, renal cysts, and tumors. Loss of FH enzymatic activity results in accumulation of intracellular fumarate which has been proposed to act as a competitive inhibitor of 2-oxoglutarate-dependent oxygenases including the hypoxia-inducible factor (HIF) hydroxylases, thus activating oncogenic HIF pathways. Interestingly, our studies have questioned the role of HIF and have highlighted other candidate mechanisms, in particular the non-enzymatic modification of cysteine residues (succination) that could lead to disruption or loss of protein functions, dysfunctional cell metabolism and cell signaling. Here, we discuss the evidence for proposing fumarate as an onco-metabolite.

18.
Mol Cell Biol ; 32(15): 3081-94, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22645311

ABSTRACT

Loss-of-function mutations of the tumor suppressor gene encoding fumarase (FH) occur in individuals with hereditary leiomyomatosis and renal cell cancer syndrome (HLRCC). We found that loss of FH activity conferred protection from apoptosis in normal human renal cells and fibroblasts. In FH-defective cells, both hypoxia-inducible factor 1α (HIF-1α) and HIF-2α accumulated, but they were not required for apoptosis protection. Conversely, AMP-activated protein kinase (AMPK) was activated and required, as evidenced by the finding that FH inactivation failed to protect AMPK-null mouse embryo fibroblasts (MEFs) and AMPK-depleted human renal cells. Activated AMPK was detected in renal cysts, which occur in mice with kidney-targeted deletion of Fh1 and in kidney cancers of HLRCC patients. In Fh1-null MEFs, AMPK activation was sustained by fumarate accumulation and not by defective energy metabolism. Addition of fumarate and succinate to kidney cells led to extracellular signal-regulated kinase 1/2 (ERK1/2) and AMPK activation, probably through a receptor-mediated mechanism. These findings reveal a new mechanism of tumorigenesis due to FH loss and an unexpected pro-oncogenic role for AMPK that is important in considering AMPK reactivation as a therapeutic strategy against cancer.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Fumarate Hydratase/genetics , Fumarates/metabolism , AMP-Activated Protein Kinases/deficiency , AMP-Activated Protein Kinases/genetics , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Fumarate Hydratase/deficiency , Fumarate Hydratase/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/genetics , Leiomyomatosis/genetics , Mice , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/biosynthesis , Mitogen-Activated Protein Kinase 3/metabolism , Neoplastic Syndromes, Hereditary/genetics , RNA Interference , RNA, Small Interfering , Reactive Oxygen Species/analysis , Signal Transduction , Skin Neoplasms , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Uterine Neoplasms
19.
Cell Metab ; 15(3): 361-71, 2012 Mar 07.
Article in English | MEDLINE | ID: mdl-22405071

ABSTRACT

The citric acid cycle (CAC) metabolite fumarate has been proposed to be cardioprotective; however, its mechanisms of action remain to be determined. To augment cardiac fumarate levels and to assess fumarate's cardioprotective properties, we generated fumarate hydratase (Fh1) cardiac knockout (KO) mice. These fumarate-replete hearts were robustly protected from ischemia-reperfusion injury (I/R). To compensate for the loss of Fh1 activity, KO hearts maintain ATP levels in part by channeling amino acids into the CAC. In addition, by stabilizing the transcriptional regulator Nrf2, Fh1 KO hearts upregulate protective antioxidant response element genes. Supporting the importance of the latter mechanism, clinically relevant doses of dimethylfumarate upregulated Nrf2 and its target genes, hence protecting control hearts, but failed to similarly protect Nrf2-KO hearts in an in vivo model of myocardial infarction. We propose that clinically established fumarate derivatives activate the Nrf2 pathway and are readily testable cytoprotective agents.


Subject(s)
Antioxidants/metabolism , Fumarates/therapeutic use , NF-E2-Related Factor 2/metabolism , Animals , Dimethyl Fumarate , Fumarate Hydratase/deficiency , Fumarate Hydratase/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Myocardial Infarction/genetics , Myocardial Infarction/prevention & control , NF-E2-Related Factor 2/genetics , Signal Transduction/genetics , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL