Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters

Country/Region as subject
Publication year range
1.
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
2.
Nature ; 477(7363): 225-8, 2011 Aug 17.
Article in English | MEDLINE | ID: mdl-21849978

ABSTRACT

Fumarate hydratase (FH) is an enzyme of the tricarboxylic acid cycle (TCA cycle) that catalyses the hydration of fumarate into malate. Germline mutations of FH are responsible for hereditary leiomyomatosis and renal-cell cancer (HLRCC). It has previously been demonstrated that the absence of FH leads to the accumulation of fumarate, which activates hypoxia-inducible factors (HIFs) at normal oxygen tensions. However, so far no mechanism that explains the ability of cells to survive without a functional TCA cycle has been provided. Here we use newly characterized genetically modified kidney mouse cells in which Fh1 has been deleted, and apply a newly developed computer model of the metabolism of these cells to predict and experimentally validate a linear metabolic pathway beginning with glutamine uptake and ending with bilirubin excretion from Fh1-deficient cells. This pathway, which involves the biosynthesis and degradation of haem, enables Fh1-deficient cells to use the accumulated TCA cycle metabolites and permits partial mitochondrial NADH production. We predicted and confirmed that targeting this pathway would render Fh1-deficient cells non-viable, while sparing wild-type Fh1-containing cells. This work goes beyond identifying a metabolic pathway that is induced in Fh1-deficient cells to demonstrate that inhibition of haem oxygenation is synthetically lethal when combined with Fh1 deficiency, providing a new potential target for treating HLRCC patients.


Subject(s)
Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , Genes, Lethal/genetics , Genes, Tumor Suppressor , Heme Oxygenase (Decyclizing)/genetics , Heme Oxygenase (Decyclizing)/metabolism , Mutation/genetics , Animals , Bilirubin/metabolism , Cell Line , Cells, Cultured , Citric Acid Cycle , Computer Simulation , Fumarate Hydratase/deficiency , Fumarates/metabolism , Glutamine/metabolism , Heme/metabolism , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Kidney Neoplasms/drug therapy , Kidney Neoplasms/enzymology , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Leiomyomatosis/congenital , Leiomyomatosis/drug therapy , Leiomyomatosis/enzymology , Leiomyomatosis/genetics , Leiomyomatosis/metabolism , Mice , Mitochondria/metabolism , NAD/metabolism , Neoplastic Syndromes, Hereditary , Skin Neoplasms , Uterine Neoplasms
3.
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
4.
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
5.
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
6.
Cancer Cell ; 11(4): 311-9, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17418408

ABSTRACT

Germline mutations in the fumarate hydratase (FH) tumor suppressor gene predispose to leiomyomatosis, renal cysts, and renal cell cancer (HLRCC). HLRCC tumors overexpress HIF1alpha and hypoxia pathway genes. We conditionally inactivated mouse Fh1 in the kidney. Fh1 mutants developed multiple clonal renal cysts that overexpressed Hif1alpha and Hif2alpha. Hif targets, such as Glut1 and Vegf, were upregulated. We found that Fh1-deficient murine embryonic stem cells and renal carcinomas from HLRCC showed similar overexpression of HIF and hypoxia pathway components to the mouse cysts. Our data have shown in vivo that pseudohypoxic drive, resulting from HIF1alpha (and HIF2alpha) overexpression, is a direct consequence of Fh1 inactivation. Our mouse may be useful for testing therapeutic interventions that target angiogenesis and HIF-prolyl hydroxylation.


Subject(s)
Carcinoma, Renal Cell/etiology , Fumarate Hydratase/genetics , Gene Silencing/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Diseases, Cystic/etiology , Kidney Neoplasms/etiology , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Cell Hypoxia , Cell Proliferation , Female , Glucose Transporter Type 1/metabolism , Kidney Diseases, Cystic/metabolism , Kidney Diseases, Cystic/pathology , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
7.
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
8.
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
9.
Biochim Biophys Acta ; 1807(11): 1432-43, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21771581

ABSTRACT

The SDHA, SDHB, SDHC, SDHD genes encode the four subunits of succinate dehydrogenase (SDH; mitochondrial complex II), a mitochondrial enzyme involved in two essential energy-producing metabolic processes of the cell, the Krebs cycle and the electron transport chain. Germline loss-of-function mutations in any of the SDH genes or assembly factor (SDHAF2) cause hereditary paraganglioma/phaeochromocytoma syndrome (HPGL/PCC) through a mechanism which is largely unknown. Owing to the central function of SDH in cellular energy metabolism it is important to understand its role in tumor suppression. Here is reported an overview of genetics, clinical and molecular progress recently performed in understanding the basis of HPGL/PCC tumorigenesis.


Subject(s)
Isoenzymes/genetics , Mutation , Neoplasms/enzymology , Neoplasms/genetics , Succinate Dehydrogenase/genetics , Animals , Genes, Tumor Suppressor , Humans , Isoenzymes/chemistry , Mitochondria/enzymology , Protein Subunits/chemistry , Protein Subunits/genetics , Succinate Dehydrogenase/chemistry
10.
Hum Mol Genet ; 19(19): 3844-51, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20660115

ABSTRACT

Mutations in the gene encoding the Krebs cycle enzyme fumarate hydratase (FH) predispose to hereditary leiomyomatosis and renal cell cancer in affected individuals. FH-associated neoplasia is characterized by defective mitochondrial function and by upregulation of transcriptional pathways mediated by hypoxia-inducible factor (HIF), although whether and by what means these processes are linked has been disputed. We analysed the HIF pathway in Fh1-/- mouse embryonic fibroblasts (MEFs), in FH-defective neoplastic tissues and in Fh1-/- MEFs re-expressing either wild-type or an extra-mitochondrial restricted form of FH. These experiments demonstrated that upregulation of HIF-1alpha occurs as a direct consequence of FH inactivation. Fh1-/- cells accumulated intracellular fumarate and manifested severe impairment of HIF prolyl but not asparaginyl hydroxylation which was corrected by provision of exogenous 2-oxoglutarate (2-OG). Re-expression of the extra-mitochondrial form of FH in Fh1-/- cells was sufficient to reduce intracellular fumarate and to correct dysregulation of the HIF pathway completely, even in cells that remained profoundly defective in mitochondrial energy metabolism. The findings indicate that upregulation of HIF-1alpha arises from competitive inhibition of the 2-OG-dependent HIF hydroxylases by fumarate and not from disruption of mitochondrial energy metabolism.


Subject(s)
Fumarate Hydratase/deficiency , Mitochondria/metabolism , Signal Transduction , Animals , Cell Hypoxia , Embryo, Mammalian/cytology , Fibroblasts/enzymology , Fibroblasts/pathology , Fumarate Hydratase/metabolism , Genetic Complementation Test , Humans , Hydroxylation , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Models, Biological , Oxygen Consumption , Proline/metabolism , Protein Processing, Post-Translational
11.
J Pathol ; 223(1): 1-3, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20938930

ABSTRACT

Autosomal dominant polycystic kidney disease (PKD) is an inherited disease that results from mutations in either polycystin (PKD1) or polycystin 2 (PKD2), both of which are large, complex, and multifunctional proteins whose loss results in the development of numerous fluid-filled cysts and fibrosis that compromise renal function. A number of spontaneous and engineered mouse models of PKD have provided some understanding of many aspects of cyst development, modifier genes, and mechanistic pathways, but fall short of reproducing the human disease accurately. Two recent papers in The Journal of Pathology set out new models using miRNA, or inducible and targeted recombination, that achieve partial or timed suppression of Pkd1. Instead of knocking out Pkd1 immediately, or completely, these more subtle approaches may help deliver more faithful models of this significant human renal disease.


Subject(s)
Disease Models, Animal , Polycystic Kidney Diseases/genetics , Animals , Gene Knockdown Techniques/methods , Haploinsufficiency , Humans , Mice , RNA, Small Interfering/genetics , TRPP Cation Channels/genetics
12.
J Pathol ; 225(1): 4-11, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21630274

ABSTRACT

Germline mutations in the FH gene encoding the Krebs cycle enzyme fumarate hydratase predispose to hereditary leiomyomatosis and renal cell cancer (HLRCC) syndrome. FH-deficient cells and tissues accumulate high levels of fumarate, which may act as an oncometabolite and contribute to tumourigenesis. A recently proposed role for fumarate in the covalent modification of cysteine residues to S-(2-succinyl) cysteine (2SC) (termed protein succination) prompted us to assess 2SC levels in our existing models of HLRCC. Herein, using a previously characterized antibody against 2SC, we show that genetic ablation of FH causes high levels of protein succination. We next hypothesized that immunohistochemistry for 2SC would serve as a metabolic biomarker for the in situ detection of FH-deficient tissues. Robust detection of 2SC was observed in Fh1 (murine FH)-deficient renal cysts and in a retrospective series of HLRCC tumours (n = 16) with established FH mutations. Importantly, 2SC was undetectable in normal tissues (n = 200) and tumour types not associated with HLRCC (n = 1342). In a prospective evaluation of cases referred for genetic testing for HLRCC, the presence of 2SC-modified proteins (2SCP) correctly predicted genetic alterations in FH in every case. In two series of unselected type II papillary renal cancer (PRCC), prospectively analysed by 2SCP staining followed by genetic analysis, the biomarker accurately identified previously unsuspected FH mutations (2/33 and 1/36). The investigation of whether metabolites in other tumour types produce protein modification signature(s) that can be assayed using similar strategies will be of interest in future studies of cancer.


Subject(s)
Carcinoma, Renal Cell/diagnosis , Fumarate Hydratase/deficiency , Kidney Neoplasms/diagnosis , Leiomyomatosis/diagnosis , Neoplastic Syndromes, Hereditary/diagnosis , Adult , Aged , Animals , Biomarkers, Tumor/deficiency , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/genetics , Disease Models, Animal , Female , Fumarate Hydratase/genetics , Fumarate Hydratase/metabolism , Genetic Predisposition to Disease , Germ-Line Mutation , Humans , Kidney Neoplasms/genetics , Leiomyomatosis/genetics , Loss of Heterozygosity , Male , Mice , Mice, Knockout , Mice, Transgenic , Middle Aged , Neoplastic Syndromes, Hereditary/genetics , Prospective Studies , Sensitivity and Specificity , Succinic Acid/metabolism
13.
J Biol Chem ; 284(25): 16767-16775, 2009 Jun 19.
Article in English | MEDLINE | ID: mdl-19386601

ABSTRACT

Hypoxia-inducible factor (HIF) controls an extensive range of adaptive responses to hypoxia. To better understand this transcriptional cascade we performed genome-wide chromatin immunoprecipitation using antibodies to two major HIF-alpha subunits, and correlated the results with genome-wide transcript profiling. Within a tiled promoter array we identified 546 and 143 sequences that bound, respectively, to HIF-1alpha or HIF-2alpha at high stringency. Analysis of these sequences confirmed an identical core binding motif for HIF-1alpha and HIF-2alpha (RCGTG) but demonstrated that binding to this motif was highly selective, with binding enriched at distinct regions both upstream and downstream of the transcriptional start. Comparison of HIF-promoter binding data with bidirectional HIF-dependent changes in transcript expression indicated that whereas a substantial proportion of positive responses (>20% across all significantly regulated genes) are direct, HIF-dependent gene suppression is almost entirely indirect. Comparison of HIF-1alpha- versus HIF-2alpha-binding sites revealed that whereas some loci bound HIF-1alpha in isolation, many bound both isoforms with similar affinity. Despite high-affinity binding to multiple promoters, HIF-2alpha contributed to few, if any, of the transcriptional responses to acute hypoxia at these loci. Given emerging evidence for biologically distinct functions of HIF-1alpha versus HIF-2alpha understanding the mechanisms restricting HIF-2alpha activity will be of interest.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Hypoxia/genetics , Cell Hypoxia/physiology , DNA, Neoplasm/genetics , DNA, Neoplasm/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Base Sequence , Binding Sites/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation , Female , Gene Expression Profiling , Genome-Wide Association Study , Humans , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Protein Binding
14.
Lancet Oncol ; 10(8): 764-71, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19576851

ABSTRACT

BACKGROUND: Phaeochromocytomas and paragangliomas are neuro-endocrine tumours that occur sporadically and in several hereditary tumour syndromes, including the phaeochromocytoma-paraganglioma syndrome. This syndrome is caused by germline mutations in succinate dehydrogenase B (SDHB), C (SDHC), or D (SDHD) genes. Clinically, the phaeochromocytoma-paraganglioma syndrome is often unrecognised, although 10-30% of apparently sporadic phaeochromocytomas and paragangliomas harbour germline SDH-gene mutations. Despite these figures, the screening of phaeochromocytomas and paragangliomas for mutations in the SDH genes to detect phaeochromocytoma-paraganglioma syndrome is rarely done because of time and financial constraints. We investigated whether SDHB immunohistochemistry could effectively discriminate between SDH-related and non-SDH-related phaeochromocytomas and paragangliomas in large retrospective and prospective tumour series. METHODS: Immunohistochemistry for SDHB was done on 220 tumours. Two retrospective series of 175 phaeochromocytomas and paragangliomas with known germline mutation status for phaeochromocytoma-susceptibility or paraganglioma-susceptibility genes were investigated. Additionally, a prospective series of 45 phaeochromocytomas and paragangliomas was investigated for SDHB immunostaining followed by SDHB, SDHC, and SDHD mutation testing. FINDINGS: SDHB protein expression was absent in all 102 phaeochromocytomas and paragangliomas with an SDHB, SDHC, or SDHD mutation, but was present in all 65 paraganglionic tumours related to multiple endocrine neoplasia type 2, von Hippel-Lindau disease, and neurofibromatosis type 1. 47 (89%) of the 53 phaeochromocytomas and paragangliomas with no syndromic germline mutation showed SDHB expression. The sensitivity and specificity of the SDHB immunohistochemistry to detect the presence of an SDH mutation in the prospective series were 100% (95% CI 87-100) and 84% (60-97), respectively. INTERPRETATION: Phaeochromocytoma-paraganglioma syndrome can be diagnosed reliably by an immunohistochemical procedure. SDHB, SDHC, and SDHD germline mutation testing is indicated only in patients with SDHB-negative tumours. SDHB immunohistochemistry on phaeochromocytomas and paragangliomas could improve the diagnosis of phaeochromocytoma-paraganglioma syndrome. FUNDING: The Netherlands Organisation for Scientific Research, Dutch Cancer Society, Vanderes Foundation, Association pour la Recherche contre le Cancer, Institut National de la Santé et de la Recherche Médicale, and a PHRC grant COMETE 3 for the COMETE network.


Subject(s)
Adrenal Gland Neoplasms/genetics , Immunohistochemistry/methods , Paraganglioma/genetics , Pheochromocytoma/genetics , Succinate Dehydrogenase/genetics , Adolescent , Adrenal Gland Neoplasms/diagnosis , Adult , Aged , Blotting, Western , Child , DNA Mutational Analysis , Female , Germ-Line Mutation , Humans , Male , Membrane Proteins/genetics , Middle Aged , Paraganglioma/diagnosis , Pheochromocytoma/diagnosis , Syndrome , Young Adult
15.
Endocr Relat Cancer ; 16(2): 515-25, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19208735

ABSTRACT

Mutations in succinate dehydrogense-B (SDHB) and the von Hippel-Lindau (VHL) genes result in an increased risk of developing chromaffin tumours via a common aetiological pathway. The aim of the present retrospective study was to compare the clinical phenotypes of disease in subjects developing chromaffin tumours as a result of SDHB mutations or VHL disease. Thirty-one subjects with chromaffin tumours were assessed; 16 subjects had SDHB gene mutations and 15 subjects had a diagnosis of VHL. VHL-related tumours were predominantly adrenal phaeochromocytomas (22/26; 84.6%), while SDHB-related tumours were predominantly extra-adrenal paragangliomas (19/25; 76%). Median age at onset of the first chromaffin tumour was similar in the two cohorts. Tumour size was significantly larger in the SDHB cohort in comparison with the VHL cohort (P=0.002). Multifocal disease was present in 9/15 (60%) of the VHL cohort (bilateral phaeochromocytomas) and only 3/16 (19%) of the SDHB cohort, while metastatic disease was found in 5/16 (31%) of the SDHB cohort but not in the VHL cohort to date. The frequency of symptoms, hypertension and the magnitude of catecholamine secretion appeared to be greater in the SDHB cohort. Renal cell carcinomas were a feature in 5/15 (33%) of the VHL cohort and 1/16 (6%) of the SDHB cohort. These data indicate that SDHB-related tumours are predominantly extra-adrenal in location and associated with higher catecholamine secretion and more malignant disease, in subjects who appear more symptomatic. VHL-related tumours tend to be adrenal phaeochromocytomas, frequently bilateral and associated with a milder phenotype.


Subject(s)
Adrenal Gland Neoplasms/genetics , Carcinoma, Renal Cell/genetics , Kidney Neoplasms/genetics , Paraganglioma, Extra-Adrenal/genetics , Pheochromocytoma/genetics , Succinate Dehydrogenase/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Adolescent , Adrenal Gland Neoplasms/metabolism , Adrenal Gland Neoplasms/pathology , Adult , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Catecholamines/metabolism , Child , Cohort Studies , Female , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Male , Middle Aged , Mutation/genetics , Paraganglioma, Extra-Adrenal/metabolism , Paraganglioma, Extra-Adrenal/secondary , Pheochromocytoma/metabolism , Pheochromocytoma/pathology , Prognosis , Retrospective Studies , Succinate Dehydrogenase/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Young Adult
16.
Biochem J ; 416(3): 387-94, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18713068

ABSTRACT

The transcription factor HIF (hypoxia-inducible factor) mediates a highly pleiotrophic response to hypoxia. Many recent studies have focused on defining the extent of this transcriptional response. In the present study we have analysed regulation by hypoxia among transcripts encoding human Fe(II)- and 2-oxoglutarate-dependent oxygenases. Our results show that many of these genes are regulated by hypoxia and define two groups of histone demethylases as new classes of hypoxia-regulated genes. Patterns of induction were consistent across a range of cell lines with JMJD1A (where JMJD is Jumonji-domain containing) and JMJD2B demonstrating robust, and JMJD2C more modest, up-regulation by hypoxia. Functional genetic and chromatin immunoprecipitation studies demonstrated the importance of HIF-1alpha in mediating these responses. Given the importance of histone methylation status in defining patterns of gene expression under different physiological and pathophysiological conditions, these findings predict a role for the HIF system in epigenetic regulation.


Subject(s)
Gene Expression Regulation, Enzymologic , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia , Neoplasm Proteins/metabolism , Oxidoreductases, N-Demethylating/metabolism , Transcription Factors/metabolism , Amino Acids, Dicarboxylic/metabolism , Cell Line , Deferoxamine/metabolism , Gene Expression Profiling , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Jumonji Domain-Containing Histone Demethylases , Ketoglutaric Acids/metabolism , Molecular Sequence Data , Neoplasm Proteins/genetics , Oligonucleotide Array Sequence Analysis , Oxidoreductases, N-Demethylating/genetics , Oxygenases/genetics , Oxygenases/metabolism , Promoter Regions, Genetic , Protein Subunits/genetics , Protein Subunits/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Siderophores/metabolism , Transcription Factors/genetics
17.
Clin Endocrinol (Oxf) ; 69(4): 587-96, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18419787

ABSTRACT

OBJECTIVE: Phaeochromocytomas and paragangliomas are familial in up to 25% of cases and can result from succinate dehydrogenase (SDH) gene mutations. The aim of this study was to describe the clinical manifestations of subjects with SDH-B gene mutations. DESIGN: Retrospective case-series. PATIENTS: Thirty-two subjects with SDH-B gene mutations followed up between 1975 and 2007. Mean follow-up of 5.8 years (SD 7.4, range 0-31). Patients seen at St Bartholomew's Hospital, London and other UK centres. MEASUREMENTS: Features of clinical presentation, genetic mutations, tumour location, catecholamine secretion, clinical course and management. RESULTS: Sixteen of 32 subjects (50%) were affected by disease. Two previously undescribed mutations in the SDH-B gene were noted. A family history of disease was apparent in only 18% of index subjects. Mean age at diagnosis was 34 years (SD 15.4, range 10-62). 50% of affected subjects had disease by the age of 26 years. 69% (11 of 16) were hypertensive and 80% (12 of 15) had elevated secretions of catecholamines/metabolites. 24% (6 of 25) of tumours were located in the adrenal and 76% (19 of 25) were extra-adrenal. 19% (3 of 16) had multifocal disease. Metastatic paragangliomas developed in 31% (5 of 16). One subject developed a metastatic type II papillary renal cell carcinoma. The cohort malignancy rate was 19% (6 of 32). Macrovascular disease was noted in two subjects without hypertension. CONCLUSION: SDH-B mutation carriers develop disease early and predominantly in extra-adrenal locations. Disease penetrance is incomplete. Metastatic disease is prominent but levels are less than previously reported. Clinical manifestations may include papillary renal cell carcinoma and macrovascular disease.


Subject(s)
Adrenal Gland Neoplasms/diagnosis , Paraganglioma/diagnosis , Pheochromocytoma/diagnosis , Succinate Dehydrogenase/genetics , Adolescent , Adrenal Gland Neoplasms/epidemiology , Adrenal Gland Neoplasms/genetics , Adrenal Gland Neoplasms/pathology , Adult , Case-Control Studies , Child , Comorbidity , Family , Female , Heterozygote , Humans , Male , Middle Aged , Mutation , Paraganglioma/epidemiology , Paraganglioma/genetics , Paraganglioma/pathology , Pedigree , Pheochromocytoma/epidemiology , Pheochromocytoma/genetics , Pheochromocytoma/pathology , Retrospective Studies , Young Adult
18.
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
19.
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
20.
J Clin Endocrinol Metab ; 91(8): 3071-5, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16757530

ABSTRACT

CONTEXT: Leydig cell tumors (LCTs) are the most common non-germ-cell neoplasms of the testis. LCTs are often hormonally active and can result in precocious virilization or in adult feminization. We identified an LCT in an affected individual from a kindred with hereditary leiomyomatosis and renal cell cancer (HLRCC) and a germline fumarate hydratase (FH) mutation (N64T). OBJECTIVE: Our objective was to investigate the role of FH mutations in predisposition to LCTs. DESIGN: We tested for pathogenic effects of the N64T mutation and screened an additional 29 unselected adult LCTs for FH alterations. We also tested these LCTs for mutations in two genes, the LH/choriogonadotropin receptor (LHCGR) and the guanine nucleotide-binding protein alpha (GNAS) that had been implicated in LCT tumorigenesis. RESULTS: No mutations were found in GNAS, and one tumor had a LHCGR somatic substitution. In addition to the HLRCC case with the N64T germline FH mutation, we identified one other LCT with a previously unreported FH mutation (M411I). Both LCTs from these patients showed loss of the wild-type FH allele. Immunohistochemical and in situ hybridization analyses demonstrated activation of the hypoxia/angiogenesis pathway not only in the tumors belonging to the FH mutation carriers but also in several other mutation-negative LCTs. CONCLUSIONS: Our study shows that some LCTs are caused by FH mutations and represents one of the first reports of germline mutations in any type of adult testicular tumor.


Subject(s)
Fumarate Hydratase/genetics , Genetic Predisposition to Disease , Germ-Line Mutation , Leydig Cell Tumor/genetics , Testicular Neoplasms/genetics , Base Sequence , Chromogranins , DNA, Complementary/chemistry , GTP-Binding Protein alpha Subunits, Gs/genetics , Humans , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/analysis , Immunohistochemistry , In Situ Hybridization , Leydig Cell Tumor/chemistry , Male , Models, Molecular , Neovascularization, Pathologic , Receptors, LH/genetics , Sequence Analysis, DNA , Testicular Neoplasms/chemistry , Vascular Endothelial Growth Factor A/analysis
SELECTION OF CITATIONS
SEARCH DETAIL