Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 136(5): 839-851, 2009 Mar 06.
Article in English | MEDLINE | ID: mdl-19217150

ABSTRACT

A key function of blood vessels, to supply oxygen, is impaired in tumors because of abnormalities in their endothelial lining. PHD proteins serve as oxygen sensors and may regulate oxygen delivery. We therefore studied the role of endothelial PHD2 in vessel shaping by implanting tumors in PHD2(+/-) mice. Haplodeficiency of PHD2 did not affect tumor vessel density or lumen size, but normalized the endothelial lining and vessel maturation. This resulted in improved tumor perfusion and oxygenation and inhibited tumor cell invasion, intravasation, and metastasis. Haplodeficiency of PHD2 redirected the specification of endothelial tip cells to a more quiescent cell type, lacking filopodia and arrayed in a phalanx formation. This transition relied on HIF-driven upregulation of (soluble) VEGFR-1 and VE-cadherin. Thus, decreased activity of an oxygen sensor in hypoxic conditions prompts endothelial cells to readjust their shape and phenotype to restore oxygen supply. Inhibition of PHD2 may offer alternative therapeutic opportunities for anticancer therapy.


Subject(s)
Blood Vessels/cytology , DNA-Binding Proteins/metabolism , Endothelial Cells/metabolism , Immediate-Early Proteins/metabolism , Neoplasm Metastasis , Neoplasms/blood supply , Oxygen/metabolism , Animals , Blood Vessels/embryology , Blood Vessels/metabolism , Cell Shape , DNA-Binding Proteins/genetics , Endothelial Cells/cytology , Glycolysis , Heterozygote , Hypoxia/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases , Immediate-Early Proteins/genetics , Mice , Neoplasms/pathology , Procollagen-Proline Dioxygenase
2.
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
3.
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
4.
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
5.
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
6.
Blood ; 122(10): 1741-5, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23894152

ABSTRACT

Local hypoxia in hematopoietic stem cell (HSC) niches is thought to regulate HSC functions. Hypoxia-inducible factor-1 (Hif-1) and Hif-2 are key mediators of cellular responses to hypoxia. Although oxygen-regulated α-subunits of Hifs, namely Hif-1α and Hif-2α, are closely related, they play overlapping and also distinct functions in nonhematopoietic tissues. Although Hif-1α-deficient HSCs lose their activity on serial transplantation, the role for Hif-2α in cell-autonomous HSC maintenance remains unknown. Here, we demonstrate that constitutive or inducible hematopoiesis-specific Hif-2α deletion does not affect HSC numbers and steady-state hematopoiesis. Furthermore, using serial transplantations and 5-fluorouracil treatment, we demonstrate that HSCs do not require Hif-2α to self-renew and recover after hematopoietic injury. Finally, we show that Hif-1α deletion has no major impact on steady-state maintenance of Hif-2α-deficient HSCs and their ability to repopulate primary recipients, indicating that Hif-1α expression does not account for normal behavior of Hif-2α-deficient HSCs.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Animals , Cell Proliferation , Cell Survival , Female , Gene Deletion , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Male , Mice
7.
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
8.
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
9.
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
10.
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
11.
J Pathol ; 226(1): 73-83, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22009253

ABSTRACT

Adenomatous polyposis coli (APC ) mutations are found in most colorectal tumours. These mutations are almost always protein-truncating, deleting both central domains that regulate Wnt signalling and C-terminal domains that interact with the cytoskeleton. The importance of Wnt dysregulation for colorectal tumourigenesis is well characterized. It is, however, unclear whether loss of C-terminal functions contributes to tumourigenesis, although this protein region has been implicated in cellular processes--including polarity, migration, mitosis, and chromosomal instability (CIN)­that have been postulated as critical for the development and progression of intestinal tumours. Since almost all APC mutations in human patients disrupt both central and C-terminal regions, we created a mouse model to test the role of the C-terminus of APC in intestinal tumourigenesis. This mouse (Apc(ΔSAMP)) carries an internal deletion within Apc that dysregulates Wnt by removing the beta-catenin-binding and SAMP repeats, but leaves the C-terminus intact. We compared Apc(ΔSAMP) mice with Apc(1322T) animals. The latter allele represented the most commonly found human APC mutation and was identical to Apc(ΔSAMP) except for absence of the entire C-terminus. Apc(ΔSAMP) mice developed numerous intestinal adenomas indistinguishable in number, location, and dysplasia from those seen in Apc(1322T) mice. No carcinomas were found in Apc(ΔSAMP) or Apc(1322T) animals. While similar disruption of the Wnt signalling pathway was observed in tumours from both mice, no evidence of differential C-terminus functions (such as cell migration, CIN, or localization of APC and EB1) was seen. We conclude that the C-terminus of APC does not influence intestinal adenoma development or progression.


Subject(s)
Adenoma/genetics , Adenomatous Polyposis Coli Protein/genetics , Intestinal Neoplasms/genetics , Adenoma/pathology , Adenomatous Polyposis Coli Protein/chemistry , Animals , Blotting, Western , Cell Movement/genetics , Disease Progression , Fluorescent Antibody Technique , Immunohistochemistry , In Situ Hybridization , Intestinal Neoplasms/pathology , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Oligonucleotide Array Sequence Analysis , Protein Structure, Tertiary/genetics , Signal Transduction/genetics , Wnt Signaling Pathway
12.
J Physiol ; 590(16): 4079-91, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22615432

ABSTRACT

This study investigated the function of each of the hypoxia inducible factor (HIF) prolyl-4-hydroxylase enzymes (PHD1­3) in the first 24 h following transient focal cerebral ischaemia by using mice with each isoform genetically suppressed. Male, 8- to 12-week old PHD1−/−, PHD2+/− and PHD3−/− mice and their wild-type (WT) littermate were subjected to 45 min of middle cerebral artery occlusion (MCAO). During the experiments, regional cerebral blood flow (rCBF) was recorded by laser Doppler flowmetry. Behaviour was assessed at both 2 h and 24 h after reperfusion with a common neuroscore. Infarct volumes, blood­brain barrier (BBB) disruption, cerebral vascular density, apoptosis, reactive oxygen species (ROS), HIF1α, and glycogen levels were then determined using histological and immunohistochemical techniques. When compared to their WT littermates, PHD2+/− mice had significantly increased cerebral microvascular density and more effective restoration of CBF upon reperfusion. PHD2+/− mice showed significantly better functional outcomes and higher activity rates at both 2 h and 24 h after MCAO, associated with significant fewer apoptotic cells in the penumbra and less BBB disruption; PHD3−/− mice had impaired rCBF upon early reperfusion but comparable functional outcomes; PHD1−/− mice did not show any significant changes following the MCAO. Production of ROS, HIF1α staining and glycogen content in the brain were not different in any comparison. Life-long genetic inhibition of PHD enzymes produces different effects on outcome in the first 24 h after transient cerebral ischaemia. These need to be considered in optimizing therapeutic effects of PHD inhibitors, particularly when isoform specific inhibitors become available.


Subject(s)
Brain Ischemia/enzymology , Procollagen-Proline Dioxygenase/metabolism , Animals , Body Temperature Regulation , Body Weight , Brain Ischemia/metabolism , Cerebral Arteries , Cerebral Cortex/blood supply , Gene Expression Regulation/physiology , Genotype , Hypoxia-Inducible Factor-Proline Dioxygenases , Male , Mice , Mice, Knockout , Procollagen-Proline Dioxygenase/genetics , Protein Isoforms
13.
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
14.
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
15.
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
16.
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
17.
18.
Gut ; 59(12): 1680-6, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20926645

ABSTRACT

BACKGROUND AND AIMS: Adenomatous polyposis coli (APC) is a tumour suppressor gene mutated in the germline of patients with familial adenomatous polyposis (FAP) and somatically in most colorectal cancers. APC mutations impair ß-catenin degradation, resulting in increased Wnt signalling. The most frequent APC mutation is a codon 1309 truncation that is associated with severe FAP. A previous study compared two mouse models of intestinal tumorigenesis, Apc(R850X) (Min) and Apc(1322T) (1322T), the latter a model of human codon 1309 changes. 1322T mice had more severe polyposis but, surprisingly, these tumours had lower levels of nuclear ß-catenin than Min tumours. The consequences of these different ß-catenin levels were investigated. METHODS: Enterocytes were isolated from 1322T and Min tumours by microdissection and gene expression profiling was performed. Differentially expressed Wnt targets and other stem cell markers were validated using quantitative PCR, in situ hybridisation and immunohistochemistry. RESULTS: As expected, lower nuclear ß-catenin levels in 1322T lesions were associated with generally lower levels of Wnt target expression. However, expression of the Wnt target and stem cell marker Lgr5 was significantly higher in 1322T tumours than in Min tumours. Other stem cell markers (Musashi1, Bmi1 and the Wnt target Cd44) were also at higher levels in 1322T tumours. In addition, expression of the Bmp antagonist Gremlin1 was higher in 1322T tumours, together with lower Bmp2 and Bmp4 expression. CONCLUSIONS: The severe phenotype caused by truncation of Apc at codon 1322 is associated with an increased number of stem cells. Thus, a submaximal level of Wnt signalling favours the stem cell phenotype and this may promote tumorigenesis. A level of Wnt signalling exists that is too high for optimal tumour growth.


Subject(s)
Adenomatous Polyposis Coli/metabolism , Genes, APC , Receptors, G-Protein-Coupled/metabolism , Wnt Proteins/metabolism , Adenomatous Polyposis Coli/genetics , Animals , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Mice , Mice, Inbred C57BL , Microdissection/methods , Neoplasm Proteins/metabolism , Signal Transduction/physiology , Wnt Proteins/genetics
19.
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
20.
Gastroenterology ; 136(7): 2204-2213.e1-13, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19248780

ABSTRACT

BACKGROUND & AIMS: We previously demonstrated that the 2 APC mutations in human colorectal tumors are coselected, because tumorigenesis requires an optimal level of Wnt signaling. We and others subsequently showed that the truncated APC proteins in colorectal tumors usually retain a total of 1-2 beta-catenin binding/degradation repeats (20AARs); very few intestinal tumors have proteins with no 20AARs. The coselection of the "2 hits" at APC makes it difficult to undertake further mechanistic studies in this area in humans. In mice, however, second hits appear to vary with the strain or genetic background used. This suggested the possibility of creating suboptimal Apc genotypes in the mouse. METHODS: We have constructed a mouse, Apc(1322T), with a mutant protein retaining one 20AAR. After repeated backcrossing to the C57BL/6J background, we compared the 1322T animals with the widely used Min mouse in which the mutant Apc protein has zero 20AARs. RESULTS: In both mice, intestinal adenomas showed copy-neutral loss of heterozygosity, making them homozygous for the mutant Apc allele. 1322T animals had markedly more severe polyposis, with earlier-onset, larger, more numerous, and more severely dysplastic adenomas. 1322T tumors also had more marked Paneth cell differentiation and higher frequencies of crypt fission. Somewhat surprisingly, nuclear beta-catenin expression was lower in 1322T than Min tumors. CONCLUSIONS: We propose that the Apc(1322T) mutation produces submaximal beta-catenin levels that promote early tumor growth more effectively than the Apc(Min) mutation.


Subject(s)
Adenomatous Polyposis Coli/genetics , Cell Transformation, Neoplastic/genetics , Mutation/genetics , Signal Transduction/genetics , beta Catenin/metabolism , Adenomatous Polyposis Coli/pathology , Alleles , Animals , Blotting, Western , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Genes, APC , Genetic Predisposition to Disease , Loss of Heterozygosity , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Models, Genetic , Paneth Cells/cytology , Paneth Cells/physiology , Signal Transduction/physiology , Species Specificity , beta Catenin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL