Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
3.
Proc Natl Acad Sci U S A ; 115(46): E10869-E10878, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30381462

ABSTRACT

Mutations in the TP53 gene and microenvironmentally driven activation of hypoxia-inducible factor-1 (HIF-1) typically occur in later stages of tumorigenesis. An ongoing challenge is the identification of molecular determinants of advanced cancer pathogenesis to design alternative last-line therapeutic options. Here, we report that p53 mutants influence the tumor microenvironment by cooperating with HIF-1 to promote cancer progression. We demonstrate that in non-small cell lung cancer (NSCLC), p53 mutants exert a gain-of-function (GOF) effect on HIF-1, thus regulating a selective gene expression signature involved in protumorigenic functions. Hypoxia-mediated activation of HIF-1 leads to the formation of a p53 mutant/HIF-1 complex that physically binds the SWI/SNF chromatin remodeling complex, promoting expression of a selective subset of hypoxia-responsive genes. Depletion of p53 mutants impairs the HIF-mediated up-regulation of extracellular matrix (ECM) components, including type VIIa1 collagen and laminin-γ2, thus affecting tumorigenic potential of NSCLC cells in vitro and in mouse models in vivo. Analysis of surgically resected human NSCLC revealed that expression of this ECM gene signature was highly correlated with hypoxic tumors exclusively in patients carrying p53 mutations and was associated with poor prognosis. Our data reveal a GOF effect of p53 mutants in hypoxic tumors and suggest synergistic activities of p53 and HIF-1. These findings have important implications for cancer progression and might provide innovative last-line treatment options for advanced NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Hypoxia-Inducible Factor 1/genetics , Lung Neoplasms/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Hypoxia/genetics , Cell Line, Tumor , Extracellular Matrix , Genes, p53 , Heterografts , Humans , Hypoxia-Inducible Factor 1/metabolism , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Mutation , Transcriptional Activation , Tumor Microenvironment , Tumor Suppressor Protein p53/genetics
5.
BMC Cancer ; 17(1): 418, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28619042

ABSTRACT

BACKGROUND: A high rate of glycolysis leading to elevated lactate content has been linked to poor clinical outcomes in patients with head and neck and cervical cancer treated with radiotherapy. Although the biological explanation for this relationship between lactate and treatment response remains unclear, there is a continued interest in evaluating strategies of targeting metabolism to enhance the effectiveness of radiotherapy. The goal of this study was to investigate the effect of metabolic-targeting through HIF-1α inhibition and the associated changes in glycolysis, oxygen consumption and response on the efficacy of high-dose single-fraction radiotherapy (HD-SFRT). METHODS: HIF-1α wild-type and HIF-1α knockdown FaDu and ME180 xenograft tumors were grown in the hind leg of mice that were placed in an environmental chamber and exposed to different oxygen conditions (air-breathing and hypoxia). Ex vivo bioluminescence microscopy was used to measure lactate and ATP levels and the hypoxic fraction was measured using EF5 immunohistochemical staining. The oxygen consumption rate (OCR) in each cell line in response to in vitro hypoxia was measured using an extracellular flux analyzer. Tumor growth delay in vivo was measured following HD-SFRT irradiation of 20 Gy. RESULTS: Targeting HIF-1α reduced lactate content, and increased both oxygen consumption and hypoxic fraction in these tumors after exposure to short-term continuous hypoxia. Tumors with intact HIF-1α subjected to HD-SFRT immediately following hypoxia exposure were less responsive to treatment than tumors without functional HIF-1α, and tumors irradiated under air breathing conditions regardless of HIF-1α status. CONCLUSIONS: Blocking the HIF1 response during transient hypoxic stress increased hypoxia, reduced lactate levels and enhanced response to HD-SFRT. This strategy of combining hypofractionated radiotherapy with metabolic reprogramming to inhibit anaerobic metabolism may increase the efficacy of HD-SFRT through increased oxygen consumption and complementary killing of radiosensitive and hypoxic, radioresistant cells.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia/metabolism , Lactic Acid/metabolism , Neoplasms/metabolism , Oxygen Consumption , Adenosine Triphosphate/metabolism , Animals , Biomarkers , Cell Line, Tumor , Disease Models, Animal , Energy Metabolism/radiation effects , Female , Gene Knockdown Techniques , Glycolysis , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Neoplasms/pathology , Neoplasms/radiotherapy , Neovascularization, Pathologic , Radiation Dosage , Tumor Burden/radiation effects , Xenograft Model Antitumor Assays
6.
Exp Cell Res ; 356(2): 204-208, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28457987

ABSTRACT

Lorenz Poellinger was a leader in understanding the effects of altered microenvironmental conditions in tumor biology and in normal physiology. His work examining the effects of hypoxia and the HIF transcription factors has expanded our understanding of the role of the microenvironment in affecting the behaviour of both normal and malignant cells. Furthermore, his work provides a model for understanding the adaptive responses to other metabolic stress conditions. By investigating the molecular mechanisms responsible for the adaptive responses to metabolic stress in normal physiological situations, across pathological conditions, and in different model organisms, his work shows the power of combining data from different model systems and physiological contexts. In cancers, it has become clear that in order to evolve to become an aggressive malignant disease, tumor cells must acquire the capacity to tolerate a host of abnormal and stressful metabolic conditions. This metabolic stress can be thought of as a fire that tumor cells must douse with enough water to survive, and may offer opportunities to exploit smoldering vulnerabilities in order to eradicate malignant cells.


Subject(s)
Adaptation, Physiological/physiology , Cell Hypoxia/physiology , Cell Transformation, Neoplastic/metabolism , Neoplasms/metabolism , Tumor Microenvironment/physiology , Animals , Humans , Stress, Physiological/physiology
8.
Proc Natl Acad Sci U S A ; 114(2): 292-297, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28011762

ABSTRACT

Although the enzymatic activity of isocitrate dehydrogenase 1 (IDH1) was defined decades ago, its functions in vivo are not yet fully understood. Cytosolic IDH1 converts isocitrate to α-ketoglutarate (α-KG), a key metabolite regulating nitrogen homeostasis in catabolic pathways. It was thought that IDH1 might enhance lipid biosynthesis in liver or adipose tissue by generating NADPH, but we show here that lipid contents are relatively unchanged in both IDH1-null mouse liver and IDH1-deficient HepG2 cells generated using the CRISPR-Cas9 system. Instead, we found that IDH1 is critical for liver amino acid (AA) utilization. Body weights of IDH1-null mice fed a high-protein diet (HPD) were abnormally low. After prolonged fasting, IDH1-null mice exhibited decreased blood glucose but elevated blood alanine and glycine compared with wild-type (WT) controls. Similarly, in IDH1-deficient HepG2 cells, glucose consumption was increased, but alanine utilization and levels of intracellular α-KG and glutamate were reduced. In IDH1-deficient primary hepatocytes, gluconeogenesis as well as production of ammonia and urea were decreased. In IDH1-deficient whole livers, expression levels of genes involved in AA metabolism were reduced, whereas those involved in gluconeogenesis were up-regulated. Thus, IDH1 is critical for AA utilization in vivo and its deficiency attenuates gluconeogenesis primarily by impairing α-KG-dependent transamination of glucogenic AAs such as alanine.


Subject(s)
Amino Acids/metabolism , Isocitrate Dehydrogenase/deficiency , Liver/metabolism , Animals , Blood Glucose/metabolism , Cell Line, Tumor , Fasting/metabolism , Gluconeogenesis , Glucose/metabolism , Glutamic Acid/metabolism , Hep G2 Cells , Hepatocytes/metabolism , Humans , Isocitrate Dehydrogenase/metabolism , Ketoglutaric Acids/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Up-Regulation/physiology
9.
Proc Natl Acad Sci U S A ; 113(52): 15084-15089, 2016 12 27.
Article in English | MEDLINE | ID: mdl-27956631

ABSTRACT

Oncogenic isocitrate dehydrogenase (IDH)1 and IDH2 mutations at three hotspot arginine residues cause an enzymatic gain of function that leads to the production and accumulation of the metabolite 2-hydroxyglutarate (2HG), which contributes to the development of a number of malignancies. In the hematopoietic system, mutations in IDH1 at arginine (R) 132 and in IDH2 at R140 and R172 are commonly observed in acute myeloid leukemia, and elevated 2HG is observed in cells and serum. However, in angioimmunoblastic T-cell lymphoma (AITL), mutations are almost exclusively restricted to IDH2 R172, and levels of 2HG have not been comprehensively measured. In this study, we investigate the expression pattern of mutant IDH2 in the AITL tumor microenvironment and measure levels of 2HG in tissue and serum of AITL patients. We find that mutant IDH2 expression is restricted to the malignant T-cell component of AITL, and that 2HG is elevated in tumor tissue and serum of patients. We also investigate the differences between the three hotspot mutation sites in IDH1 and IDH2 using conditional knock-in mouse models. These studies show that in the lymphoid system, mutations in IDH2 at R172 produce high levels of 2HG compared with mutations at the other two sites and that lymphoid development is impaired in these animals. These data provide evidence that IDH2 R172 mutations may be the only variants present in AITL because of their capacity to produce significant amounts of the oncometabolite 2HG in the cell of origin of this disease.


Subject(s)
Glutarates/metabolism , Isocitrate Dehydrogenase/genetics , Lymphoma, T-Cell/immunology , Animals , Biomarkers, Tumor , Flow Cytometry , Gene Expression Regulation, Neoplastic , Genotype , Humans , Isocitrate Dehydrogenase/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Lymphocytes/metabolism , Lymphoma, T-Cell/metabolism , Mice , Mice, Knockout , Mutation
10.
Cancer Cell ; 30(5): 660-662, 2016 11 14.
Article in English | MEDLINE | ID: mdl-27846386

ABSTRACT

Although altered glucose metabolism is a well-studied feature of malignant cells, little is known about the direct metabolism of fructose. In this issue of Cancer Cell, Chen et al. report that AML cells consume fructose and use it to maintain viability, especially when glucose is scarce.


Subject(s)
Fructose/metabolism , Glucose/metabolism , Humans
11.
Cancer Cell ; 30(2): 337-348, 2016 08 08.
Article in English | MEDLINE | ID: mdl-27424808

ABSTRACT

Mutations in the isocitrate dehydrogenase-1 gene (IDH1) are common drivers of acute myeloid leukemia (AML) but their mechanism is not fully understood. It is thought that IDH1 mutants act by inhibiting TET2 to alter DNA methylation, but there are significant unexplained clinical differences between IDH1- and TET2-mutant diseases. We have discovered that mice expressing endogenous mutant IDH1 have reduced numbers of hematopoietic stem cells (HSCs), in contrast to Tet2 knockout (TET2-KO) mice. Mutant IDH1 downregulates the DNA damage (DD) sensor ATM by altering histone methylation, leading to impaired DNA repair, increased sensitivity to DD, and reduced HSC self-renewal, independent of TET2. ATM expression is also decreased in human IDH1-mutated AML. These findings may have implications for treatment of IDH-mutant leukemia.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/genetics , DNA Damage , DNA Repair , DNA-Binding Proteins/genetics , Hematopoietic Stem Cells/enzymology , Isocitrate Dehydrogenase/genetics , Proto-Oncogene Proteins/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , DNA-Binding Proteins/metabolism , Dioxygenases , Down-Regulation , Hematopoietic Stem Cells/cytology , Humans , Isocitrate Dehydrogenase/metabolism , Mice , Mutation , Proto-Oncogene Proteins/metabolism
12.
Cancer Res ; 76(16): 4708-19, 2016 08 15.
Article in English | MEDLINE | ID: mdl-27325644

ABSTRACT

Proliferating cancer cells are characterized by high rates of glycolysis, lactate production, and altered mitochondrial metabolism. This metabolic reprogramming provides important metabolites for proliferation of tumor cells, including glioblastoma. These biological processes, however, generate oxidative stress that must be balanced through detoxification of reactive oxygen species (ROS). Using an unbiased retroviral loss-of-function screen in nontransformed human astrocytes, we demonstrate that mitochondrial PTEN-induced kinase 1 (PINK1) is a regulator of the Warburg effect and negative regulator of glioblastoma growth. We report that loss of PINK1 contributes to the Warburg effect through ROS-dependent stabilization of hypoxia-inducible factor-1A and reduced pyruvate kinase muscle isozyme 2 activity, both key regulators of aerobic glycolysis. Mechanistically, PINK1 suppresses ROS and tumor growth through FOXO3a, a master regulator of oxidative stress and superoxide dismutase 2. These findings highlight the importance of PINK1 and ROS balance in normal and tumor cells. PINK1 loss was observed in a significant number of human brain tumors including glioblastoma (n > 900) and correlated with poor patient survival. PINK1 overexpression attenuates in vivo glioblastoma growth in orthotopic mouse xenograft models and a transgenic glioblastoma model in Drosophila Cancer Res; 76(16); 4708-19. ©2016 AACR.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Animals , Astrocytes/metabolism , Blotting, Western , Cell Proliferation , Drosophila , Glycolysis/physiology , Heterografts , Humans , Immunohistochemistry , Mice , Mice, Inbred NOD , Mice, SCID , Oxidative Stress/physiology
13.
Cell Metab ; 23(5): 767-9, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27166941

ABSTRACT

The impact of a tumor on distant organs is not well characterized but may be important for understanding immune interactions and the process of metastasis. Masri and colleagues (2016) now identify an interesting effect of lung tumor development on circadian regulation of liver metabolism.


Subject(s)
Circadian Clocks , Liver/metabolism , Lung Neoplasms/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Humans , Lung Neoplasms/pathology , Models, Biological , Mutation/genetics , Signal Transduction
14.
Proc Natl Acad Sci U S A ; 113(5): 1387-92, 2016 Feb 02.
Article in English | MEDLINE | ID: mdl-26787889

ABSTRACT

Gain-of-function mutations in isocitrate dehydrogenase 1 (IDH1) are key drivers of hematopoietic malignancies. Although these mutations are most commonly associated with myeloid diseases, they also occur in malignancies of the T-cell lineage. To investigate their role in these diseases and provide tractable disease models for further investigation, we analyzed the T-cell compartment in a conditional knock-in (KI) mouse model of mutant Idh1. We observed the development of a spontaneous T-cell acute lymphoblastic leukemia (T-ALL) in these animals. The disease was transplantable and maintained expression of mutant IDH1. Whole-exome sequencing revealed the presence of a spontaneous activating mutation in Notch1, one of the most common mutations in human T-ALL, suggesting Idh1 mutations may have the capacity to cooperate with Notch1 to drive T-ALL. To further investigate the Idh1 mutation as an oncogenic driver in the T-cell lineage, we crossed Idh1-KI mice with conditional Trp53 null mice, a well-characterized model of T-cell malignancy, and found that T-cell lymphomagenesis was accelerated in mice bearing both mutations. Because both IDH1 and p53 are known to affect cellular metabolism, we compared the requirements for glucose and glutamine in cells derived from these tumors and found that cells bearing the Idh1 mutation have an increased dependence on both glucose and glutamine. These data suggest that mutant IDH1 contributes to malignancy in the T-cell lineage and may alter the metabolic profile of malignant T cells.


Subject(s)
Isocitrate Dehydrogenase/genetics , Mutation , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Exome , Genes, p53 , Mice
15.
Proc Natl Acad Sci U S A ; 112(29): 9088-93, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26150517

ABSTRACT

Mitochondrial aldehyde dehydrogenase 2 (ALDH2) in the liver removes toxic aldehydes including acetaldehyde, an intermediate of ethanol metabolism. Nearly 40% of East Asians inherit an inactive ALDH2*2 variant, which has a lysine-for-glutamate substitution at position 487 (E487K), and show a characteristic alcohol flush reaction after drinking and a higher risk for gastrointestinal cancers. Here we report the characterization of knockin mice in which the ALDH2(E487K) mutation is inserted into the endogenous murine Aldh2 locus. These mutants recapitulate essentially all human phenotypes including impaired clearance of acetaldehyde, increased sensitivity to acute or chronic alcohol-induced toxicity, and reduced ALDH2 expression due to a dominant-negative effect of the mutation. When treated with a chemical carcinogen, these mutants exhibit increased DNA damage response in hepatocytes, pronounced liver injury, and accelerated development of hepatocellular carcinoma (HCC). Importantly, ALDH2 protein levels are also significantly lower in patient HCC than in peritumor or normal liver tissues. Our results reveal that ALDH2 functions as a tumor suppressor by maintaining genomic stability in the liver, and the common human ALDH2 variant would present a significant risk factor for hepatocarcinogenesis. Our study suggests that the ALDH2*2 allele-alcohol interaction may be an even greater human public health hazard than previously appreciated.


Subject(s)
Aldehyde Dehydrogenase/genetics , Carcinoma, Hepatocellular/enzymology , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Mutation/genetics , Alcoholic Intoxication/enzymology , Alcoholic Intoxication/pathology , Aldehyde Dehydrogenase, Mitochondrial , Amino Acid Substitution , Animals , Base Sequence , Carcinogenesis/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Ethanol/adverse effects , Gene Knock-In Techniques , Genotyping Techniques , Hepatocytes/enzymology , Hepatocytes/pathology , Humans , Hyperpigmentation/pathology , Immunohistochemistry , Liver/enzymology , Liver/pathology , Liver Neoplasms/pathology , Mice, Inbred C57BL , Mutant Proteins/metabolism , Polymorphism, Genetic , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Skin/pathology , Survival Analysis
16.
Cancer J ; 21(2): 56-61, 2015.
Article in English | MEDLINE | ID: mdl-25815844

ABSTRACT

The Warburg effect was first described by Otto Warburg in the 1920s and describes the preferential conversion of glucose to lactate as opposed to its metabolism through the citric acid cycle to fuel oxidative phosphorylation in the mitochondria, even in the presence of oxygen. This phenotype is a common feature of malignant cells and is also observed in some highly proliferative normal tissues. The selective advantage provided by this phenotype is not entirely clear. Adopting this metabolic state may allow tumor cells to balance their need for ATP, biosynthetic precursor molecules, and reducing power in order to respond to growth and proliferation signals and may provide a selective advantage in the hypoxic and acidic microenvironments that are often a feature of solid tumors. Oncogenic signaling pathways and responses to the local microenvironment combine to produce this metabolic phenotype via a number of molecular mechanisms. A better understanding of these mechanisms in both tumor and normal tissues and a more complete understanding of how the Warburg effect interacts with the rest of the tumor metabolic network should provide opportunities for novel clinical intervention.


Subject(s)
Glucose/metabolism , Mitochondria/metabolism , Neoplasms/metabolism , Oxidative Phosphorylation , Phenotype , Animals , Biological Transport , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Glycolysis , Humans , Neoplasms/genetics , Signal Transduction
17.
Proc Natl Acad Sci U S A ; 112(9): 2829-34, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25730874

ABSTRACT

Enchondromas are benign cartilage tumors and precursors to malignant chondrosarcomas. Somatic mutations in the isocitrate dehydrogenase genes (IDH1 and IDH2) are present in the majority of these tumor types. How these mutations cause enchondromas is unclear. Here, we identified the spectrum of IDH mutations in human enchondromas and chondrosarcomas and studied their effects in mice. A broad range of mutations was identified, including the previously unreported IDH1-R132Q mutation. These mutations harbored enzymatic activity to catalyze α-ketoglutarate to d-2-hydroxyglutarate (d-2HG). Mice expressing Idh1-R132Q in one allele in cells expressing type 2 collagen showed a disordered growth plate, with persistence of type X-expressing chondrocytes. Chondrocyte cell cultures from these animals or controls showed that there was an increase in proliferation and expression of genes characteristic of hypertrophic chondrocytes with expression of Idh1-R132Q or 2HG treatment. Col2a1-Cre;Idh1-R132Q mutant knock-in mice (mutant allele expressed in chondrocytes) did not survive after the neonatal stage. Col2a1-Cre/ERT2;Idh1-R132 mutant conditional knock-in mice, in which Cre was induced by tamoxifen after weaning, developed multiple enchondroma-like lesions. Taken together, these data show that mutant IDH or d-2HG causes persistence of chondrocytes, giving rise to rests of growth-plate cells that persist in the bone as enchondromas.


Subject(s)
Chondrocytes , Enchondromatosis , Gene Expression Regulation, Enzymologic , Isocitrate Dehydrogenase , Mutation, Missense , Amino Acid Substitution , Animals , Chondrocytes/enzymology , Chondrocytes/pathology , Collagen Type II/biosynthesis , Collagen Type II/genetics , Enchondromatosis/enzymology , Enchondromatosis/genetics , Enchondromatosis/pathology , Glutarates/adverse effects , Glutarates/pharmacology , Humans , Isocitrate Dehydrogenase/biosynthesis , Isocitrate Dehydrogenase/genetics , Mice , Mice, Mutant Strains
18.
Cancer Discov ; 3(7): 730-41, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23796461

ABSTRACT

Heterozygous mutations in catalytic arginine residues of isocitrate dehydrogenases 1 and 2 (IDH1 and IDH2) are common in glioma, acute myeloid leukemia, chondrosarcoma, cholangiocarcinoma, and angioimmunoblastic T-cell lymphoma. The mutant enzymes acquire a neomorphic activity that converts α-ketoglutarate (α-KG) to D-2-hydroxyglutarate (D2HG), a rare metabolite. In cells and tissues expressing mutant IDH, D2HG concentrations are highly elevated. D2HG may act as an "oncometabolite" by inhibiting a class of α-KG-dependent enzymes involved in epigenetic regulation, collagen synthesis, and cell signaling. Knock-in mouse models of IDH1 mutations have shed light on these mechanisms and will provide valuable animal models for further investigation.


Subject(s)
Glutarates/metabolism , Isocitrate Dehydrogenase/biosynthesis , Neoplasms/genetics , Animals , Arginine/genetics , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Gene Knock-In Techniques , Humans , Isocitrate Dehydrogenase/genetics , Mice , Mutation , Neoplasms/drug therapy , Neoplasms/pathology
19.
Genes Dev ; 26(18): 2038-49, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22925884

ABSTRACT

Isocitrate dehydrogenase-1 (IDH1) R132 mutations occur in glioma, but their physiological significance is unknown. Here we describe the generation and characterization of brain-specific Idh1 R132H conditional knock-in (KI) mice. Idh1 mutation results in hemorrhage and perinatal lethality. Surprisingly, intracellular reactive oxygen species (ROS) are attenuated in Idh1-KI brain cells despite an apparent increase in the NADP(+)/NADPH ratio. Idh1-KI cells also show high levels of D-2-hydroxyglutarate (D2HG) that are associated with inhibited prolyl-hydroxylation of hypoxia-inducible transcription factor-1α (Hif1α) and up-regulated Hif1α target gene transcription. Intriguingly, D2HG also blocks prolyl-hydroxylation of collagen, causing a defect in collagen protein maturation. An endoplasmic reticulum (ER) stress response induced by the accumulation of immature collagens may account for the embryonic lethality of these mutants. Importantly, D2HG-mediated impairment of collagen maturation also led to basement membrane (BM) aberrations that could play a part in glioma progression. Our study presents strong in vivo evidence that the D2HG produced by the mutant Idh1 enzyme is responsible for the above effects.


Subject(s)
Basement Membrane/pathology , Collagen/metabolism , Glutarates/metabolism , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Animals , Basement Membrane/metabolism , Brain/cytology , Brain/pathology , Gene Knock-In Techniques , Genotype , Glioma/pathology , Mice , Mutation , Protein Stability , Reactive Oxygen Species/metabolism , Stress, Physiological
20.
Blood ; 120(7): 1466-9, 2012 Aug 16.
Article in English | MEDLINE | ID: mdl-22760778

ABSTRACT

Inactivating mutations of the Ten-Eleven Translocation 2 (TET2) gene were first identified in myeloid malignancies and more recently in peripheral T-cell lymphomas (PTCLs). In the present study, we investigated the presence of TET2 coding sequence mutations and their clinical relevance in a large cohort of 190 PTCL patients. TET2 mutations were identified in 40 of 86 (47%) cases of angioimmunoblastic T-cell lymphoma (AITL) and in 22 of 58 (38%) cases of peripheral T-cell lymphoma, not otherwise specified (PTCL-NOS), but were absent in all other PTCL entities, with the exception of 2 of 10 cases of enteropathy-associated T-cell lymphoma. Among PTCL-NOS, a heterogeneous group of lymphoma-comprising cases likely to derive from Th follicular (T(FH)) cells similarly to AITL, TET2 mutations were more frequent when PTCL-NOS expressed T(FH) markers and/or had features reminiscent of AITL (58% vs 24%, P = .01). In the AITL and PTCL-NOS subgroups, TET2 mutations were associated with advanced-stage disease, thrombocytopenia, high International Prognostic Index scores, and a shorter progression-free survival.


Subject(s)
DNA-Binding Proteins/genetics , Lymphoma, T-Cell, Peripheral/genetics , Lymphoma, T-Cell, Peripheral/immunology , Mutation/genetics , Proto-Oncogene Proteins/genetics , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/pathology , Aged , Dioxygenases , Female , Humans , Male , Recurrence
SELECTION OF CITATIONS
SEARCH DETAIL
...