Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Article in English | MEDLINE | ID: mdl-38191174

ABSTRACT

Isocitrate dehydrogenase 1 and 2 (IDH1 and IDH2) are metabolic enzymes that interconvert isocitrate and 2-oxoglutarate (2OG). Gain-of-function mutations in IDH1 and IDH2 occur in a number of cancers, including acute myeloid leukemia, glioma, cholangiocarcinoma, and chondrosarcoma. These mutations cripple the wild-type activity of IDH and cause the enzymes to catalyze a partial reverse reaction in which 2OG is reduced but not carboxylated, resulting in production of the (R)-enantiomer of 2-hydroxyglutarate ((R)-2HG). (R)-2HG accumulation in IDH-mutant tumors results in profound dysregulation of cellular metabolism. The most well-characterized oncogenic effects of (R)-2HG involve the dysregulation of 2OG-dependent epigenetic tumor-suppressor enzymes. However, (R)-2HG has many other effects in IDH-mutant cells, some that promote transformation and others that induce metabolic dependencies. Herein, we review how cancer-associated IDH mutations impact epigenetic regulation and cellular metabolism and discuss how these effects can potentially be leveraged to therapeutically target IDH-mutant tumors.


Subject(s)
Isocitrate Dehydrogenase , Mutation , Neoplasms , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Humans , Neoplasms/genetics , Epigenesis, Genetic , Glutarates/metabolism , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Animals
2.
Blood ; 143(11): 1032-1044, 2024 Mar 14.
Article in English | MEDLINE | ID: mdl-38096369

ABSTRACT

ABSTRACT: Extreme disease phenotypes can provide key insights into the pathophysiology of common conditions, but studying such cases is challenging due to their rarity and the limited statistical power of existing methods. Herein, we used a novel approach to pathway-based mutational burden testing, the rare variant trend test (RVTT), to investigate genetic risk factors for an extreme form of sepsis-induced coagulopathy, infectious purpura fulminans (PF). In addition to prospective patient sample collection, we electronically screened over 10.4 million medical records from 4 large hospital systems and identified historical cases of PF for which archived specimens were available to perform germline whole-exome sequencing. We found a significantly increased burden of low-frequency, putatively function-altering variants in the complement system in patients with PF compared with unselected patients with sepsis (P = .01). A multivariable logistic regression analysis found that the number of complement system variants per patient was independently associated with PF after controlling for age, sex, and disease acuity (P = .01). Functional characterization of PF-associated variants in the immunomodulatory complement receptors CR3 and CR4 revealed that they result in partial or complete loss of anti-inflammatory CR3 function and/or gain of proinflammatory CR4 function. Taken together, these findings suggest that inherited defects in CR3 and CR4 predispose to the maladaptive hyperinflammation that characterizes severe sepsis with coagulopathy.


Subject(s)
Purpura Fulminans , Sepsis , Humans , Purpura Fulminans/genetics , Prospective Studies , Receptors, Complement
3.
Cancer Discov ; 13(6): 1478-1497, 2023 06 02.
Article in English | MEDLINE | ID: mdl-36847506

ABSTRACT

Oncogenic mutations in isocitrate dehydrogenase 1 (IDH1) and IDH2 occur in a wide range of cancers, including acute myeloid leukemia (AML) and glioma. Mutant IDH enzymes convert 2-oxoglutarate (2OG) to (R)-2-hydroxyglutarate [(R)-2HG], an oncometabolite that is hypothesized to promote cellular transformation by dysregulating 2OG-dependent enzymes. The only (R)-2HG target that has been convincingly shown to contribute to transformation by mutant IDH is the myeloid tumor suppressor TET2. However, there is ample evidence to suggest that (R)-2HG has other functionally relevant targets in IDH-mutant cancers. Here, we show that (R)-2HG inhibits KDM5 histone lysine demethylases and that this inhibition contributes to cellular transformation in IDH-mutant AML and IDH-mutant glioma. These studies provide the first evidence of a functional link between dysregulation of histone lysine methylation and transformation in IDH-mutant cancers. SIGNIFICANCE: Mutant IDH is known to induce histone hypermethylation. However, it is not known if this hypermethylation is functionally significant or is a bystander effect of (R)-2HG accumulation in IDH-mutant cells. Here, we provide evidence that KDM5 inhibition by (R)-2HG contributes to mutant IDH-mediated transformation in AML and glioma. This article is highlighted in the In This Issue feature, p. 1275.


Subject(s)
Glioma , Leukemia, Myeloid, Acute , Humans , Histones/metabolism , Histone Demethylases/genetics , Mutation , Glutarates , Cell Transformation, Neoplastic/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Glioma/genetics , DNA Methylation , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism
4.
NPJ Precis Oncol ; 6(1): 61, 2022 Sep 02.
Article in English | MEDLINE | ID: mdl-36056177

ABSTRACT

The mutant IDH1 inhibitor ivosidenib improves outcomes for patients with IDH1-mutated cholangiocarcinoma, but resistance inevitably develops. Mechanisms of resistance and strategies to overcome resistance are poorly understood. Here we describe two patients with IDH1 R132C-mutated metastatic cholangiocarcinoma who developed acquired resistance to ivosidenib. After disease progression, one patient developed an oncogenic IDH2 mutation, and the second patient acquired a secondary IDH1 D279N mutation. To characterize the putative IDH1 resistance mutation, cells expressing the double-mutant were generated. In vitro, IDH1 R132H/D279N produces (R)-2HG less efficiently than IDH1 R132H. However, its binding to ivosidenib is impaired and it retains the ability to produce (R)-2HG and promote cellular transformation in the presence of ivosidenib. The irreversible mutant IDH1 inhibitor LY3410738 binds and blocks (R)-2HG production and cellular transformation by IDH1 R132H/D279N. These resistance mechanisms suggest that IDH1-mutated cholangiocarcinomas remain dependent on (R)-2HG even after prolonged ivosidenib treatment. Sequential mutant IDH inhibitor therapy should be explored as a strategy to overcome acquired resistance to mutant IDH inhibitors.

5.
Cancer Cell ; 40(9): 939-956.e16, 2022 09 12.
Article in English | MEDLINE | ID: mdl-35985343

ABSTRACT

Mutations affecting isocitrate dehydrogenase (IDH) enzymes are prevalent in glioma, leukemia, and other cancers. Although mutant IDH inhibitors are effective against leukemia, they seem to be less active in aggressive glioma, underscoring the need for alternative treatment strategies. Through a chemical synthetic lethality screen, we discovered that IDH1-mutant glioma cells are hypersensitive to drugs targeting enzymes in the de novo pyrimidine nucleotide synthesis pathway, including dihydroorotate dehydrogenase (DHODH). We developed a genetically engineered mouse model of mutant IDH1-driven astrocytoma and used it and multiple patient-derived models to show that the brain-penetrant DHODH inhibitor BAY 2402234 displays monotherapy efficacy against IDH-mutant gliomas. Mechanistically, this reflects an obligate dependence of glioma cells on the de novo pyrimidine synthesis pathway and mutant IDH's ability to sensitize to DNA damage upon nucleotide pool imbalance. Our work outlines a tumor-selective, biomarker-guided therapeutic strategy that is poised for clinical translation.


Subject(s)
Brain Neoplasms , Glioma , Leukemia , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/genetics , Enzyme Inhibitors/therapeutic use , Glioma/drug therapy , Glioma/genetics , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mice , Mutation , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Salicylanilides , Triazoles
6.
Nat Commun ; 13(1): 4998, 2022 08 25.
Article in English | MEDLINE | ID: mdl-36008402

ABSTRACT

Some small cell lung cancers (SCLCs) are highly sensitive to inhibitors of the histone demethylase LSD1. LSD1 inhibitors are thought to induce their anti-proliferative effects by blocking neuroendocrine differentiation, but the mechanisms by which LSD1 controls the SCLC neuroendocrine phenotype are not well understood. To identify genes required for LSD1 inhibitor sensitivity in SCLC, we performed a positive selection genome-wide CRISPR/Cas9 loss of function screen and found that ZFP36L1, an mRNA-binding protein that destabilizes mRNAs, is required for LSD1 inhibitor sensitivity. LSD1 binds and represses ZFP36L1 and upon LSD1 inhibition, ZFP36L1 expression is restored, which is sufficient to block the SCLC neuroendocrine differentiation phenotype and induce a non-neuroendocrine "inflammatory" phenotype. Mechanistically, ZFP36L1 binds and destabilizes SOX2 and INSM1 mRNAs, two transcription factors that are required for SCLC neuroendocrine differentiation. This work identifies ZFP36L1 as an LSD1 target gene that controls the SCLC neuroendocrine phenotype and demonstrates that modulating mRNA stability of lineage transcription factors controls neuroendocrine to non-neuroendocrine plasticity.


Subject(s)
Butyrate Response Factor 1/metabolism , Lung Neoplasms , Small Cell Lung Carcinoma , Histone Demethylases/genetics , Histone Demethylases/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , RNA-Binding Proteins/genetics , Repressor Proteins/metabolism , Small Cell Lung Carcinoma/metabolism , Small Cell Lung Carcinoma/pathology , Transcription Factors/metabolism
7.
Surg Neurol Int ; 11: 403, 2020.
Article in English | MEDLINE | ID: mdl-33365166

ABSTRACT

BACKGROUND: Alterations in normal coagulation and hemostasis are critical issues that require special attention in the neurosurgical patient. These disorders pose unique challenges in the management of these patients who often have concurrent acute ischemic and hemorrhagic injuries. Although neurosurgical intervention in such cases may be unavoidable and potentially life-saving, these patients should be closely observed after instrumentation. CASE DESCRIPTION: A 57-year-old male with liver cirrhosis secondary to amyloid light-chain amyloidosis was admitted to the intensive care unit for the management of delayed hydrocephalus. An external ventricular drain (EVD) was placed for the treatment and monitoring of hydrocephalus. Five days after EVD placement, a head computed tomography scan revealed a tract hemorrhage. However, on repeated imaging, the size of the hemorrhage continued to increase despite aggressive blood pressure control and several doses of phytonadione. Extensive coagulopathy workup was remarkable for low factor VII levels. In that setting, recombinant activated factor VII was administered to normalize factor VII levels, and the tract hemorrhage stabilized. CONCLUSION: To the best of our knowledge, this is the first case of spontaneous hemorrhage after EVD placement in the setting of liver cirrhosis-associated factor VII deficiency. Our case highlights the importance of identifying coagulation disorders in neurosurgical patients at high risk for coagulopathy and closely monitoring them postoperatively.

8.
Nat Rev Cancer ; 20(12): 710-726, 2020 12.
Article in English | MEDLINE | ID: mdl-33087883

ABSTRACT

2-Oxoglutarate-dependent dioxygenases (2OGDDs) are a superfamily of enzymes that play diverse roles in many biological processes, including regulation of hypoxia-inducible factor-mediated adaptation to hypoxia, extracellular matrix formation, epigenetic regulation of gene transcription and the reprogramming of cellular metabolism. 2OGDDs all require oxygen, reduced iron and 2-oxoglutarate (also known as α-ketoglutarate) to function, although their affinities for each of these co-substrates, and hence their sensitivity to depletion of specific co-substrates, varies widely. Numerous 2OGDDs are recurrently dysregulated in cancer. Moreover, cancer-specific metabolic changes, such as those that occur subsequent to mutations in the genes encoding succinate dehydrogenase, fumarate hydratase or isocitrate dehydrogenase, can dysregulate specific 2OGDDs. This latter observation suggests that the role of 2OGDDs in cancer extends beyond cancers that harbour mutations in the genes encoding members of the 2OGDD superfamily. Herein, we review the regulation of 2OGDDs in normal cells and how that regulation is corrupted in cancer.


Subject(s)
Dioxygenases/physiology , Ketoglutaric Acids/metabolism , Neoplasms/metabolism , Cell Hypoxia , Citric Acid Cycle , DNA Damage , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , Hypoxia-Inducible Factor-Proline Dioxygenases/physiology , Neoplasms/drug therapy , Neoplasms/etiology , Reactive Oxygen Species/metabolism
9.
Leukemia ; 33(10): 2403-2415, 2019 10.
Article in English | MEDLINE | ID: mdl-30940908

ABSTRACT

Acute myeloid leukemia (AML) is a devastating disease, with the majority of patients dying within a year of diagnosis. For patients with relapsed/refractory AML, the prognosis is particularly poor with currently available treatments. Although genetically heterogeneous, AML subtypes share a common differentiation arrest at hematopoietic progenitor stages. Overcoming this differentiation arrest has the potential to improve the long-term survival of patients, as is the case in acute promyelocytic leukemia (APL), which is characterized by a chromosomal translocation involving the retinoic acid receptor alpha gene. Treatment of APL with all-trans retinoic acid (ATRA) induces terminal differentiation and apoptosis of leukemic promyelocytes, resulting in cure rates of over 80%. Unfortunately, similarly efficacious differentiation therapies have, to date, been lacking outside of APL. Inhibition of dihydroorotate dehydrogenase (DHODH), a key enzyme in the de novo pyrimidine synthesis pathway, was recently reported to induce differentiation of diverse AML subtypes. In this report we describe the discovery and characterization of BAY 2402234 - a novel, potent, selective and orally bioavailable DHODH inhibitor that shows monotherapy efficacy and differentiation induction across multiple AML subtypes. Herein, we present the preclinical data that led to initiation of a phase I evaluation of this inhibitor in myeloid malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Enzyme Inhibitors/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Oxidoreductases Acting on CH-CH Group Donors/antagonists & inhibitors , Animals , Apoptosis/drug effects , Cell Line, Tumor , Dihydroorotate Dehydrogenase , Female , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/metabolism , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Pyrimidines/metabolism , THP-1 Cells , Translocation, Genetic/drug effects
10.
J Med Genet ; 56(1): 39-42, 2019 01.
Article in English | MEDLINE | ID: mdl-30287596

ABSTRACT

BACKGROUND: Immune checkpoint inhibitor therapy is a modern breakthrough in medical oncology, but it can precipitate inflammatory and autoimmune adverse effects. Among the most serious of these toxicities is haemophagocytic lymphohistiocytosis (HLH), a life-threatening disorder of unbridled immune activation that results in injury to multiple organ systems. OBJECTIVE: Description of a case of pembrolizumab-associated HLH in a patient with a proposed underlying genetic risk factor for its occurrence. METHODS AND RESULTS: We describe a patient with aggressive metastatic breast cancer who developed HLH while undergoing experimental treatment with pembrolizumab, resulting in critical illness and multiorgan system failure. Pembrolizumab discontinuation and high-dose corticosteroids were effective in managing HLH. Subsequent next-generation sequencing of 15 genes associated with HLH revealed a germline polymorphism in perforin-1 (PRF1), PRFA91V, that may have predisposed the patient to develop HLH. The patient has had no evidence of malignancy for 2 years following recovery despite receiving no further cancer-directed treatment. CONCLUSIONS: HLH is a rare but serious complication of immune checkpoint blockade. Patients with underlying hypomorphic alleles in PRF1 may be predisposed to develop this toxicity. Further studies are necessary to confirm a possible link between perforin gene mutations and immune checkpoint blockade-associated HLH.


Subject(s)
Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Breast Neoplasms/complications , Breast Neoplasms/genetics , Lymphohistiocytosis, Hemophagocytic/etiology , Perforin/genetics , Polymorphism, Single Nucleotide , Alleles , Amino Acid Substitution , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Disease Susceptibility , Female , Humans , Immunosuppressive Agents/therapeutic use , Lymphohistiocytosis, Hemophagocytic/diagnosis , Lymphohistiocytosis, Hemophagocytic/drug therapy , Middle Aged , Treatment Outcome
12.
Proc Natl Acad Sci U S A ; 115(16): E3741-E3748, 2018 04 17.
Article in English | MEDLINE | ID: mdl-29610306

ABSTRACT

Inactivation of the retinoblastoma gene (RB1) product, pRB, is common in many human cancers. Targeting downstream effectors of pRB that are central to tumorigenesis is a promising strategy to block the growth of tumors harboring loss-of-function RB1 mutations. One such effector is retinoblastoma-binding protein 2 (RBP2, also called JARID1A or KDM5A), which encodes an H3K4 demethylase. Binding of pRB to RBP2 has been linked to the ability of pRB to promote senescence and differentiation. Importantly, genetic ablation of RBP2 is sufficient to phenocopy pRB's ability to induce these cellular changes in cell culture experiments. Moreover, germline Rbp2 deletion significantly impedes tumorigenesis in Rb1+/- mice. The value of RBP2 as a therapeutic target in cancer, however, hinges on whether loss of RBP2 could block the growth of established tumors as opposed to simply delaying their onset. Here we show that conditional, systemic ablation of RBP2 in tumor-bearing Rb1+/- mice is sufficient to slow tumor growth and significantly extend survival without causing obvious toxicity to the host. These findings show that established Rb1-null tumors require RBP2 for growth and further credential RBP2 as a therapeutic target in human cancers driven by RB1 inactivation.


Subject(s)
DNA-Binding Proteins/physiology , Histone Code/physiology , Jumonji Domain-Containing Histone Demethylases/physiology , Molecular Targeted Therapy/methods , Neoplasm Proteins/physiology , Pituitary Neoplasms/enzymology , Retinoblastoma Protein/deficiency , Thyroid Neoplasms/enzymology , Alleles , Animals , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Echocardiography , Enzyme Activation/drug effects , Fibroblasts , Genes, Retinoblastoma , Heart Septal Defects/genetics , Histone Code/drug effects , Integrases/drug effects , Jumonji Domain-Containing Histone Demethylases/deficiency , Jumonji Domain-Containing Histone Demethylases/genetics , Mice , Mice, Inbred C57BL , Pituitary Neoplasms/genetics , Pituitary Neoplasms/therapy , Recombinant Fusion Proteins/biosynthesis , Recombinant Fusion Proteins/genetics , Tamoxifen/pharmacology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/therapy , Transgenes/drug effects
13.
Hematol Oncol Clin North Am ; 31(4): 565-576, 2017 08.
Article in English | MEDLINE | ID: mdl-28673388

ABSTRACT

Clonal myeloid disorders are characterized by genetic alterations that activate cytokine signaling pathways and stimulate cell proliferation. These activated signaling pathways have been extensively studied as potential therapeutic targets, and tyrosine kinase inhibitors have indeed had extraordinary success in treating BCR/ABL-positive chronic myeloiud leukemia. However, although inhibitors of other activated kinases have been developed that perform well in preclinical studies, the therapeutic efficacy of these drugs in patients has been unimpressive. This article discusses potential reasons for these discordant results and outlines recent scientific advances that are informing future efforts to target activated kinases in clonal myeloid disorders.


Subject(s)
Antineoplastic Agents/therapeutic use , Molecular Targeted Therapy , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/metabolism , Signal Transduction/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor , Disease Progression , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Humans , Mutation , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Oncogenes , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use
14.
Nature ; 543(7647): 634-635, 2017 03 29.
Article in English | MEDLINE | ID: mdl-28358070
15.
Cancer Discov ; 6(4): 368-81, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26951227

ABSTRACT

UNLABELLED: Somatic mutations in calreticulin (CALR) are present in approximately 40% of patients with myeloproliferative neoplasms (MPN), but the mechanism by which mutant CALR is oncogenic remains unclear. Here, we demonstrate that expression of mutant CALR alone is sufficient to engender MPN in mice and recapitulates the disease phenotype of patients with CALR-mutant MPN. We further show that the thrombopoietin receptor MPL is required for mutant CALR-driven transformation through JAK-STAT pathway activation, thus rendering mutant CALR-transformed hematopoietic cells sensitive to JAK2 inhibition. Finally, we demonstrate that the oncogenicity of mutant CALR is dependent on the positive electrostatic charge of the C-terminus of the mutant protein, which is necessary for physical interaction between mutant CALR and MPL. Together, our findings elucidate a novel paradigm of cancer pathogenesis and reveal how CALR mutations induce MPN. SIGNIFICANCE: The mechanism by which CALR mutations induce MPN remains unknown. In this report, we show that the positive charge of the CALR mutant C-terminus is necessary to transform hematopoietic cells by enabling binding between mutant CALR and the thrombopoietin receptor MPL.


Subject(s)
Calreticulin/genetics , Cell Transformation, Neoplastic/genetics , Mutation , Protein Interaction Domains and Motifs/genetics , Receptors, Thrombopoietin/genetics , Animals , Base Sequence , Bone Marrow Transplantation , Calreticulin/chemistry , Calreticulin/metabolism , Cell Line , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Female , Frameshift Mutation , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Mice , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Phenotype , Protein Binding , Protein Kinase Inhibitors/pharmacology , Receptors, Thrombopoietin/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Structure Collapse
16.
Genes Dev ; 27(8): 836-52, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23630074

ABSTRACT

Mutations in metabolic enzymes, including isocitrate dehydrogenase 1 (IDH1) and IDH2, in cancer strongly implicate altered metabolism in tumorigenesis. IDH1 and IDH2 catalyze the interconversion of isocitrate and 2-oxoglutarate (2OG). 2OG is a TCA cycle intermediate and an essential cofactor for many enzymes, including JmjC domain-containing histone demethylases, TET 5-methylcytosine hydroxylases, and EglN prolyl-4-hydroxylases. Cancer-associated IDH mutations alter the enzymes such that they reduce 2OG to the structurally similar metabolite (R)-2-hydroxyglutarate [(R)-2HG]. Here we review what is known about the molecular mechanisms of transformation by mutant IDH and discuss their implications for the development of targeted therapies to treat IDH mutant malignancies.


Subject(s)
Glutarates/metabolism , Isocitrate Dehydrogenase/genetics , Isocitrate Dehydrogenase/metabolism , Mutation , Neoplasms/enzymology , Neoplasms/genetics , Animals , Catalysis , Humans , Neoplasms/therapy , Oncogenes/genetics
17.
Science ; 339(6127): 1621-5, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23393090

ABSTRACT

Mutations in IDH1 and IDH2, the genes coding for isocitrate dehydrogenases 1 and 2, are common in several human cancers, including leukemias, and result in overproduction of the (R)-enantiomer of 2-hydroxyglutarate [(R)-2HG]. Elucidation of the role of IDH mutations and (R)-2HG in leukemogenesis has been hampered by a lack of appropriate cell-based models. Here, we show that a canonical IDH1 mutant, IDH1 R132H, promotes cytokine independence and blocks differentiation in hematopoietic cells. These effects can be recapitulated by (R)-2HG, but not (S)-2HG, despite the fact that (S)-2HG more potently inhibits enzymes, such as the 5'-methylcytosine hydroxylase TET2, that have previously been linked to the pathogenesis of IDH mutant tumors. We provide evidence that this paradox relates to the ability of (S)-2HG, but not (R)-2HG, to inhibit the EglN prolyl hydroxylases. Additionally, we show that transformation by (R)-2HG is reversible.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Glutarates/metabolism , Hematopoiesis , Isocitrate Dehydrogenase/metabolism , Leukemia/enzymology , Procollagen-Proline Dioxygenase/antagonists & inhibitors , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Humans , Isocitrate Dehydrogenase/genetics , Leukemia/genetics , Models, Biological
19.
Am J Med ; 117(5): 353-6, 2004 Sep 01.
Article in English | MEDLINE | ID: mdl-15336585

ABSTRACT

PRESENTING FEATURES: A 53-year-old man who had human immunodeficiency virus (HIV) presented to the Johns Hopkins Hospital with a 3-month history of increasing dysphagia, cough, dyspnea, chest pain, and an episode of syncope. His past medical history was notable for oral and presumptive esophageal candidiasis that was treated with fluconazole 6 months prior to presentation. Three months prior to presentation, he discontinued his medications, and his symptoms of dysphagia recurred. During that time he developed intermittent fevers and chills, progressively worsening dyspnea on exertion, and a cough productive of white sputum. He also reported a 40-lb weight loss over the past 3 months. On the day prior to presentation, he had chest pain and shortness of breath followed by weakness, dizziness, and a brief syncopal episode. He denied orthopnea, paroxysmal nocturnal dyspnea, lower extremity edema, jaundice, hemoptysis, hematemesis, melena, hematochezia, or diarrhea. There was no history of alcohol use, and he stopped smoking tobacco approximately 1 month previously. He smoked cocaine but denied injection drug use. The patient had never been on antiretroviral therapy and had never had his CD4 count or viral load measured. On physical examination, the patient was a thin, cachectic man who appeared older than his stated age. His vital signs were notable for blood pressure of 102/69 mm Hg, resting tachycardia of 102 beats per minute, resting oxygen saturation of 92% on room air, normal resting respiratory rate, and a temperature of 38.1 degrees C. His oropharynx was clear, with no signs of thrush or mucosal ulcers. His pulmonary examination was notable for diminished breath sounds in the lower lung fields bilaterally. Cardiac, abdominal, and neurologic examinations were normal. His skin was intact, with no visible petechiae, rashes, nodules, or ulcers. Laboratory studies showed a total white blood cell count of 3.2 x 10(3)/microL, with a total lymphocyte count of 330/microL, hematocrit of 30.2%, a serum sodium level of 129 mEq/L, and a serum lactate dehydrogenase level of 219 IU/L. The patient had an absolute CD4 count of 8 cells/mm3 and a HIV viral load of 86,457 copies/mL. His arterial blood gas on room air had a pH of 7.51, a PCO2 of 33 mm Hg, and a PO2 of 55 mm Hg. Electrocardiogram and serial serum cardiac enzymes were normal. A chest radiograph showed bilateral upper lobe patchy infiltrates with left upper lobe consolidation. Computed tomographic (CT) scan of the chest with contrast showed bilateral ground glass infiltrates with focal consolidation (Figure 1) and no evidence of pulmonary embolism. Induced sputum was negative for Pneumocystis carinii, fungi, or acid-fast bacilli. A bronchoalveolar lavage was performed. What is the diagnosis?


Subject(s)
AIDS-Related Opportunistic Infections/diagnosis , Lung Diseases, Fungal/diagnosis , Pneumonia, Pneumocystis/diagnosis , Sporotrichosis/diagnosis , AIDS-Related Opportunistic Infections/complications , AIDS-Related Opportunistic Infections/drug therapy , Anti-Infective Agents/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Bronchoalveolar Lavage Fluid/microbiology , Chest Pain/microbiology , Cough/microbiology , Deglutition Disorders/microbiology , Diagnosis, Differential , Drug Therapy, Combination , Dyspnea/microbiology , Humans , Lung Diseases, Fungal/complications , Lung Diseases, Fungal/drug therapy , Male , Middle Aged , Pneumonia, Pneumocystis/complications , Pneumonia, Pneumocystis/drug therapy , Prednisone/therapeutic use , Sporotrichosis/complications , Sporotrichosis/drug therapy , Syncope/microbiology , Tomography, X-Ray Computed , Trimethoprim, Sulfamethoxazole Drug Combination/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...