Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
BMC Cancer ; 18(1): 286, 2018 03 13.
Article in English | MEDLINE | ID: mdl-29534684

ABSTRACT

BACKGROUND: The role of the hypoxia signaling pathway in the pathogenesis of pheochromocytoma/paraganglioma (PPGL)-polycythemia syndrome has been elucidated. Novel somatic mutations in hypoxia-inducible factor type 2A (HIF2A) and germline mutations in prolyl hydroxylase type 1 and type 2 (PHD1 and PHD2) have been identified to cause upregulation of the hypoxia signaling pathway and its target genes including erythropoietin (EPO) and its receptor (EPOR). However, in a minority of patients presenting with this syndrome, the genetics and molecular pathogenesis remain unexplained. The aim of the present study was to uncover novel genetic causes of PPGL-polycythemia syndrome. CASE PRESENTATION: A female presented with a history of JAK2V617F positive PV, diagnosed in 2007, and right adrenal pheochromocytoma diagnosed and resected in 2011. Her polycythemia symptoms and hematocrit levels continued to worsen from 2007 to 2011, with an increased frequency of phlebotomies. Postoperatively, until early 2013, her hematocrit levels remained normalized. Following this, the hematocrit levels ranged between 46.4 and 48.9% [35-45%]. Tumor tissue from the patient was further tested for mutations in genes related to upregulation of the hypoxia signaling pathway including iron regulatory protein 1 (IRP1), which is a known regulator of HIF-2α mRNA translation. Functional studies were performed to investigate the consequences of these mutations, especially their effect on the HIF signaling pathway and EPO. Indel mutations (c.267-1_267delGGinsTA) were discovered at the exon 3 splicing site of IRP1. Minigene construct and splicing site analysis showed that the mutation led to a new splicing site and a frameshift mutation of IRP1, which caused a truncated protein. Fluorescence in situ hybridization analysis demonstrated heterozygous IRP1 deletions in tumor cells. Immunohistochemistry results confirmed the truncated IRP1 and overexpressed HIF-2α, EPO and EPOR in tumor cells. CONCLUSIONS: This is the first report which provides direct molecular genetic evidence of association between a somatic IRP1 loss-of-function mutation and PHEO and secondary polycythemia. In patients diagnosed with PHEO/PGL and polycythemia with negative genetic testing for mutations in HIF2A, PHD1/2, and VHL, IRP1 should be considered as a candidate gene.


Subject(s)
Adrenal Gland Neoplasms/genetics , Germ-Line Mutation , Iron Regulatory Protein 1/genetics , Janus Kinase 2/genetics , Pheochromocytoma/genetics , Polycythemia Vera/genetics , RNA Splicing , Adrenal Gland Neoplasms/complications , Adrenal Gland Neoplasms/pathology , Adult , Female , Humans , Pheochromocytoma/complications , Pheochromocytoma/pathology , Polycythemia Vera/complications , Polycythemia Vera/pathology , Prognosis
3.
J Biol Chem ; 288(47): 33571-33584, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24121508

ABSTRACT

The central pathway for controlling red cell mass is the PHD (prolyl hydroxylase domain protein):hypoxia-inducible factor (HIF) pathway. HIF, which is negatively regulated by PHD, activates numerous genes, including ones involved in erythropoiesis, such as the ERYTHROPOIETIN (EPO) gene. Recent studies have implicated PHD2 as the key PHD isoform regulating red cell mass. Studies of humans have identified erythrocytosis-associated, heterozygous point mutations in the PHD2 gene. A key question concerns the mechanism by which human mutations lead to phenotypes. In the present report, we generated and characterized a mouse line in which a P294R knock-in mutation has been introduced into the mouse Phd2 locus to model the first reported human PHD2 mutation (P317R). Phd2(P294R/+) mice display a degree of erythrocytosis equivalent to that seen in Phd2(+/-) mice. The Phd2(P294R/+)-associated erythrocytosis is reversed in a Hif2a(+/-), but not a Hif1a(+/-) background. Additional studies using various conditional knock-outs of Phd2 reveal that erythrocytosis can be induced by homozygous and heterozygous knock-out of Phd2 in renal cortical interstitial cells using a Pax3-Cre transgene or by homozygous knock-out of Phd2 in hematopoietic progenitors driven by a Vav1-Cre transgene. These studies formally prove that a missense mutation in PHD2 is the cause of the erythrocytosis, show that this occurs through haploinsufficiency, and point to multifactorial control of red cell mass by PHD2.


Subject(s)
Haploinsufficiency , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Mutation, Missense , Polycythemia/metabolism , Amino Acid Substitution , Animals , Disease Models, Animal , Gene Knock-In Techniques , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Mice , Mice, Transgenic , Polycythemia/genetics , Polycythemia/pathology
4.
J Biol Chem ; 288(24): 17134-44, 2013 Jun 14.
Article in English | MEDLINE | ID: mdl-23640890

ABSTRACT

The central pathway for oxygen-dependent control of red cell mass is the prolyl hydroxylase domain protein (PHD):hypoxia inducible factor (HIF) pathway. PHD site specifically prolyl hydroxylates the transcription factor HIF-α, thereby targeting the latter for degradation. Under hypoxia, this modification is attenuated, allowing stabilized HIF-α to activate target genes, including that for erythropoietin (EPO). Studies employing genetically modified mice point to Hif-2α, one of two main Hif-α isoforms, as being the critical regulator of Epo in the adult mouse. More recently, erythrocytosis patients with heterozygous point mutations in the HIF2A gene have been identified; whether these mutations were polymorphisms unrelated to the phenotype could not be ruled out. In the present report, we characterize a mouse line bearing a G536W missense mutation in the Hif2a gene that corresponds to the first such human mutation identified (G537W). We obtained mice bearing both heterozygous and homozygous mutations at this locus. We find that these mice display, in a mutation dose-dependent manner, erythrocytosis and pulmonary hypertension with a high degree of penetrance. These findings firmly establish missense mutations in HIF-2α as a cause of erythrocytosis, highlight the importance of this HIF-α isoform in erythropoiesis, and point to physiologic consequences of HIF-2α dysregulation.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Hypertension, Pulmonary/genetics , Mutation, Missense , Polycythemia/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blood Gas Analysis , Cells, Cultured , Disease Models, Animal , Endothelin-1/genetics , Endothelin-1/metabolism , Erythropoiesis , Erythropoietin/blood , Erythropoietin/genetics , Gene Expression , Gene Knock-In Techniques , Genetic Association Studies , Humans , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/blood , Hypertrophy, Right Ventricular/genetics , Hypertrophy, Right Ventricular/physiopathology , Kidney/metabolism , Lung/metabolism , Lung/pathology , Lung/physiopathology , Mice , Mice, Inbred C57BL , Mutagenesis , Polycythemia/blood , Polycythemia/physiopathology , Proto-Oncogene Proteins c-sis/genetics , Proto-Oncogene Proteins c-sis/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Respiratory Rate , Up-Regulation , Vascular Endothelial Growth Factor A/blood
5.
Stem Cells ; 31(7): 1434-45, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23592435

ABSTRACT

The incidence of refractory acute myeloid leukemia (AML) is on the increase due in part to an aging population that fails to respond to traditional therapies. High throughput genomic analysis promises better diagnosis, prognosis, and therapeutic intervention based on improved patient stratification. Relevant preclinical models are urgently required to advance drug development in this area. The collaborating oncogenes, HOXA9 and MEIS1, are frequently co-overexpressed in cytogenetically normal AML (CN-AML), and a conditional transplantation mouse model was developed that demonstrated oncogene dependency and expression levels comparable to CN-AML patients. Integration of gene signatures obtained from the mouse model and a cohort of CN-AML patients using statistically significant connectivity map analysis identified Entinostat as a drug with the potential to alter the leukemic condition toward the normal state. Ex vivo treatment of leukemic cells, but not age-matched normal bone marrow controls, with Entinostat validated the gene signature and resulted in reduced viability in liquid culture, impaired colony formation, and loss of the leukemia initiating cell. Furthermore, in vivo treatment with Entinostat resulted in prolonged survival of leukemic mice. This study demonstrates that the HDAC inhibitor Entinostat inhibits disease maintenance and prolongs survival in a clinically relevant murine model of cytogenetically normal AML.


Subject(s)
Benzamides/pharmacology , Histone Deacetylase Inhibitors/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Pyridines/pharmacology , Animals , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Immunophenotyping , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Mice, Inbred C57BL
7.
Diabetes Care ; 35(3): 574-80, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22301123

ABSTRACT

OBJECTIVE: To compare associations of maternal glucose and A1C with adverse outcomes in the multinational Hyperglycemia and Adverse Pregnancy Outcome (HAPO) Study and determine, based on those comparisons, if A1C measurement can provide an alternative to an oral glucose tolerance test (OGTT) in pregnant women. RESEARCH DESIGN AND METHODS: Eligible pregnant women underwent a 75-g OGTT at 24-32 weeks' gestation. A sample for A1C was also collected. Neonatal anthropometrics and cord serum C-peptide were measured. Associations with outcomes were assessed using multiple logistic regression with adjustment for potential confounders. RESULTS: Among 23,316 HAPO Study participants with glucose levels blinded to caregivers, 21,064 had a nonvariant A1C result. The mean ± SD A1C was 4.79 ± 0.40%. Associations were significantly stronger with glucose measures than with A1C for birth weight, sum of skinfolds, and percent body fat >90th percentile and for fasting and 1-h glucose for cord C-peptide (all P < 0.01). For example, in fully adjusted models, odds ratios (ORs) for birth weight >90th percentile for each measure higher by 1 SD were 1.39, 1.45, and 1.38, respectively, for fasting, 1-, and 2-h plasma glucose and 1.15 for A1C. ORs for cord C-peptide >90th percentile were 1.56, 1.45, and 1.35 for glucose, respectively, and 1.32 for A1C. ORs were similar for glucose and A1C for primary cesarean section, preeclampsia, and preterm delivery. CONCLUSIONS: On the basis of associations with adverse outcomes, these findings suggest that A1C measurement is not a useful alternative to an OGTT in pregnant women.


Subject(s)
Glycated Hemoglobin/metabolism , Hyperglycemia/physiopathology , Adult , Blood Glucose/metabolism , Diabetes, Gestational/blood , Female , Glucose Tolerance Test , Humans , Hyperglycemia/complications , Pregnancy , Pregnancy Outcome
8.
FASEB J ; 25(6): 2001-11, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21389259

ABSTRACT

The hypoxia-inducible factors (HIFs; isoforms HIF-1α, HIF-2α, HIF-3α) mediate many responses to hypoxia. Their regulation is principally by oxygen-dependent degradation, which is initiated by hydroxylation of specific proline residues followed by binding of von Hippel-Lindau (VHL) protein. Chuvash polycythemia is a disorder with elevated HIF. It arises through germline homozygosity for hypomorphic VHL alleles and has a phenotype of hematological, cardiopulmonary, and metabolic abnormalities. This study explores the phenotype of two other HIF pathway diseases: classic VHL disease and HIF-2α gain-of-function mutation. No cardiopulmonary abnormalities were detected in classic VHL disease. HIF-2α gain-of-function mutations were associated with pulmonary hypertension, increased cardiac output, increased heart rate, and increased pulmonary ventilation relative to metabolism. Comparison of the HIF-2α gain-of-function responses with data from studies of Chuvash polycythemia suggested that other aspects of the Chuvash phenotype were diminished or absent. In classic VHL disease, patients are germline heterozygous for mutations in VHL, and the present results suggest that a single wild-type allele for VHL is sufficient to maintain normal cardiopulmonary function. The HIF-2α gain-of-function phenotype may be more limited than the Chuvash phenotype either because HIF-1α is not elevated in the former condition, or because other HIF-independent functions of VHL are perturbed in Chuvash polycythemia.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carbon Dioxide/blood , Cardiovascular Physiological Phenomena/genetics , Gene Expression Regulation/physiology , Oxygen/blood , von Hippel-Lindau Disease/metabolism , Basic Helix-Loop-Helix Transcription Factors/blood , Basic Helix-Loop-Helix Transcription Factors/genetics , Case-Control Studies , Exercise Test , Female , Humans , Male , Mutation , von Hippel-Lindau Disease/blood , von Hippel-Lindau Disease/genetics
9.
Int J Cancer ; 129(11): 2566-76, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21442620

ABSTRACT

Erythropoietin (Epo) regulates erythropoiesis by binding to its receptor (EpoR) and promoting cell proliferation, differentiation and inhibition of apoptosis. Epo is widely used to treat cervical cancer-related anaemia. However, there are data suggesting that administration of Epo is associated with an increment in recurrence rate, and decreased disease-free and overall survival. In the present study, we investigated the expression of Epo and EpoR on cervical cancer cell lines. We observed that both EpoR and extracellular Epo are constitutively expressed in cervical cancer cells. Inhibition of either Epo or EpoR expression with siRNA attenuated cell proliferation, whereas addition of exogenous Epo led to a significant increase in cell growth, both in vitro and in vivo. Epo-induced proliferation was associated with the activation of JAK2, JAK3, STAT3 and STAT5 but not JAK1 and STAT1. Our results are consistent with the existence of a functional, endogenous Epo/EpoR system in cervical cancer with the capacity to activate the transduction of signals resulting in an increased proliferation potential.


Subject(s)
Autocrine Communication , Cell Proliferation , Erythropoietin/pharmacology , Janus Kinases/metabolism , Paracrine Communication , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , STAT5 Transcription Factor/metabolism , Uterine Cervical Neoplasms/metabolism , Animals , Apoptosis , Blotting, Western , Female , Flow Cytometry , Humans , Immunoenzyme Techniques , Immunoprecipitation , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Janus Kinase 3/genetics , Janus Kinase 3/metabolism , Janus Kinases/genetics , Mice , Mice, Nude , RNA, Messenger/genetics , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , Reverse Transcriptase Polymerase Chain Reaction , STAT1 Transcription Factor/genetics , STAT3 Transcription Factor/genetics , STAT5 Transcription Factor/genetics , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/pathology
10.
Proc Natl Acad Sci U S A ; 107(28): 12722-7, 2010 Jul 13.
Article in English | MEDLINE | ID: mdl-20616028

ABSTRACT

The hypoxia-inducible factor (HIF) family of transcription factors directs a coordinated cellular response to hypoxia that includes the transcriptional regulation of a number of metabolic enzymes. Chuvash polycythemia (CP) is an autosomal recessive human disorder in which the regulatory degradation of HIF is impaired, resulting in elevated levels of HIF at normal oxygen tensions. Apart from the polycythemia, CP patients have marked abnormalities of cardiopulmonary function. No studies of integrated metabolic function have been reported. Here we describe the response of these patients to a series of metabolic stresses: exercise of a large muscle mass on a cycle ergometer, exercise of a small muscle mass (calf muscle) which allowed noninvasive in vivo assessments of muscle metabolism using (31)P magnetic resonance spectroscopy, and a standard meal tolerance test. During exercise, CP patients had early and marked phosphocreatine depletion and acidosis in skeletal muscle, greater accumulation of lactate in blood, and reduced maximum exercise capacities. Muscle biopsy specimens from CP patients showed elevated levels of transcript for pyruvate dehydrogenase kinase, phosphofructokinase, and muscle pyruvate kinase. In cell culture, a range of experimental manipulations have been used to study the effects of HIF on cellular metabolism. However, these approaches provide no potential to investigate integrated responses at the level of the whole organism. Although CP is relatively subtle disorder, our study now reveals a striking regulatory role for HIF on metabolism during exercise in humans. These findings have significant implications for the development of therapeutic approaches targeting the HIF pathway.


Subject(s)
Gene Expression Regulation/physiology , Hypoxia/genetics , Hypoxia/metabolism , Transcription Factors/metabolism , Adult , Exercise/physiology , Female , Humans , Lactates/metabolism , Lactic Acid/metabolism , Male , Muscle, Skeletal/metabolism , Muscles/metabolism , Oxygen/metabolism , Polycythemia/genetics , Polycythemia/metabolism , Transcription Factors/genetics
11.
Neonatology ; 96(1): 50-60, 2009.
Article in English | MEDLINE | ID: mdl-19204410

ABSTRACT

BACKGROUND: Genetic processes underlying fetal lung development and maturation are incompletely understood. Better knowledge of these processes would provide insights into the causes of lung malformations and prevention of respiratory distress syndrome and the potential adverse effects of glucocorticoids. Hox genes are involved in the lung branching morphogenesis and maturation of respiratory epithelium, but their expression pattern remains to be defined. OBJECTIVES: We hypothesized that genes involved in lung branching would be downregulated during early development, whereas those involved in maturation would be unchanged or upregulated. METHODS: TaqMan real-time primers and probes were designed for all 39 murine Hox genes, and the murine SP-B gene and transcription profiles of these genes were obtained from whole lungs isolated at e14.5, e16.5, e18.5, e19.5 and postnatal days 1 and 20. RESULTS: Hox genes in clusters A and B, specifically those between paralog groups 3 and 7, were the most represented, with Hoxa4 and Hoxa5 being the most highly transcribed. A wave of reduced transcription in 16 Hox genes, coincident with increased SP-B transcription, was observed with advancing gestation. Consistently high transcription of Hoxa5 from e14.5 to postnatal day 20 may indicate that sustained transcription is required for normal lung maturation. When e15.5 lungs were cultured with dexamethasone, Hoxb6, Hoxb7 and Hoxb8 levels were significantly upregulated, creating the potential for modulation of diverse downstream target genes. CONCLUSIONS: Improved understanding of the genetic processes underlying lung development afforded by our Q-PCR platform may allow development of more specific methods for inducing fetal lung maturation.


Subject(s)
Homeodomain Proteins/genetics , Lung/chemistry , Lung/embryology , Pulmonary Surfactant-Associated Protein B/genetics , Animals , DNA-Binding Proteins/genetics , Dexamethasone/pharmacology , Female , Fetal Organ Maturity/drug effects , Gene Expression Regulation, Developmental , Gestational Age , Glucocorticoids/pharmacology , Lung/growth & development , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Phosphoproteins/genetics , Polymerase Chain Reaction , RNA, Messenger/analysis , Transcription Factors
12.
J Biol Chem ; 284(14): 9050-8, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19208626

ABSTRACT

A classic physiologic response to hypoxia in humans is the up-regulation of the ERYTHROPOIETIN (EPO) gene, which is the central regulator of red blood cell mass. The EPO gene, in turn, is activated by hypoxia inducible factor (HIF). HIF is a transcription factor consisting of an alpha subunit (HIF-alpha) and a beta subunit (HIF-beta). Under normoxic conditions, prolyl hydroxylase domain protein (PHD, also known as HIF prolyl hydroxylase and egg laying-defective nine protein) site specifically hydroxylates HIF-alpha in a conserved LXXLAP motif (where underlining indicates the hydroxylacceptor proline). This provides a recognition motif for the von Hippel Lindau protein, a component of an E3 ubiquitin ligase complex that targets hydroxylated HIF-alpha for degradation. Under hypoxic conditions, this inherently oxygen-dependent modification is arrested, thereby stabilizing HIF-alpha and allowing it to activate the EPO gene. We previously identified and characterized an erythrocytosis-associated HIF2A mutation, G537W. More recently, we reported two additional erythrocytosis-associated HIF2A mutations, G537R and M535V. Here, we describe the functional characterization of these two mutants as well as a third novel erythrocytosis-associated mutation, P534L. These mutations affect residues C-terminal to the LXXLAP motif. We find that all result in impaired degradation and thus aberrant stabilization of HIF-2alpha. However, each exhibits a distinct profile with respect to their effects on PHD2 binding and von Hippel Lindau interaction. These findings reinforce the importance of HIF-2alpha in human EPO regulation, demonstrate heterogeneity of functional defects arising from these mutations, and point to a critical role for residues C-terminal to the LXXLAP motif in HIF-alpha.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Polycythemia/metabolism , Amino Acid Sequence , Basic Helix-Loop-Helix Transcription Factors/chemistry , Basic Helix-Loop-Helix Transcription Factors/genetics , Biocatalysis , Cell Line , Humans , Hydroxylation , Molecular Sequence Data , Mutation/genetics , Polycythemia/genetics , Procollagen-Proline Dioxygenase/genetics , Procollagen-Proline Dioxygenase/metabolism , Proline/genetics , Proline/metabolism , Protein Binding , Sequence Alignment , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
13.
Exp Hematol ; 37(3): 322-33.e3, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19157684

ABSTRACT

Hemopoietic progenitor cells express clustered homeobox (Hox) genes in a pattern characteristic of their lineage and stage of differentiation. In general, HOX expression tends to be higher in more primitive and lower in lineage-committed cells. These trends have led to the hypothesis that self-renewal of hemopoietic stem/progenitor cells is HOX-dependent and that dysregulated HOX expression underlies maintenance of the leukemia-initiating cell. Gene expression profile studies support this hypothesis and specifically highlight the importance of the HOXA cluster in hemopoiesis and leukemogenesis. Within this cluster HOXA6 and HOXA9 are highly expressed in patients with acute myeloid leukemia and form part of the "Hox code" identified in murine models of this disease. We have examined endogenous expression of Hoxa6 and Hoxa9 in purified primary progenitors as well as four growth factor-dependent cell lines FDCP-Mix, EML, 32Dcl3, and Ba/F3, representative of early multipotential and later committed precursor cells respectively. Hoxa6 was consistently higher expressed than Hoxa9, preferentially expressed in primitive cells and was both growth-factor and cell-cycle regulated. Enforced overexpression of HOXA6 or HOXA9 in FDCP-Mix resulted in increased proliferation and colony formation but had negligible effect on differentiation. In both FDCP-Mix and the more committed Ba/F3 precursor cells overexpression of HOXA6 potentiated factor-independent proliferation. These findings demonstrate that Hoxa6 is directly involved in fundamental processes of hemopoietic progenitor cell development.


Subject(s)
Cell Proliferation , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/physiology , Animals , Cell Differentiation , Cell Line , DNA, Complementary , Hematopoiesis , Humans , Mice , Stem Cells/cytology , Stem Cells/metabolism , Transfection
14.
Front Biosci ; 13: 4297-308, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18508512

ABSTRACT

The mammalian HOX gene network encodes a family of proteins which act as master regulators of developmental processes such as embryogenesis and hematopoiesis. The complex arrangement, regulation and co-factor association of HOX has been an area of intense research, particularly in cancer biology, for over a decade. The concept of redeployment of embryonic regulators in the neoplastic arena has received support from many quarters. Observations of altered HOX gene expression in various solid tumours and leukemia appear to support the thesis that 'oncology recapitulates ontogeny' but the identification of critical HOX subsets and their functional role in cancer onset and maintenance requires further investigation. The application of novel techniques and model systems will continue to enhance our understanding of the HOX network in the years to come. Better understanding of the intricacy of the complex as well as identification of functional pathways and direct targets of the encoded proteins will permit harnessing of this family of genes for clinical application.


Subject(s)
Genes, Homeobox , Hematopoiesis/physiology , Leukemia/genetics , Animals , Chromosome Mapping , Evolution, Molecular , Gene Expression Regulation , Gene Expression Regulation, Neoplastic , Hematopoiesis/genetics , Humans , Leukemia/classification , Leukemia/physiopathology , Mammals , Mice , Mice, Knockout , Signal Transduction , Transcription, Genetic , Translocation, Genetic
15.
Blood ; 111(11): 5400-2, 2008 Jun 01.
Article in English | MEDLINE | ID: mdl-18378852

ABSTRACT

Erythrocytosis can arise from deregulation of the erythropoietin (Epo) axis resulting from defects in the oxygen-sensing pathway. Epo synthesis is controlled by the hypoxia inducible factor (HIF) complex, composed of an alpha and a beta subunit. There are 2 main alpha subunits, HIF-1 alpha and HIF-2 alpha. Recently, a HIF-2 alpha Gly537Trp mutation was identified in a family with erythrocytosis. This raises the possibility of HIF2A mutations being associated with other cases of erythrocytosis. We now report a subsequent analysis of HIF2A in a cohort of 75 erythrocytosis patients and identify 4 additional patients with novel heterozygous Met535Val and Gly537Arg mutations. All patients presented at a young age with elevated serum Epo. Mutations at Gly-537 account for 4 of 5 HIF2A mutations associated with erythrocytosis. These findings support the importance of HIF-2 alpha in human Epo regulation and warrant investigation of HIF2A in patients with unexplained erythrocytosis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Polycythemia/genetics , Adolescent , Adult , Base Sequence , DNA Mutational Analysis , Erythropoietin/blood , Female , Humans , Male , Molecular Sequence Data , Mutation , Polymerase Chain Reaction
16.
N Engl J Med ; 358(2): 162-8, 2008 Jan 10.
Article in English | MEDLINE | ID: mdl-18184961

ABSTRACT

Hypoxia-inducible factor (HIF) alpha, which has three isoforms, is central to the continuous balancing of the supply and demand of oxygen throughout the body. HIF-alpha is a transcription factor that modulates a wide range of processes, including erythropoiesis, angiogenesis, and cellular metabolism. We describe a family with erythrocytosis and a mutation in the HIF2A gene, which encodes the HIF-2alpha protein. Our functional studies indicate that this mutation leads to stabilization of the HIF-2alpha protein and suggest that wild-type HIF-2alpha regulates erythropoietin production in adults.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Erythropoiesis/genetics , Erythropoietin/biosynthesis , Point Mutation , Polycythemia/genetics , Adult , Basic Helix-Loop-Helix Transcription Factors/metabolism , DNA Mutational Analysis , Female , Genotype , Hematocrit , Hemoglobins/analysis , Humans , Male , Pedigree , Polycythemia/metabolism , Polymerase Chain Reaction
17.
Blood ; 110(6): 2193-6, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17579185

ABSTRACT

The molecular basis of the erythrocytosis group of red cell disorders is incompletely defined. Some cases are due to dysregulation of erythropoietin (Epo) synthesis. The hypoxia inducible transcription factor (HIF) tightly regulates Epo synthesis. HIF in turn is regulated through its alpha subunit, which under normoxic conditions is hydroxylated on specific prolines and targeted for degradation by the von Hippel Lindau (VHL) protein. Several mutations in VHL have been reported in erythrocytosis, but only 1 mutation in the HIF prolyl hydroxylase PHD2 (prolyl hydroxylase domain protein 2) has been described. Here, we report a novel PHD2 mutation, Arg371His, which causes decreased HIF binding, HIF hydroxylase, and HIF inhibitory activities. In the tertiary structure of PHD2, Arg371 lies close to the previously described Pro317Arg mutation site. These findings substantiate PHD2 as a critical enzyme controlling HIF and therefore Epo in humans, and furthermore suggest the location of an active site groove in PHD2 that binds HIF.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mutation/genetics , Polycythemia/pathology , Procollagen-Proline Dioxygenase/genetics , Adult , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor-Proline Dioxygenases , Male , Polycythemia/genetics , Polycythemia/metabolism , Procollagen-Proline Dioxygenase/metabolism , Receptors, Erythropoietin/genetics , Receptors, Erythropoietin/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
18.
Stem Cells ; 25(2): 380-4, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17038666

ABSTRACT

Recent evidence confirms the presence of erythropoietin receptors on a variety of cancer cells. This has raised concerns about the use of erythropoiesis-stimulating agents in the treatment of cancer-related anemia. Having previously identified expression of functional erythropoietin receptors in a non-small cell lung carcinoma cell line, H838, which activated key signaling pathways in response to erythropoietin stimulation, we now demonstrate impaired downregulation of the erythropoietin receptor in these tumor cells. The erythropoietin receptor is not ubiquitinated following erythropoietin stimulation in this cancer cell line, and there is no turnover of the receptor in either unstimulated or stimulated cells. Compounding this blunted response is impaired SOCS3 induction downstream of erythropoietin stimulation and an extremely delayed SOCS1 response. If this finding in non-small cell lung carcinoma is a widespread phenomenon, then impaired erythropoietin receptor downregulation and degradation in tumor cells has clinical implications for those patients receiving erythropoiesis-stimulating agents for cancer-related anemia.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Down-Regulation , Lung Neoplasms/metabolism , Receptors, Erythropoietin/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Cycloheximide/pharmacology , Down-Regulation/drug effects , Erythropoietin/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/genetics , Lysosomes/drug effects , Lysosomes/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Biosynthesis/drug effects , Protein Processing, Post-Translational/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Erythropoietin/genetics , Recombinant Proteins , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Ubiquitins/metabolism
19.
Stem Cells ; 25(3): 718-22, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17110616

ABSTRACT

Immunohistochemical studies on formalin-fixed, paraffin-embedded (FFPE) tissue utilizing polyclonal antibodies form the cornerstone of many reports claiming to demonstrate erythropoietin receptor (EPOR) expression in malignant tissue. Recently, Elliott et al. (Blood 2006;107:1892-1895) reported that the antibodies commonly used to detect EPOR expression also detect non-EPOR proteins, and that their binding to EPOR was severely abrogated by two synthetic peptides based on the sequence of heat shock protein (HSP) 70, HSP70-2, and HSP70-5. We have investigated the specificity of the C20 antibody for detecting EPOR expression in non-small cell lung carcinoma (NSCLC) utilizing tissue microarrays. A total of 34 cases were available for study. Antibody absorbed with peptide resulted in marked suppression of cytoplasmic staining compared with nonabsorbed antibody. Four tumors that initially showed a membranous pattern of staining retained this pattern with absorbed antibody. Positive membranous immunoreactivity was also observed in 6 of 30 tumors that originally showed a predominantly cytoplasmic pattern of staining. Using the C20 antibody for Western blots, we detected three main bands, at 100, 66, and 59 kDa. Preincubation with either peptide caused abolition of the 66-kDa band, which contains non-EPOR sequences including heat shock peptides. These results call into question the significance of previous immunohistochemical studies of EPOR expression in malignancy and emphasize the need for more specific anti-EPOR antibodies to define the true extent of EPOR expression in neoplastic tissue.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Receptors, Erythropoietin/genetics , Amino Acid Sequence , Antibody Specificity , Carcinoma, Non-Small-Cell Lung/immunology , Cell Line, Tumor , HSP70 Heat-Shock Proteins/genetics , Heat-Shock Proteins/genetics , Humans , Lung Neoplasms/immunology , Molecular Sequence Data , Peptide Fragments/chemistry , Receptors, Erythropoietin/immunology
20.
Clin Trials ; 3(4): 397-407, 2006.
Article in English | MEDLINE | ID: mdl-17060214

ABSTRACT

BACKGROUND: Measurement of glucose in multiple Field Laboratories requires rigorous standardization when patients and caregivers are masked, unless predefined thresholds are met. Local misclassification of participants at the thresholds can introduce recruitment bias and adversely affect the integrity of study findings. PURPOSE: To describe the challenges and the approach to meeting them in measuring glucose, HbA1c, and C-peptide in the Hyperglycemia and Adverse Pregnancy Outcome (HAPO) Study. HAPO is an observational epidemiologic study of 25 000 pregnant women from 15 centres in 10 countries, designed to clarify unanswered questions on associations of maternal glycemia, less severe than overt diabetes mellitus, with risks of adverse pregnancy outcome. METHODS: Glucose tolerance (75 g two-hour OGTT) is assessed locally at 24-32 weeks' gestation, with results masked if fasting and two-hour plasma glucose are

Subject(s)
Blood Glucose/analysis , C-Peptide/blood , Clinical Laboratory Information Systems/organization & administration , Hemoglobins/analysis , Hyperglycemia/blood , Pregnancy Complications/blood , Pregnancy Complications/metabolism , Pregnancy Outcome , Female , Global Health , Glycated Hemoglobin , Humans , Infant, Newborn , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...