Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Mais filtros

Bases de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 117(22): 12230-12238, 2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32414920

RESUMO

Tibetans have adapted to the chronic hypoxia of high altitude and display a distinctive suite of physiologic adaptations, including augmented hypoxic ventilatory response and resistance to pulmonary hypertension. Genome-wide studies have consistently identified compelling genetic signatures of natural selection in two genes of the Hypoxia Inducible Factor pathway, PHD2 and HIF2A The product of the former induces the degradation of the product of the latter. Key issues regarding Tibetan PHD2 are whether it is a gain-of-function or loss-of-function allele, and how it might contribute to high-altitude adaptation. Tibetan PHD2 possesses two amino acid changes, D4E and C127S. We previously showed that in vitro, Tibetan PHD2 is defective in its interaction with p23, a cochaperone of the HSP90 pathway, and we proposed that Tibetan PHD2 is a loss-of-function allele. Here, we report that additional PHD2 mutations at or near Asp-4 or Cys-127 impair interaction with p23 in vitro. We find that mice with the Tibetan Phd2 allele display augmented hypoxic ventilatory response, supporting this loss-of-function proposal. This is phenocopied by mice with a mutation in p23 that abrogates the PHD2:p23 interaction. Hif2a haploinsufficiency, but not the Tibetan Phd2 allele, ameliorates hypoxia-induced increases in right ventricular systolic pressure. The Tibetan Phd2 allele is not associated with hemoglobin levels in mice. We propose that Tibetans possess genetic alterations that both activate and inhibit selective outputs of the HIF pathway to facilitate successful adaptation to the chronic hypoxia of high altitude.


Assuntos
Adaptação Fisiológica , Proteínas de Ligação a DNA/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Prolina Dioxigenases do Fator Induzível por Hipóxia/fisiologia , Hipóxia/fisiopatologia , Mutação com Perda de Função , Alelos , Altitude , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Ligação a DNA/genética , Humanos , Camundongos , Camundongos Knockout , Fenótipo , Seleção Genética , Tibet
2.
Ann Hematol ; 98(5): 1111-1118, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-30535576

RESUMO

External quality assurance (EQA) programs are vital to ensure high quality and standardized results in molecular diagnostics. It is important that EQA for quantitative analysis takes into account the variation in methodology. Results cannot be expected to be more accurate than limits of the technology used, and it is essential to recognize factors causing substantial outlier results. The present study aimed to identify parameters of specific importance for JAK2 V617F quantification by quantitative PCR, using different starting materials, assays, and technical platforms. Sixteen samples were issued to participating laboratories in two EQA rounds. In the first round, 19 laboratories from 11 European countries analyzing JAK2 V617F as part of their routine diagnostics returned results from in-house assays. In the second round, 25 laboratories from 17 countries participated. Despite variations in starting material, assay set-up and instrumentation the laboratories were generally well aligned in the EQA program. However, EQA based on a single technology appears to be a valuable tool to achieve standardization of the quantification of JAK2 V617F allelic burden.


Assuntos
Janus Quinase 2/genética , Mutação de Sentido Incorreto , Patologia Molecular/normas , Garantia da Qualidade dos Cuidados de Saúde , Reação em Cadeia da Polimerase em Tempo Real/normas , Substituição de Aminoácidos , Feminino , Humanos , Masculino
3.
BMC Cancer ; 18(1): 286, 2018 03 13.
Artigo em Inglês | MEDLINE | ID: mdl-29534684

RESUMO

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.


Assuntos
Neoplasias das Glândulas Suprarrenais/genética , Mutação em Linhagem Germinativa , Proteína 1 Reguladora do Ferro/genética , Janus Quinase 2/genética , Feocromocitoma/genética , Policitemia Vera/genética , Splicing de RNA , Neoplasias das Glândulas Suprarrenais/complicações , Neoplasias das Glândulas Suprarrenais/patologia , Adulto , Feminino , Humanos , Feocromocitoma/complicações , Feocromocitoma/patologia , Policitemia Vera/complicações , Policitemia Vera/patologia , Prognóstico
4.
Blood ; 123(3): 366-76, 2014 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-24196071

RESUMO

Neutrophil lifespan and function are regulated by hypoxia via components of the hypoxia inducible factor (HIF)/von Hippel Lindau/hydroxylase pathway, including specific roles for HIF-1α and prolyl hydroxylase-3. HIF-2α has both distinct and overlapping biological roles with HIF-1α and has not previously been studied in the context of neutrophil biology. We investigated the role of HIF-2α in regulating key neutrophil functions. Human and murine peripheral blood neutrophils expressed HIF-2α, with expression up-regulated by acute and chronic inflammatory stimuli and in disease-associated inflammatory neutrophil. HIF2A gain-of-function mutations resulted in a reduction in neutrophil apoptosis both ex vivo, through the study of patient cells, and in vivo in a zebrafish tail injury model. In contrast, HIF-2α-deficient murine inflammatory neutrophils displayed increased sensitivity to nitrosative stress induced apoptosis ex vivo and increased neutrophil apoptosis in vivo, resulting in a reduction in neutrophilic inflammation and reduced tissue injury. Expression of HIF-2α was temporally dissociated from HIF-1α in vivo and predominated in the resolution phase of inflammation. These data support a critical and selective role for HIF-2α in persistence of neutrophilic inflammation and provide a platform to dissect the therapeutic utility of targeting HIF-2α in chronic inflammatory diseases.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Regulação da Expressão Gênica , Inflamação , Neutrófilos/metabolismo , Animais , Apoptose , Hipóxia Celular , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Muramidase , Neutrófilos/citologia , Fagocitose , Fenótipo , RNA/metabolismo , Explosão Respiratória , Peixe-Zebra
5.
Blood Cells Mol Dis ; 54(1): 123-31, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25128083

RESUMO

DEK is important in regulating cellular processes including proliferation, differentiation and maintenance of stem cell phenotype. The translocation t(6;9) in Acute Myeloid Leukemia (AML), which fuses DEK with NUP214, confers a poor prognosis and a higher risk of relapse. The over-expression of DEK in AML has been reported, but different studies have shown diminished levels in pediatric and promyelocytic leukemias. This study has characterized DEK expression, in silico, using a large multi-center cohort of leukemic and normal control cases. Overall, DEK was under-expressed in AML compared to normal bone marrow (NBM). Studying specific subtypes of AML confirmed either no significant change or a significant reduction in DEK expression compared to NBM. Importantly, the similarity of DEK expression between AML and NBM was confirmed using immunohistochemistry analysis of tissue mircorarrays. In addition, stratification of AML patients based on median DEK expression levels indicated that DEK showed no effect on the overall survival of patients. DEK expression during normal hematopoiesis did reveal a relationship with specific cell types implicating a distinct function during myeloid differentiation. Whilst DEK may play a potential role in hematopoiesis, it remains to be established whether it is important for leukemagenesis, except when involved in the t(6;9) translocation.


Assuntos
Proteínas Cromossômicas não Histona/biossíntese , Proteínas de Ligação a DNA/biossíntese , Bases de Dados Genéticas , Regulação Leucêmica da Expressão Gênica , Hematopoese , Leucemia Mieloide Aguda/metabolismo , Proteínas Oncogênicas/biossíntese , Animais , Proteínas Cromossômicas não Histona/genética , Cromossomos Humanos Par 6/genética , Cromossomos Humanos Par 9/genética , Estudos de Coortes , Proteínas de Ligação a DNA/genética , Intervalo Livre de Doença , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Estudos Multicêntricos como Assunto , Proteínas Oncogênicas/genética , Proteínas de Ligação a Poli-ADP-Ribose , Taxa de Sobrevida , Translocação Genética
6.
Eur J Haematol ; 95(4): 270-9, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25951317

RESUMO

Since the discovery of the JAK2 V617F mutation in the majority of the myeloproliferative neoplasms (MPN) of polycythemia vera, essential thrombocythemia and primary myelofibrosis ten years ago, further MPN-specific mutational events, notably in JAK2 exon 12, MPL exon 10 and CALR exon 9 have been identified. These discoveries have been rapidly incorporated into evolving molecular diagnostic algorithms. Whilst many of these mutations appear to have prognostic implications, establishing MPN diagnosis is of immediate clinical importance with selection, implementation and the continual evaluation of the appropriate laboratory methodology to achieve this diagnosis similarly vital. The advantages and limitations of these approaches in identifying and quantitating the common MPN-associated mutations are considered herein with particular regard to their clinical utility. The evolution of molecular diagnostic applications and platforms has occurred in parallel with the discovery of MPN-associated mutations, and it therefore appears likely that emerging technologies such as next-generation sequencing and digital PCR will in the future play an increasing role in the molecular diagnosis of MPN.


Assuntos
Transtornos Mieloproliferativos/diagnóstico , Transtornos Mieloproliferativos/genética , Calreticulina/genética , Éxons , Humanos , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Técnicas de Diagnóstico Molecular/métodos , Técnicas de Diagnóstico Molecular/normas , Mutação , Transtornos Mieloproliferativos/metabolismo , Garantia da Qualidade dos Cuidados de Saúde , Receptores de Trombopoetina/genética
7.
Hum Mutat ; 35(1): 15-26, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24115288

RESUMO

Congenital erythrocytosis (CE), or congenital polycythemia, represents a rare and heterogeneous clinical entity. It is caused by deregulated red blood cell production where erythrocyte overproduction results in elevated hemoglobin and hematocrit levels. Primary congenital familial erythrocytosis is associated with low erythropoietin (Epo) levels and results from mutations in the Epo receptor gene (EPOR). Secondary CE arises from conditions causing tissue hypoxia and results in increased Epo production. These include hemoglobin variants with increased affinity for oxygen (HBB, HBA mutations), decreased production of 2,3-bisphosphoglycerate due to BPGM mutations, or mutations in the genes involved in the hypoxia sensing pathway (VHL, EPAS1, and EGLN1). Depending on the affected gene, CE can be inherited either in an autosomal dominant or recessive mode, with sporadic cases arising de novo. Despite recent important discoveries in the molecular pathogenesis of CE, the molecular causes remain to be identified in about 70% of the patients. With the objective of collecting all the published and unpublished cases of CE the COST action MPN&MPNr-Euronet developed a comprehensive Internet-based database focusing on the registration of clinical history, hematological, biochemical, and molecular data (http://www.erythrocytosis.org/). In addition, unreported mutations are also curated in the corresponding Leiden Open Variation Database.


Assuntos
Bases de Dados Genéticas , Predisposição Genética para Doença , Mutação , Policitemia/congênito , Receptores da Eritropoetina/genética , Hipóxia Celular/genética , Eritropoetina/metabolismo , Humanos , Internet , Policitemia/genética , Policitemia/metabolismo , Transdução de Sinais/genética
8.
J Biol Chem ; 288(24): 17134-44, 2013 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-23640890

RESUMO

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.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Hipertensão Pulmonar/genética , Mutação de Sentido Incorreto , Policitemia/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Gasometria , Células Cultivadas , Modelos Animais de Doenças , Endotelina-1/genética , Endotelina-1/metabolismo , Eritropoese , Eritropoetina/sangue , Eritropoetina/genética , Expressão Gênica , Técnicas de Introdução de Genes , Estudos de Associação Genética , Humanos , Hipertensão Pulmonar/sangue , Hipertensão Pulmonar/fisiopatologia , Hipertrofia Ventricular Direita/sangue , Hipertrofia Ventricular Direita/genética , Hipertrofia Ventricular Direita/fisiopatologia , Rim/metabolismo , Pulmão/metabolismo , Pulmão/patologia , Pulmão/fisiopatologia , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese , Policitemia/sangue , Policitemia/fisiopatologia , Proteínas Proto-Oncogênicas c-sis/genética , Proteínas Proto-Oncogênicas c-sis/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Taxa Respiratória , Regulação para Cima , Fator A de Crescimento do Endotélio Vascular/sangue
9.
J Biol Chem ; 288(47): 33571-33584, 2013 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-24121508

RESUMO

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.


Assuntos
Haploinsuficiência , Prolina Dioxigenases do Fator Induzível por Hipóxia/metabolismo , Mutação de Sentido Incorreto , Policitemia/metabolismo , Substituição de Aminoácidos , Animais , Modelos Animais de Doenças , Técnicas de Introdução de Genes , Humanos , Prolina Dioxigenases do Fator Induzível por Hipóxia/genética , Camundongos , Camundongos Transgênicos , Policitemia/genética , Policitemia/patologia
10.
Br J Haematol ; 165(4): 519-28, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24533580

RESUMO

Primary familial and congenital polycythaemia (PFCP) is a disease characterized by increased red blood cell mass, and can be associated with mutations in the intracellular region of the erythropoietin (EPO) receptor (EPOR). Here we explore the mechanisms by which EPOR mutations induce PFCP, using an experimental system based on chimeric receptors between epidermal growth factor receptor (EGFR) and EPOR. The design of the chimeras enabled EPOR signalling to be triggered by EGF binding. Using this system we analysed three novel EPOR mutations discovered in PFCP patients: a deletion mutation (Del1377-1411), a nonsense mutation (C1370A) and a missense mutation (G1445A). Three different chimeras, bearing these mutations in the cytosolic, EPOR region were generated; Hence, the differences in the chimera-related effects are specifically attributed to the mutations. The results show that the different mutations affect various aspects related to the signalling and metabolism of the chimeric receptors. These include slower degradation rate, higher levels of glycan-mature chimeric receptors, increased sensitivity to low levels of EGF (replacing EPO in this system) and extended signalling cascades. This study provides a novel experimental system to study polycythaemia-inducing mutations in the EPOR, and sheds new light on underlying mechanisms of EPOR over-activation in PFCP patients.


Assuntos
Policitemia/congênito , Receptores da Eritropoetina/genética , Linhagem Celular , Membrana Celular/metabolismo , Códon sem Sentido , Análise Mutacional de DNA , Receptores ErbB/genética , Glicosilação , Humanos , Técnicas In Vitro , Janus Quinase 2/metabolismo , Sistema de Sinalização das MAP Quinases , Mutação de Sentido Incorreto , Policitemia/genética , Polissacarídeos/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Receptores da Eritropoetina/química , Receptores da Eritropoetina/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Fator de Transcrição STAT5/fisiologia , Deleção de Sequência , Transdução de Sinais/genética , Relação Estrutura-Atividade , Transfecção
11.
Pediatr Blood Cancer ; 60(11): 1734-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23776154

RESUMO

During recent years, the increasing knowledge of genetic and physiological changes in polycythemia vera (PV) and of different types of congenital erythrocytosis has led to fundamental changes in recommendations for the diagnostic approach to patients with erythrocytosis. Although widely accepted for adult patients this approach may not be appropriate with regard to children and adolescents affected by erythrocytosis. The "congenital erythrocytosis" working group established within the framework of the MPN&MPNr-EuroNet (COST action BM0902) addressed this question in a consensus finding process and developed a specific algorithm for the diagnosis of erythrocytosis in childhood and adolescence which is presented here.


Assuntos
Algoritmos , Policitemia/classificação , Policitemia/diagnóstico , Adolescente , Adulto , Criança , Humanos
12.
Proc Natl Acad Sci U S A ; 107(28): 12722-7, 2010 Jul 13.
Artigo em Inglês | MEDLINE | ID: mdl-20616028

RESUMO

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.


Assuntos
Regulação da Expressão Gênica/fisiologia , Hipóxia/genética , Hipóxia/metabolismo , Fatores de Transcrição/metabolismo , Adulto , Exercício Físico/fisiologia , Feminino , Humanos , Lactatos/metabolismo , Ácido Láctico/metabolismo , Masculino , Músculo Esquelético/metabolismo , Músculos/metabolismo , Oxigênio/metabolismo , Policitemia/genética , Policitemia/metabolismo , Fatores de Transcrição/genética
13.
Blood ; 115(22): 4517-23, 2010 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-20304805

RESUMO

The 46/1 JAK2 haplotype predisposes to V617F-positive myeloproliferative neoplasms, but the underlying mechanism is obscure. We analyzed essential thrombocythemia patients entered into the PT-1 studies and, as expected, found that 46/1 was overrepresented in V617F-positive cases (n = 404) versus controls (n = 1492, P = 3.9 x 10(-11)). The 46/1 haplotype was also overrepresented in cases without V617F (n = 347, P = .009), with an excess seen for both MPL exon 10 mutated and V617F, MPL exon 10 nonmutated cases. Analysis of further MPL-positive, V617F-negative cases confirmed an excess of 46/1 (n = 176, P = .002), but no association between MPL mutations and MPL haplotype was seen. An excess of 46/1 was also seen in JAK2 exon 12 mutated cases (n = 69, P = .002), and these mutations preferentially arose on the 46/1 chromosome (P = .029). No association between 46/1 and clinical or laboratory features was seen in the PT-1 cohort either with or without V617F. The excess of 46/1 in JAK2 exon 12 cases is compatible with both the "hypermutability" and "fertile ground" hypotheses, but the excess in MPL-mutated cases argues against the former. No difference in sequence, splicing, or expression of JAK2 was found on 46/1 compared with other haplotypes, suggesting that any functional difference of JAK2 on 46/1, if it exists, must be relatively subtle.


Assuntos
Janus Quinase 2/genética , Mutação , Transtornos Mieloproliferativos/genética , Receptores de Trombopoetina/genética , Adulto , Idoso , Substituição de Aminoácidos , Sequência de Bases , Estudos de Casos e Controles , Estudos de Coortes , Primers do DNA/genética , Éxons , Feminino , Frequência do Gene , Predisposição Genética para Doença , Haplótipos , Humanos , Desequilíbrio de Ligação , Masculino , Pessoa de Meia-Idade , Modelos Genéticos , Mutação de Sentido Incorreto , Policitemia Vera/genética , Polimorfismo de Nucleotídeo Único , Estudos Prospectivos , Trombocitemia Essencial/tratamento farmacológico , Trombocitemia Essencial/genética
14.
FASEB J ; 25(6): 2001-11, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21389259

RESUMO

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.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Dióxido de Carbono/sangue , Fenômenos Fisiológicos Cardiovasculares/genética , Regulação da Expressão Gênica/fisiologia , Oxigênio/sangue , Doença de von Hippel-Lindau/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/sangue , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Estudos de Casos e Controles , Teste de Esforço , Feminino , Humanos , Masculino , Mutação , Doença de von Hippel-Lindau/sangue , Doença de von Hippel-Lindau/genética
15.
J Pediatr Hematol Oncol ; 34(6): 457-60, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22627575

RESUMO

BACKGROUND: Cytochrome b5 reductase (CB5R) deficiency is a recessively inherited autosomal disorder that is either benign (type I) or associated with severe neurological problems (type II). Specific mutations in the CYB5R gene are not exclusive to each type. OBSERVATION: Two cyanotic children with developmental delay but with slow progression were investigated for CB5R deficiency. A novel mutation, p.Arg58Pro, was independently detected in both cases. CONCLUSIONS: The clinical variability and severity of the disease reflect the combined effects of impaired function of the 2 mutant enzymes. As illustrated by these 2 cases, inheritance of p.Arg58Pro with either p.Gly76Ser or pLeu188del causes a clinical condition more severe than type I and less severe than the type II cases reported to date.


Assuntos
Citocromo-B(5) Redutase/deficiência , Citocromo-B(5) Redutase/genética , Deficiências do Desenvolvimento/etiologia , Metemoglobinemia/enzimologia , Metemoglobinemia/genética , Mutação/genética , Pré-Escolar , Deficiências do Desenvolvimento/patologia , Feminino , Humanos , Lactente , Masculino , Metemoglobinemia/complicações , Fenótipo , Prognóstico
16.
N Engl J Med ; 358(2): 162-8, 2008 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-18184961

RESUMO

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.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Eritropoese/genética , Eritropoetina/biossíntese , Mutação Puntual , Policitemia/genética , Adulto , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Análise Mutacional de DNA , Feminino , Genótipo , Hematócrito , Hemoglobinas/análise , Humanos , Masculino , Linhagem , Policitemia/metabolismo , Reação em Cadeia da Polimerase
17.
Hemoglobin ; 35(4): 428-9, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21797710

RESUMO

We describe a high oxygen affinity hemoglobin (Hb) variant (Hb Vanderbilt) as a result of a heterozygous novel base change from T to A at codon 89 (AGT>AGA) leading to an amino acid change from serine to arginine.


Assuntos
Hemoglobinas Anormais/genética , Mutação de Sentido Incorreto , Globinas beta/genética , Adulto , Ligação Competitiva , Análise Mutacional de DNA , Hemoglobinas Anormais/metabolismo , Humanos , Masculino , Oxigênio/metabolismo , Ligação Proteica , Globinas beta/metabolismo
18.
Blood ; 111(11): 5400-2, 2008 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-18378852

RESUMO

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.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Policitemia/genética , Adolescente , Adulto , Sequência de Bases , Análise Mutacional de DNA , Eritropoetina/sangue , Feminino , Humanos , Masculino , Dados de Sequência Molecular , Mutação , Reação em Cadeia da Polimerase
19.
Haematologica ; 95(5): 829-32, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20007141

RESUMO

The ERYTHROPOIETIN (EPO) gene is regulated by the transcription factor Hypoxia Inducible Factor-alpha (HIF-alpha). In this pathway, Prolyl Hydroxylase Domain protein 2 (PHD2) hydroxylates two prolyl residues in HIF-alpha, which in turn promotes HIF-alpha degradation by the von Hippel Lindau (VHL) protein. Evidence that HIF-2alpha is the important isoform for EPO regulation in humans comes from the recent observation that mutations in the HIF2A gene are associated with cases of erythrocytosis. We report here a new erythrocytosis-associated mutation, p.Asp539Glu, in the HIF2A gene. Similar to all reported cases, the affected residue is in close vicinity and C-terminal to the primary hydroxylation site in HIF-2alpha, Pro531. This mutation, however, is notable in producing a rather subtle amino acid substitution. Nonetheless, we find that this mutation compromises binding of HIF-2alpha to both PHD2 and VHL, and we propose that this mutation is the cause of erythrocytosis in this individual.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Mutação de Sentido Incorreto/genética , Policitemia/diagnóstico , Policitemia/genética , Adolescente , Fatores de Transcrição Hélice-Alça-Hélice Básicos/química , Feminino , Humanos
20.
Br J Haematol ; 147(4): 450-8, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19735488

RESUMO

Recently several different JAK2 exon12 mutations have been identified in V617F negative polycythaemia vera (PV) or idiopathic erythrocytosis (IE) patients. The patients present with erythrocytosis, ligand-independent cell growth and low serum erythropoietin (EPO) levels. Within this group, a deletion of amino acids 542-543 (N542-E543del) of JAK2 is most prevalent. We have previously shown that in the presence of JAK2(V617F), suppressor of cytokine signalling 3 (SOCS3) is unable to negatively regulate EPO signalling and proliferation of V617F-expressing cells. Here we report a PV patient heterozygous for the somatic JAK2(N542-E543del) mutation and a previously unreported germline mutation within the SH2 domain of SOCS3 (F136L). Interestingly, the SOCS3(F136L) mutation was detected in a Japanese myeloproliferative disorder patient cohort at double the frequency of healthy controls. Cells expressing SOCS3(F136L) had markedly elevated EPO-induced proliferation and extended EPO-induced JAK2 phosphorylation. Additionally, compared to wild-type SOCS3, mutant SOCS3 had an extended half-life in the presence of JAK2 and JAK2(N542-E543del). Our findings suggest that this loss-of-function SOCS3 mutation may have contributed to disease onset by causing deregulated JAK2 signalling in the presence of a constitutively active JAK2(N542-E543del) mutant.


Assuntos
Eritropoetina/fisiologia , Mutação em Linhagem Germinativa , Policitemia Vera/genética , Proteínas Supressoras da Sinalização de Citocina/genética , Adulto , Sequência de Bases , Proliferação de Células , Células Cultivadas , Eritropoetina/sangue , Feminino , Humanos , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Mutação , Fosforilação , Proteína 3 Supressora da Sinalização de Citocinas , Proteínas Supressoras da Sinalização de Citocina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA