Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Blood ; 141(18): 2261-2274, 2023 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-36790527

RESUMO

Pathogenic missense variants in SLFN14, which encode an RNA endoribonuclease protein that regulates ribosomal RNA (rRNA) degradation, are known to cause inherited thrombocytopenia (TP) with impaired platelet aggregation and adenosine triphosphate secretion. Despite mild laboratory defects, the patients displayed an obvious bleeding phenotype. However, the function of SLFN14 in megakaryocyte (MK) and platelet biology remains unknown. This study aimed to model the disease in an immortalized MK cell line (imMKCL) and to characterize the platelet transcriptome in patients with the SLFN14 K219N variant. MK derived from heterozygous and homozygous SLFN14 K219N imMKCL and stem cells of blood from patients mainly presented with a defect in proplatelet formation and mitochondrial organization. SLFN14-defective platelets and mature MK showed signs of rRNA degradation; however, this was absent in undifferentiated imMKCL cells and granulocytes. Total platelet RNA was sequenced in 2 patients and 19 healthy controls. Differential gene expression analysis yielded 2999 and 2888 significantly (|log2 fold change| >1, false discovery rate <0.05) up- and downregulated genes, respectively. Remarkably, these downregulated genes were not enriched in any biological pathway, whereas upregulated genes were enriched in pathways involved in (mitochondrial) translation and transcription, with a significant upregulation of 134 ribosomal protein genes (RPGs). The upregulation of mitochondrial RPGs through increased mammalian target of rapamycin complex 1 (mTORC1) signaling in SLFN14 K219N MK seems to be a compensatory response to rRNA degradation. mTORC1 inhibition with rapamycin resulted in further enhanced rRNA degradation in SLFN14 K219N MK. Taken together, our study indicates dysregulation of mTORC1 coordinated ribosomal biogenesis is the disease mechanism for SLFN14-related TP.


Assuntos
Trombocitopenia , Humanos , Trombocitopenia/patologia , Plaquetas/metabolismo , Ribossomos/metabolismo , Megacariócitos/patologia , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , RNA/metabolismo
2.
Blood Adv ; 5(18): 3568-3580, 2021 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34546355

RESUMO

Brain-derived neurotrophic factor (BDNF) has both autocrine and paracrine roles in neurons, and its release and signaling mechanisms have been extensively studied in the central nervous system. Large quantities of BDNF have been reported in circulation, essentially stored in platelets with concentrations reaching 100- to 1000-fold those of neurons. Despite this abundance, the function of BDNF in platelet biology has not been explored. At low concentrations, BDNF primed platelets, acting synergistically with classical agonists. At high concentrations, BDNF induced complete biphasic platelet aggregation that in part relied on amplification from secondary mediators. Neurotrophin-4, but not nerve growth factor, and an activating antibody against the canonical BDNF receptor tropomyosin-related kinase B (TrkB) induced similar platelet responses to BDNF, suggesting TrkB could be the mediator. Platelets expressed, both at their surface and in their intracellular compartment, a truncated form of TrkB lacking its tyrosine kinase domain. BDNF-induced platelet aggregation was prevented by inhibitors of Ras-related C3 botulinum toxin substrate 1 (Rac1), protein kinase C, and phosphoinositide 3-kinase. BDNF-stimulated platelets secreted a panel of angiogenic and inflammatory cytokines, which may play a role in maintaining vascular homeostasis. Two families with autism spectrum disorder were found to carry rare missense variants in the BDNF gene. Platelet studies revealed defects in platelet aggregation to low concentrations of collagen, as well as reduced adenosine triphosphate secretion in response to adenosine diphosphate. In summary, circulating BDNF levels appear to regulate platelet activation, aggregation, and secretion through activation of a truncated TrkB receptor and downstream kinase-dependent signaling.


Assuntos
Transtorno do Espectro Autista , Fator Neurotrófico Derivado do Encéfalo , Humanos , Fosfatidilinositol 3-Quinases , Ativação Plaquetária , Agregação Plaquetária
3.
Nature ; 583(7814): 96-102, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32581362

RESUMO

Most patients with rare diseases do not receive a molecular diagnosis and the aetiological variants and causative genes for more than half such disorders remain to be discovered1. Here we used whole-genome sequencing (WGS) in a national health system to streamline diagnosis and to discover unknown aetiological variants in the coding and non-coding regions of the genome. We generated WGS data for 13,037 participants, of whom 9,802 had a rare disease, and provided a genetic diagnosis to 1,138 of the 7,065 extensively phenotyped participants. We identified 95 Mendelian associations between genes and rare diseases, of which 11 have been discovered since 2015 and at least 79 are confirmed to be aetiological. By generating WGS data of UK Biobank participants2, we found that rare alleles can explain the presence of some individuals in the tails of a quantitative trait for red blood cells. Finally, we identified four novel non-coding variants that cause disease through the disruption of transcription of ARPC1B, GATA1, LRBA and MPL. Our study demonstrates a synergy by using WGS for diagnosis and aetiological discovery in routine healthcare.


Assuntos
Internacionalidade , Programas Nacionais de Saúde , Doenças Raras/diagnóstico , Doenças Raras/genética , Sequenciamento Completo do Genoma , Complexo 2-3 de Proteínas Relacionadas à Actina/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Alelos , Bases de Dados Factuais , Eritrócitos/metabolismo , Fator de Transcrição GATA1/genética , Humanos , Fenótipo , Locos de Características Quantitativas , Receptores de Trombopoetina/genética , Medicina Estatal , Reino Unido
4.
JIMD Rep ; 47(1): 9-16, 2019 May.
Artigo em Inglês | MEDLINE | ID: mdl-31240161

RESUMO

BACKGROUND: Brain monoamine vesicular transport disease is an infantile onset neurodevelopmental disorder caused by variants in SLC18A2, which codes for the vesicular monoamine transporter 2 (VMAT2) protein, involved in the transport of monoamines into synaptic vesicles and of serotonin into platelet dense granules. CASE PRESENTATION: The presented case is of a child, born of healthy consanguineous parents, who exhibited hypotonia, mental disability, epilepsy, uncontrolled movements, and gastrointestinal problems. A trial treatment with L-DOPA proved unsuccessful and the exact neurological involvement could not be discerned due to normal metabolic and brain magnetic resonance imaging results.Platelet studies and whole genome sequencing were performed. At age 4, the child's platelets showed a mild aggregation and adenosine triphosphate secretion defect that could be explained by dysmorphic dense granules observed by electron microscopy. Interestingly, the dense granules were almost completely depleted of serotonin. A novel homozygous p.P316A missense variant in VMAT2 was detected in the patient and the consanguineous parents were found to be heterozygous for this variant. Although the presence of VMAT2 on platelet dense granules has been demonstrated before, this is the first report of defective platelet dense granule function related to absent serotonin storage in a patient with VMAT2 deficiency but without obvious clinical bleeding problems. CONCLUSIONS: This study illustrates the homology between serotonin metabolism in brain and platelets, suggesting that these blood cells can be model cells for some pathways relevant for neurological diseases. The literature on VMAT2 deficiency is reviewed.

5.
Platelets ; 30(7): 931-934, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31204551

RESUMO

A germline heterozygous gain-of-function p.E527K variant in tyrosine kinase SRC was previously found to cause thrombocytopenia, myelofibrosis, bleeding, bone pathologies, premature edentulism and mild facial dysmorphia in nine patients of a single pedigree. Because of this variant, SRC loses its self-inhibitory capacity, causing constitutively active SRC expression in platelets. These patients have fewer and heterogeneous-sized platelets that are hyporeactive to collagen. We now report a 5-year-old girl with syndromic thrombocytopenia due to the same SRC-E527K variant that occurs de novo. A bone marrow biopsy, blood smear analysis, platelet aggregations, flow cytometric analysis of P-selectin, SRC expression and tyrosine phosphorylation studies were performed to confirm the similarities between the two families. This study strengthens our previous finding that hyperactive SRC kinase results in mild platelet dysfunction and thrombocytopenia with hypogranular platelets and further expands the clinical description of this syndrome to improve early recognition.


Assuntos
Trombocitopenia/metabolismo , Quinases da Família src/metabolismo , Pré-Escolar , Feminino , Humanos
6.
Haematologica ; 104(5): 1036-1045, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30467204

RESUMO

Sphingolipids are fundamental to membrane trafficking, apoptosis, and cell differentiation and proliferation. KDSR or 3-keto-dihydrosphingosine reductase is an essential enzyme for de novo sphingolipid synthesis, and pathogenic mutations in KDSR result in the severe skin disorder erythrokeratodermia variabilis et progressiva-4 Four of the eight reported cases also had thrombocytopenia but the underlying mechanism has remained unexplored. Here we expand upon the phenotypic spectrum of KDSR deficiency with studies in two siblings with novel compound heterozygous variants associated with thrombocytopenia, anemia, and minimal skin involvement. We report a novel phenotype of progressive juvenile myelofibrosis in the propositus, with spontaneous recovery of anemia and thrombocytopenia in the first decade of life. Examination of bone marrow biopsies showed megakaryocyte hyperproliferation and dysplasia. Megakaryocytes obtained by culture of CD34+ stem cells confirmed hyperproliferation and showed reduced proplatelet formation. The effect of KDSR insufficiency on the sphingolipid profile was unknown, and was explored in vivo and in vitro by a broad metabolomics screen that indicated activation of an in vivo compensatory pathway that leads to normalization of downstream metabolites such as ceramide. Differentiation of propositus-derived induced pluripotent stem cells to megakaryocytes followed by expression of functional KDSR showed correction of the aberrant cellular and biochemical phenotypes, corroborating the critical role of KDSR in proplatelet formation. Finally, Kdsr depletion in zebrafish recapitulated the thrombocytopenia and showed biochemical changes similar to those observed in the affected siblings. These studies support an important role for sphingolipids as regulators of cytoskeletal organization during megakaryopoiesis and proplatelet formation.


Assuntos
Oxirredutases do Álcool/deficiência , Plaquetas/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Megacariócitos/patologia , Esfingolipídeos/metabolismo , Trombocitopenia/etiologia , Oxirredutases do Álcool/genética , Animais , Plaquetas/metabolismo , Diferenciação Celular , Células Cultivadas , Criança , Feminino , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Megacariócitos/metabolismo , Metabolômica , Mutação , Linhagem , Prognóstico , Trombocitopenia/metabolismo , Trombocitopenia/patologia , Peixe-Zebra
7.
J Allergy Clin Immunol ; 142(2): 630-646, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29391254

RESUMO

BACKGROUND: Roifman syndrome is a rare inherited disorder characterized by spondyloepiphyseal dysplasia, growth retardation, cognitive delay, hypogammaglobulinemia, and, in some patients, thrombocytopenia. Compound heterozygous variants in the small nuclear RNA gene RNU4ATAC, which is necessary for U12-type intron splicing, were identified recently as driving Roifman syndrome. OBJECTIVE: We studied 3 patients from 2 unrelated kindreds harboring compound heterozygous or homozygous stem II variants in RNU4ATAC to gain insight into the mechanisms behind this disorder. METHODS: We systematically profiled the immunologic and hematologic compartments of the 3 patients with Roifman syndrome and performed RNA sequencing to unravel important splicing defects in both cell lineages. RESULTS: The patients exhibited a dramatic reduction in B-cell numbers, with differentiation halted at the transitional B-cell stage. Despite abundant B-cell activating factor availability, development past this B-cell activating factor-dependent stage was crippled, with disturbed minor splicing of the critical mitogen-activated protein kinase 1 signaling component. In the hematologic compartment patients with Roifman syndrome demonstrated defects in megakaryocyte differentiation, with inadequate generation of proplatelets. Platelets from patients with Roifman syndrome were rounder, with increased tubulin and actin levels, and contained increased α-granule and dense granule markers. Significant minor intron retention in 354 megakaryocyte genes was observed, including DIAPH1 and HPS1, genes known to regulate platelet and dense granule formation, respectively. CONCLUSION: Together, our results provide novel molecular and cellular data toward understanding the immunologic and hematologic features of Roifman syndrome.


Assuntos
Linfócitos B/fisiologia , Plaquetas/fisiologia , Cardiomiopatias/genética , Síndromes de Imunodeficiência/genética , Megacariócitos/fisiologia , Deficiência Intelectual Ligada ao Cromossomo X/genética , Proteína Quinase 1 Ativada por Mitógeno/genética , Osteocondrodisplasias/genética , Células Precursoras de Linfócitos B/fisiologia , RNA Nuclear Pequeno/genética , Doenças Retinianas/genética , Adolescente , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Criança , Pré-Escolar , Humanos , Lactente , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Linhagem , Doenças da Imunodeficiência Primária , Processamento de Proteína/genética , Transdução de Sinais/genética , Sequenciamento do Exoma
8.
Haematologica ; 102(4): 695-706, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28082341

RESUMO

Gray platelet syndrome is named after the gray appearance of platelets due to the absence of α-granules. It is caused by recessive mutations in NBEAL2, resulting in macrothrombocytopenia and myelofibrosis. Though using the term gray platelets for GATA1 deficiency has been debated, a reduced number of α-granules has been described for macrothrombocytopenia due to GATA1 mutations. We compared platelet size and number of α-granules for two NBEAL2 and two GATA1-deficient patients and found reduced numbers of α-granules for all, with the defect being more pronounced for NBEAL2 deficiency. We further hypothesized that the granule defect for GATA1 is due to a defective control of NBEAL2 expression. Remarkably, platelets from two patients, and Gata1-deficient mice, expressed almost no NBEAL2. The differentiation of GATA1 patient-derived CD34+ stem cells to megakaryocytes showed defective proplatelet and α-granule formation with strongly reduced NBEAL2 protein and ribonucleic acid expression. Chromatin immunoprecipitation sequencing revealed 5 GATA binding sites in a regulatory region 31 kb upstream of NBEAL2 covered by a H3K4Me1 mark indicative of an enhancer locus. Luciferase reporter constructs containing this region confirmed its enhancer activity in K562 cells, and mutagenesis of the GATA1 binding sites resulted in significantly reduced enhancer activity. Moreover, DNA binding studies showed that GATA1 and GATA2 physically interact with this enhancer region. GATA1 depletion using small interfering ribonucleic acid in K562 cells also resulted in reduced NBEAL2 expression. In conclusion, we herein show a long-distance regulatory region with GATA1 binding sites as being a strong enhancer for NBEAL2 expression.


Assuntos
Proteínas Sanguíneas/genética , Elementos Facilitadores Genéticos , Fator de Transcrição GATA1/metabolismo , Regulação da Expressão Gênica , Alelos , Plaquetas/metabolismo , Diferenciação Celular/genética , Fator de Transcrição GATA1/deficiência , Fator de Transcrição GATA1/genética , Expressão Gênica , Genes Recessivos , Genes Reporter , Genes Ligados ao Cromossomo X , Estudos de Associação Genética , Predisposição Genética para Doença , Homozigoto , Humanos , Megacariócitos/citologia , Megacariócitos/metabolismo , Megacariócitos/ultraestrutura , Mutação , Fenótipo , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno/genética , Trombocitopenia/sangue , Trombocitopenia/genética , Trombocitopenia/patologia
9.
Sci Transl Med ; 8(328): 328ra30, 2016 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-26936507

RESUMO

The Src family kinase (SFK) member SRC is a major target in drug development because it is activated in many human cancers, yet deleterious SRC germline mutations have not been reported. We used genome sequencing and Human Phenotype Ontology patient coding to identify a gain-of-function mutation in SRC causing thrombocytopenia, myelofibrosis, bleeding, and bone pathologies in nine cases. Modeling of the E527K substitution predicts loss of SRC's self-inhibitory capacity, which we confirmed with in vitro studies showing increased SRC kinase activity and enhanced Tyr(419) phosphorylation in COS-7 cells overexpressing E527K SRC. The active form of SRC predominates in patients' platelets, resulting in enhanced overall tyrosine phosphorylation. Patients with myelofibrosis have hypercellular bone marrow with trilineage dysplasia, and their stem cells grown in vitro form more myeloid and megakaryocyte (MK) colonies than control cells. These MKs generate platelets that are dysmorphic, low in number, highly variable in size, and have a paucity of α-granules. Overactive SRC in patient-derived MKs causes a reduction in proplatelet formation, which can be rescued by SRC kinase inhibition. Stem cells transduced with lentiviral E527K SRC form MKs with a similar defect and enhanced tyrosine phosphorylation levels. Patient-derived and E527K-transduced MKs show Y419 SRC-positive stained podosomes that induce altered actin organization. Expression of mutated src in zebrafish recapitulates patients' blood and bone phenotypes. Similar studies of platelets and MKs may reveal the mechanism underlying the severe bleeding frequently observed in cancer patients treated with next-generation SFK inhibitors.


Assuntos
Osso e Ossos/patologia , Hemorragia/genética , Mutação/genética , Mielofibrose Primária/genética , Trombocitopenia/genética , Quinases da Família src/genética , Animais , Plaquetas/patologia , Células COS , Chlorocebus aethiops , Feminino , Hematopoese , Hemorragia/complicações , Humanos , Masculino , Linhagem , Fenótipo , Mielofibrose Primária/complicações , Trombocitopenia/complicações , Transfecção , Peixe-Zebra
10.
J Proteomics ; 86: 53-69, 2013 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-23681173

RESUMO

Alternating hemiplegia of childhood (AHC) is a rare syndrome with repeated hemiplegic episodes, paroxysmal events and global neurological impairment. Recently, heterozygous de novo ATP1A3 missense mutations have been identified in AHC patients, but the underlying pathogenesis mechanism remains unknown. Mutation analysis of ATP1A3 in 9 unrelated AHC cases revealed mostly D801N or E815K variants. As platelets represent a good cellular model to study defects in neuropathologies, morphological and functional experiments were performed in these subjects. Platelets from the AHC patients presented with structural and functional abnormalities of granules positive for the lysosomal marker CD63. Similar structural granule abnormalities were detected in patients' fibroblasts. Proteomic analysis of platelets and fibroblasts showed a total of 93 differentially expressed proteins in AHC mainly involved in metabolism. Interestingly, 7 of these proteins were detected in both cell types, including the lysosomal protein cathepsin. AHC fibroblasts revealed significantly increased levels of activated cathepsin B, which induces a stronger activation of apoptosis. Our study is the first to link ATP1A3 defects in AHC to a platelet and fibroblast lysosomal defect with evidence of increased apoptosis. Further studies are needed to define how this lysosomal defect is related to decreased ATPase activity. Biological Significance Only recently, the genetic cause of AHC was identified as heterozygous ATP1A3 mutations, but the underlying pathophysiological mechanism still remains unknown. By performing functional, morphological and proteomic studies in AHC patients we found a structural and functional granule defect in AHC platelets and fibroblasts that was specifically found in granules positive for the lysosomal marker CD63. In particular, proteomics identified several differentially expressed proteins in fibroblasts and platelets from AHC cases that are predicted to have an important role in cell function and maintenance, a pathway typically attributed to lysosomes. The lysosomal protein cathepsin was found to be differentially expressed in both platelets and fibroblasts of AHC patients, inducing a stronger activation of mainly the intrinsic apoptosis. Despite the precise mechanism for the increased lysosomal cathepsin B-dependent apoptosis detected in AHC in relation to impaired ATP1A3 deserves further studies, we could here show some evidence for a defective regulation of apoptosis in AHC, a disease that still has no biochemical or neuroradiological parameters for diagnosis.


Assuntos
Plaquetas/metabolismo , Catepsina B/metabolismo , Hemiplegia/genética , ATPase Trocadora de Sódio-Potássio/genética , Apoptose/genética , Catepsina B/genética , Eletroforese em Gel Bidimensional , Feminino , Fibroblastos/metabolismo , Hemiplegia/metabolismo , Hemiplegia/patologia , Humanos , Lactente , Recém-Nascido , Lisossomos/enzimologia , Masculino , Transcriptoma
11.
Genet Med ; 15(1): 55-63, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22899094

RESUMO

PURPOSE: Aquaporin 7 (AQP7) belongs to the aquaglyceroporin family, which transports glycerol and water. AQP7-deficient mice develop obesity, insulin resistance, and hyperglyceroluria. However, AQP7's pathophysiologic role in humans is not yet known. METHODS: Three children with psychomotor retardation and hyperglyceroluria were screened for AQP7 mutations. The children were from unrelated families. Urine and plasma glycerol levels were measured using a three-step enzymatic approach. Platelet morphology and function were studied using electron microscopy, aggregations, and adenosine triphosphate (ATP) secretion tests. RESULTS: The index patients were homozygous for AQP7 G264V, which has previously been shown to inhibit transport of glycerol in Xenopus oocytes. We also detected a subclinical platelet secretion defect with reduced ATP secretion, and the absence of a secondary aggregation wave after epinephrine stimulation. Electron microscopy revealed round platelets with centrally located granules. Immunostaining showed AQP7 colocalization, with dense granules that seemed to be released after strong platelet activation. Healthy relatives of these patients, who were homozygous (not heterozygous) for G264V, also had hyperglyceroluria and platelet granule abnormalities. CONCLUSION: The discovery of an association between urine glycerol loss and a platelet secretion defect is a novel one, and our findings imply the involvement of AQPs in platelet secretion. Additional studies are needed to define whether AQP7 G264V is also a risk factor for mental disability.


Assuntos
Aquaporinas/genética , Transtornos Plaquetários/genética , Homozigoto , Mutação , Adolescente , Adulto , Substituição de Aminoácidos , Aquaporina 3/genética , Aquaporinas/metabolismo , Plaquetas/metabolismo , Plaquetas/ultraestrutura , Criança , Pré-Escolar , Códon , Feminino , Glicerol/sangue , Glicerol/urina , Humanos , Lactente , Masculino , Pessoa de Meia-Idade , Linhagem , Transporte Proteico , Adulto Jovem
12.
PLoS One ; 7(6): e38579, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22679513

RESUMO

BACKGROUND: Pseudohypoparathyroidism (PHP) indicates a group of heterogeneous disorders whose common feature is represented by impaired signaling of hormones that activate Gsalpha, encoded by the imprinted GNAS gene. PHP-Ib patients have isolated Parathormone (PTH) resistance and GNAS epigenetic defects while PHP-Ia cases present with hormone resistance and characteristic features jointly termed as Albright's Hereditary Osteodystrophy (AHO) due to maternally inherited GNAS mutations or similar epigenetic defects as found for PHP-Ib. Pseudopseudohypoparathyroidism (PPHP) patients with an AHO phenotype and no hormone resistance and progressive osseous heteroplasia (POH) cases have inactivating paternally inherited GNAS mutations. METHODOLOGY/PRINCIPAL FINDINGS: We here describe 17 subjects with an AHO-like phenotype that could be compatible with having PPHP but none of them carried Gsalpha mutations. Functional platelet studies however showed an obvious Gs hypofunction in the 13 patients that were available for testing. Methylation for the three differentially methylated GNAS regions was quantified via the Sequenom EpiTYPER. Patients showed significant hypermethylation of the XL amplicon compared to controls (36 ± 3 vs. 29 ± 3%; p<0.001); a pattern that is reversed to XL hypomethylation found in PHPIb. Interestingly, XL hypermethylation was associated with reduced XLalphaS protein levels in the patients' platelets. Methylation for NESP and ExonA/B was significantly different for some but not all patients, though most patients have site-specific CpG methylation abnormalities in these amplicons. Since some AHO features are present in other imprinting disorders, the methylation of IGF2, H19, SNURF and GRB10 was quantified. Surprisingly, significant IGF2 hypermethylation (20 ± 10 vs. 14 ± 7%; p<0.05) and SNURF hypomethylation (23 ± 6 vs. 32 6%; p<0.001) was found in patients vs. controls, while H19 and GRB10 methylation was normal. CONCLUSION/SIGNIFICANCE: In conclusion, this is the first report of methylation defects including GNAS in patients with an AHO-like phenotype without endocrinological abnormalities. Additional studies are still needed to correlate the methylation defect with the clinical phenotype.


Assuntos
Metilação de DNA/genética , Displasia Fibrosa Poliostótica/genética , Cromograninas , Proteína Adaptadora GRB10/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Humanos , Fator de Crescimento Insulin-Like II/genética , Proteínas Nucleares/genética , Fenótipo , Pseudo-Hipoparatireoidismo
13.
Nat Genet ; 44(4): 435-9, S1-2, 2012 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-22366785

RESUMO

The exon-junction complex (EJC) performs essential RNA processing tasks. Here, we describe the first human disorder, thrombocytopenia with absent radii (TAR), caused by deficiency in one of the four EJC subunits. Compound inheritance of a rare null allele and one of two low-frequency SNPs in the regulatory regions of RBM8A, encoding the Y14 subunit of EJC, causes TAR. We found that this inheritance mechanism explained 53 of 55 cases (P < 5 × 10(-228)) of the rare congenital malformation syndrome. Of the 53 cases with this inheritance pattern, 51 carried a submicroscopic deletion of 1q21.1 that has previously been associated with TAR, and two carried a truncation or frameshift null mutation in RBM8A. We show that the two regulatory SNPs result in diminished RBM8A transcription in vitro and that Y14 expression is reduced in platelets from individuals with TAR. Our data implicate Y14 insufficiency and, presumably, an EJC defect as the cause of TAR syndrome.


Assuntos
Predisposição Genética para Doença , Proteínas de Ligação a RNA/genética , Trombocitopenia/genética , Deformidades Congênitas das Extremidades Superiores/genética , Regiões 5' não Traduzidas/genética , Adolescente , Adulto , Sequência de Aminoácidos , Animais , Sequência de Bases , Criança , Pré-Escolar , Síndrome Congênita de Insuficiência da Medula Óssea , Feminino , Variação Genética , Humanos , Lactente , Recém-Nascido , Masculino , Mutação , Contagem de Plaquetas , Polimorfismo de Nucleotídeo Único , Rádio (Anatomia)/anormalidades , Alinhamento de Sequência , Análise de Sequência de DNA , Trombocitopenia/congênito , Adulto Jovem , Peixe-Zebra/genética
14.
FASEB J ; 26(5): 2125-36, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22308195

RESUMO

RGS18 was originally identified as a R4 subfamily member of regulators of G-protein signaling (RGS) with specific expression in hematopoietic progenitors, myeloerythroid cells, and megakaryocytes, though its physiological role in hematopoiesis remained unknown. Here, we show that lentiviral RGS18 overexpression during differentiation of mouse Sca1(+) hematopoietic stem cells induced a 50% increase of megakaryocyte proliferation. RGS18 depletion in zebrafish results in thrombocytopenia, as 66 to 88% of the embryos lack thrombocytes after injection of an ATG or splice-blocking morpholino, respectively. These embryos have no defects in early hematopoiesis, erythropoiesis, or leukocyte number and migration. In addition, all RGS18 depleted embryos have curly tails and an almost absent response to acoustic stimuli. In situ hybridization in zebrafish, Xenopus, and mouse embryos shows RGS18 expression in thrombocytes and/or hematological tissues but also in brain and otic vesicles. RGS18 interferes with development of cilia in hair cells of the inner ear and neuromast cells. On the basis of literature evidence that RGS-R4 members interact with the G-protein-modulated Wnt/calcium pathway, Wnt5b- but not Wnt5a-depleted embryos phenocopy all RGS18 knockdown effects. In summary, our study is the first to show that RGS18 regulates megakaryopoiesis but also reveals its unexpected role in ciliogenesis, at least in lower vertebrates, via interference with Wnt signaling.


Assuntos
Cílios/fisiologia , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Mecanotransdução Celular , Megacariócitos/fisiologia , Animais , Sequência de Bases , Primers do DNA , Citometria de Fluxo , Técnicas de Silenciamento de Genes , Peptídeos e Proteínas de Sinalização Intracelular/genética , Ligantes , Camundongos , Proteínas RGS , Proteínas Wnt/metabolismo
15.
J Hum Genet ; 57(4): 277-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22277900

RESUMO

Albright's hereditary osteodystrophy (AHO) is characterized by short stature, round face, calcifications, obesity, brachydactyly and intellectual disability. AHO without hormone resistance is called pseudopseudohypoparathyroidism (PPHP), a rare clinical condition difficult to diagnose with highly variable features. PPHP is caused by paternally inherited loss-of-function mutations in the GNAS. Patients with 2q37 microdeletions or HDAC4 mutations are also defined as having an AHO-like phenotype with normal stimulatory G (Gs) function. We have studied 256 patients with AHO features but no other diagnosis. Their platelet Gs activity was determined via the aggregation-inhibition test showing Gs hypo- or hyperfuncton in 24% and 15% of the patients, respectively. Before initiating with detailed (epi)genetic GNAS studies, we here wanted to excluded copy number variants (CNVs) in GNAS as cause of AHO with a novel large-scale screening technique. Multiplex amplicon quantification (MAQ) for CNVs screening was developed for the 20q13.3 region including GNAS and potential long-range imprinting control elements such as STX16. This is the first large-scale GNAS CNV study in patients with common AHO features but no CNVs were detected. In conclusion, CNVs in the GNAS region are not likely to cause an AHO-like phenotype with or without abnormal platelet Gs activity. Future studies will be undertaken to find out whether these AHO patients with abnormal Gs function are characterized by GNAS coding or methylation defects.


Assuntos
Variações do Número de Cópias de DNA , Displasia Fibrosa Poliostótica/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Plaquetas/patologia , Cromograninas , Deleção Cromossômica , Cromossomos Humanos Par 20/genética , Epigênese Genética , Displasia Fibrosa Poliostótica/patologia , Testes Genéticos/métodos , Humanos , Fenótipo , Agregação Plaquetária , Reação em Cadeia da Polimerase/métodos , Pseudo-Hipoparatireoidismo , Sintaxina 16/genética
16.
Pediatr Res ; 67(3): 314-9, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19924028

RESUMO

We describe a patient, who developed during the first week of life petechiae and hematomas caused by severe thrombocytopenia and gastrointestinal bleeding due to multiple small gastric hemangiomata. Bone marrow examination showed hypermegakaryocytosis and dysmegakaryopoiesis. Alloimmune thrombocytopenia was excluded. Only 3 y later, platelet counts normalized and bleedings disappeared but small skin hemangiomata remained. Electron microscopy showed enlarged round platelets with a paucity of alpha granules similar as in GATA1-deficient patients but no GATA1 mutation was found. Immunoblot analysis showed a strong interaction between patient Igs and recombinant GATA1, GATA2, and the N finger (Nf) of GATA1. The lymphocyte transformation test with recombinant GATA1Nf was positive. In vitro culturing of normal CD34 cells with purified patient Igs showed a decreased number of megakaryocyte colonies but an increased overall size of the colonies compared with control Igs. Mice injected with patient Igs showed a reduced platelet count compared with mice injected with control Igs. Thrombopoiesis was also reduced after injection of patient Igs in transgenic zebrafish compared with control Igs. In conclusion, this study is the first report of an anti-GATA1 autoantibody leading to severe thrombocytopenia and gastrointestinal bleeding from multiple pinpoint hemangiomata.


Assuntos
Autoanticorpos/sangue , Autoimunidade , Fator de Transcrição GATA1/imunologia , Hemorragia Gastrointestinal/imunologia , Neoplasias Gastrointestinais/imunologia , Hemangioma/imunologia , Trombocitopenia/imunologia , Animais , Animais Geneticamente Modificados , Células Cultivadas , Pré-Escolar , Feminino , Fator de Transcrição GATA1/genética , Hemorragia Gastrointestinal/sangue , Hemorragia Gastrointestinal/terapia , Neoplasias Gastrointestinais/sangue , Neoplasias Gastrointestinais/terapia , Hemangioma/sangue , Humanos , Recém-Nascido , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Contagem de Plaquetas , Índice de Gravidade de Doença , Trombocitopenia/sangue , Trombocitopenia/terapia , Trombopoese , Transfecção , Peixe-Zebra/genética
17.
J Clin Endocrinol Metab ; 93(12): 4844-9, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18796523

RESUMO

CONTEXT: Pseudohypoparathyroidism type Ia and pseudopseudohypoparathyroidism are characterized by Albright's hereditary osteodystrophy (AHO), respectively, with and without hormone resistance. Both clinical conditions result from decreased expression or function of the alpha-subunit of the stimulatory G protein (Gsalpha) of adenylyl cyclase due to heterozygous inactivating mutations in GNAS. Homozygous GNAS defects have not been described. OBJECTIVE: A genetic and functional GNAS study was undertaken in a boy with morbid obesity (body mass index Z-score of 5 at the age of 3 yr, with a body fat fraction of 40%, which is more than twice normal), TSH resistance, pseudohypoparathyroidism, and a prothrombotic state. RESULTS: The boy was found to be a first case with a compound heterozygous GNAS defect: a de novo R231C mutation on the paternal allele and on the other allele a maternally inherited unique combination of three C to T nucleotide substitutions in exon 7 (I185I), intron 7 (IVS7 + 31), and exon 13 (N371N) leading to aberrant splicing of GNAS. Platelets of this boy displayed a pronounced Gsalpha hypofunction and were spontaneously hyperreactive resulting in a prothrombotic state due to extremely low cAMP levels. CONCLUSION: This report expands the human GNAS genotype-phenotype spectrum to include compound heterozygosity and a prothrombotic state.


Assuntos
Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Obesidade Mórbida/genética , Pseudo-Hipoparatireoidismo/genética , Trombose/genética , Síndrome da Resistência aos Hormônios Tireóideos/genética , Adiposidade/genética , Adiposidade/fisiologia , Western Blotting , Índice de Massa Corporal , Peso Corporal , Cromograninas , AMP Cíclico/sangue , DNA/genética , Fibroblastos/metabolismo , Humanos , Hiperfosfatemia/etiologia , Hipocalcemia/etiologia , Lactente , Masculino , Mutação , Obesidade Mórbida/complicações , Obesidade Mórbida/patologia , Agregação Plaquetária , Testes de Função Plaquetária , Pseudo-Hipoparatireoidismo/complicações , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Trombose/complicações , Síndrome da Resistência aos Hormônios Tireóideos/complicações
18.
Hum Mol Genet ; 17(3): 357-66, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-17981813

RESUMO

Controversy exists regarding the cause of the significantly increased blood loss during spinal surgery in Duchenne muscular dystrophy (DMD) patients compared with similar surgery in other patients. DMD is caused by a mutation in the cytoskeletal dystrophin, which binds to extracellular matrix laminin and which has been described as a G-protein-coupled receptor. We hypothesized that disturbed cytoskeleton organization in DMD patients would alter Gs protein and collagen signalling in platelets, leading to dysfunctional platelets and a haemorrhagic tendency during surgery. In the present study, we found that platelets and skin fibroblasts, respectively, express the Dp71 and Dp116 dystrophin isoforms. Absent or decreased expression of these isoforms in DMD patients correlates with significant Gs alpha upregulation. Moreover, dysfunctional dystrophin in these cells is accompanied with increased Gs signalling and higher cAMP levels after Gs stimulation. Functional analysis showed that DMD platelets responded slower to collagen with an extensive shape change in the aggregometer and with a significantly reduced platelet adhesion to coated collagen under flow. The decreased collagen activation was shown to result from both Gs activation and cytoskeletal disruption and not from decreased expression of platelet membrane receptors or impaired von Willebrand factor (vWF) activity. In conclusion, DMD platelets have a disorganized cytoskeleton and manifest Gs hyperactivity and reduced platelet collagen reactivity. Their increased bleeding during surgery will, at least partly, result from the increased platelet Gs activity after the release of natural Gs agonists as prostacyclin during surgery and an ineffective reactivity to collagen.


Assuntos
Plaquetas/metabolismo , Distrofina/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/sangue , Distrofia Muscular de Duchenne/sangue , Distrofia Muscular de Duchenne/genética , Fusão Vertebral/efeitos adversos , Perda Sanguínea Cirúrgica/fisiopatologia , Plaquetas/efeitos dos fármacos , Plaquetas/ultraestrutura , Estudos de Casos e Controles , Criança , Colágeno/farmacologia , Citoesqueleto/metabolismo , Humanos , Técnicas In Vitro , Masculino , Mutação , Ativação Plaquetária/efeitos dos fármacos , Ativação Plaquetária/genética , Transdução de Sinais
19.
Blood ; 111(4): 1885-93, 2008 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-18000164

RESUMO

Megakaryocytes and platelets express the Gs-coupled VPAC1 receptor, for which the pituitary adenylyl cyclase-activating peptide (PACAP) and the vasointestinal peptide (VIP) are agonists. We here demonstrate a regulatory role for VPAC1 signaling during megakaryopoiesis. A total of 2 patients with trisomy 18p with PACAP overexpression and transgenic mice overexpressing PACAP in megakaryocytes have thrombopathy, a mild thrombocytopenia, and a reduced number of mature megakaryocytes in their bone marrow. In vitro differentiation of hematopoietic stem cells from the patient and transgenic mice shows a reduced number of megakaryocyte colonies compared with controls. The addition of PACAP, VIP, or the adenylyl cyclase activator forskolin to CD34(+) cells inhibits megakaryocyte differentiation. In contrast, neutralizing monoclonal anti-PACAP (PP1A4) or anti-VPAC1 (23A11) antibodies inhibit cAMP formation and stimulate megakaryopoiesis in a thrombopoietin-independent manner. Moreover, wild-type mice obtain an increased platelet count after subcutaneous injection of PP1A4 or 23A11. These antibodies also elevate platelet numbers in animal models of myelosuppressive therapy and in GATA1-deficient mice with congenital thrombocytopenia. Furthermore, 23A11 stimulates the in vitro megakaryocyte differentiation of both normal and GATA1-deficient human CD34(+) cells. Together, our data strongly suggest that VPAC1 signaling tempers normal megakaryopoiesis, and that inhibition of this pathway stimulates megakaryocyte differentiation, enhancing platelet recovery after myelosuppressive therapy and in GATA1 deficiency.


Assuntos
Megacariócitos/citologia , Megacariócitos/fisiologia , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/genética , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/genética , Animais , Animais Geneticamente Modificados , Antígenos CD/análise , Antígenos CD34/análise , Linhagem Celular Tumoral , Cromossomos Humanos Par 18 , AMP Cíclico/fisiologia , Humanos , Camundongos , Camundongos Transgênicos , Polipeptídeo Hipofisário Ativador de Adenilato Ciclase/fisiologia , Coelhos , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/fisiologia , Trombocitopenia/genética , Trissomia/genética
20.
Blood ; 106(7): 2356-62, 2005 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-15956286

RESUMO

The discoid form of platelets is maintained by a marginal band of tightly coiled microtubules. beta1-tubulin is the major isoform within platelet and megakaryocyte microtubules. In 24.2% of 33 unrelated inherited macrothrombocytopenia patients and in 10.6% of 272 subjects of a healthy population a P for Q substitution in beta1-tubulin was found in the highly conserved residue 43. Heterozygous carriers of the Q43P variant showed a reduced platelet protein beta1-tubulin expression. Transfection of green fluorescent protein (GFP)-tagged Q43P beta1-tubulin in megakaryocytic MEG01 cells resulted in a disturbed tubulin organization. Electron microscopy revealed enlarged spherocytic platelets with a disturbed marginal band and organelle-free zones. In addition, platelets with the Q43P beta1-tubulin variant had reduced adenosine triphosphate (ATP) secretion, thrombin receptor activating peptide (TRAP)-induced aggregation and collagen adhesion. The prevalence of the Q43P beta1-tubulin variant was also 2 times higher (odds ratio, [OR] = 2.1;95% confidence interval [CI], 1.22-3.59) among control subjects than among patients with cardiovascular disease (10.4% versus 5.2%, P < .001). By analyzing this protective factor in men and women separately, this association was only found in men. This study thus presents the functional consequences of the platelet Q43P beta1-tubulin substitution that is frequent in the healthy population and may protect men against arterial thrombosis.


Assuntos
Plaquetas/citologia , Doenças Cardiovasculares/sangue , Doenças Cardiovasculares/genética , Predisposição Genética para Doença , Polimorfismo Genético , Trombocitopenia/genética , Tubulina (Proteína)/genética , Trifosfato de Adenosina/metabolismo , Adulto , Idoso , Sequência de Aminoácidos , Sequência de Bases , Plaquetas/fisiologia , Plaquetas/ultraestrutura , Adesão Celular , Colágeno/metabolismo , Análise Mutacional de DNA , DNA Complementar/metabolismo , Feminino , Genótipo , Proteínas de Fluorescência Verde/metabolismo , Heterozigoto , Humanos , Immunoblotting , Masculino , Megacariócitos/metabolismo , Microscopia Eletrônica , Microscopia de Contraste de Fase , Pessoa de Meia-Idade , Dados de Sequência Molecular , Razão de Chances , Perfusão , Adesividade Plaquetária , Isoformas de Proteínas , Fatores Sexuais , Trombocitopenia/sangue , Trombose/genética , Trombose/prevenção & controle , Transfecção , Tubulina (Proteína)/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA