Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 137
Filter
1.
Semin Thromb Hemost ; 2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38499192

ABSTRACT

Glanzmann thrombasthenia (GT) is the most common inherited platelet disorder (IPD) with mucocutaneous bleeding and a failure of platelets to aggregate when stimulated. The molecular cause is insufficient or defective αIIbß3, an integrin encoded by the ITGA2B and ITGB3 genes. On activation αIIbß3 undergoes conformational changes and binds fibrinogen (Fg) and other proteins to join platelets in the aggregate. The application of next-generation sequencing (NGS) to patients with IPDs has accelerated genotyping for GT; progress accompanied by improved mutation curation. The evaluation by NGS of variants in other hemostasis and vascular genes is a major step toward understanding why bleeding varies so much between patients. The recently discovered role for glycoprotein VI in thrombus formation, through its binding to fibrin and surface-bound Fg, may offer a mechanosensitive back-up for αIIbß3, especially at sites of inflammation. The setting up of national networks for IPDs and GT is improving patient care. Hematopoietic stem cell therapy provides a long-term cure for severe cases; however, prophylaxis by monoclonal antibodies designed to accelerate fibrin formation at injured sites in the vasculature is a promising development. Gene therapy using lentil-virus vectors remains a future option with CRISPR/Cas9 technologies offering a promising alternative route.

2.
Haematologica ; 108(3): 772-784, 2023 03 01.
Article in English | MEDLINE | ID: mdl-35638551

ABSTRACT

Vascular homeostasis is impaired in various diseases thereby contributing to the progression of their underlying pathologies. The endothelial immediate early gene Apolipoprotein L domain-containing 1 (APOLD1) helps to regulate endothelial function. However, its precise role in endothelial cell biology remains unclear. We have localized APOLD1 to endothelial cell contacts and to Weibel-Palade bodies (WPB) where it associates with von Willebrand factor (VWF) tubules. Silencing of APOLD1 in primary human endothelial cells disrupted the cell junction-cytoskeletal interface, thereby altering endothelial permeability accompanied by spontaneous release of WPB contents. This resulted in an increased presence of WPB cargoes, notably VWF and angiopoietin-2 in the extracellular medium. Autophagy flux, previously recognized as an essential mechanism for the regulated release of WPB, was impaired in the absence of APOLD1. In addition, we report APOLD1 as a candidate gene for a novel inherited bleeding disorder across three generations of a large family in which an atypical bleeding diathesis was associated with episodic impaired microcirculation. A dominant heterozygous nonsense APOLD1:p.R49* variant segregated to affected family members. Compromised vascular integrity resulting from an excess of plasma angiopoietin-2, and locally impaired availability of VWF may explain the unusual clinical profile of APOLD1:p.R49* patients. In summary, our findings identify APOLD1 as an important regulator of vascular homeostasis and raise the need to consider testing of endothelial cell function in patients with inherited bleeding disorders without apparent platelet or coagulation defects.


Subject(s)
Vascular Diseases , Weibel-Palade Bodies , Humans , von Willebrand Factor/genetics , Endothelial Cells/physiology , Angiopoietin-2/genetics , Exocytosis/physiology , Hemostasis , Intercellular Junctions
3.
Blood ; 135(14): 1146-1160, 2020 04 02.
Article in English | MEDLINE | ID: mdl-32040544

ABSTRACT

Increasing evidence suggests that platelets play a predominant role in colon and breast cancer metastasis, but the underlying molecular mechanisms remain elusive. Glycoprotein VI (GPVI) is a platelet-specific receptor for collagen and fibrin that triggers platelet activation through immunoreceptor tyrosine-based activation motif (ITAM) signaling and thereby regulates diverse functions, including platelet adhesion, aggregation, and procoagulant activity. GPVI has been proposed as a safe antithrombotic target, because its inhibition is protective in models of arterial thrombosis, with only minor effects on hemostasis. In this study, the genetic deficiency of platelet GPVI in mice decreased experimental and spontaneous metastasis of colon and breast cancer cells. Similar results were obtained with mice lacking the spleen-tyrosine kinase Syk in platelets, an essential component of the ITAM-signaling cascade. In vitro and in vivo analyses supported that mouse, as well as human GPVI, had platelet adhesion to colon and breast cancer cells. Using a CRISPR/Cas9-based gene knockout approach, we identified galectin-3 as the major counterreceptor of GPVI on tumor cells. In vivo studies demonstrated that the interplay between platelet GPVI and tumor cell-expressed galectin-3 uses ITAM-signaling components in platelets and favors the extravasation of tumor cells. Finally, we showed that JAQ1 F(ab')2-mediated inhibition of GPVI efficiently impairs platelet-tumor cell interaction and tumor metastasis. Our study revealed a new mechanism by which platelets promote the metastasis of colon and breast cancer cells and suggests that GPVI represents a promising target for antimetastatic therapies.


Subject(s)
Blood Platelets/pathology , Breast Neoplasms/pathology , Colonic Neoplasms/pathology , Galectin 3/metabolism , Platelet Membrane Glycoproteins/metabolism , Animals , Blood Platelets/metabolism , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Movement , Colonic Neoplasms/metabolism , Female , Humans , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Metastasis/pathology , Platelet Activation , Platelet Membrane Glycoproteins/genetics , Protein Interaction Maps
4.
Haematologica ; 106(2): 337-350, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33147934

ABSTRACT

Inherited platelet disorders resulting from platelet function defects and a normal platelet count cause a moderate or severe bleeding diathesis. Since the description of Glanzmann thrombasthenia resulting from defects of ITGA2B and ITGB3, new inherited platelet disorders have been discovered, facilitated by the use of high throughput sequencing and genomic analyses. Defects of RASGRP2 and FERMT3 responsible for severe bleeding syndromes and integrin activation have illustrated the critical role of signaling molecules. Important are mutations of P2RY12 encoding the major ADP receptor causal for an inherited platelet disorder with inheritance characteristics that depend on the variant identified. Interestingly, variants of GP6 encoding the major subunit of the collagen receptor GPVI/FcRγ associate only with mild bleeding. The numbers of genes involved in dense granule defects including Hermansky-Pudlak and Chediak Higashi syndromes continue to progress and are updated. The ANO6 gene encoding a Ca2+-activated ion channel required for phospholipid scrambling is responsible for the rare Scott syndrome and decreased procoagulant activity. A novel EPHB2 defect in a familial bleeding syndrome demonstrates a role for this tyrosine kinase receptor independent of the classical model of its interaction with ephrins. Such advances highlight the large diversity of variants affecting platelet function but not their production, despite the difficulties in establishing a clear phenotype when few families are affected. They have provided insights into essential pathways of platelet function and have been at the origin of new and improved therapies for ischemic disease. Nevertheless, many patients remain without a diagnosis and requiring new strategies that are now discussed.


Subject(s)
Blood Platelet Disorders , Thrombasthenia , Blood Platelet Disorders/diagnosis , Blood Platelet Disorders/genetics , Blood Platelets , Genotype , Guanine Nucleotide Exchange Factors , Humans , Phenotype , Thrombasthenia/diagnosis , Thrombasthenia/genetics
5.
Blood ; 132(19): 2067-2077, 2018 11 08.
Article in English | MEDLINE | ID: mdl-30213874

ABSTRACT

The ephrin transmembrane receptor family of tyrosine kinases is involved in platelet function. We report the first EPHB2 variant affecting platelets in 2 siblings (P1 and P2) from a consanguineous family with recurrent bleeding and normal platelet counts. Whole-exome sequencing identified a c.2233C>T variant (missense p.R745C) of the EPHB2 gene. P1 and P2 were homozygous for this variant, while their asymptomatic parents were heterozygous. The p.R745C variant within the tyrosine kinase domain was associated with defects in platelet aggregation, αIIbß3 activation, and granule secretion induced by G-protein-coupled receptor (GPCR) agonists and convulxin, as well as in thrombus formation on collagen under flow. In contrast, clot retraction, flow-dependent platelet adhesion, and spreading on fibrinogen were only mildly affected, indicating limited effects on αIIbß3 outside-in signaling. Most importantly, Lyn, Syk, and FcRγ phosphorylation, the initial steps in glycoprotein VI (GPVI) platelet signaling were drastically impaired in the absence of platelet-platelet contact, indicating a positive role for EPHB2 in GPVI activation. Likewise platelet activation by PAR4-AP showed defective Src activation, as opposed to normal protein kinase C activity and Ca2+ mobilization. Overexpression of wild-type and R745C EPHB2 variant in RBL-2H3 (rat basophilic leukemia) cells stably expressing human GPVI confirmed that EPHB2 R745C mutation impaired EPHB2 autophosphorylation but had no effect on ephrin ligand-induced EPHB2 clustering, suggesting it did not interfere with EPHB2-ephrin-mediated cell-to-cell contact. In conclusion, this novel inherited platelet disorder affecting EPHB2 demonstrates this tyrosine kinase receptor plays an important role in platelet function through crosstalk with GPVI and GPCR signaling.


Subject(s)
Blood Platelets/pathology , Mutation, Missense , Platelet Activation , Receptor, EphB2/genetics , Adolescent , Blood Platelets/metabolism , Blood Platelets/ultrastructure , Child , Female , Humans , Male , Pedigree , Platelet Adhesiveness , Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Membrane Glycoproteins/metabolism , Receptor, EphB2/metabolism , Signal Transduction , Young Adult
6.
Haematologica ; 105(8): 2004-2019, 2020 08.
Article in English | MEDLINE | ID: mdl-32527953

ABSTRACT

Over the last 100 years the role of platelets in hemostatic events and their production by megakaryocytes have gradually been defined. Progressively, thrombocytopenia was recognized as a cause of bleeding, first through an acquired immune disorder; then, since 1948, when Bernard-Soulier syndrome was first described, inherited thrombocytopenia became a fascinating example of Mendelian disease. The platelet count is often severely decreased and platelet size variable; associated platelet function defects frequently aggravate bleeding. Macrothrombocytopenia with variable proportions of enlarged platelets is common. The number of circulating platelets will depend on platelet production, consumption and lifespan. The bulk of macrothrombocytopenias arise from defects in megakaryopoiesis with causal variants in transcription factor genes giving rise to altered stem cell differentiation and changes in early megakaryocyte development and maturation. Genes encoding surface receptors, cytoskeletal and signaling proteins also feature prominently and Sanger sequencing associated with careful phenotyping has allowed their early classification. It quickly became apparent that many inherited thrombocytopenias are syndromic while others are linked to an increased risk of hematologic malignancies. In the last decade, the application of next-generation sequencing, including whole exome sequencing, and the use of gene platforms for rapid testing have greatly accelerated the discovery of causal genes and extended the list of variants in more common disorders. Genes linked to an increased platelet turnover and apoptosis have also been identified. The current challenges are now to use next-generation sequencing in first-step screening and to define bleeding risk and treatment better.


Subject(s)
Bernard-Soulier Syndrome , Thrombocytopenia , Blood Platelets , Humans , Megakaryocytes , Thrombocytopenia/diagnosis , Thrombocytopenia/genetics , Thrombopoiesis/genetics
7.
Blood ; 127(23): 2903-14, 2016 06 09.
Article in English | MEDLINE | ID: mdl-26912466

ABSTRACT

Macrothrombocytopenia (MTP) is a heterogeneous group of disorders characterized by enlarged and reduced numbers of circulating platelets, sometimes resulting in abnormal bleeding. In most MTP, this phenotype arises because of altered regulation of platelet formation from megakaryocytes (MKs). We report the identification of DIAPH1, which encodes the Rho-effector diaphanous-related formin 1 (DIAPH1), as a candidate gene for MTP using exome sequencing, ontological phenotyping, and similarity regression. We describe 2 unrelated pedigrees with MTP and sensorineural hearing loss that segregate with a DIAPH1 R1213* variant predicting partial truncation of the DIAPH1 diaphanous autoregulatory domain. The R1213* variant was linked to reduced proplatelet formation from cultured MKs, cell clustering, and abnormal cortical filamentous actin. Similarly, in platelets, there was increased filamentous actin and stable microtubules, indicating constitutive activation of DIAPH1. Overexpression of DIAPH1 R1213* in cells reproduced the cytoskeletal alterations found in platelets. Our description of a novel disorder of platelet formation and hearing loss extends the repertoire of DIAPH1-related disease and provides new insight into the autoregulation of DIAPH1 activity.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Hearing Loss/genetics , Mutation , Thrombocytopenia/genetics , A549 Cells , Adolescent , Adult , Aged , Case-Control Studies , Cells, Cultured , Child , Female , Formins , Genetic Association Studies , Genetic Predisposition to Disease , HEK293 Cells , Hearing Loss/complications , Humans , Male , Middle Aged , Pedigree , Polymorphism, Single Nucleotide , Syndrome , Thrombocytopenia/complications , Young Adult
9.
Am J Hematol ; 93(2): 195-204, 2018 02.
Article in English | MEDLINE | ID: mdl-29090484

ABSTRACT

Rare gain-of-function mutations within the ITGA2B or ITGB3 genes have been recognized to cause macrothrombocytopenia (MTP). Here we report three new families with autosomal dominant (AD) MTP, two harboring the same mutation of ITGA2B, αIIbR995W, and a third family with an ITGB3 mutation, ß3D723H. In silico analysis shows how the two mutated amino acids directly modify the salt bridge linking the intra-cytoplasmic part of αIIb to ß3 of the integrin αIIbß3. For all affected patients, the bleeding syndrome and MTP was mild to moderate. Platelet aggregation tended to be reduced but not absent. Electron microscopy associated with a morphometric analysis revealed large round platelets; a feature being the presence of abnormal large α-granules with some giant forms showing signs of fusion. Analysis of the maturation and development of megakaryocytes reveal no defect in their early maturation but abnormal proplatelet formation was observed with increased size of the tips. Interestingly, this study revealed that in addition to the classical phenotype of patients with αIIbß3 intracytoplasmic mutations there is an abnormal maturation of α-granules. It is now necessary to determine if this feature is a characteristic of all mutations disturbing the αIIb R995/ß3 D723 salt bridge.


Subject(s)
Cytoplasmic Granules/pathology , Integrin alpha2/genetics , Integrin beta3/genetics , Thrombocytopenia/etiology , Blood Platelets/ultrastructure , Computer Simulation , Family , Humans , Megakaryocytes , Platelet Aggregation , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry
10.
Platelets ; : 1-5, 2018 Sep 05.
Article in English | MEDLINE | ID: mdl-30183446

ABSTRACT

Professor GVR Born, Gus to his friends, was one of the great pioneers of platelet research. My early memories of him have enabled me to look back at his early years in Oxford and London. A brilliant and generous man with always the time to discuss and advise he was instrumental in deciphering the principle stages of the aggregation of blood platelets by ADP, a path aided by his development and use of the platelet aggregometer. He applied his knowledge to the real time analysis of platelet and leukocyte involvement in thrombus formation in animal models and to the development of atherosclerosis and thrombosis and their pharmacological inhibition. What follows is a personal account of the major steps in this early work and of the actors involved.

11.
Platelets ; 29(7): 737-738, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29985732

ABSTRACT

The number of genes involved in the identification of macrothrombocytopenia (MTP) is growing but the clinical consequences for the affected patients are not well determined. Here, we report the management of the bleeding risk for a patient with the newly reported and rare DIAPH1-related disease during surgery for infertility and then during her subsequent pregnancy. The R1213* DIAPH1 variant responsible for a mild bleeding syndrome in six families was considered a potential risk factor for our patient. Preliminary laparoscopic surgery was followed by neosalpingostomy to open the obstructed fallopian tube that was followed by an ectopic pregnancy requiring further surgery, tranexamic acid was used on each occasion and no bleeding complications were observed. A second pregnancy proceeded to term; the mother's platelet count was controlled throughout the gestation period and remained close to her basal values. No bleeding occurred at delivery or during the postpartum period. In conclusion, with strict repeated assessments of blood parameters and maintenance of the platelet count, the bleeding risk in pregnancy in DIAPH1-related disease can be successfully controlled.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Pregnancy Complications , Thrombocytopenia/diagnosis , Thrombocytopenia/genetics , Thrombocytopenia/therapy , Amino Acid Substitution , Biomarkers , Blood Transfusion , Disease Management , Female , Formins , Genetic Variation , Gestational Age , Hemorrhage/etiology , Hemorrhage/prevention & control , Humans , Mutation , Platelet Count , Pregnancy , Pregnancy Outcome , Syndrome
12.
Platelets ; 29(1): 84-86, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28726538

ABSTRACT

Congenital platelet function disorders are often the result of defects in critical signal transduction pathways required for platelet adhesion and clot formation. Mutations affecting RASGRP2, the gene encoding the Rap GTPase activator, CalDAG-GEFI, give rise to a novel, and rare, group of platelet signal transduction abnormalities. We here report platelet function studies for two brothers (P1 and P2) expressing a novel variant of RASGRP2, CalDAG-GEFI(p.Gly305Asp). P1 and P2 have a lifelong history of bleeding with severe epistaxis successfully treated with platelet transfusions or rFVIIa. Other bleedings include extended hemorrhage from minor wounds. Platelet counts and plasma coagulation were normal, as was αIIbß3 and GPIb expression on the platelet surface. Aggregation of patients' platelets was significantly impaired in response to select agonists including ADP, epinephrine, collagen, and calcium ionophore A23187. Integrin αIIbß3 activation and granule release were also impaired. CalDAG-GEFI protein expression was markedly reduced but not absent. Homology modeling places the Gly305Asp substitution at the GEF-Rap1 interface, suggesting that the mutant protein has very limited catalytic activity. In summary, we here describe a novel mutation in RASGRP2 that affects both expression and function of CalDAG-GEFI and that causes impaired platelet adhesive function and significant bleeding in humans.


Subject(s)
Blood Platelet Disorders/blood , Blood Platelet Disorders/genetics , Blood Platelets/metabolism , Guanine Nucleotide Exchange Factors/genetics , Hemorrhage/etiology , Biomarkers , Blood Platelet Disorders/complications , Child , Erythrocyte Indices , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/metabolism , Hemorrhage/diagnosis , Humans , Male , Models, Molecular , Pedigree , Phenotype , Platelet Aggregation/genetics , Platelet Count , Protein Conformation , Signal Transduction
13.
Haematologica ; 102(2): 282-294, 2017 02.
Article in English | MEDLINE | ID: mdl-27663637

ABSTRACT

Variants in ETV6, which encodes a transcription repressor of the E26 transformation-specific family, have recently been reported to be responsible for inherited thrombocytopenia and hematologic malignancy. We sequenced the DNA from cases with unexplained dominant thrombocytopenia and identified six likely pathogenic variants in ETV6, of which five are novel. We observed low repressive activity of all tested ETV6 variants, and variants located in the E26 transformation-specific binding domain (encoding p.A377T, p.Y401N) led to reduced binding to corepressors. We also observed a large expansion of megakaryocyte colony-forming units derived from variant carriers and reduced proplatelet formation with abnormal cytoskeletal organization. The defect in proplatelet formation was also observed in control CD34+ cell-derived megakaryocytes transduced with lentiviral particles encoding mutant ETV6. Reduced expression levels of key regulators of the actin cytoskeleton CDC42 and RHOA were measured. Moreover, changes in the actin structures are typically accompanied by a rounder platelet shape with a highly heterogeneous size, decreased platelet arachidonic response, and spreading and retarded clot retraction in ETV6 deficient platelets. Elevated numbers of circulating CD34+ cells were found in p.P214L and p.Y401N carriers, and two patients from different families suffered from refractory anemia with excess blasts, while one patient from a third family was successfully treated for acute myeloid leukemia. Overall, our study provides novel insights into the role of ETV6 as a driver of cytoskeletal regulatory gene expression during platelet production, and the impact of variants resulting in platelets with altered size, shape and function and potentially also in changes in circulating progenitor levels.


Subject(s)
Blood Platelets/metabolism , Germ-Line Mutation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Proto-Oncogene Proteins c-ets/genetics , Repressor Proteins/genetics , Thrombopoiesis/genetics , Antigens, CD34/metabolism , Blood Cell Count , Cell Differentiation , Family , Female , Gene Expression Regulation , Genotype , Humans , Hyperplasia , Male , Megakaryocytes/cytology , Megakaryocytes/metabolism , Megakaryocytes/pathology , Pedigree , Phenotype , Platelet Count , Proto-Oncogene Proteins c-ets/metabolism , Repressor Proteins/metabolism , Transcription, Genetic , ETS Translocation Variant 6 Protein
14.
Br J Haematol ; 175(4): 686-695, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27469266

ABSTRACT

We recently reported mutation analysis of the largest cohort of Glanzmann thrombasthenia (GT) patients so far examined. Sanger sequencing of coding regions, splice sites, upstream and downstream regions of the ITGA2B and ITGB3 genes identified 78 causal genetic variants (55 novel); 4 large deletions or duplications were also detected. We have now analysed the expression of non-causal gene polymorphisms in the sequenced regions of both genes in selected members of this cohort. We identified 10 mostly silent variants in ITGA2B and 37 in ITGB3; all were present in control donor databases. Three non-synonymous single nucleotide polymorphisms present were human platelet alloantigen (HPA) variants. A series of haplogroups, often including HPA-3b in ITGA2B, repeated with little variation across unrelated families of wide geographical origins and with different GT-causing mutations whether in ITGA2B or ITGB3. In contrast, a deleterious heterozygous c.1440-13_c.1440-1del in intron 14 of ITGA2B shared a common ITGA2B haplogroup composed of at least five gene polymorphisms and re-occurred in seven European families with no known family relationships. Our results highlight the value of gene polymorphism analysis in GT and are consistent with the bulk of disease-causing mutations in GT being of recent origin.


Subject(s)
Genetic Variation , Integrin alpha2/genetics , Integrin beta3/genetics , Linkage Disequilibrium , Thrombasthenia/diagnosis , Thrombasthenia/genetics , Alleles , Cohort Studies , Gene Frequency , Genetic Drift , Genotype , Haplotypes , Humans , Mutation , Polymorphism, Genetic
15.
Am J Hematol ; 91(7): 714-8, 2016 07.
Article in English | MEDLINE | ID: mdl-26971401

ABSTRACT

There is much current interest in the role of the platelet storage pool of α-granule proteins both in hemostasis and non-hemostatic events. As well as in the arrest of bleeding, the secreted proteins participate in wound healing, inflammation, and innate immunity while in pathology they may be actors in arterial thrombosis and atherosclerosis as well as cancer and metastasis. For a long time, gray platelet syndrome (GPS) has been regarded as the classic inherited platelet disorder caused by an absence of α-granules and their contents. While NBEAL2 is the major source of mutations in GPS, other gene variants may give rise to significant α-granule deficiencies in platelets. These include GATA1, VPS33B, or VIPAS39 in the arthrogryposis, renal dysfunction, and cholestasis (ARC) syndrome and now GFI1B. Nevertheless, many phenotypic differences are associated with mutations in these genes. This critical review was aimed to assess genotype/phenotype variability in disorders of platelet α-granule biogenesis and to urge caution in grouping all genetic defects of α-granules as GPS. Am. J. Hematol. 91:714-718, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Blood Platelets/ultrastructure , Cytoplasmic Granules/genetics , Gray Platelet Syndrome/diagnosis , Cytoplasmic Granules/pathology , Humans , Mutation
16.
Hum Mutat ; 36(5): 548-61, 2015 May.
Article in English | MEDLINE | ID: mdl-25728920

ABSTRACT

We report the largest international study on Glanzmann thrombasthenia (GT), an inherited bleeding disorder where defects of the ITGA2B and ITGB3 genes cause quantitative or qualitative defects of the αIIbß3 integrin, a key mediator of platelet aggregation. Sequencing of the coding regions and splice sites of both genes in members of 76 affected families identified 78 genetic variants (55 novel) suspected to cause GT. Four large deletions or duplications were found by quantitative real-time PCR. Families with mutations in either gene were indistinguishable in terms of bleeding severity that varied even among siblings. Families were grouped into type I and the rarer type II or variant forms with residual αIIbß3 expression. Variant forms helped identify genes encoding proteins mediating integrin activation. Splicing defects and stop codons were common for both ITGA2B and ITGB3 and essentially led to a reduced or absent αIIbß3 expression; included was a heterozygous c.1440-13_c.1440-1del in intron 14 of ITGA2B causing exon skipping in seven unrelated families. Molecular modeling revealed how many missense mutations induced subtle changes in αIIb and ß3 domain structure across both subunits, thereby interfering with integrin maturation and/or function. Our study extends knowledge of GT and the pathophysiology of an integrin.


Subject(s)
Mutation , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Thrombasthenia/genetics , Cohort Studies , DNA Mutational Analysis , Exons , Gene Rearrangement , Genetic Association Studies , Genetic Testing , Genotype , Humans , Integrin alpha2/chemistry , Integrin alpha2/genetics , Integrin beta3/chemistry , Integrin beta3/genetics , Models, Molecular , Phenotype , Platelet Glycoprotein GPIIb-IIIa Complex/chemistry , Protein Conformation , Protein Interaction Domains and Motifs , RNA Splice Sites , RNA Splicing , Sequence Deletion , Thrombasthenia/diagnosis
17.
Hum Mutat ; 35(9): 1033-45, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24934643

ABSTRACT

Bernard-Soulier syndrome (BSS) is a rare autosomal recessive bleeding disorder characterized by defects of the GPIb-IX-V complex, a platelet receptor for von Willebrand factor (VWF). Most of the mutations identified in the genes encoding for the GP1BA (GPIbα), GP1BB (GPIbß), and GP9 (GPIX) subunits prevent expression of the complex at the platelet membrane or more rarely its interaction with VWF. As a consequence, platelets are unable to adhere to the vascular subendothelium and agglutinate in response to ristocetin. In order to collect information on BSS patients, we established an International Consortium for the study of BSS, allowing us to enrol and genotype 132 families (56 previously unreported). With 79 additional families for which molecular data were gleaned from the literature, the 211 families characterized so far have mutations in the GP1BA (28%), GP1BB (28%), or GP9 (44%) genes. There is a wide spectrum of mutations with 112 different variants, including 22 novel alterations. Consistent with the rarity of the disease, 85% of the probands carry homozygous mutations with evidence of founder effects in some geographical areas. This overview provides the first global picture of the molecular basis of BSS and will lead to improve patient diagnosis and management.


Subject(s)
Bernard-Soulier Syndrome/genetics , Genetic Variation , Mutation , Alleles , Bernard-Soulier Syndrome/diagnosis , Databases, Nucleic Acid , Founder Effect , Humans , Platelet Glycoprotein GPIb-IX Complex/genetics , Polymorphism, Single Nucleotide , Web Browser , von Willebrand Diseases/genetics
18.
J Biol Chem ; 288(41): 29621-32, 2013 Oct 11.
Article in English | MEDLINE | ID: mdl-23995838

ABSTRACT

Strongly activated "coated" platelets are characterized by increased phosphatidylserine (PS) surface expression, α-granule protein retention, and lack of active integrin αIIbß3. To study how they are incorporated into thrombi despite a lack of free activated integrin, we investigated the structure, function, and formation of the α-granule protein "coat." Confocal microscopy revealed that fibrin(ogen) and thrombospondin colocalized as "cap," a single patch on the PS-positive platelet surface. In aggregates, the cap was located at the point of attachment of the PS-positive platelets. Without fibrin(ogen) retention, their ability to be incorporated in aggregates was drastically reduced. The surface fibrin(ogen) was strongly decreased in the presence of a fibrin polymerization inhibitor GPRP and also in platelets from a patient with dysfibrinogenemia and a fibrinogen polymerization defect. In contrast, a fibrinogen-clotting protease ancistron increased the amount of fibrin(ogen) and thrombospondin on the surface of the PS-positive platelets stimulated with collagen-related peptide. Transglutaminases are also involved in fibrin(ogen) retention. However, platelets from patients with factor XIII deficiency had normal retention, and a pan-transglutaminase inhibitor T101 had only a modest inhibitory effect. Fibrin(ogen) retention was normal in Bernard-Soulier syndrome and kindlin-3 deficiency, but not in Glanzmann thrombasthenia lacking the platelet pool of fibrinogen and αIIbß3. These data show that the fibrin(ogen)-covered cap, predominantly formed as a result of fibrin polymerization, is a critical mechanism that allows coated (or rather "capped") platelets to become incorporated into thrombi despite their lack of active integrins.


Subject(s)
Blood Platelets/metabolism , Fibrin/metabolism , Fibrinogen/metabolism , Platelet Aggregation , Thrombospondins/metabolism , Blood Coagulation/drug effects , Blood Platelets/drug effects , Blotting, Western , Female , Flow Cytometry , Humans , Microscopy, Confocal , Oligopeptides/pharmacology , Phosphatidylserines/metabolism , Polymerization/drug effects , Thrombasthenia/blood , Thrombasthenia/metabolism , Thrombosis/metabolism , Transglutaminases/metabolism
19.
Br J Haematol ; 165(2): 165-78, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24286193

ABSTRACT

Genetic defects of platelets constitute rare diseases that include bleeding syndromes of autosomal dominant, recessive or X-linked inheritance. They affect platelet production, resulting in a low circulating platelet count and changes in platelet morphology, platelet function, or a combination of both with altered megakaryopoiesis and a defective platelet response. As a result, blood platelets fail to fulfil their haemostatic function. Most studied of the platelet function disorders are deficiencies of glycoprotein mediators of adhesion and aggregation while defects of primary receptors for stimuli include the P2Y12 ADP receptor. Studies on inherited defects of (i) secretion from storage organelles (dense granules, α-granules), (ii) the platelet cytoskeleton and (iii) the generation of pro-coagulant activity have identified genes indirectly controlling the functional response. Signalling pathway defects leading to agonist-specific modifications of platelet aggregation are the current target of exome-sequencing strategies. We now review recent advances in the molecular characterization of platelet function defects.


Subject(s)
Blood Coagulation Disorders, Inherited/genetics , Blood Coagulation Disorders, Inherited/metabolism , Blood Platelet Disorders/genetics , Blood Platelet Disorders/metabolism , Blood Platelet Disorders/congenital , Blood Platelets/drug effects , Blood Platelets/metabolism , Humans , Platelet Adhesiveness , Purpura, Thrombotic Thrombocytopenic/genetics , Purpura, Thrombotic Thrombocytopenic/metabolism , Receptors, Cell Surface/metabolism , Signal Transduction , Thrombasthenia/etiology , Thrombasthenia/genetics , Thrombasthenia/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL