Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
1.
J Cell Biochem ; 124(7): 989-1001, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37210732

RESUMO

Mutations in the αIIb ß-propeller domain have long been known to disrupt heterodimerization and intracellular trafficking of αIIbß3 complexes leading to diminished surface expression and/or function, resulting in Glanzmann thrombasthenia. Our previous study on three ß-propeller mutations, namely G128S, S287L, and G357S, showed variable defects in protein transport correlated with the patient's clinical phenotypes. Pulse-chase experiments revealed differences in αIIbß3 complex maturation among the three mutations. Hence, the current study aims to correlate conformational changes caused by each one of them. Evolutionary conservation analysis, stability analysis, and molecular dynamics simulations of the three mutant structures were carried out. Stability analysis revealed that, while G128S and G357S mutations destabilized the ß-propeller structure, S287L retained the stability. Wild-type and mutant ß-propeller structures, when subjected to molecular dynamics simulations, confirmed that G128S and G357S were both destabilizing in nature when compared with the wild-type and S287L based on several parameters studied, like RMSD, RMSF, Rg, FEL, PCA, secondary structure, and hydrogen bonds. In our previous study, we demonstrated that mutant S287L αIIbß3 complexes were more stable than the wild-type αIIbß3 complexes, as evidenced in pulse-chase experiments. These findings corroborate variable intracellular fates of mutant αIIbß3 complexes as a result of these ß-propeller mutations.


Assuntos
Integrina alfa2 , Integrina beta3 , Complexo Glicoproteico GPIIb-IIIa de Plaquetas , Trombastenia , Humanos , Integrina beta3/genética , Simulação de Dinâmica Molecular , Mutação , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Estrutura Secundária de Proteína , Trombastenia/genética , Trombastenia/metabolismo , Integrina alfa2/genética , Integrina alfa2/metabolismo
2.
Blood Adv ; 7(13): 3180-3191, 2023 07 11.
Artigo em Inglês | MEDLINE | ID: mdl-36884296

RESUMO

This report identifies a novel variant form of the inherited bleeding disorder Glanzmann thrombasthenia, exhibiting only mild bleeding in a physically active individual. The platelets cannot aggregate ex vivo with physiologic agonists of activation, although microfluidic analysis with whole blood displays moderate ex vivo platelet adhesion and aggregation consistent with mild bleeding. Immunocytometry shows reduced expression of αIIbß3 on quiescent platelets that spontaneously bind/store fibrinogen, and activation-dependent antibodies (ligand-induced binding site-319.4 and PAC-1) report ß3 extension suggesting an intrinsic activation phenotype. Genetic analysis reveals a single F153Sß3 substitution within the ßI-domain from a heterozygous T556C nucleotide substitution of ITGB3 exon 4 in conjunction with a previously reported IVS5(+1)G>A splice site mutation with undetectable platelet messenger RNA accounting for hemizygous expression of S153ß3. F153 is completely conserved among ß3 of several species and all human ß-integrin subunits suggesting that it may play a vital role in integrin structure/function. Mutagenesis of αIIb-F153Sß3 also displays reduced levels of a constitutively activated αIIb-S153ß3 on HEK293T cells. The overall structural analysis suggests that a bulky aromatic, nonpolar amino acid (F,W)153ß3 is critical for maintaining the resting conformation of α2- and α1-helices of the ßI-domain because small amino acid substitutions (S,A) facilitate an unhindered inward movement of the α2- and α1-helices of the ßI-domain toward the constitutively active αIIbß3 conformation, while a bulky aromatic, polar amino acid (Y) hinders such movements and restrains αIIbß3 activation. The data collectively demonstrate that disruption of F153ß3 can significantly alter normal integrin/platelet function, although reduced expression of αIIb-S153ß3 may be compensated by a hyperactive conformation that promotes viable hemostasis.


Assuntos
Complexo Glicoproteico GPIIb-IIIa de Plaquetas , Trombastenia , Humanos , Aminoácidos/genética , Células HEK293 , Mutação , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Trombastenia/genética , Trombastenia/metabolismo
4.
Platelets ; 33(4): 551-561, 2022 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-34275420

RESUMO

Glanzmann thrombasthenia (GT) is a rare autosomal recessive bleeding disorder characterized by impaired platelet aggregation due to defects in integrin αIIbß3, a fibrinogen receptor. Platelet phenotypes and allelic variations in 28 Turkish GT patients are reported. Platelets αIIbß3 expression was evaluated by flow cytometry. Sequence analyzes of ITGA2B and ITGB3 genes allowed identifying nine variants. Non-sense variation effect on αIIbß3 expression was studied by using transfected cell lines. 3D molecular dynamics (MDs) simulations allowed characterizing structural alterations. Five new alleles were described. αIIb:p.Gly423Asp, p.Asp560Ala and p.Tyr784Cys substitutions impaired αIIbß3 expression. The αIIb:p.Gly128Val substitution allowed normal expression; however, the corresponding NM_000419.3:c.476G>T variation would create a cryptic donor splicing site altering mRNA processing. The ß3:p.Gly540Asp substitution allowed αIIbß3 expression in HEK-293 cells but induced its constitutive activation likely by impairing αIIb and ß3 legs interaction. The substitution alters the ß3 I-EGF-3 domain flexibility as shown by MDs simulations. GT variations are mostly unique although the NM_000419.3:c.1752 + 2 T > C and NM_000212.2:c.1697 G > A variations identified in 4 and 8 families, respectively, might be a current cause of GT in Turkey. MD simulations suggested how some subtle structural variations in the ß3 I-EGF domains might induce constitutive activation of αIIbß3 without altering the global domain structure.


Assuntos
Integrina alfa2 , Integrina beta3 , Complexo Glicoproteico GPIIb-IIIa de Plaquetas , Trombastenia , Fator de Crescimento Epidérmico , Células HEK293 , Humanos , Integrina alfa2/genética , Integrina beta3/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Trombastenia/genética , Trombastenia/metabolismo , Turquia
5.
Mol Med Rep ; 23(6)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33880575

RESUMO

Itgb3­integrin­deficient (Itgb3­/­) mice have been reported as a Glanzmann thrombasthenia (GT) model and have been used for platelet research. However, it remains unclear whether this mouse model can fully simulate patients with GT or whether it has different characteristics from these patients. The present study aimed to answer this question. Itgb3­/­ mice were tested for platelet function, tail bleeding, whole­blood count, bone marrow hematopoiesis and organ enlargement. Itgb3­/­ platelets showed impaired functions, including fibrinogen binding, aggregation, adhesion or spreading. Itgb3­/­ mice demonstrated decreased platelet count and microcytic hypochromic anemia. Reduced iron staining of bone marrow and decreased plasma ferritin level confirmed the diagnosis of iron deficiency anemia. Evident splenomegaly was observed in Itgb3­/­ mice. Immunohistochemical analysis of spleen biopsy revealed normal expression of CD3 and CD19, but elevated expression of CD71, which suggested that the splenomegaly in Itgb3­/­ mice may be associated with extramedullary hematopoiesis. In conclusion, Itgb3­/­ mice exhibited some unique characteristics that differed from those of human patients with GT and thus cannot completely simulate patients with GT.


Assuntos
Integrina beta3/genética , Integrina beta3/metabolismo , Trombastenia/genética , Trombastenia/metabolismo , Anemia , Animais , Plaquetas/metabolismo , Modelos Animais de Doenças , Feminino , Hematopoese , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Testes de Função Plaquetária , Baço , Esplenomegalia , Transcriptoma
6.
Blood Coagul Fibrinolysis ; 32(2): 146-150, 2021 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-33196509

RESUMO

Glanzmann thrombasthenia is a rare autosomal recessive genetic disease characterized by platelet aggregation dysfunction caused by a congenital defect of platelet membrane glycoprotein IIb/IIIa (integrin αIIbß3). Integrin αIIbß3, a calcium-dependent heterodimer, plays a critical role in platelet aggregation. We described a boy who was hospitalized with serious epistaxis at 10 months of age who had a history of repeated petechiae and spontaneous epistaxis since birth. Flow cytometry showed normal surface expression of platelet antigens. Genetic analysis and sequencing revealed the novel missense mutation c.G1252>T (p.Gly418Cys) in ITGA2B. This heterozygous amino acid mutation flanked the fourth calcium-binding domain and may interfere with integrin biogenesis via mechanisms other than merely altering cell surface expression. We discuss the heterogeneity of the genotype and phenotype with this atypical case and review the relevant literature on mutations adjacent to or within the calcium-binding domains in Glanzmann thrombasthenia.


Assuntos
Integrina alfa2/genética , Mutação de Sentido Incorreto , Trombastenia/genética , Cálcio/metabolismo , Heterozigoto , Humanos , Lactente , Integrina alfa2/química , Integrina alfa2/metabolismo , Masculino , Domínios Proteicos , Trombastenia/metabolismo
7.
Int J Mol Sci ; 21(24)2020 Dec 14.
Artigo em Inglês | MEDLINE | ID: mdl-33327658

RESUMO

Patients affected by the rare Glanzmann thrombasthenia (GT) suffer from defective or low levels of the platelet-associated glycoprotein (GP) IIb/IIIa, which acts as a fibrinogen receptor, and have therefore an impaired ability to aggregate platelets. Because the procoagulant activity is a dichotomous facet of platelet activation, diverging from the aggregation endpoint, we were interested in characterizing the ability to generate procoagulant platelets in GT patients. Therefore, we investigated, by flow cytometry analysis, platelet functions in three GT patients as well as their ability to generate procoagulant collagen-and-thrombin (COAT) platelets upon combined activation with convulxin-plus-thrombin. In addition, we further characterized intracellular ion fluxes during the procoagulant response, using specific probes to monitor by flow cytometry kinetics of cytosolic calcium, sodium, and potassium ion fluxes. GT patients generated higher percentages of procoagulant COAT platelets compared to healthy donors. Moreover, they were able to mobilize higher levels of cytosolic calcium following convulxin-plus-thrombin activation, which is congruent with the greater procoagulant activity. Further investigations will dissect the role of GPIIb/IIIa outside-in signalling possibly implicated in the regulation of platelet procoagulant activity.


Assuntos
Plaquetas/metabolismo , Trombastenia/metabolismo , Plaquetas/fisiologia , Cálcio/metabolismo , Colágeno/metabolismo , Citometria de Fluxo , Humanos , Ativação Plaquetária/fisiologia , Potássio/metabolismo , Sódio/metabolismo , Trombina/metabolismo
9.
Thromb Haemost ; 119(6): 906-915, 2019 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-30934104

RESUMO

Factor XIIIa (fXIIIa) is a transglutaminase that plays a crucial role in fibrin clot stabilization and regulation of fibrinolysis. It is known to bind to procoagulant platelets. In contrast, the zymogen fXIII interaction with platelets is not well characterized. We investigated the interaction of zymogen fXIII with activated platelet subpopulations. Confocal microscopy and flow cytometry using fluorescently labelled factors and antibodies. Phosphatidylserine (PS)-positive activated platelets bound 700 to 800 molecules/cell of fXIII at 100 nM, while both PS-negative activated platelets and resting platelets bound 200 to 400 molecules/cell. The binding was reversible, calcium-independent and linear within the fXIII concentration range of up to 1,000 nM. fXIII predominantly bound to the caps of procoagulant platelets and co-localized with fibrinogen. Exogenous fibrinogen promoted fXIII binding by activated PS-negative platelets; this effect was abolished by the integrin αIIbß3 antagonist monafram. The fXIII binding was 1.5- to 3-fold decreased for platelets from four patients with grey platelet syndrome, and was variable for platelets from six patients with Glanzmann's thrombasthenia. Strong platelet stimulation, fibrinogen and αIIbß3 play essential roles in fXIII binding, without any of them fXIII does not bind to platelets. The preferential binding in the cap-like structures might be important for increasing local fXIII concentration in platelet thrombi.


Assuntos
Afibrinogenemia/metabolismo , Plaquetas/fisiologia , Fator VIII/metabolismo , Fibrinogênio/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Trombastenia/metabolismo , Afibrinogenemia/genética , Coagulação Sanguínea , Precursores Enzimáticos , Fibrinólise , Humanos , Ativação Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Ligação Proteica
10.
Sci Rep ; 8(1): 10300, 2018 07 09.
Artigo em Inglês | MEDLINE | ID: mdl-29985398

RESUMO

Mass cytometry (MC) uses mass spectrometry to simultaneously detect multiple metal-conjugated antibodies on single cells, thereby enabling the detailed study of cellular function. Here, for the first time, we applied MC to the analysis of platelets. We developed a panel of 14 platelet-specific metal-tagged antibodies (targeting cluster of differentiation [CD] 9, CD29, CD31, CD36, CD41, CD42a, CD42b, CD61, CD62P, CD63, CD107a, CD154, glycoprotein [GP] VI and activated integrin αIIbß3) and compared this panel with two fluorescence flow cytometry (FFC) panels (CD41, CD42b, and CD61; or CD42b, CD62P, and activated integrin αIIbß3) in the evaluation of activation-dependent changes in glycoprotein expression on healthy subject and Glanzmann thrombasthenia (GT) platelets. High-dimensional analysis of surface markers detected by MC identified previously unappreciated subpopulations of platelets in healthy donors. As expected, MC and FFC revealed that GT platelets had significantly reduced CD41, CD61, and activated integrin αIIbß3 surface expression. MC also revealed that surface expression of CD9, CD42a and CD63 were elevated, CD31, CD154 and GPVI were reduced and CD29, CD36, CD42b, CD62P and CD107a were similar on GT platelets compared to healthy donor platelets. In summary, MC revealed distinct platelet subtypes in healthy subjects and novel alterations in surface glycoproteins on GT platelets.


Assuntos
Plaquetas/metabolismo , Citometria de Fluxo/métodos , Glicoproteínas de Membrana/metabolismo , Trombastenia/patologia , Plaquetas/citologia , Estudos de Casos e Controles , Humanos , Integrina beta3/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Trombastenia/metabolismo
11.
Sci Rep ; 8(1): 5428, 2018 04 03.
Artigo em Inglês | MEDLINE | ID: mdl-29615672

RESUMO

Haemostatic platelet function is intimately linked to cellular mechanics and cytoskeletal morphology. How cytoskeletal reorganizations give rise to a highly contractile phenotype that is necessary for clot contraction remains poorly understood. To elucidate this process in vitro, we developed a morphometric screen to quantify the spatial organization of actin fibres and vinculin adhesion sites in single spread platelets. Platelets from healthy donors predominantly adopted a bipolar morphology on fibrinogen and fibronectin, whereas distinguishable, more isotropic phenotypes on collagen type I or laminin. Specific integrin αIIbß3 inhibitors induced an isotropic cytoskeletal organization in a dose-dependent manner. The same trend was observed with decreasing matrix stiffness. Circular F-actin arrangements in platelets from a patient with type II Glanzmann thrombasthenia (GT) were consistent with the residual activity of a small number of αIIbß3 integrins. Cytoskeletal morphologies in vitro thus inform about platelet adhesion receptor identity and functionality, and integrin αIIbß3 mechanotransduction fundamentally determines the adoption of a bipolar phenotype associated with contraction. Super-resolution microscopy and electron microscopies further confirmed the stress fibre-like contractile actin architecture. For the first time, our assay allows the unbiased and quantitative assessment of platelet morphologies and could help to identify defective platelet behaviour contributing to elusive bleeding phenotypes.


Assuntos
Plaquetas/citologia , Fenômenos Mecânicos , Fenótipo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Adulto , Fenômenos Biomecânicos , Plaquetas/metabolismo , Citoesqueleto/metabolismo , Fibrinogênio/metabolismo , Hemostasia , Humanos , Ligantes , Transdução de Sinais , Trombastenia/sangue , Trombastenia/metabolismo
12.
Mol Genet Genomic Med ; 6(2): 249-260, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29385657

RESUMO

BACKGROUND: Studies on the inherited bleeding disorder, Glanzmann thrombasthenia (GT), have helped define the role of the αIIbß3 integrin in platelet aggregation. Stable bent αIIbß3 undergoes conformation changes on activation allowing fibrinogen binding and its taking an extended form. The αIIb genu assures the fulcrum of the bent state. Our goal was to determine how structural changes induced by missense mutations in the αIIb genu define GT phenotype. METHODS: Sanger sequencing of ITGA2B and ITGB3 in the index case followed by in silico modeling of all known GT-causing missense mutations extending from the lower part of the ß-propeller, and through the thigh and upper calf-1 domains. RESULTS: A homozygous c.1772A>C transversion in exon 18 of ITGA2B coding for a p.Asp591Ala substitution in an interconnecting loop of the lower thigh domain of αIIb in a patient with platelets lacking αIIbß3 led us to extend our in silico modeling to all 16 published disease-causing missense variants potentially affecting the αIIb genu. Modifications of structuring H-bonding were the major cause in the thigh domain although one mutation gave mRNA decay. In contrast, short-range changes induced in calf-1 appeared minor suggesting long-range effects. All result in severe to total loss of αIIbß3 in platelets. The absence of mutations within a key Ca2+-binding loop in the genu led us to scan public databases; three potential single allele variants giving major structural changes were identiffied suggesting that this key region is not protected from genetic variation. CONCLUSIONS: It appears that the αIIb genu is the object of stringent quality control to prevent platelets from circulating with activated and extended integrin.


Assuntos
Integrina alfa2/genética , Trombastenia/genética , Adulto , Alelos , Plaquetas/metabolismo , Plaquetas/fisiologia , Simulação por Computador , Éxons , Feminino , Humanos , Integrina alfa2/metabolismo , Integrina beta3/genética , Articulação do Joelho , Agregação Plaquetária , Testes de Função Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/química , Glicoproteína IIb da Membrana de Plaquetas/genética , Trombastenia/metabolismo
14.
Stem Cell Res ; 20: 14-20, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28232155

RESUMO

Glanzmann thrombasthenia (GT) is a rare monogenic hemorrhagic disorder involving aggregation defect of non-nuclear platelets. In this study we generated induced pluripotent stem cells (iPSCs) from skin fibroblasts of a GT patient with complex heterogeneous mutations of ITGA2B gene. GT-iPSCs could be successfully differentiated into platelets (GT-iPS-platelets). GT-iPS-platelets were CD41-/CD42b+/CD61- and were platelet activation marker (PAC-1) negative after adenosine diphosphate (ADP) activation. Furthermore, GT-iPS-platelets were defective in platelet aggregation tests in vitro. Moreover, exogenous expression of the wild-type ITGA2B gene in GT-iPS platelets restored CD41 expression and normal platelet aggregation. Our study suggested that patient-specific iPSCs could be a potential target of stem cell based gene therapy for platelet diseases.


Assuntos
Reprogramação Celular , Células-Tronco Pluripotentes Induzidas/metabolismo , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Trombastenia/patologia , Animais , Sequência de Bases , Plaquetas/citologia , Plaquetas/metabolismo , Diferenciação Celular , Linhagem Celular , Análise Mutacional de DNA , Fosfatase 2 de Especificidade Dupla/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/transplante , Integrina alfa2/genética , Cariótipo , Camundongos , Camundongos Nus , Agregação Plaquetária , Glicoproteína IIb da Membrana de Plaquetas/genética , Polimorfismo Genético , Teratoma/metabolismo , Teratoma/patologia , Trombastenia/genética , Trombastenia/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
15.
Blood ; 128(9): 1282-9, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27235135

RESUMO

In addition to mutations in ITG2B or ITGB3 genes that cause defective αIIbß3 expression and/or function in Glanzmann's thrombasthenia patients, platelet dysfunction can be a result of genetic variability in proteins that mediate inside-out activation of αIIbß3 The RASGRP2 gene is strongly expressed in platelets and neutrophils, where its encoded protein CalDAG-GEFI facilitates the activation of Rap1 and subsequent activation of integrins. We used next-generation sequencing (NGS) and whole-exome sequencing (WES) to identify 2 novel function-disrupting mutations in RASGRP2 that account for bleeding diathesis and platelet dysfunction in 2 unrelated families. By using a panel of 71 genes, we identified a homozygous change (c.1142C>T) in exon 10 of RASGRP2 in a 9-year-old child of Chinese origin (family 1). This variant led to a p.Ser381Phe substitution in the CDC25 catalytic domain of CalDAG-GEFI. In 2 Spanish siblings from family 2, WES identified a nonsense homozygous variation (c.337C>T) (p.Arg113X) in exon 5 of RASGRP2 CalDAG-GEFI expression was markedly reduced in platelets from all patients, and by using a novel in vitro assay, we found that the nucleotide exchange activity was dramatically reduced in CalDAG-GEFI p.Ser381Phe. Platelets from homozygous patients exhibited agonist-specific defects in αIIbß3 integrin activation and aggregation. In contrast, α- and δ-granule secretion, platelet spreading, and clot retraction were not markedly affected. Integrin activation in the patients' neutrophils was also impaired. These patients are the first cases of a CalDAG-GEFI deficiency due to homozygous RASGRP2 mutations that are linked to defects in both leukocyte and platelet integrin activation.


Assuntos
Plaquetas/metabolismo , Éxons , Fatores de Troca do Nucleotídeo Guanina , Mutação de Sentido Incorreto , Ativação Plaquetária/genética , Trombastenia , Proteínas rap1 de Ligação ao GTP/metabolismo , Substituição de Aminoácidos , Plaquetas/patologia , Criança , Ativação Enzimática/genética , Feminino , Fatores de Troca do Nucleotídeo Guanina/genética , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Integrina beta3/genética , Integrina beta3/metabolismo , Masculino , Pessoa de Meia-Idade , Glicoproteína IIb da Membrana de Plaquetas/genética , Glicoproteína IIb da Membrana de Plaquetas/metabolismo , Vesículas Secretórias/genética , Vesículas Secretórias/metabolismo , Trombastenia/genética , Trombastenia/metabolismo , Trombastenia/patologia
16.
Haematologica ; 101(1): 46-56, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26452979

RESUMO

Several patients have been reported to have variant dominant forms of Glanzmann thrombasthenia, associated with macrothrombocytopenia and caused by gain-of-function mutations of ITGB3 or ITGA2B leading to reduced surface expression and constitutive activation of integrin αIIbß3. The mechanisms leading to a bleeding phenotype of these patients have never been addressed. The aim of this study was to unravel the mechanism by which ITGB3 mutations causing activation of αIIbß3 lead to platelet dysfunction and macrothrombocytopenia. Using platelets from two patients carrying the ß3 del647-686 mutation and Chinese hamster ovary cells expressing different αIIbß3-activating mutations, we showed that reduced surface expression of αIIbß3 is due to receptor internalization. Moreover, we demonstrated that permanent triggering of αIIbß3-mediated outside-in signaling causes an impairment of cytoskeletal reorganization arresting actin turnover at the stage of polymerization. The induction of actin polymerization by jasplakinolide, a natural toxin that promotes actin nucleation and prevents depolymerization of stress fibers, in control platelets produced an impairment of platelet function similar to that of patients with variant forms of dominant Glanzmann thrombasthenia. del647-686ß3-transduced murine megakaryocytes generated proplatelets with a reduced number of large tips and asymmetric barbell-proplatelets, suggesting that impaired cytoskeletal rearrangement is the cause of macrothrombocytopenia. These data show that impaired cytoskeletal remodeling caused by a constitutively activated αIIbß3 is the main effector of platelet dysfunction and macrothrombocytopenia, and thus of bleeding, in variant forms of dominant Glanzmann thrombasthenia.


Assuntos
Plaquetas/metabolismo , Citoesqueleto/metabolismo , Mutação , Trombastenia/metabolismo , Trombocitopenia/metabolismo , Animais , Plaquetas/patologia , Células CHO , Cricetinae , Cricetulus , Citoesqueleto/genética , Citoesqueleto/patologia , Feminino , Humanos , Integrina alfa2/genética , Integrina alfa2/metabolismo , Integrina beta3/genética , Integrina beta3/metabolismo , Masculino , Trombastenia/genética , Trombastenia/patologia , Trombocitopenia/genética , Trombocitopenia/patologia
17.
Proc Natl Acad Sci U S A ; 112(15): E1898-907, 2015 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-25827233

RESUMO

Next-generation sequencing is transforming our understanding of human genetic variation but assessing the functional impact of novel variants presents challenges. We analyzed missense variants in the integrin αIIbß3 receptor subunit genes ITGA2B and ITGB3 identified by whole-exome or -genome sequencing in the ThromboGenomics project, comprising ∼32,000 alleles from 16,108 individuals. We analyzed the results in comparison with 111 missense variants in these genes previously reported as being associated with Glanzmann thrombasthenia (GT), 20 associated with alloimmune thrombocytopenia, and 5 associated with aniso/macrothrombocytopenia. We identified 114 novel missense variants in ITGA2B (affecting ∼11% of the amino acids) and 68 novel missense variants in ITGB3 (affecting ∼9% of the amino acids). Of the variants, 96% had minor allele frequencies (MAF) < 0.1%, indicating their rarity. Based on sequence conservation, MAF, and location on a complete model of αIIbß3, we selected three novel variants that affect amino acids previously associated with GT for expression in HEK293 cells. αIIb P176H and ß3 C547G severely reduced αIIbß3 expression, whereas αIIb P943A partially reduced αIIbß3 expression and had no effect on fibrinogen binding. We used receiver operating characteristic curves of combined annotation-dependent depletion, Polyphen 2-HDIV, and sorting intolerant from tolerant to estimate the percentage of novel variants likely to be deleterious. At optimal cut-off values, which had 69-98% sensitivity in detecting GT mutations, between 27% and 71% of the novel αIIb or ß3 missense variants were predicted to be deleterious. Our data have implications for understanding the evolutionary pressure on αIIbß3 and highlight the challenges in predicting the clinical significance of novel missense variants.


Assuntos
Sequenciamento de Nucleotídeos em Larga Escala/métodos , Mutação de Sentido Incorreto , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Trombastenia/genética , Alelos , Bases de Dados de Ácidos Nucleicos , Exoma/genética , Fibrinogênio/química , Fibrinogênio/metabolismo , Frequência do Gene , Células HEK293 , Humanos , Immunoblotting , Modelos Moleculares , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/química , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Ligação Proteica , Conformação Proteica , Estrutura Terciária de Proteína , Trombastenia/metabolismo
18.
Cell Signal ; 26(2): 279-86, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24240055

RESUMO

The specific TLR2/1 complex activator Pam3CSK4 has been shown to provoke prominent activation and aggregation of human non-nucleated platelets. As Pam3CSK4-evoked platelet activation does not employ the major signalling pathway established in nucleated immune cells, we investigated if the TLR2/1 complex on platelets may initiate signalling pathways known to be induced by physiological agonists such as collagen via GPVI or thrombin via PARs. We found that triggering TLR2/1 complex-signalling with Pam3CSK4, in common with that induced via GPVI, and in contrast to that provoked by PARs, involves tyrosine phosphorylation of the adaptor protein LAT as well as of PLCγ2 in a src- and Syk-dependent manner. In this respect, we provide evidence that Pam3CSK4 does not cross-activate GPVI. Further, by the use of platelets from a Glanzmann's thrombasthenia patient lacking ß(3), in contrast to findings in nucleated immune cells, we show that the initiation of platelet activation by Pam3CSK4 does not involve integrin ß(3) signalling; whereas the latter, subsequent to intermediate TXA2 synthesis and signalling, was found to be indispensable for proper dense granule secretion and full platelet aggregation. Together, our findings reveal that triggering the TLR2/1 complex with Pam3CSK4 initiates human platelet activation by engaging tyrosine kinases of the src family and Syk, the adaptor protein LAT, as well as the key mediator PLCγ2.


Assuntos
Plaquetas/metabolismo , Ativação Plaquetária , Transdução de Sinais , Receptor 1 Toll-Like/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Cálcio/metabolismo , Venenos de Crotalídeos/farmacologia , Humanos , Integrinas/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lectinas Tipo C , Lipopeptídeos/farmacologia , Proteínas de Membrana/metabolismo , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Fosfolipase C gama/metabolismo , Fosforilação/efeitos dos fármacos , Ativação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Glicoproteínas da Membrana de Plaquetas/metabolismo , Proteínas Tirosina Quinases/metabolismo , Pirimidinas/farmacologia , Receptores de Tromboxano A2 e Prostaglandina H2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Quinase Syk , Trombastenia/metabolismo , Trombastenia/patologia , Receptor 1 Toll-Like/química , Receptor 2 Toll-Like/química , Receptor 2 Toll-Like/metabolismo , Quinases da Família src/antagonistas & inibidores , Quinases da Família src/metabolismo
19.
Br J Haematol ; 165(2): 165-78, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24286193

RESUMO

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.


Assuntos
Transtornos Herdados da Coagulação Sanguínea/genética , Transtornos Herdados da Coagulação Sanguínea/metabolismo , Transtornos Plaquetários/genética , Transtornos Plaquetários/metabolismo , Transtornos Plaquetários/congênito , Plaquetas/efeitos dos fármacos , Plaquetas/metabolismo , Humanos , Adesividade Plaquetária , Púrpura Trombocitopênica Trombótica/genética , Púrpura Trombocitopênica Trombótica/metabolismo , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Trombastenia/etiologia , Trombastenia/genética , Trombastenia/metabolismo
20.
PLoS One ; 8(11): e78683, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24236036

RESUMO

Mutations in ITGA2B and ITGB3 cause Glanzmann thrombasthenia, an inherited bleeding disorder in which platelets fail to aggregate when stimulated. Whereas an absence of expression or qualitative defects of αIIbß3 mainly affect platelets and megakaryocytes, αvß3 has a widespread tissue distribution. Little is known of how amino acid substitutions of ß3 comparatively affect the expression and structure of both integrins. We now report computer modelling including molecular dynamics simulations of extracellular head domains of αIIbß3 and αvß3 to determine the role of a novel ß3 Pro189Ser (P163S in the mature protein) substitution that abrogates αIIbß3 expression in platelets while allowing synthesis of αvß3. Transfection of wild-type and mutated integrins in CHO cells confirmed that only αvß3 surface expression was maintained. Modeling initially confirmed that replacement of αIIb by αv in the dimer results in a significant decrease in surface contacts at the subunit interface. For αIIbß3, the presence of ß3S163 specifically displaces an α-helix starting at position 259 and interacting with ß3R261 while there is a moderate 11% increase in intra-subunit H-bonds and a very weak decrease in the global H-bond network. In contrast, for αvß3, S163 has different effects with ß3R261 coming deeper into the propeller with a 43% increase in intra-subunit H-bonds but with little effect on the global H-bond network. Compared to the WT integrins, the P163S mutation induces a small increase in the inter-subunit fluctuations for αIIbß3 but a more rigid structure for αvß3. Overall, this mutation stabilizes αvß3 despite preventing αIIbß3 expression.


Assuntos
Integrina alfa2/genética , Integrina alfaV/genética , Integrina beta3/química , Simulação de Dinâmica Molecular , Trombastenia/genética , Animais , Células CHO , Cricetinae , Cricetulus , Feminino , Expressão Gênica , Humanos , Ligação de Hidrogênio , Integrina alfa2/metabolismo , Integrina alfaV/metabolismo , Integrina beta3/genética , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Domínios e Motivos de Interação entre Proteínas , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Trombastenia/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...