Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 301
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 180(5): 862-877.e22, 2020 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-32142679

RESUMO

Using untargeted metabolomics (n = 1,162 subjects), the plasma metabolite (m/z = 265.1188) phenylacetylglutamine (PAGln) was discovered and then shown in an independent cohort (n = 4,000 subjects) to be associated with cardiovascular disease (CVD) and incident major adverse cardiovascular events (myocardial infarction, stroke, or death). A gut microbiota-derived metabolite, PAGln, was shown to enhance platelet activation-related phenotypes and thrombosis potential in whole blood, isolated platelets, and animal models of arterial injury. Functional and genetic engineering studies with human commensals, coupled with microbial colonization of germ-free mice, showed the microbial porA gene facilitates dietary phenylalanine conversion into phenylacetic acid, with subsequent host generation of PAGln and phenylacetylglycine (PAGly) fostering platelet responsiveness and thrombosis potential. Both gain- and loss-of-function studies employing genetic and pharmacological tools reveal PAGln mediates cellular events through G-protein coupled receptors, including α2A, α2B, and ß2-adrenergic receptors. PAGln thus represents a new CVD-promoting gut microbiota-dependent metabolite that signals via adrenergic receptors.


Assuntos
Doenças Cardiovasculares/sangue , Microbioma Gastrointestinal/genética , Glutamina/análogos & derivados , Trombose/metabolismo , Animais , Artérias/lesões , Artérias/metabolismo , Artérias/microbiologia , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Plaquetas/metabolismo , Plaquetas/microbiologia , Doenças Cardiovasculares/genética , Doenças Cardiovasculares/microbiologia , Doenças Cardiovasculares/patologia , Morte Súbita Cardíaca/patologia , Glutamina/sangue , Glutamina/genética , Humanos , Masculino , Metaboloma/genética , Metabolômica/métodos , Camundongos , Infarto do Miocárdio/sangue , Infarto do Miocárdio/microbiologia , Ativação Plaquetária/genética , Receptores Adrenérgicos alfa/sangue , Receptores Adrenérgicos alfa/genética , Receptores Adrenérgicos beta/sangue , Receptores Adrenérgicos beta/genética , Fatores de Risco , Acidente Vascular Cerebral/sangue , Acidente Vascular Cerebral/microbiologia , Acidente Vascular Cerebral/patologia , Trombose/genética , Trombose/microbiologia , Trombose/patologia
2.
Cell Commun Signal ; 22(1): 332, 2024 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-38886790

RESUMO

BACKGROUND: HRASKO/NRASKO double knockout mice exhibit exceedingly high rates of perinatal lethality due to respiratory failure caused by a significant lung maturation delay. The few animals that reach adulthood have a normal lifespan, but present areas of atelectasis mixed with patches of emphysema and normal tissue in the lung. METHODS: Eight double knockout and eight control mice were analyzed using micro-X-ray computerized tomography and a Small Animal Physiological Monitoring system. Tissues and samples from these mice were analyzed using standard histological and Molecular Biology methods and the significance of the results analyzed using a Student´s T-test. RESULTS: The very few double knockout mice surviving up to adulthood display clear craniofacial abnormalities reminiscent of those seen in RASopathy mouse models, as well as thrombocytopenia, bleeding anomalies, and reduced platelet activation induced by thrombin. These surviving mice also present heart and spleen hyperplasia, and elevated numbers of myeloid-derived suppressor cells in the spleen. Mechanistically, we observed that these phenotypic alterations are accompanied by increased KRAS-GTP levels in heart, platelets and primary mouse embryonic fibroblasts from these animals. CONCLUSIONS: Our data uncovers a new, previously unidentified mechanism capable of triggering a RASopathy phenotype in mice as a result of the combined removal of HRAS and NRAS.


Assuntos
GTP Fosfo-Hidrolases , Camundongos Knockout , Fenótipo , Proteínas Proto-Oncogênicas p21(ras) , Animais , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Camundongos , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Ativação Plaquetária/genética , Baço/patologia , Baço/metabolismo , Proteínas Monoméricas de Ligação ao GTP
3.
Int J Mol Sci ; 25(11)2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38892129

RESUMO

This study focuses on understanding the transcriptional heterogeneity of activated platelets and its impact on diseases such as sepsis, COVID-19, and systemic lupus erythematosus (SLE). Recognizing the limited knowledge in this area, our research aims to dissect the complex transcriptional profiles of activated platelets to aid in developing targeted therapies for abnormal and pathogenic platelet subtypes. We analyzed single-cell transcriptional profiles from 47,977 platelets derived from 413 samples of patients with these diseases, utilizing Deep Neural Network (DNN) and eXtreme Gradient Boosting (XGB) to distinguish transcriptomic signatures predictive of fatal or survival outcomes. Our approach included source data annotations and platelet markers, along with SingleR and Seurat for comprehensive profiling. Additionally, we employed Uniform Manifold Approximation and Projection (UMAP) for effective dimensionality reduction and visualization, aiding in the identification of various platelet subtypes and their relation to disease severity and patient outcomes. Our results highlighted distinct platelet subpopulations that correlate with disease severity, revealing that changes in platelet transcription patterns can intensify endotheliopathy, increasing the risk of coagulation in fatal cases. Moreover, these changes may impact lymphocyte function, indicating a more extensive role for platelets in inflammatory and immune responses. This study identifies crucial biomarkers of platelet heterogeneity in serious health conditions, paving the way for innovative therapeutic approaches targeting platelet activation, which could improve patient outcomes in diseases characterized by altered platelet function.


Assuntos
Plaquetas , COVID-19 , Lúpus Eritematoso Sistêmico , Aprendizado de Máquina , SARS-CoV-2 , Sepse , Análise de Célula Única , Transcriptoma , Humanos , COVID-19/sangue , COVID-19/genética , COVID-19/virologia , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/sangue , Plaquetas/metabolismo , Análise de Célula Única/métodos , Sepse/genética , Sepse/sangue , Perfilação da Expressão Gênica/métodos , Ativação Plaquetária/genética
4.
Am J Hum Genet ; 107(2): 211-221, 2020 08 06.
Artigo em Inglês | MEDLINE | ID: mdl-32649856

RESUMO

Dual antiplatelet therapy reduces ischemic events in cardiovascular disease, but it increases bleeding risk. Thrombin receptors PAR1 and PAR4 are drug targets, but the role of thrombin in platelet aggregation remains largely unexplored in large populations. We performed a genome-wide association study (GWAS) of platelet aggregation in response to full-length thrombin, followed by clinical association analyses, Mendelian randomization, and functional characterization including iPSC-derived megakaryocyte and platelet experiments. We identified a single sentinel variant in the GRK5 locus (rs10886430-G, p = 3.0 × 10-42) associated with increased thrombin-induced platelet aggregation (ß = 0.70, SE = 0.05). We show that disruption of platelet GRK5 expression by rs10886430-G is associated with enhanced platelet reactivity. The proposed mechanism of a GATA1-driven megakaryocyte enhancer is confirmed in allele-specific experiments. Utilizing further data, we demonstrate that the allelic effect is highly platelet- and thrombin-specific and not likely due to effects on thrombin levels. The variant is associated with increased risk of cardiovascular disease outcomes in UK BioBank, most strongly with pulmonary embolism. The variant associates with increased risk of stroke in the MEGASTROKE, UK BioBank, and FinnGen studies. Mendelian randomization analyses in independent samples support a causal role for rs10886430-G in increasing risk for stroke, pulmonary embolism, and venous thromboembolism through its effect on thrombin-induced platelet reactivity. We demonstrate that G protein-coupled receptor kinase 5 (GRK5) promotes platelet activation specifically via PAR4 receptor signaling. GRK5 inhibitors in development for the treatment of heart failure and cancer could have platelet off-target deleterious effects. Common variants in GRK5 may modify clinical outcomes with PAR4 inhibitors, and upregulation of GRK5 activity or signaling in platelets may have therapeutic benefits.


Assuntos
Plaquetas/fisiologia , Doenças Cardiovasculares/genética , Receptores de Trombina/genética , Transdução de Sinais/genética , Trombina/genética , Alelos , Embolia/genética , Feminino , Estudo de Associação Genômica Ampla/métodos , Insuficiência Cardíaca/genética , Humanos , Pulmão/fisiologia , Masculino , Pessoa de Meia-Idade , Neoplasias/genética , Ativação Plaquetária/genética , Agregação Plaquetária/genética , Receptor PAR-1/genética , Acidente Vascular Cerebral/genética
5.
Blood ; 137(24): 3443-3453, 2021 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-33512486

RESUMO

Glycoprotein VI (GPVI) is the major signaling receptor for collagen on platelets. We have raised 54 nanobodies (Nb), grouped into 33 structural classes based on their complementary determining region 3 loops, against recombinant GPVI-Fc (dimeric GPVI) and have characterized their ability to bind recombinant GPVI, resting and activated platelets, and to inhibit platelet activation by collagen. Nbs from 6 different binding classes showed the strongest binding to recombinant GPVI-Fc, suggesting that there was not a single dominant class. The most potent 3, Nb2, 21, and 35, inhibited collagen-induced platelet aggregation with nanomolar half maximal inhibitory concentration (IC50) values and inhibited platelet aggregation under flow. The binding KD of the most potent Nb, Nb2, against recombinant monomeric and dimeric GPVI was 0.6 and 0.7 nM, respectively. The crystal structure of monomeric GPVI in complex with Nb2 revealed a binding epitope adjacent to the collagen-related peptide (CRP) binding groove within the D1 domain. In addition, a novel conformation of GPVI involving a domain swap between the D2 domains was observed. The domain swap is facilitated by the outward extension of the C-C' loop, which forms the domain swap hinge. The functional significance of this conformation was tested by truncating the hinge region so that the domain swap cannot occur. Nb2 was still able to displace collagen and CRP binding to the mutant, but signaling was abolished in a cell-based NFAT reporter assay. This demonstrates that the C-C' loop region is important for GPVI signaling but not ligand binding and suggests the domain-swapped structure may represent an active GPVI conformation.


Assuntos
Complexo Antígeno-Anticorpo , Plaquetas , Glicoproteínas da Membrana de Plaquetas , Multimerização Proteica , Anticorpos de Domínio Único , Complexo Antígeno-Anticorpo/química , Complexo Antígeno-Anticorpo/metabolismo , Plaquetas/química , Plaquetas/metabolismo , Humanos , Ativação Plaquetária/efeitos dos fármacos , Ativação Plaquetária/genética , Glicoproteínas da Membrana de Plaquetas/química , Glicoproteínas da Membrana de Plaquetas/genética , Glicoproteínas da Membrana de Plaquetas/metabolismo , Domínios Proteicos , Multimerização Proteica/efeitos dos fármacos , Multimerização Proteica/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/farmacologia
6.
BMC Vet Res ; 19(1): 271, 2023 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-38087280

RESUMO

BACKGROUND: Peripheral blood carries a reservoir of mRNAs that regulate cardiac structure and function potential. Although it is well recognized that the typical symptoms of Myxomatous Mitral Valve Disease (MMVD) stage B2 are long-standing hemodynamic disorder and cardiac structure remodeling caused by mitral regurgitation, the transcriptomic alterations in blood from such dogs are not understood. RESULTS: In the present study, comparative high-throughput transcriptomic profiling of blood was performed from normal control (NC) and naturally-occurring MMVD stage B2 (MMVD) dogs. Using Weighted Gene Co-expression Network Analyses (WGCNA), Gene Ontology (GO), and Kyoto Encyclopedia of Gene and Genomes (KEGG), we identified that the turquoise module was the most highly correlated with echocardiographic features and found 64 differentially expressed genes (DEGs) that were significantly enriched in platelet activation related pathways. Therefore, from the turquoise module, we selected five DEGs (MDM2, ROCK1, RIPK1, SNAP23, and ARHGAP35) that, according to real-time qPCR, exhibited significant enrichment in platelet activation related pathways for validation. The results showed that the blood transcriptional abundance of MDM2, ROCK1, RIPK1, and SNAP23 differed significantly (P < 0.01) between NC and MMVD dogs. On the other hand, Correlation Analysis revealed that MDM2, ROCK1, RIPK1, and SNAP23 genes negatively regulated the heart structure parameters, and followed the same trend as observed in WGCNA. CONCLUSION: We screened four platelet activation related genes, MDM2, ROCK1, RIPK1, and SNAP23, which may be considered as the candidate biomarkers for the diagnosis of MMVD stage B2. These findings provided new insights into MMVD pathogenesis.


Assuntos
Doenças do Cão , Doenças das Valvas Cardíacas , Insuficiência da Valva Mitral , Cães , Animais , Valva Mitral/patologia , Doenças das Valvas Cardíacas/genética , Doenças das Valvas Cardíacas/veterinária , Insuficiência da Valva Mitral/genética , Insuficiência da Valva Mitral/veterinária , Ativação Plaquetária/genética , Ecocardiografia/veterinária
7.
Int J Mol Sci ; 24(8)2023 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-37108819

RESUMO

It is currently believed that plaque complication, with the consequent superimposed thrombosis, is a key factor in the clinical occurrence of acute coronary syndromes (ACSs). Platelets are major players in this process. Despite the considerable progress made by the new antithrombotic strategies (P2Y12 receptor inhibitors, new oral anticoagulants, thrombin direct inhibitors, etc.) in terms of a reduction in major cardiovascular events, a significant number of patients with previous ACSs treated with these drugs continue to experience events, indicating that the mechanisms of platelet remain largely unknown. In the last decade, our knowledge of platelet pathophysiology has improved. It has been reported that, in response to physiological and pathological stimuli, platelet activation is accompanied by de novo protein synthesis, through a rapid and particularly well-regulated translation of resident mRNAs of megakaryocytic derivation. Although the platelets are anucleate, they indeed contain an important fraction of mRNAs that can be quickly used for protein synthesis following their activation. A better understanding of the pathophysiology of platelet activation and the interaction with the main cellular components of the vascular wall will open up new perspectives in the treatment of the majority of thrombotic disorders, such as ACSs, stroke, and peripheral artery diseases before and after the acute event. In the present review, we will discuss the novel role of noncoding RNAs in modulating platelet function, highlighting the possible implications in activation and aggregation.


Assuntos
Síndrome Coronariana Aguda , Trombose , Humanos , Plaquetas/metabolismo , Anticoagulantes/farmacologia , Ativação Plaquetária/genética , Hemostasia , Trombose/metabolismo , RNA não Traduzido/metabolismo , Síndrome Coronariana Aguda/metabolismo , Inibidores da Agregação Plaquetária/farmacologia , Agregação Plaquetária
8.
Cytometry A ; 101(4): 290-297, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34997669

RESUMO

Platelets are small anucleate blood cells that contribute to hemostasis, immunity, and inflammation. Circulating platelets are heterogeneous in size, age, receptor expression, and reactivity. They inherit many features from megakaryocytes and are further modified on exposure to bioactive substances in the bloodstream. Among these substances, prothrombotic agonists, vasodilators, and bloodborne pathogens modulate platelet phenotypes via distinct signaling cascades. The ability of platelets to respond to (patho)physiologic signals is incompletely understood but likely depends on their repertoire of surface receptors, which may partition them into discrete subsets with specialized functions and divergent abilities. The single-cell resolution of flow and mass cytometry is ideal for immunophenotyping and allows the identification of platelet subsets in remarkable detail. In this report, we describe the surface markers and gating strategies needed for the comprehensive characterization of platelets.


Assuntos
Plaquetas , Megacariócitos , Biomarcadores/metabolismo , Citometria de Fluxo , Hemostasia , Humanos , Imunofenotipagem , Ativação Plaquetária/genética
9.
Blood ; 136(25): 2933-2945, 2020 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-33331924

RESUMO

Systemic lupus erythematosus (SLE) is an autoimmune inflammatory disease characterized by deposits of immune complexes (ICs) in organs and tissues. The expression of FcγRIIA by human platelets, which is their unique receptor for immunoglobulin G antibodies, positions them to ideally respond to circulating ICs. Whereas chronic platelet activation and thrombosis are well-recognized features of human SLE, the exact mechanisms underlying platelet activation in SLE remain unknown. Here, we evaluated the involvement of FcγRIIA in the course of SLE and platelet activation. In patients with SLE, levels of ICs are associated with platelet activation. Because FcγRIIA is absent in mice, and murine platelets do not respond to ICs in any existing mouse model of SLE, we introduced the FcγRIIA (FCGR2A) transgene into the NZB/NZWF1 mouse model of SLE. In mice, FcγRIIA expression by bone marrow cells severely aggravated lupus nephritis and accelerated death. Lupus onset initiated major changes to the platelet transcriptome, both in FcγRIIA-expressing and nonexpressing mice, but enrichment for type I interferon response gene changes was specifically observed in the FcγRIIA mice. Moreover, circulating platelets were degranulated and were found to interact with neutrophils in FcγRIIA-expressing lupus mice. FcγRIIA expression in lupus mice also led to thrombosis in lungs and kidneys. The model recapitulates hallmarks of human SLE and can be used to identify contributions of different cellular lineages in the manifestations of SLE. The study further reveals a role for FcγRIIA in nephritis and in platelet activation in SLE.


Assuntos
Autoanticorpos/imunologia , Plaquetas/imunologia , Imunoglobulina G/imunologia , Nefrite Lúpica/imunologia , Ativação Plaquetária/imunologia , Receptores de IgG/imunologia , Animais , Autoanticorpos/genética , Plaquetas/patologia , Modelos Animais de Doenças , Imunoglobulina G/genética , Nefrite Lúpica/genética , Nefrite Lúpica/patologia , Camundongos , Camundongos Transgênicos , Ativação Plaquetária/genética , Receptores de IgG/genética
10.
Blood ; 136(8): 974-988, 2020 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-32584951

RESUMO

Several adaptor molecules bind to cytoplasmic tails of ß-integrins and facilitate bidirectional signaling, which is critical in thrombosis and hemostasis. Interfering with integrin-adaptor interactions spatially or temporally to inhibit thrombosis without affecting hemostasis is an attractive strategy for the development of safe antithrombotic drugs. We show for the first time that the 14-3-3ζ-c-Src-integrin-ß3 complex is formed during platelet activation. 14-3-3ζ-c-Src interaction is mediated by the -PIRLGLALNFSVFYYE- fragment (PE16) on the 14-3-3ζ and SH2-domain on c-Src, whereas the 14-3-3ζ-integrin-ß3 interaction is mediated by the -ESKVFYLKMKGDYYRYL- fragment (EL17) on the 14-3-3ζ and -KEATSTF- fragment (KF7) on the ß3-integrin cytoplasmic tail. The EL17-motif inhibitor, or KF7 peptide, interferes with the formation of the 14-3-3ζ-c-Src-integrin-ß3 complex and selectively inhibits ß3 outside-in signaling without affecting the integrin-fibrinogen interaction, which suppresses thrombosis without causing significant bleeding. This study characterized a previously unidentified 14-3-3ζ-c-Src-integrin-ß3 complex in platelets and provided a novel strategy for the development of safe and effective antithrombotic treatments.


Assuntos
Proteínas 14-3-3/metabolismo , Integrina beta3/metabolismo , Ativação Plaquetária , Proteínas Proto-Oncogênicas pp60(c-src)/metabolismo , Proteínas 14-3-3/genética , Adulto , Animais , Feminino , Células HEK293 , Humanos , Integrina beta3/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Complexos Multiproteicos/metabolismo , Complexos Multiproteicos/fisiologia , Ativação Plaquetária/genética , Proteínas Proto-Oncogênicas pp60(c-src)/genética , Transdução de Sinais/fisiologia
11.
Blood ; 135(22): 1969-1982, 2020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32276277

RESUMO

Anticoagulant protein S (PS) in platelets (PSplt) resembles plasma PS and is released on platelet activation, but its role in thrombosis has not been elucidated. Here we report that inactivation of PSplt expression using the Platelet factor 4 (Pf4)-Cre transgene (Pros1lox/loxPf4-Cre+) in mice promotes thrombus propensity in the vena cava, where shear rates are low, but not in the carotid artery, where shear rates are high. At a low shear rate, PSplt functions as a cofactor for both activated protein C and tissue factor pathway inhibitor, thereby limiting factor X activation and thrombin generation within the growing thrombus and ensuring that highly activated platelets and fibrin remain localized at the injury site. In the presence of high thrombin concentrations, clots from Pros1lox/loxPf4-Cre- mice contract, but not clots from Pros1lox/loxPf4-Cre+ mice, because of highly dense fibrin networks. Thus, PSplt controls platelet activation as well as coagulation in thrombi in large veins, but not in large arteries.


Assuntos
Plaquetas/metabolismo , Proteína S/metabolismo , Trombose/sangue , Animais , Tempo de Sangramento , Coagulação Sanguínea/genética , Coagulação Sanguínea/fisiologia , Proteínas de Ligação ao Cálcio/deficiência , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Ativação Plaquetária/genética , Ativação Plaquetária/fisiologia , Agregação Plaquetária/genética , Agregação Plaquetária/fisiologia , Fator Plaquetário 4/genética , Fator Plaquetário 4/metabolismo , Proteína S/genética , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Trombose/etiologia , Trombose/genética , Trombose Venosa/sangue , Trombose Venosa/etiologia , Trombose Venosa/genética
12.
Blood ; 136(15): 1773-1782, 2020 10 08.
Artigo em Inglês | MEDLINE | ID: mdl-32542378

RESUMO

G protein-coupled receptors are critical mediators of platelet activation whose signaling can be modulated by members of the regulator of G protein signaling (RGS) family. The 2 most abundant RGS proteins in human and mouse platelets are RGS10 and RGS18. While each has been studied individually, critical questions remain about the overall impact of this mode of regulation in platelets. Here, we report that mice missing both proteins show reduced platelet survival and a 40% decrease in platelet count that can be partially reversed with aspirin and a P2Y12 antagonist. Their platelets have increased basal (TREM)-like transcript-1 expression, a leftward shift in the dose/response for a thrombin receptor-activating peptide, an increased maximum response to adenosine 5'-diphosphate and TxA2, and a greatly exaggerated response to penetrating injuries in vivo. Neither of the individual knockouts displays this constellation of findings. RGS10-/- platelets have an enhanced response to agonists in vitro, but platelet count and survival are normal. RGS18-/- mice have a 15% reduction in platelet count that is not affected by antiplatelet agents, nearly normal responses to platelet agonists, and normal platelet survival. Megakaryocyte number and ploidy are normal in all 3 mouse lines, but platelet recovery from severe acute thrombocytopenia is slower in RGS18-/- and RGS10-/-18-/- mice. Collectively, these results show that RGS10 and RGS18 have complementary roles in platelets. Removing both at the same time discloses the extent to which this regulatory mechanism normally controls platelet reactivity in vivo, modulates the hemostatic response to injury, promotes platelet production, and prolongs platelet survival.


Assuntos
Plaquetas/metabolismo , Ativação Plaquetária/genética , Proteínas RGS/genética , Trombopoese/genética , Animais , Plaquetas/efeitos dos fármacos , Sobrevivência Celular/genética , Camundongos , Camundongos Knockout , Fosforilação , Fator de Ativação de Plaquetas/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Contagem de Plaquetas , Proteínas RGS/metabolismo , Trombopoese/efeitos dos fármacos
13.
Platelets ; 33(4): 512-519, 2022 May 19.
Artigo em Inglês | MEDLINE | ID: mdl-35264060

RESUMO

Platelets are essential mediators of physiological hemostasis and pathological thrombosis. Currently available tests and markers of platelet activation did not prove successful in guiding treatment decisions for patients with cardiovascular disease, justifying further research into novel markers of platelet reactivity. Platelets contain a variety of microRNAs (miRNAs) and are a major contributor to the extracellular circulating miRNA pool. Levels of platelet-derived miRNAs in the circulation have been associated with different measures of platelet activation as well as antiplatelet therapy and have therefore been implied as potential new markers of platelet reactivity. In contrast to the ex vivo assessment of platelet reactivity by current platelet function tests, miRNA measurements may enable assessment of platelet reactivity in vivo. It remains to be seen however, whether miRNAs may aid clinical diagnostics. Major limitations in the platelet miRNA research field remain the susceptibility to preanalytical variation, non-standardized sample preparation and data normalization that hampers inter-study comparisons. In this review, we provide an overview of the literature on circulating miRNAs as biomarkers of platelet activation, highlighting the underlying biology, the application in patients with cardiovascular disease and antiplatelet therapy and elaborating on technical limitations regarding their quantification in the circulation.


Assuntos
Doenças Cardiovasculares , MicroRNA Circulante , MicroRNAs , Biomarcadores , Plaquetas , Doenças Cardiovasculares/diagnóstico , Doenças Cardiovasculares/tratamento farmacológico , MicroRNA Circulante/genética , Humanos , MicroRNAs/genética , Ativação Plaquetária/genética , Inibidores da Agregação Plaquetária/uso terapêutico
14.
Int J Mol Sci ; 23(2)2022 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-35054930

RESUMO

Inhalation of particulate matter in polluted air causes direct, size-restricted passage in the circulation and pronounced lung inflammation, provoking platelet activation and (non)-fatal cardiovascular complications. To determine potency and mechanism of platelet sensitization via neutrophil enzymes, we performed in vitro aggregation studies in washed human platelets and in murine and human blood, in the presence of elastase, cathepsin G and regular platelet agonists, present in damaged arteries. The impact of both enzymes on in vivo thrombogenicity was studied in the same thrombosis mouse model, previously having demonstrated that neutrophil activation enhances peripheral thrombogenicity. At 0.05 U/mL, cathepsin G activated washed human platelets via PAR1, whereas at 0.35 U/mL, aggregation occurred via PAR4. In Swiss mouse platelet-rich plasma no aggregation occurred by cathepsin G at 0.4 U/mL. In human and murine blood, aggregations by 0.05-0.1 U/mL cathepsin G were similar and not PAR-mediated, but platelet aggregation was inhibited by ADP antagonists, advocating cathepsin G-released ADP in blood as the true agonist of sustained platelet activation. In the mouse thrombosis model, cathepsin G and elastase amplified mild thrombogenicity at blood concentrations that activated platelets in vitro. This study shows that cathepsin G and elastase secreted in the circulation during mild air pollution-induced lung inflammation lyse red blood cell membrane proteins, leading to ADP-leakage into plasma, sensitizing platelets and amplifying their contribution to cardiovascular complications of ambient particle inhalation.


Assuntos
Artérias/metabolismo , Plaquetas/metabolismo , Catepsina G/metabolismo , Neutrófilos/metabolismo , Ativação Plaquetária , Trombose/etiologia , Trombose/metabolismo , Difosfato de Adenosina/metabolismo , Animais , Artérias/patologia , Biomarcadores , Catepsina G/genética , Suscetibilidade a Doenças , Humanos , Camundongos , Camundongos Knockout , Ativação de Neutrófilo , Elastase Pancreática/metabolismo , Ativação Plaquetária/genética , Agregação Plaquetária/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Trombose/patologia
15.
Ann Hum Genet ; 85(2): 80-91, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33249558

RESUMO

Phosphodiesterase 3A (PDE3A) is an enzyme that plays an important role in the regulation of cyclic adenosine monophosphate (cAMP)-mediated intracellular signaling in cardiac myocytes and platelets. PDE3A hydrolyzes cAMP, which results in a decrease in intracellular cAMP levels and leads to platelet activation. Whole-exome sequencing of 50 DNA samples from a healthy Korean population revealed a total of 13 single nucleotide polymorphisms including five missense variants, D12N, Y497C, H504Q, C707R, and A980V. Recombinant proteins for the five variants of PDE3A (and wild-type protein) were expressed in a FreeStyle 293 expression system with site-directed mutagenesis. The expression of the recombinant PDE3A proteins was confirmed with Western blotting. Catalytic activity of the PDE3A missense variants and wild-type enzyme was measured with a PDE-based assay. Effects of the missense variants on the inhibition of PDE3A activity by cilostazol were also investigated. All variant proteins showed reduced activity (33-53%; p < .0001) compared to the wild-type protein. In addition, PDE3A activity was inhibited by cilostazol in a dose-dependent manner and was further suppressed in the missense variants. Specifically, the PDE3A Y497C showed significantly reduced activity, consistent with the predictions of in silico analyses. The present study provides evidence that individuals carrying the PDE3A Y497C variant may have lower enzyme activity for cAMP hydrolysis, which could cause interindividual variation in cAMP-mediated physiological functions.


Assuntos
Cilostazol/administração & dosagem , AMP Cíclico/genética , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/genética , Ativação Plaquetária/efeitos dos fármacos , Adulto , Plaquetas/efeitos dos fármacos , Cilostazol/efeitos adversos , Relação Dose-Resposta a Droga , Feminino , Humanos , Masculino , Mutação de Sentido Incorreto/genética , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Ativação Plaquetária/genética , Inibidores da Agregação Plaquetária/administração & dosagem , Inibidores da Agregação Plaquetária/efeitos adversos , Polimorfismo Genético/genética , Transdução de Sinais/efeitos dos fármacos , Sequenciamento do Exoma
16.
Clin Immunol ; 228: 108755, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33984497

RESUMO

Platelet-bound complement activation products (PC4d) are associated with thrombosis in Systemic Lupus Erythematosus (SLE). This study investigated the effect of PC4d on platelet function, as a mechanistic link to arterial thrombosis. In a cohort of 150 SLE patients, 13 events had occurred within five years of enrollment. Patients with arterial events had higher PC4d levels (13.6 [4.4-24.0] vs. 4.0 [2.5-8.3] net MFI), with PC4d 10 being the optimal cutoff for event detection. The association of arterial events with PC4d remained significant after adjusting for antiphospholipid status, smoking, and prednisone use (p = 0.045). PC4d levels correlated with lower platelet counts (r = -0.26, p = 0.002), larger platelet volumes (r = 0.22, p = 0.009) and increased platelet aggregation: the adenosine diphosphate (ADP) concentration to achieve 50% maximal aggregation (EC50) was lower in patients with PC4d 10 compared with PC4d < 10 (1.6 vs. 3.7, p = 0.038, respectively). These results suggest that PC4d may be a mechanistic marker for vascular disease in SLE.


Assuntos
Plaquetas/metabolismo , Ativação do Complemento/imunologia , Complemento C4/imunologia , Lúpus Eritematoso Sistêmico/complicações , Lúpus Eritematoso Sistêmico/metabolismo , Ativação Plaquetária/genética , Doenças Vasculares/etiologia , Difosfato de Adenosina/metabolismo , Autoanticorpos/imunologia , Autoimunidade , Biomarcadores , Plaquetas/imunologia , Complemento C4/metabolismo , Suscetibilidade a Doenças , Humanos , Lúpus Eritematoso Sistêmico/imunologia , Ativação Plaquetária/imunologia , Agregação Plaquetária , Contagem de Plaquetas , Trombose/etiologia , Trombose/metabolismo , Doenças Vasculares/metabolismo
17.
FASEB J ; 34(5): 6871-6887, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32248623

RESUMO

This study used constitutive CD226 gene knockout (KO) mice as a model to investigate the functions and mechanisms of CD226 in megakaryocyte (MK) maturation and platelet activation. Although CD226 deficiency did not cause MK polyploidization or platelet granule abnormalities, increased MK counts were detected in the femora bone marrow (BM) and spleen of CD226 KO mice. Particularly, CD226 KO mice have a more extensive membrane system in MKs and platelets than wild-type (WT) mice. We also demonstrated that CD226 KO mice displayed increased platelet counts, shortened bleeding time, and enhanced platelet aggregation. CD226 KO platelets had an increased mature platelet ratio compared to the control platelets. In addition, the observed reduction in bleeding time may be due to decreased nitric oxide (NO) production in the platelets. Platelet-specific CD226-deficient mice showed similar increased MK counts, shortened bleeding time, enhanced platelet aggregation, and decreased NO production in platelets. Furthermore, we performed middle cerebral artery occlusion-reperfusion surgery on WT and CD226 KO mice to explore the potential effect of CD226 on acute ischemia-reperfusion injury; the results revealed that CD226 deficiency led to significantly increased infarct area. Thus, CD226 is a promising candidate for the treatment of thrombotic disorders.


Assuntos
Antígenos de Diferenciação de Linfócitos T/sangue , Megacariócitos/citologia , Megacariócitos/fisiologia , Ativação Plaquetária/fisiologia , Animais , Antígenos de Diferenciação de Linfócitos T/genética , Plaquetas/fisiologia , Plaquetas/ultraestrutura , Isquemia Encefálica/sangue , Isquemia Encefálica/genética , Isquemia Encefálica/patologia , Modelos Animais de Doenças , Feminino , Integrina beta3/sangue , Masculino , Megacariócitos/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Ativação Plaquetária/genética , Adesividade Plaquetária/genética , Adesividade Plaquetária/fisiologia , Agregação Plaquetária/genética , Agregação Plaquetária/fisiologia , Contagem de Plaquetas , Trombopoese/genética , Trombopoese/fisiologia
18.
Acta Haematol ; 144(3): 337-344, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33227791

RESUMO

Differentiating the aetiology of thrombocytosis is limited yet crucial in patients with essential thrombocythaemia (ET). MicroRNAs (miRNAs) regulate haematopoiesis and lineage commitment; aberrant expression of miRNAs plays an important role in myeloproliferative neoplasms. However, the miRNA profile has been poorly explored in ET patients compared to patients with reactive thrombocytosis (RT). A total of 9 samples, including 5 ET patient samples, 2 RT patient samples, and 2 healthy control samples, were analysed in this study. We produced 81.43 million reads from transcripts and 59.60 million reads from small RNAs. We generated a comprehensive miRNA-mRNA regulatory network and identified unique 14 miRNA expression patterns associated with ET. Among the 14 miRNAs, miR-1268a was downregulated in ET and showed an inverse correlation with its 8 putative target genes, including genes associated with thrombus formation and platelet activation (CDH6, EHD2, FUT1, KIF26A, LINC00346, PTPRN, SERF1A, and SLC6A9). Principal component analysis (PCA) showed ET and non-ET groups well clustered in space, suggesting each group had a distinctive expression pattern of mRNAs and miRNAs. These results suggest that the significant dysregulation of miR-1268a and its 8 target genes could be a unique expression of platelet mi-RNAs and miRNA/mRNA regulatory network in ET patients.


Assuntos
MicroRNAs/química , RNA/química , Trombocitemia Essencial/patologia , Transcriptoma , Plaquetas/citologia , Plaquetas/metabolismo , Estudos de Casos e Controles , Análise por Conglomerados , Regulação para Baixo , Redes Reguladoras de Genes , Humanos , Janus Quinase 2/genética , MicroRNAs/metabolismo , Ativação Plaquetária/genética , Polimorfismo de Nucleotídeo Único , Análise de Componente Principal , RNA/metabolismo , RNA-Seq , Trombocitemia Essencial/metabolismo
19.
Mol Biol Rep ; 48(5): 4573-4580, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-34146200

RESUMO

Leukocyte and platelet rich fibrin (L-PRF) is one of the platelet concentrates used to support regeneration and healing process. Many studies showed possible immunological and antibacterial properties of L-PRF. We perform an in vitro study to analyze the effect of L-PRF on platelet activation, platelet-leukocytes interactions and antimicrobial activity, important components in the healing process. Molecular biomarkers related with platelet activation and platelet-leukocyte interactions were analyzed by means of flow cytometry when L-PRF exudate was added to whole blood platelets. L-PRF membrane was used to evaluate antimicrobial activity using Enterococcus faecalis (ATCC 29212), Pseudomonas aeruginosa (ATCC 27853) and Candida albicans (ATCC 90028). Our experimental design allows to evaluate platelet activation and analyze molecular biomarkers of other immune cells and platelet-leukocyte interactions. From the results obtained we can conclude that L-PRF can be a valuable tool in healing process, efficient in activating platelets of whole blood and inhibiting microbial growth. In our opinion, the use of L-PRF exudate, in addition to L-PRF membrane, presents some advantages that have to be considered in clinical trials. Additional research on the characterization and quantification of cells and its products present in the L-PRF exudate, as well as on the temporal factor released. Also, further studies using strains isolated from clinical cases are needed.


Assuntos
Anti-Infecciosos/farmacologia , Plaquetas/metabolismo , Ativação Plaquetária/genética , Fibrina Rica em Plaquetas/química , Cicatrização/genética , Anti-Infecciosos/sangue , Anti-Infecciosos/química , Candida albicans/efeitos dos fármacos , Enterococcus faecalis/efeitos dos fármacos , Humanos , Leucócitos/química , Plasma Rico em Plaquetas/metabolismo , Pseudomonas aeruginosa/efeitos dos fármacos
20.
Brain ; 143(1): 210-221, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31755939

RESUMO

Thrombosis and platelet activation play a central role in stroke pathogenesis, and antiplatelet and anticoagulant therapies are central to stroke prevention. However, whether haematological traits contribute equally to all ischaemic stroke subtypes is uncertain. Furthermore, identification of associations with new traits may offer novel treatment opportunities. The aim of this research was to ascertain causal relationships between a wide range of haematological traits and ischaemic stroke and its subtypes. We obtained summary statistics from 27 published genome-wide association studies of haematological traits involving over 375 000 individuals, and genetic associations with stroke from the MEGASTROKE Consortium (n = 67 000 stroke cases). Using two-sample Mendelian randomization we analysed the association of genetically elevated levels of 36 blood cell traits (platelets, mature/immature red cells, and myeloid/lymphoid/compound white cells) and 49 haemostasis traits (including clotting cascade factors and markers of platelet function) with risk of developing ischaemic (AIS), cardioembolic (CES), large artery (LAS), and small vessel stroke (SVS). Several factors on the intrinsic clotting pathway were significantly associated (P < 3.85 × 10-4) with CES and LAS, but not with SVS (e.g. reduced factor VIII activity with AIS/CES/LAS; raised factor VIII antigen with AIS/CES; and increased factor XI activity with AIS/CES). On the common pathway, increased gamma (γ') fibrinogen was significantly associated with AIS/CES. Furthermore, elevated plateletcrit was significantly associated with AIS/CES, eosinophil percentage of white cells with LAS, and thrombin-activatable fibrinolysis inhibitor activation peptide antigen with AIS. We also conducted a follow-up analysis in UK Biobank, which showed that amongst individuals with atrial fibrillation, those with genetically lower levels of factor XI are at reduced risk of AIS compared to those with normal levels of factor XI. These results implicate components of the intrinsic and common pathways of the clotting cascade, as well as several other haematological traits, in the pathogenesis of CES and possibly LAS, but not SVS. The lack of associations with SVS suggests thrombosis may be less important for this stroke subtype. Plateletcrit and factor XI are potentially tractable new targets for secondary prevention of ischaemic stroke, while factor VIII and γ' fibrinogen require further population-based studies to ascertain their possible aetiological roles.


Assuntos
Coagulação Sanguínea/genética , Fibrinólise/genética , Embolia Intracraniana/sangue , Trombose Intracraniana/sangue , Ativação Plaquetária/genética , Acidente Vascular Cerebral/sangue , Contagem de Células Sanguíneas , Isquemia Encefálica/sangue , Isquemia Encefálica/epidemiologia , Causalidade , Fator VIII/metabolismo , Fator XI/metabolismo , Fibrinogênio/metabolismo , Estudo de Associação Genômica Ampla , Humanos , Embolia Intracraniana/epidemiologia , Trombose Intracraniana/epidemiologia , Análise da Randomização Mendeliana , Fatores de Risco , Acidente Vascular Cerebral/epidemiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA