Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Arterioscler Thromb Vasc Biol ; 40(2): 335-349, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31941383

RESUMO

OBJECTIVE: Cardiovascular disease is a major public health problem. Among cardiovascular disease's risk factors, tobacco smoking is considered the single most preventable cause of death, with thrombosis being the main mechanism of cardiovascular disease mortality in smokers. While tobacco smoking has been on the decline, the use of waterpipes/hookah has been rising, mainly due to the perception that they are less harmful than regular cigarettes. Strikingly, there are few studies on the negative effects of waterpipes on the cardiovascular system, and none regarding their direct contribution to thrombus formation. Approach and Results: We used a waterpipe whole-body exposure protocol that mimics real-life human exposure scenarios and investigated its effects, relative to clean air, on platelet function, hemostasis, and thrombogenesis. We found that waterpipe smoke (WPS)-exposed mice exhibited both shortened thrombus occlusion and bleeding times. Further, our results show that platelets from WPS-exposed mice are hyperactive, with enhanced agonist-induced aggregation, dense and α-granule secretion, αIIbß3 integrin activation, phosphatidylserine expression, and platelet spreading, when compared with clean air-exposed platelets. Finally, at the molecular level, it was found that Akt (protein kinase B) and ERK (extracellular signal-regulated kinases) phosphorylation are enhanced in the WPS and in nicotine-treated platelets. CONCLUSIONS: Our findings demonstrate that WPS exposure directly modulates hemostasis and increases the risk of thrombosis and that this is mediated, in part, via a state of platelet hyperactivity. The negative health impact of WPS/hookah, therefore, should not be underestimated. Moreover, this study should also help in raising public awareness of the toxic effects of waterpipe/hookah.


Assuntos
Plaquetas/efeitos dos fármacos , Artérias Carótidas/efeitos dos fármacos , Ativação Plaquetária/fisiologia , Cachimbos de Água , Fumar/efeitos adversos , Trombose/metabolismo , Animais , Plaquetas/metabolismo , Artérias Carótidas/patologia , Cotinina/toxicidade , Modelos Animais de Doenças , Citometria de Fluxo , Seguimentos , Immunoblotting , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Nicotina/toxicidade , Contagem de Plaquetas , Fumaça/efeitos adversos , Trombose/induzido quimicamente , Fatores de Tempo
2.
Environ Health Prev Med ; 24(1): 58, 2019 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-31521105

RESUMO

Hookah or waterpipe smoking or use is an emerging trend in the US population, especially among the youth. The misperception of hookah being less harmful than cigarettes and the availability of different but "appealing" flavors are considered among the main reasons for this trend. Hookah users however are exposed to many of the same toxic compounds/by-products as cigarette users, but at dramatically higher levels, which might lead to more severe negative health effects. In fact, hookah users are at risks of infections, cancers, lung disease, and other medical conditions. Moreover, because of the overlapping toxicant/chemical profile to conventional cigarettes, hookah smoke effects on the cardiovascular system are thought to be comparable to those of conventional cigarettes. A major source of tobacco addiction is nicotine, whose levels in hookah are extremely variable as they depend on the type of tobacco used. Taken together, in this review of literature, we will provide insights on the negative health effects of hookah in general, with a focus on what is known regarding its impact on the cardiovascular system.


Assuntos
Doenças Cardiovasculares/etiologia , Exposição por Inalação/efeitos adversos , Cachimbos de Água , Poluição por Fumaça de Tabaco/efeitos adversos , Fumar Cachimbo de Água/efeitos adversos , Sistemas Eletrônicos de Liberação de Nicotina , Substâncias Perigosas/análise , Substâncias Perigosas/toxicidade , Humanos , Nicotina/análise , Nicotina/toxicidade , Nicotiana/química , Poluição por Fumaça de Tabaco/análise , Fumar Cachimbo de Água/epidemiologia
3.
Biochem Biophys Res Commun ; 500(2): 268-274, 2018 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-29649481

RESUMO

The current work investigates the notion that inducible clustering of signaling mediators of the IKK pathway is important for platelet activation. Thus, while the CARMA1, Bcl10, and MALT1 (CBM) complex is essential for triggering IKK/NF-κB activation upon platelet stimulation, the signals that elicit its formation and downstream effector activation remain elusive. We demonstrate herein that IKKß is involved in membrane fusion, and serves as a critical protein kinase required for initial formation and the regulation of the CARMA1/MALT1/Bcl10/CBM complex in platelets. We also show that IKKß regulates these processes via modulation of phosphorylation of Bcl10 and IKKγ polyubiquitination. Collectively, our data demonstrate that IKKß regulates membrane fusion and the remodeling of the CBM complex formation.


Assuntos
Proteína 10 de Linfoma CCL de Células B/metabolismo , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Quinase I-kappa B/metabolismo , Proteína de Translocação 1 do Linfoma de Tecido Linfoide Associado à Mucosa/metabolismo , Animais , Plaquetas/metabolismo , Plaquetas/ultraestrutura , Deleção de Genes , Fusão de Membrana , Camundongos , Fosforilação , Proteína Quinase C-delta/metabolismo , Ubiquitinação
4.
Biochim Biophys Acta ; 1863(2): 314-21, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26628381

RESUMO

The regulators of G protein signaling (RGS) protein superfamily negatively controls G protein-coupled receptor signal transduction pathways. One of the members of this family, RGS16, is highly expressed in megakaryocytes and platelets. Studies of its function in platelet and megakaryocyte biology have been limited, in part, due to lack of pharmacological inhibitors. For example, RGS16 overexpression inhibited CXC chemokine receptor 4 (CXCR4)-mediated megakaryocyte migration. More recent studies showed that the chemokine stromal cell-derived factor (SDF1α or CXCL12) regulates platelet function via CXCR4. Based on these considerations, the present study investigated the capacity of RGS16 to regulate CXCL12-dependent platelet function, using the RGS16 knockout mouse model (Rgs16(-/-)). RGS16-deficient platelets had increased protease activated receptor 4 and collagen-induced aggregation, as well as increased CXCL12-dependent agonist-induced aggregation, dense and alpha granule secretion, integrin αIIbß3 activation and phosphatidylserine exposure compared to those from WT littermates. CXCL12 alone did not stimulate aggregation or secretion in either RGS16-deficient or WT platelets. Furthermore, platelets from Rgs16(-/-) mice displayed enhanced phosphorylation of ERK and Akt following CXCL12 stimulation relative to controls. Finally, we also found that PKCδ is involved in regulating CXCL12-dependent activation of ERK and Akt, in the Rgs16-deficient platelets. Collectively, our findings provide the first evidence that RGS16 plays an important role in platelet function by modulating CXCL12-dependent platelet activation.


Assuntos
Quimiocina CXCL12/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Proteínas RGS/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Colágeno/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Immunoblotting , Camundongos Knockout , Fosfatidilserinas/metabolismo , Fosforilação/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas RGS/genética , Receptores Ativados por Proteinase/metabolismo , Transdução de Sinais/genética
5.
Biochem Biophys Res Commun ; 493(2): 1069-1074, 2017 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-28928091

RESUMO

Given that platelet hyperactivity is known to give rise to thrombotic disorders, new and/or novel antiplatelet therapies are constantly being developed to add to, or to complement the current arsenal of agents. To this end, adenosine diphosphate (ADP) is an important platelet activator that acts by binding to the G-protein coupled P2Y1 and P2Y12 receptors. Although the contribution of the P2Y12 receptor to the genesis of thrombosis is well established, the parenteral arsenal of drugs targeting this receptor in clinical use is limited to cangrelor. In this study, we investigated the potential antiplatelet activity of an antibody targeting the ligand-binding domain of the P2Y12 receptor (abbreviated P2Y12Ab). Our in vitro studies revealed that the P2Y12Ab could effectively inhibit aggregation induced by ADP, as well as that triggered by the thromboxane receptor agonist U46619. Additionally, using FACS analysis, we observed reduced P-selectin, phosphatidylserine exposure and integrin activation in the presence of P2Y12Ab. As for its in vivo effects, the P2Y12Ab also demonstrated protection against thrombus formation; albeit this was accompanied with a bleeding diathesis (longer bleeding time). Notably, this inhibitory profile is consistent with that observed with oral anti-P2Y12 agents. Collectively, our findings demonstrate that the P2Y12Ab functionally blocks platelet activity in vitro and in vivo, and support the notion that it can be purposed as a parenteral antiplatelet agent, to be used in conjunction with and/or as a complement to current antiplatelet therapies.


Assuntos
Anticorpos/uso terapêutico , Plaquetas/efeitos dos fármacos , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/uso terapêutico , Antagonistas do Receptor Purinérgico P2Y/uso terapêutico , Trombose/prevenção & controle , Animais , Anticorpos/farmacologia , Plaquetas/citologia , Plaquetas/metabolismo , Hemostasia/efeitos dos fármacos , Humanos , Integrinas/metabolismo , Camundongos Endogâmicos C57BL , Inibidores da Agregação Plaquetária/farmacologia , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Receptores Purinérgicos P2Y12/metabolismo , Trombose/metabolismo
7.
Arterioscler Thromb Vasc Biol ; 35(3): 637-44, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25593131

RESUMO

OBJECTIVE: Platelet hyperactivity is associated with vascular disease and contributes to the genesis of thrombotic disorders. ADP plays an important role in platelet activation and activates platelets through 2 G-protein-coupled receptors, the Gq-coupled P2Y1 receptor (P2Y1R), and the Gi-coupled P2Y12 receptor. Although the involvement of the P2Y1R in thrombogenesis is well established, there are no antagonists that are currently available for clinical use. APPROACH AND RESULTS: Our goal is to determine whether a novel antibody targeting the ligand-binding domain, ie, second extracellular loop (EL2) of the P2Y1R (EL2Ab) could inhibit platelet function and protect against thrombogenesis. Our results revealed that the EL2Ab does indeed inhibit ADP-induced platelet aggregation, in a dose-dependent manner. Furthermore, EL2Ab was found to inhibit integrin GPIIb-IIIa activation, dense and α granule secretion, and phosphatidylserine exposure. These inhibitory effects translated into protection against thrombus formation, as evident by a prolonged time for occlusion in a FeCl3-induced thrombosis model, but this was accompanied by a prolonged tail bleeding time. We also observed a dose-dependent displacement of the radiolabeled P2Y1R antagonist [(3)H]MRS2500 from its ligand-binding site by EL2Ab. CONCLUSIONS: Collectively, our findings demonstrate that EL2Ab binds to and exhibits P2Y1R-dependent function-blocking activity in the context of platelets. These results add further evidence for a role of the P2Y1R in thrombosis and validate the concept that targeting it is a relevant alternative or complement to current antiplatelet strategies.


Assuntos
Anticorpos/farmacologia , Plaquetas/efeitos dos fármacos , Fibrinolíticos/farmacologia , Ativação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/farmacologia , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Receptores Purinérgicos P2Y1/efeitos dos fármacos , Animais , Anticorpos/metabolismo , Anticorpos/toxicidade , Sítios de Ligação , Ligação Competitiva , Plaquetas/metabolismo , Lesões das Artérias Carótidas/sangue , Lesões das Artérias Carótidas/tratamento farmacológico , Nucleotídeos de Desoxiadenina/metabolismo , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Epitopos , Fibrinolíticos/metabolismo , Fibrinolíticos/toxicidade , Hemorragia/induzido quimicamente , Hemostasia/efeitos dos fármacos , Humanos , Ligantes , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfatidilserinas/sangue , Agregação Plaquetária/efeitos dos fármacos , Inibidores da Agregação Plaquetária/metabolismo , Inibidores da Agregação Plaquetária/toxicidade , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/antagonistas & inibidores , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Antagonistas do Receptor Purinérgico P2Y/metabolismo , Antagonistas do Receptor Purinérgico P2Y/toxicidade , Receptores Purinérgicos P2Y1/sangue , Receptores Purinérgicos P2Y1/deficiência , Receptores Purinérgicos P2Y1/genética , Receptores Purinérgicos P2Y1/imunologia , Receptores Purinérgicos P2Y1/metabolismo , Vesículas Secretórias/efeitos dos fármacos , Vesículas Secretórias/metabolismo , Trombose/sangue , Trombose/prevenção & controle , Fatores de Tempo
8.
Biochem Biophys Res Commun ; 462(4): 378-82, 2015 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-25969426

RESUMO

Regulators of G protein signaling (RGS) proteins are known to interact with and negatively regulate/turn-off G protein activation. RGS18 is identified as an R4 subfamily member of this family with specific expression in hematopoietic progenitors, myeloerythroid cells, megakaryocytes and platelets. Studies focused on understanding its function in platelet biology have been limited, in part, due to lack of pharmacological inhibitors. Thus, the present study investigated the function of RGS18 in platelets, using the RGS18 knockout mouse model (RGS18(-/-)). We identified phenotypic differences between RGS18(-/-) and wild-type (WT) mice, and show that RGS18 plays a significant role in hemostasis and thrombosis. Hence, RGS18 deficiency markedly shortened bleeding as well as occlusion times (in vivo). Furthermore, RGS18(-/-) platelets displayed hyper-responsiveness with regards to agonist induced aggregation (in vitro). This gain of function phenotype may serve as the mechanism or explain, at least in part, the enhanced hemostasis and thrombosis phenotype observed in the RGS18 deletion mice. Collectively, our findings provide valuable insight and highlight a critical and direct role for RGS18 in modulating platelet function.


Assuntos
Hemostasia/fisiologia , Agregação Plaquetária/fisiologia , Proteínas RGS/fisiologia , Trombose/fisiopatologia , Animais , Camundongos , Camundongos Knockout , Proteínas RGS/genética
9.
Blood ; 121(22): 4567-74, 2013 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-23613522

RESUMO

Platelet secretion plays a key role in thrombosis, thus the platelet secretory machinery offers a unique target to modulate hemostasis. We report the regulation of platelet secretion via phosphorylation of SNAP-23 at Ser95. Phosphorylation of this t-soluble N-ethylmaleimide sensitive factor attachment protein receptor (SNARE) occurs upon activation of known elements of the platelet signaling cascades (ie, phospholipase C, [Ca(2+)]i, protein kinase C) and requires IκB kinase (IKK)-ß. Other elements of the nuclear factor κB/IκB cascade (ie, IKK-α,-ß,-γ/NEMO and CARMA/MALT1/Bcl10 complex) are present in anucleate platelets and IκB is phosphorylated upon activation, suggesting that this pathway is active in platelets and implying a nongenomic role for IKK. Inhibition of IKK-ß, either pharmacologically (with BMS-345541, BAY11-7082, or TPCA-1) or by genetic manipulation (platelet factor 4 Cre:IKK-ß(flox/flox)), blocked SNAP-23 phosphorylation, platelet secretion, and SNARE complex formation; but, had no effect on platelet morphology or other metrics of platelet activation. Consistently, SNAP-23 phosphorylation enhanced membrane fusion of SNARE-containing proteoliposomes. In vivo studies with IKK inhibitors or platelet-specific IKK-ß knockout mice showed that blocking IKK-ß activity significantly prolonged tail bleeding times, suggesting that currently available IKK inhibitors may affect hemostasis.


Assuntos
Plaquetas/metabolismo , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Ativação Plaquetária/fisiologia , Proteínas Qb-SNARE/metabolismo , Proteínas Qc-SNARE/metabolismo , Animais , Grânulos Citoplasmáticos/metabolismo , Ativação Enzimática/fisiologia , Hemostasia/fisiologia , Fusão de Membrana/fisiologia , Camundongos , Camundongos Knockout , Fosforilação/fisiologia , Proteínas SNARE/metabolismo , Transdução de Sinais/fisiologia
10.
Wound Repair Regen ; 23(5): 644-56, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26135854

RESUMO

Very little is known about lipid function during wound healing, and much less during impaired healing. Such understanding will help identify what roles lipid signaling plays in the development of impaired/chronic wounds. We took a lipidomics approach to study the alterations in lipid profile in the LIGHT(-/-) mouse model of impaired healing which has characteristics that resemble those of impaired/chronic wounds in humans, including high levels of oxidative stress, excess inflammation, increased extracellular matrix degradation and blood vessels with fibrin cuffs. The latter suggests excess coagulation and potentially increased platelet aggregation. We show here that in these impaired wounds there is an imbalance in the arachidonic acid (AA) derived eicosonoids that mediate or modulate inflammatory reactions and platelet aggregation. In the LIGHT(-/-) impaired wounds there is a significant increase in enzymatically derived breakdown products of AA. We found that early after injury there was a significant increase in the eicosanoids 11-, 12-, and 15-hydroxyeicosa-tetranoic acid, and the proinflammatory leukotrienes (LTD4 and LTE) and prostaglandins (PGE2 and PGF2α ). Some of these eicosanoids also promote platelet aggregation. This led us to examine the levels of other eicosanoids known to be involved in the latter process. We found that thromboxane (TXA2 /B2 ), and prostacyclins 6kPGF1α are elevated shortly after wounding and in some cases during healing. To determine whether they have an impact in platelet aggregation and hemostasis, we tested LIGHT(-/-) mouse wounds for these two parameters and found that, indeed, platelet aggregation and hemostasis are enhanced in these mice when compared with the control C57BL/6 mice. Understanding lipid signaling in impaired wounds can potentially lead to development of new therapeutics or in using existing nonsteroidal anti-inflammatory agents to help correct the course of healing.


Assuntos
Ácido Araquidônico/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Estresse Oxidativo , Pele/lesões , Cicatrização/efeitos dos fármacos , Ferimentos e Lesões/tratamento farmacológico , Animais , Modelos Animais de Doenças , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Elastase Pancreática/metabolismo , Transdução de Sinais/efeitos dos fármacos , Pele/metabolismo , Pele/patologia , Suínos , Ferimentos e Lesões/metabolismo , Ferimentos e Lesões/patologia
11.
J Cardiovasc Pharmacol ; 66(2): 177-82, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25853992

RESUMO

Cigarette smoking is a major risk factor for acute coronary thrombosis. In fact, both active/first-hand smoke and passive/second-hand smoke exposure are known to increase the risk of coronary thrombosis. Although recently a new risk has been identified and termed third-hand smoke (THS), which is the residual tobacco smoke contaminant that remains after a cigarette is extinguished, it remains to be determined whether it can also enhance the risk of thrombogenesis, much like first-hand smoke and second-hand smoke. Therefore, the present studies investigated the impact of THS exposure in the context of platelet biology and related disease states. It was found that THS-exposed mice exhibited an enhanced platelet aggregation and secretion responses as well as enhanced integrin GPIIb-IIIa activation. Furthermore, it was found that THS exposure shortens the tail bleeding time and the occlusion time in a model of thrombosis. Thus, our data demonstrate for the first time (at least in mice) that THS exposure increases the risk of thrombosis-based disease states, which is attributed, at least in part, to their hyperactive platelets.


Assuntos
Trombose das Artérias Carótidas/induzido quimicamente , Hemostasia/efeitos dos fármacos , Exposição por Inalação/efeitos adversos , Agregação Plaquetária/efeitos dos fármacos , Produtos do Tabaco/efeitos adversos , Poluição por Fumaça de Tabaco/efeitos adversos , Animais , Trombose das Artérias Carótidas/sangue , Hemostasia/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Agregação Plaquetária/fisiologia
12.
Blood ; 120(12): 2493-500, 2012 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-22791290

RESUMO

Platelets are vital for hemostasis because they release their granule contents in response to vascular damage. Platelet exocytosis is mediated by soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs), whose interactions are governed by regulators, eg, Sec/Munc18 proteins. These proteins chaperone syntaxin t-SNAREs and are required for exocytosis. Platelets contain 3 Munc18 isoforms: Munc18a, Munc18b, and Munc18c. We report that Munc18b is the major isoform and is required for platelet secretion. Familial hemophagocytic lymphohistiocytosis type 5 (FHL5) is caused by defects in the Munc18b/STXBP2 gene. We confirm a previous report showing that platelets from FHL5 patients have defective secretion. Serotonin, ADP/ATP, and platelet factor 4 release was profoundly affected in the 2 biallelic patients and partially in a heterozygous patient. Release of lysosomal contents was only affected in the biallelic platelets. Platelets from the FHL5 biallelic patients showed decreased Munc18b and syntaxin-11 levels were significantly reduced; other syntaxins were unaffected. Munc18b formed complexes with syntaxin-11, SNAP-23, and vesicle-associated membrane protein-8 in human platelets. Other potential secretion regulators, Munc13-4 and Rab27, were also found associated. These data demonstrate a key role for Munc18b, perhaps as a limiting factor, in platelet exocytosis and suggest that it regulates syntaxin-11.


Assuntos
Plaquetas/metabolismo , Exocitose/fisiologia , Linfo-Histiocitose Hemofagocítica/metabolismo , Proteínas Munc18/metabolismo , Proteínas SNARE/metabolismo , Plaquetas/ultraestrutura , Western Blotting , Estudos de Casos e Controles , Grânulos Citoplasmáticos/metabolismo , Citometria de Fluxo , Humanos , Imunoprecipitação , Linfo-Histiocitose Hemofagocítica/genética , Linfo-Histiocitose Hemofagocítica/patologia , Agregação Plaquetária/fisiologia , Proteínas Qa-SNARE/metabolismo
13.
Blood ; 120(12): 2484-92, 2012 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-22767500

RESUMO

The platelet release reaction plays a critical role in thrombosis and contributes to the events that follow hemostasis. Previous studies have shown that platelet secretion is mediated by Soluble NSF Attachment Protein Receptor (SNARE) proteins from granule and plasma membranes. The SNAREs form transmembrane complexes that mediate membrane fusion and granule cargo release. Although VAMP-8 (v-SNARE) and SNAP-23 (a t-SNARE class) are important for platelet secretion, the identity of the functional syntaxin (another t-SNARE class) has been controversial. Previous studies using anti-syntaxin Abs in permeabilized platelets have suggested roles for both syntaxin-2 and syntaxin-4. In the present study, we tested these conclusions using platelets from syntaxin-knockout mouse strains and from a Familial Hemophagocytic Lymphohistiocytosis type 4 (FHL4) patient. Platelets from syntaxin-2 and syntaxin-4 single- or double-knockout mice had no secretion defect. Platelets from a FHL4 patient deficient in syntaxin-11 had a robust defect in agonist-induced secretion although their morphology, activation, and cargo levels appeared normal. Semiquantitative Western blotting showed that syntaxin-11 is the more abundant syntaxin in both human and murine platelets. Coimmunoprecipitation experiments showed that syntaxin-11 can form SNARE complexes with both VAMP-8 and SNAP-23. The results of the present study indicate that syntaxin-11, but not syntaxin-2 or syntaxin-4, is required for platelet exocytosis.


Assuntos
Plaquetas/metabolismo , Exocitose/fisiologia , Linfo-Histiocitose Hemofagocítica/metabolismo , Proteínas Qa-SNARE/fisiologia , Sintaxina 1/fisiologia , Animais , Plaquetas/ultraestrutura , Membrana Celular/metabolismo , Grânulos Citoplasmáticos/metabolismo , Feminino , Humanos , Imunoprecipitação , Linfo-Histiocitose Hemofagocítica/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Agregação Plaquetária , Proteínas Qa-SNARE/genética , Proteínas Qa-SNARE/metabolismo
14.
SAGE Open Med ; 12: 20503121241228446, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38322582

RESUMO

Acute kidney injury is a prevalent disease in hospitalized patients and is continuously increasing worldwide. Various efforts have been made to define and classify acute kidney injury to understand the progression of this disease. Furthermore, deviations from structure and kidney function and the current diagnostic guidelines are not adequately placed due to baseline serum creatinine values, which are rarely known and estimated based on glomerular function rate, resulting in misclassification of acute kidney injury staging. Hence, the current guidelines are still developing to improve and understand the clinical implications of risk factors and earlier predictive biomarkers of acute kidney injury. Yet, studies have indicated disadvantages and limitations with the current acute kidney injury biomarkers, including lack of sensitivity and specificity. Therefore, the present narrative review brings together the most current evidenced-based practice and literature associated with the limitations of the gold standard for acute kidney injury diagnoses, the need for novel acute kidney injury biomarkers, and the process for biomarkers to be qualified for diagnostic use under the following sections and themes. The introduction section situates the anatomy and normal and abnormal kidney functions related to acute kidney injury disorders. Guidelines in providing acute kidney injury definitions and classification are then considered, followed by a discussion of the disadvantages of standard markers used to diagnose acute kidney injury. Characteristics of an ideal acute kidney injury biomarker are discussed concerning sensitivity, specificity, and anatomic location of injury. A particular focus on the role and function of emerging biomarkers is discussed in relation to their applications and significance to the prognosis and severity of acute kidney injury. Findings show emerging markers are early indicators of acute kidney injury prediction in different clinical settings. Finally, the process required for a biomarker to be applied for diagnostic use is explained.

15.
J Clin Invest ; 119(2): 399-407, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19147982

RESUMO

Platelets are central players in atherothrombosis development in coronary artery disease. The PKC family provides important intracellular mechanisms for regulating platelet activity, and platelets express several members of this family, including the classical isoforms PKCalpha and PKCbeta and novel isoforms PKCdelta and PKCtheta. Here, we used a genetic approach to definitively demonstrate the role played by PKCalpha in regulating thrombus formation and platelet function. Thrombus formation in vivo was attenuated in Prkca-/- mice, and PKCalpha was required for thrombus formation in vitro, although this PKC isoform did not regulate platelet adhesion to collagen. The ablation of in vitro thrombus formation in Prkca-/- platelets was rescued by the addition of ADP, consistent with the key mechanistic finding that dense-granule biogenesis and secretion depend upon PKCalpha expression. Furthermore, defective platelet aggregation in response to either collagen-related peptide or thrombin could be overcome by an increase in agonist concentration. Evidence of overt bleeding, including gastrointestinal and tail bleeding, was not seen in Prkca-/- mice. In summary, the effects of PKCalpha ablation on thrombus formation and granule secretion may implicate PKCalpha as a drug target for antithrombotic therapy.


Assuntos
Plaquetas/metabolismo , Grânulos Citoplasmáticos/metabolismo , Proteína Quinase C-alfa/fisiologia , Trombose/etiologia , Difosfato de Adenosina/farmacologia , Animais , Plaquetas/ultraestrutura , Integrinas/fisiologia , Camundongos , Agregação Plaquetária/efeitos dos fármacos , Proteína Quinase C/fisiologia , Proteína Quinase C beta , Transdução de Sinais
16.
Platelets ; 21(3): 167-75, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20196629

RESUMO

Platelets contain an invaginated, tubular membranous structure called the surface-connected open canalicular system (SCCS or OCS), which is contiguous with the plasma membrane and serves as a site for granule fusion and as a reservoir of membrane for platelet spreading. According to ultrastructural studies, platelets from some species lack OCS. In an attempt to correlate biochemical and functional attributes with the presence of an OCS, platelets from human, mouse and dog (OCS(+)), and from cow, camel and horse (OCS(-)) were analysed for differential protein expression and aggregation in response to thrombin. Among the 18 different cytoskeletal and regulatory proteins examined, five (Rac1, RhoA, Ras, calmodulin and Src) were expressed at higher levels in OCS(+) platelets (p < 0.05). Given the role of Arf6 in the formation of tubular invaginations in nucleated cells, the levels of Arf6-GTP were analysed in OCS(+) and OCS(-) platelets. There was no significant correlation between the presence of OCS and total Arf6 or Arf6-GTP levels. Comparison of platelet aggregation between different species suggests that OCS(-) platelets have delayed responses. This comparison of platelets from six different species, which differ in their OCS, shows the differential expression of known signaling components and foreshadows future studies focusing on OCS formation and function.


Assuntos
Plaquetas/citologia , Plaquetas/metabolismo , Proteínas Sanguíneas/biossíntese , Membrana Celular/fisiologia , Fator 6 de Ribosilação do ADP , Fatores de Ribosilação do ADP/metabolismo , Animais , Proteínas Sanguíneas/análise , Camelus , Bovinos , Sequência Conservada , Cães , Guanosina Trifosfato/metabolismo , Cavalos , Humanos , Camundongos , Agregação Plaquetária , Especificidade da Espécie
17.
Mol Biol Cell ; 18(1): 24-33, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17065550

RESUMO

Platelet secretion is critical to hemostasis. Release of granular cargo is mediated by soluble NSF attachment protein receptors (SNAREs), but despite consensus on t-SNAREs usage, it is unclear which Vesicle Associated Membrane Protein (VAMPs: synaptobrevin/VAMP-2, cellubrevin/VAMP-3, TI-VAMP/VAMP-7, and endobrevin/VAMP-8) is required. We demonstrate that VAMP-8 is required for release from dense core granules, alpha granules, and lysosomes. Platelets from VAMP-8-/- mice have a significant defect in agonist-induced secretion, though signaling, morphology, and cargo levels appear normal. In contrast, VAMP-2+/-, VAMP-3-/-, and VAMP-2+/-/VAMP-3-/- platelets showed no defect. Consistently, tetanus toxin had no effect on secretion from permeabilized mouse VAMP-3-/- platelets or human platelets, despite cleavage of VAMP-2 and/or -3. Tetanus toxin does block the residual release from permeabilized VAMP-8-/- platelets, suggesting a secondary role for VAMP-2 and/or -3. These data imply a ranked redundancy of v-SNARE usage in platelets and suggest that VAMP-8-/- mice will be a useful in vivo model to study platelet exocytosis in hemostasis and vascular inflammation.


Assuntos
Plaquetas/metabolismo , Proteínas R-SNARE/metabolismo , Proteínas SNARE/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Plaquetas/efeitos dos fármacos , Plaquetas/ultraestrutura , Cálcio/metabolismo , Exocitose/efeitos dos fármacos , Humanos , Metaloendopeptidases/farmacologia , Camundongos , Camundongos Knockout , Agregação Plaquetária/efeitos dos fármacos , Proteínas Tirosina Quinases/metabolismo , Proteínas R-SNARE/deficiência , Transdução de Sinais/efeitos dos fármacos , Toxina Tetânica/farmacologia , Trombina/farmacologia , Proteína 2 Associada à Membrana da Vesícula/deficiência , Proteína 2 Associada à Membrana da Vesícula/metabolismo , Proteína 3 Associada à Membrana da Vesícula/deficiência
18.
Life Sci ; 262: 118481, 2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-32971104

RESUMO

AIMS: G-protein coupled receptors (GPCRs) tightly regulate platelet function by interacting with various physiological agonists. An essential mediator of GPCR signaling is the G protein αßγ heterotrimers, in which the ßγ subunits are central players in downstream signaling. Herein, we investigated the role of Gßγ subunits in platelet function, hemostasis and thrombogenesis. METHODS: To achieve this goal, platelets from both mice and humans were employed in the context of a small molecule inhibitor of Gßγ, namely gallein. We used an aggregometer to examine aggregation and dense granules secretion. We also used flow cytometry for P-selectin and PAC1 to determine the impact of inhibiting Gßγ on α -granule secretion and αIIbß3 activation. Clot retraction and the platelet spreading assay were used to examine Gßγ role in outside-in platelet signaling, whereas Western blot was employed to examine its role in Akt activation. Finally, we used the bleeding time assay and the FeCl3-induced carotid-artery injury thrombosis model to determine Gßγ contribution to in vivo platelet function. RESULTS: We observed that gallein inhibits platelet aggregation and secretion in response to agonist stimulation, in both mouse and human platelets. Furthermore, gallein also exerted inhibitory effects on integrin αIIbß3 activation, clot retraction, platelet spreading and Akt activation/phosphorylation. Finally, gallein's inhibitory effects manifested in vivo, as documented by its ability to modulate physiological hemostasis and delay thrombus formation. CONCLUSION: Our findings demonstrate, for the first time, that Gßγ subunits directly regulate GPCR-dependent platelet function, in vitro and in vivo. Moreover, these data highlight Gßγ as a novel therapeutic target for managing thrombotic disorders.


Assuntos
Plaquetas/fisiologia , Subunidades beta da Proteína de Ligação ao GTP/metabolismo , Subunidades gama da Proteína de Ligação ao GTP/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Trombose/patologia , Animais , Retração do Coágulo/fisiologia , Modelos Animais de Doenças , Hemostasia/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Agregação Plaquetária/fisiologia , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Xantenos/farmacologia
19.
J Cardiovasc Pharmacol Ther ; 25(6): 578-586, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32691614

RESUMO

BACKGROUND: Smoking is the main preventable cause of death in the United States and worldwide and is associated with serious cardiovascular health consequences, including thrombotic diseases. Recently, electronic cigarettes (e-cigarettes) and, in particular JUUL, have attained wide popularity among smokers, nonsmokers, pregnant females, and even the youth, which is alarming. Interestingly, there is/are no information/studies regarding the effect of JUUL on cardiovascular diseases, specifically in the context of modulation of platelet activation. Thus, it is important to discern the cardiovascular disease health risks associated with JUUL. METHODS AND RESULTS: We used a passive e-vape vapor inhalation system where C57BL/6J mice (10-12 weeks old) were exposed to JUUL e-cigarette vape. Menthol flavored JUUL pods containing 5% nicotine by weight were used as the e-liquid. Mice were exposed to a total of 70 puffs daily for 2 weeks; 3-second puff duration, and 25-second puff interval. The effects of JUUL relative to clean air were analyzed, on mouse platelet function in vitro (eg, aggregation) and in vivo (eg, FeCl3-induced carotid artery injury thrombosis model). Our results indicate that short-term exposure to JUUL e-cigarette causes hyperactivation of platelets and shortens the thrombus occlusion as well as hemostasis/bleeding times, relative to clean air (medians of 14 vs. 200 seconds, P < .01 and 35 vs. 295 seconds, P < .001, respectively). CONCLUSION: Our findings document-for the first time-that short-term exposure to the JUUL e-cigarette increases the risk of thrombotic events, in part by modulating platelet function, such as aggregation and secretion, in mice.


Assuntos
Plaquetas/metabolismo , Trombose das Artérias Carótidas/etiologia , Vapor do Cigarro Eletrônico/efeitos adversos , Sistemas Eletrônicos de Liberação de Nicotina , Ativação Plaquetária , Vaping/efeitos adversos , Animais , Trombose das Artérias Carótidas/sangue , Modelos Animais de Doenças , Masculino , Camundongos Endogâmicos C57BL , Fosfatidilserinas/sangue , Agregação Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Transdução de Sinais , Vaping/sangue
20.
Biochem Biophys Rep ; 20: 100685, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31508510

RESUMO

While changes in intracellular calcium levels is a central step in platelet activation and thrombus formation, the contribution and mechanism of receptor-operated calcium entry (ROCE) via transient receptor potential channels (TRPCs) in platelets remains poorly defined. In previous studies, we have shown that TRPC6 regulates hemostasis and thrombosis, in mice. In the present studies, we employed a knockout mouse model system to characterize the role of TRPC6 in ROCE and platelet activation. It was observed that the TRPC6 deletion (Trpc6 -/- ) platelets displayed impaired elevation of intracellular calcium, i.e., defective ROCE. Moreover, these platelets also exhibited defects in a host of functional responses, namely aggregation, granule secretion, and integrin αIIbß3. Interestingly, the aforementioned defects were specific to the thromboxane receptor (TPR), as no impaired responses were observed in response to ADP or the thrombin receptor-activating peptide 4 (TRAP4). The defect in ROCE in the Trpc6 -/- was also observed with 1-oleoyl-2-acetyl-sn-glycerol (OAG). Finally, our studies also revealed that TRPC6 regulates clot retraction. Taken together, our findings demonstrate that TRPC6 directly regulates TPR-dependent ROCE and platelet function. Thus, TRPC6 may serve as a novel target for the therapeutic management of thrombotic diseases.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA