Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
J Pharmacol Exp Ther ; 2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38719479

RESUMO

Substantial clinical and preclinical evidence indicates that transient receptor potential vanilloid 1 (TRPV1) receptors are expressed on terminals of colorectal chemoreceptors and mechanoreceptors and are involved in various rectal hypersensitivity disorders with common features of colorectal overactivity. These stimulatory properties of TRPV1 receptors on colorectal function suggested that brief stimulation of TRPV1 might provide a means of pharmacologically activating the colorectum to induce defecation in patients with an "unresponsive" colorectum. The current studies explored the basic features of TRPV1 receptor-induced contractions of the colorectum in anesthetized rats with and without acute spinal cord injury (aSCI). Cumulative concentration-response curves to intrarectal (IR) capsaicin (CAP) solutions (0.003% to 3.0%) were performed in anesthetized aSCI and spinal intact rats. CAP produced an "inverted U", cumulative concentration-response curve with a threshold for inducing colorectal contractions at 0.01% and a peak response at 0.1% and slight decreases in responses up to 3%. Decreases in responses with concentrations > 0.1% are due to a rapid desensitization (i.e. < 30 min) of TRPV1 receptors to each successive dose. Desensitization appeared fully recovered within 24 hours in spinal intact rats. Colorectal contractions were completely blocked by atropine, indicating a reflexogenic activation of parasympathetic neurons, and responses were completely unaffected by a neurokinin 2 receptor antagonist, indicating that release of neurokinin A (NKA) from afferent terminals and subsequent direct contractions of the smooth muscle was not involved. IR administration of 3 other TRPV1 receptor agonists produced similar results as CAP. Significance Statement Individuals with spinal cord injury often lose control of defecation. Time consuming and inconvenient bowel programs using digital stimulation of the rectum and manual extraction of stool are used to empty the bowel. We show that intrarectal administration of the TRPV1 receptor agonist, capsaicin, can induce rapid onset, short duration colorectal contractions capable of inducing defecation in spinal cord injured and intact rats. Therefore, TRPV1 agonists show promise as a potential therapeutics to induce defecation in individuals with neurogenic bowel.

2.
Spinal Cord ; 59(9): 1013-1017, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34404911

RESUMO

STUDY DESIGN: Animal proof of principle study. OBJECTIVES: Bladder and bowel dysfunction are common after spinal cord injury (SCI) and in the elderly. Neurokinin 2 receptor agonists are known to produce on-demand urination and defecation in adult SCI rats. This study compared the ability of a neurokinin 2 receptor (NK2R) agonist to produce bladder and colorectal contractions in both young adult and aged SCI rats. SETTING: Dignify Therapeutics and Integrated Laboratory Systems, Durham, NC USA. METHODS: Bladder and colorectal pressure and voiding efficiency were measured in response to the NK2R agonist, [Lys5,Me,Leu9,Nle10]-NKA(4-10) (LMN-NKA), in anesthetized animals. The potency and efficacy of LMN-NKA was examined in young adult and aged SCI (T3 or T9 transected) rats, with young adult and aged spinal intact rats included as controls. RESULTS: LMN-NKA (3-300 µg/kg i.v.) produced dose-dependent increases in bladder and colorectal pressure in all anesthetized rats. No differences in the bladder or colorectal pressure responses or voiding efficiency were observed with age or after SCI. The level of SCI did not change the pharmacodynamic responses to the agonist. CONCLUSIONS: An NK2R agonist produced similar responses in young adult and aged SCI rats, suggesting this class of agonists could be used as a potential therapy to induce on-demand urination and defecation in aged populations, with or without SCI.


Assuntos
Neoplasias Colorretais , Traumatismos da Medula Espinal , Animais , Ratos , Ratos Sprague-Dawley , Receptores da Neurocinina-2 , Medula Espinal , Traumatismos da Medula Espinal/complicações , Traumatismos da Medula Espinal/tratamento farmacológico , Bexiga Urinária , Micção
3.
Blood ; 132(18): 1951-1962, 2018 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-30131434

RESUMO

RAP GTPases, important regulators of cellular adhesion, are abundant signaling molecules in the platelet/megakaryocytic lineage. However, mice lacking the predominant isoform, RAP1B, display a partial platelet integrin activation defect and have a normal platelet count, suggesting the existence of a RAP1-independent pathway to integrin activation in platelets and a negligible role for RAP GTPases in megakaryocyte biology. To determine the importance of individual RAP isoforms on platelet production and on platelet activation at sites of mechanical injury or vascular leakage, we generated mice with megakaryocyte-specific deletion (mKO) of Rap1a and/or Rap1b Interestingly, Rap1a/b-mKO mice displayed a marked macrothrombocytopenia due to impaired proplatelet formation by megakaryocytes. In platelets, RAP isoforms had redundant and isoform-specific functions. Deletion of RAP1B, but not RAP1A, significantly reduced α-granule secretion and activation of the cytoskeleton regulator RAC1. Both isoforms significantly contributed to thromboxane A2 generation and the inside-out activation of platelet integrins. Combined deficiency of RAP1A and RAP1B markedly impaired platelet aggregation, spreading, and clot retraction. Consistently, thrombus formation in physiological flow conditions was abolished in Rap1a/b-mKO, but not Rap1a-mKO or Rap1b-mKO, platelets. Rap1a/b-mKO mice were strongly protected from experimental thrombosis and exhibited a severe defect in hemostasis after mechanical injury. Surprisingly, Rap1a/b-mKO platelets were indistinguishable from controls in their ability to prevent blood-lymphatic mixing during development and hemorrhage at sites of inflammation. In summary, our studies demonstrate an essential role for RAP1 signaling in platelet integrin activation and a critical role in platelet production. Although important for hemostatic/thrombotic plug formation, platelet RAP1 signaling is dispensable for vascular integrity during development and inflammation.


Assuntos
Plaquetas/citologia , Deleção de Genes , Adesividade Plaquetária , Trombopoese , Proteínas rap de Ligação ao GTP/genética , Proteínas rap1 de Ligação ao GTP/genética , Animais , Plaquetas/metabolismo , Hemostasia , Integrinas/metabolismo , Camundongos , Camundongos Knockout , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Trombocitopenia/genética , Trombocitopenia/metabolismo , Proteínas rap de Ligação ao GTP/metabolismo , Proteínas rap1 de Ligação ao GTP/metabolismo
4.
J Biol Chem ; 292(5): 1691-1704, 2017 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-27903653

RESUMO

The class I PI3K family of lipid kinases plays an important role in integrin αIIbß3 function, thereby supporting thrombus growth and consolidation. Here, we identify Ras/Rap1GAP Rasa3 (GAP1IP4BP) as a major phosphatidylinositol 3,4,5-trisphosphate-binding protein in human platelets and a key regulator of integrin αIIbß3 outside-in signaling. We demonstrate that cytosolic Rasa3 translocates to the plasma membrane in a PI3K-dependent manner upon activation of human platelets. Expression of wild-type Rasa3 in integrin αIIbß3-expressing CHO cells blocked Rap1 activity and integrin αIIbß3-mediated spreading on fibrinogen. In contrast, Rap1GAP-deficient (P489V) and Ras/Rap1GAP-deficient (R371Q) Rasa3 had no effect. We furthermore show that two Rasa3 mutants (H794L and G125V), which are expressed in different mouse models of thrombocytopenia, lack both Ras and Rap1GAP activity and do not affect integrin αIIbß3-mediated spreading of CHO cells on fibrinogen. Platelets from thrombocytopenic mice expressing GAP-deficient Rasa3 (H794L) show increased spreading on fibrinogen, which in contrast to wild-type platelets is insensitive to PI3K inhibitors. Together, these results support an important role for Rasa3 in PI3K-dependent integrin αIIbß3-mediated outside-in signaling and cell spreading.


Assuntos
Proteínas Ativadoras de GTPase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transdução de Sinais/fisiologia , Substituição de Aminoácidos/genética , Animais , Plaquetas/metabolismo , Plaquetas/patologia , Células CHO , Cricetinae , Cricetulus , Modelos Animais de Doenças , Proteínas Ativadoras de GTPase/genética , Humanos , Camundongos , Camundongos Mutantes , Mutação de Sentido Incorreto , Fosfatidilinositol 3-Quinases/genética , Fosfatos de Fosfatidilinositol/genética , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/genética , Receptores Citoplasmáticos e Nucleares/genética , Trombocitopenia/genética , Trombocitopenia/metabolismo , Trombocitopenia/patologia
5.
Arterioscler Thromb Vasc Biol ; 36(9): 1838-46, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27417588

RESUMO

OBJECTIVE: The tight regulation of platelet adhesiveness, mediated by the αIIbß3 integrin, is critical for hemostasis and prevention of thrombosis. We recently demonstrated that integrin affinity in platelets is controlled by the guanine nucleotide exchange factor, CalDAG-GEFI (CD-GEFI), and its target, RAP1. In this study, we investigated whether low-level expression of CD-GEFI leads to protection from thrombosis without pathological bleeding in mice. APPROACH AND RESULTS: Cdg1(low) mice were generated by knockin of human CD-GEFI cDNA into the mouse Cdg1 locus. CD-GEFI expression in platelets from Cdg1(low) mice was reduced by ≈90% when compared with controls. Activation of RAP1 and αIIbß3 was abolished at low agonist concentrations and partially inhibited at high agonist concentrations in Cdg1(low) platelets. Consistently, the aggregation response of Cdg1(low) platelets was weaker than that of wild-type platelets, but more efficient than that observed in Cdg1(-/-) platelets. Importantly, Cdg1(low) mice were strongly protected from arterial and immune complex-mediated thrombosis, with only minimal impact on primary hemostasis. CONCLUSIONS: Together, our studies suggest the partial inhibition of CD-GEFI function as a powerful new approach to safely prevent thrombotic complications.


Assuntos
Plaquetas/metabolismo , Fatores de Troca do Nucleotídeo Guanina/deficiência , Hemostasia , Ativação Plaquetária , Trombose/prevenção & controle , Animais , Modelos Animais de Doenças , Genótipo , Fatores de Troca do Nucleotídeo Guanina/sangue , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Camundongos Transgênicos , Mutação , Fenótipo , Agregação Plaquetária , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Transdução de Sinais , Trombose/sangue , Trombose/genética , Fatores de Tempo , Proteínas rap1 de Ligação ao GTP/sangue
6.
Arterioscler Thromb Vasc Biol ; 36(5): 792-9, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-26988592

RESUMO

OBJECTIVE: Platelets are important for the development and progression of atherosclerotic lesions. However, relatively little is known about the contribution of platelet signaling to this pathological process. Our recent work identified 2 independent, yet synergistic, signaling pathways that lead to the activation of the small GTPase Rap1; one mediated by the guanine nucleotide exchange factor, CalDAG-GEFI (CDGI), the other by P2Y12, a platelet receptor for adenosine diphosphate and the target of antiplatelet drugs. In this study, we evaluated lesion formation in atherosclerosis-prone low-density lipoprotein receptor deficient (Ldlr(-/-)) mice lacking CDGI or P2Y12 in hematopoietic cells. APPROACH AND RESULTS: Lethally irradiated Ldlr(-/-) mice were reconstituted with bone marrow from wild-type (WT), Caldaggef1(-/-) (cdgI(-/-)), p2y12(-/-), or cdgI(-/-)p2y12(-/-) (double knockout [DKO]) mice and fed a high-fat diet for 12 weeks. Ldlr(-/-) chimeras deficient for CDGI or P2Y12 developed significantly smaller atherosclerotic lesions in the aortic sinus and in aortas when compared with the Ldlr(-/-)/WT controls. We also observed a significant reduction in platelet-leukocyte aggregates in blood from hypercholesterolemic Ldlr(-/-)/cdgI(-/-) and Ldlr(-/-)/p2y12(-/-) chimeras. Consistently, fewer macrophages and neutrophils were detected in the aortic sinus of Ldlr(-/-)/cdgI(-/-) and Ldlr(-/-)/ p2y12(-/-) chimeras. Compared with controls, the plaque collagen content was significantly higher in Ldlr(-/-) chimeras lacking CDGI. Interestingly, no statistically significant additive effects were seen in Ldlr(-/-)/DKO chimeras when compared with chimeras lacking only CDGI. CONCLUSIONS: Our findings suggest that CDGI is critical for atherosclerotic plaque development in hypercholesterolemic Ldlr(-/-) mice because of its contribution to platelet-leukocyte aggregate formation and leukocyte recruitment to the lesion area.


Assuntos
Aorta/metabolismo , Doenças da Aorta/prevenção & controle , Aterosclerose/prevenção & controle , Fatores de Troca do Nucleotídeo Guanina/deficiência , Placa Aterosclerótica , Animais , Aorta/patologia , Doenças da Aorta/genética , Doenças da Aorta/metabolismo , Doenças da Aorta/patologia , Aterosclerose/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Plaquetas/metabolismo , Quimiotaxia de Leucócito , Colágeno/metabolismo , Dieta Hiperlipídica , Modelos Animais de Doenças , Predisposição Genética para Doença , Fatores de Troca do Nucleotídeo Guanina/genética , Hipercolesterolemia/genética , Hipercolesterolemia/metabolismo , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Inflamação/prevenção & controle , Leucócitos/metabolismo , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infiltração de Neutrófilos , Fenótipo , Adesividade Plaquetária , Receptores de LDL/deficiência , Receptores de LDL/genética , Receptores Purinérgicos P2Y12/deficiência , Receptores Purinérgicos P2Y12/genética , Fatores de Tempo , Proteínas rap1 de Ligação ao GTP/sangue
7.
Blood ; 123(17): 2722-31, 2014 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-24585775

RESUMO

Tight regulation of integrin affinity is critical for hemostasis. A final step of integrin activation is talin binding to 2 sites within the integrin ß cytoplasmic domain. Binding of talin to a membrane-distal NPxY sequence facilitates a second, weaker interaction of talin with an integrin membrane-proximal region (MPR) that is critical for integrin activation. To test the functional significance of these distinct interactions on platelet function in vivo, we generated knock-in mice expressing talin1 mutants with impaired capacity to interact with the ß3 integrin MPR (L325R) or NPLY sequence (W359A). Both talin1(L325R) and talin1(W359A) mice were protected from experimental thrombosis. Talin1(L325R) mice, but not talin(W359A) mice, exhibited a severe bleeding phenotype. Activation of αIIbß3 was completely blocked in talin1(L325R) platelets, whereas activation was reduced by approximately 50% in talin1(W359A) platelets. Quantitative biochemical measurements detected talin1(W359A) binding to ß3 integrin, albeit with a 2.9-fold lower affinity than wild-type talin1. The rate of αIIbß3 activation was slower in talin1(W359A) platelets, which consequently delayed aggregation under static conditions and reduced thrombus formation under physiological flow conditions. Together our data indicate that reduction of talin-ß3 integrin binding affinity results in decelerated αIIbß3 integrin activation and protection from arterial thrombosis without pathological bleeding.


Assuntos
Plaquetas/metabolismo , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Talina/genética , Talina/metabolismo , Trombose/metabolismo , Animais , Cálcio/metabolismo , Citometria de Fluxo , Deleção de Genes , Regulação da Expressão Gênica , Hemostasia , Camundongos , Camundongos Transgênicos , Mutação , Ligação Proteica , Resistência ao Cisalhamento , Estresse Mecânico , Ressonância de Plasmônio de Superfície
8.
J Basic Clin Physiol Pharmacol ; 34(2): 227-233, 2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36377965

RESUMO

OBJECTIVES: Neurokinin 2 receptor (NK2R) agonists may be useful for treating bladder and bowel dysfunction via direct contraction of detrusor and gastrointestinal smooth muscle. The NK2R agonist [Lys5, MeLeu9, Nle10]-NKA(4-10) (LMN-NKA) induces urination and defecation, but also produces the potential side effect of dermal flushing in rats. Although LMN-NKA is a NK2R agonist, it also has affinity for neurokinin 1 receptors (NK1R). Therefore, the goal of this study was to determine the neurokinin receptor (NKR) subtypes responsible for LMN-NKA-induced urination, defecation, and flushing by blocking either NK2Rs or NK1Rs before LMN-NKA administration. METHODS: To accomplish this goal, we developed a simple high-throughput 'rapid detection voiding assay' to detect rapid-onset drug-induced urination and defecation in rats. In LMN-NKA dose-response experiments, LMN-NKA (10-100 µg/kg, subcutaneous) was injected and urination, defecation, and flushing were monitored for 30 min. For NKR antagonist experiments, vehicle, the NK2R antagonist GR159897, or the NK1R antagonist CP-99,994 were injected before an acclimation period. Following acclimation, saline or 100 µg/kg LMN-NKA were injected, and behavior was observed for 30 min. RESULTS: LMN-NKA produced dose-related increases in urination, defecation, and flushing. Blocking NK2Rs reduced urination and blocked defecation, without affecting flushing. Blocking NK1Rs did not change LMN-NKA-induced urination or defecation but reduced LMN-NKA-induced flushing. CONCLUSIONS: Using the rapid detection voiding assay we show that LMN-NKA-induced urination and defecation are mediated by NK2Rs, while flushing is mediated by NK1Rs. Therefore, drugs that are more selective for NK2 vs. NK1Rs should produce rapid-onset urination and defecation without producing the potential side effect of flushing.


Assuntos
Receptores da Neurocinina-2 , Micção , Ratos , Animais , Receptores da Neurocinina-2/agonistas , Neurocinina A/farmacologia , Receptores da Neurocinina-1 , Defecação
10.
J Neurotrauma ; 37(6): 868-876, 2020 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-31642371

RESUMO

Acute administration of [Lys5,Me,Leu9,Nle10]-NKA(4-10) (LMN-NKA) produces contractions of the detrusor and rectum with voiding in intact and acutely spinal cord injured (SCI) rats. In the current study, the ability of LMN-NKA (10 µg/kg or 100 µg/kg, subcutaneous [SC], twice a day [bid]) or vehicle to induce voiding and defecation in chronic SCI rats was examined across 30 days. After the last day of administration, voiding response rates and bladder pressure (BP) responses to LMN-NKA (intravenous [IV] and SC) were evaluated under anesthesia. In conscious rats, LMN-NKA (100 µg/kg) produced dose-dependent micturition within 5 min, with response rates >90%, and voiding efficiency >80% in males and >60% in females, which remained stable across the 1-month test period. Similarly, LMN-NKA administration rapidly induced defecation, which also remained stable. Under anesthesia, LMN-NKA increased BP, voiding efficiency, and voiding response rates, which reached 100% at 3 and 10 µg/kg IV in males and females, respectively. SC administration produced 100% response rates in males (30 µg/kg) but only 71% in females (100 µg/kg). Efficacy in rats chronically treated with LMN-NKA was similar to naïve and vehicle-treated rats, except for reduced voiding efficiency in chronically dosed female rats (100 µg/kg). No differences in bladder weights or collagen-to-smooth muscle ratios in histological sections were seen between the groups. Thus neither tolerance, nor sensitization, to LMN-NKA-induced micturition and defecation occurs with chronic administration in rats with chronic SCI. Efficacy was higher in male than in female rats.


Assuntos
Defecação/efeitos dos fármacos , Fragmentos de Peptídeos/administração & dosagem , Receptores da Neurocinina-2/agonistas , Traumatismos da Medula Espinal/tratamento farmacológico , Micção/efeitos dos fármacos , Animais , Defecação/fisiologia , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Masculino , Ratos , Ratos Sprague-Dawley , Receptores da Neurocinina-2/metabolismo , Traumatismos da Medula Espinal/metabolismo , Traumatismos da Medula Espinal/fisiopatologia , Vértebras Torácicas/lesões , Micção/fisiologia
11.
Sci Transl Med ; 11(522)2019 12 11.
Artigo em Inglês | MEDLINE | ID: mdl-31826978

RESUMO

Platelet transfusions can fail to prevent bleeding in patients with inherited platelet function disorders (IPDs), such as Glanzmann's thrombasthenia (GT; integrin αIIbß3 dysfunction), Bernard-Soulier syndrome [BSS; glycoprotein (GP) Ib/V/IX dysfunction], and the more recently identified nonsyndromic RASGRP2 variants. Here, we used IPD mouse models and real-time imaging of hemostatic plug formation to investigate whether dysfunctional platelets impair the hemostatic function of healthy donor [wild-type (WT)] platelets. In Rasgrp2-/- mice or mice with platelet-specific deficiency in the integrin adaptor protein TALIN1 ("GT-like"), WT platelet transfusion was ineffective unless the ratio between mutant and WT platelets was ~2:1. In contrast, thrombocytopenic mice or mice lacking the extracellular domain of GPIbα ("BSS-like") required very few transfused WT platelets to normalize hemostasis. Both Rasgrp2-/- and GT-like, but not BSS-like, platelets effectively localized to the injury site. Mechanistic studies identified at least two mechanisms of interference by dysfunctional platelets in IPDs: (i) delayed adhesion of WT donor platelets due to reduced access to GPIbα ligands exposed at sites of vascular injury and (ii) impaired consolidation of the hemostatic plug. We also investigated the hemostatic activity of transfused platelets in the setting of dual antiplatelet therapy (DAPT), an acquired platelet function disorder (APD). "DAPT" platelets did not prolong the time to initial hemostasis, but plugs were unstable and frequent rebleeding was observed. Thus, we propose that the endogenous platelet count and the ratio of transfused versus endogenous platelets should be considered when treating select IPD and APD patients with platelet transfusions.


Assuntos
Transtornos Plaquetários/patologia , Plaquetas/patologia , Hemostasia , Transfusão de Plaquetas , Animais , Sítios de Ligação , Terapia Antiplaquetária Dupla , Fatores de Troca do Nucleotídeo Guanina/deficiência , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Humanos , Integrinas/metabolismo , Ligantes , Camundongos Endogâmicos C57BL , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Trombastenia/patologia , Doadores de Tecidos
12.
Blood Adv ; 3(22): 3818-3828, 2019 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-31770438

RESUMO

We previously reported the discovery of a novel lipid deacetylase in platelets, arylacetamide deacetylase-like 1 (AADACL1/NCEH1), and that its inhibition impairs agonist-induced platelet aggregation, Rap1 GTP loading, protein kinase C (PKC) activation, and ex vivo thrombus growth. However, precise mechanisms by which AADACL1 impacts platelet signaling and function in vivo are currently unknown. Here, we demonstrate that AADACL1 regulates the accumulation of ether lipids that impact PKC signaling networks crucial for platelet activation in vitro and in vivo. Human platelets treated with the AADACL1 inhibitor JW480 or the AADACL1 substrate 1-O-hexadecyl-2-acetyl-sn-glycerol (HAG) exhibited decreased platelet aggregation, granule secretion, Ca2+ flux, and PKC phosphorylation. Decreased aggregation and secretion were rescued by exogenous adenosine 5'-diphosphate, indicating that AADACL1 likely functions to induce dense granule secretion. Experiments with P2Y12-/- and CalDAG GEFI-/- mice revealed that the P2Y12 pathway is the predominate target of HAG-mediated inhibition of platelet aggregation. HAG itself displayed weak agonist properties and likely mediates its inhibitory effects via conversion to a phosphorylated metabolite, HAGP, which directly interacted with the C1a domains of 2 distinct PKC isoforms and blocked PKC kinase activity in vitro. Finally, AADACL1 inhibition in rats reduced platelet aggregation, protected against FeCl3-induced arterial thrombosis, and delayed tail bleeding time. In summary, our data support a model whereby AADACL1 inhibition shifts the platelet ether lipidome to an inhibitory axis of HAGP accumulation that impairs PKC activation, granule secretion, and recruitment of platelets to sites of vascular damage.


Assuntos
Plaquetas/metabolismo , Metabolismo dos Lipídeos , Esterol Esterase/metabolismo , Trombose/etiologia , Trombose/metabolismo , Animais , Plaquetas/efeitos dos fármacos , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Camundongos , Modelos Biológicos , Fosforilação , Ativação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Testes de Função Plaquetária , Ligação Proteica , Proteína Quinase C/metabolismo , Ratos , Receptores Purinérgicos P2Y12/metabolismo , Transdução de Sinais/efeitos dos fármacos , Esterol Esterase/antagonistas & inibidores , Especificidade por Substrato , Trombose/tratamento farmacológico
13.
Blood Adv ; 2(12): 1417-1428, 2018 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-29925524

RESUMO

von Willebrand disease (VWD) type 2B is characterized by gain-of-function mutations in von Willebrand factor (VWF), enhancing its binding affinity for the platelet receptor glycoprotein (GP)Ibα. VWD type 2B patients display a bleeding tendency associated with loss of high-molecular-weight VWF multimers and variable thrombocytopenia. We recently demonstrated that a marked defect in agonist-induced activation of the small GTPase, Rap1, and integrin αIIbß3 in VWD (p.V1316M) type 2B platelets also contributes to the bleeding tendency. Here, we investigated the molecular mechanisms underlying impaired platelet Rap1 signaling in this disease. Two distinct pathways contribute to Rap1 activation in platelets: rapid activation mediated by the calcium-sensing guanine nucleotide exchange factor CalDAG-GEF-I (CDGI) and sustained activation that is dependent on signaling by protein kinase C (PKC) and the adenosine 5'-diphosphate receptor P2Y12. To investigate which Rap1 signaling pathway is affected, we expressed VWF/p.V1316M by hydrodynamic gene transfer in wild-type and Caldaggef1-/- mice. Using αIIbß3 integrin activation as a read-out, we demonstrate that platelet dysfunction in VWD (p.V1316M) type 2B affects PKC-mediated, but not CDGI-mediated, activation of Rap1. Consistently, we observed decreased PKC substrate phosphorylation and impaired granule release in stimulated VWD type 2B platelets. Interestingly, the defect in PKC signaling was caused by a significant increase in baseline PKC substrate phosphorylation in circulating VWD (p.V1316M) type 2B platelets, suggesting that the VWF-GPIbα interaction leads to preactivation and exhaustion of the PKC pathway. Consistent with PKC preactivation, VWD (p.V1316M) type 2B mice also exhibited marked shedding of platelet GPIbα. In summary, our studies identify altered PKC signaling as the underlying cause of platelet hypofunction in p.V1316M-associated VWD type 2B.


Assuntos
Plaquetas/patologia , Proteína Quinase C/metabolismo , Doença de von Willebrand Tipo 2/sangue , Animais , Humanos , Camundongos , Mutação de Sentido Incorreto , Complexo Glicoproteico GPIb-IX de Plaquetas/metabolismo , Transdução de Sinais , Proteínas rap1 de Ligação ao GTP/metabolismo , Doença de von Willebrand Tipo 2/genética , Fator de von Willebrand/genética , Fator de von Willebrand/metabolismo
14.
Blood Adv ; 1(18): 1398-1408, 2017 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-29104956

RESUMO

Actin reorganization regulates key processes in platelet activation. Here we examined the role of the Arp2/3 complex, an essential component in actin filament branching, in platelet function. The Arpc2 gene, encoding the p34 subunit of the Arp2/3 complex, was deleted in the megakaryocyte lineage (Arpc2fl/flPF4-Cre). Deletion of the Arp2/3 complex resulted in marked microthrombocytopenia in mice, caused by premature platelet release into the bone marrow compartment and impaired platelet survival in circulation. Arpc2fl/flPF4-Cre platelets exhibited alterations in their actin cytoskeleton and their peripheral microtubule coil. Thrombocytopenia was alleviated following clodronate liposome-induced macrophage depletion in Arpc2fl/flPF4-Cre mice. Arpc2fl/flPF4-Cre platelets failed to spread and showed a mild defect in integrin activation and aggregation. However, no significant differences in hemostasis or thrombosis were observed between Arpc2fl/flPF4-Cre and control mice. Thus, Arp2/3 is critical for platelet homeostasis but plays only a minor role for vascular hemostasis.

15.
J Clin Invest ; 125(4): 1419-32, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25705885

RESUMO

The small GTPase RAP1 is critical for platelet activation and thrombus formation. RAP1 activity in platelets is controlled by the GEF CalDAG-GEFI and an unknown regulator that operates downstream of the adenosine diphosphate (ADP) receptor, P2Y12, a target of antithrombotic therapy. Here, we provide evidence that the GAP, RASA3, inhibits platelet activation and provides a link between P2Y12 and activation of the RAP1 signaling pathway. In mice, reduced expression of RASA3 led to premature platelet activation and markedly reduced the life span of circulating platelets. The increased platelet turnover and the resulting thrombocytopenia were reversed by concomitant deletion of the gene encoding CalDAG-GEFI. Rasa3 mutant platelets were hyperresponsive to agonist stimulation, both in vitro and in vivo. Moreover, activation of Rasa3 mutant platelets occurred independently of ADP feedback signaling and was insensitive to inhibitors of P2Y12 or PI3 kinase. Together, our results indicate that RASA3 ensures that circulating platelets remain quiescent by restraining CalDAG-GEFI/RAP1 signaling and suggest that P2Y12 signaling is required to inhibit RASA3 and enable sustained RAP1-dependent platelet activation and thrombus formation at sites of vascular injury. These findings provide insight into the antithrombotic effect of P2Y12 inhibitors and may lead to improved diagnosis and treatment of platelet-related disorders.


Assuntos
Proteínas Ativadoras de GTPase/fisiologia , Ativação Plaquetária/fisiologia , Proteínas rap1 de Ligação ao GTP/antagonistas & inibidores , Animais , Senescência Celular , Clopidogrel , Proteínas Ativadoras de GTPase/genética , Fatores de Troca do Nucleotídeo Guanina/deficiência , Hemostasia , Linfopenia/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ativação Plaquetária/efeitos dos fármacos , Ativação Plaquetária/genética , Inibidores da Agregação Plaquetária/farmacologia , Proteínas Proto-Oncogênicas c-akt/fisiologia , Receptores Purinérgicos P2Y12/fisiologia , Veia Safena/lesões , Esplenectomia , Trombocitopenia/genética , Trombocitopenia/cirurgia , Trombopoese , Ticlopidina/análogos & derivados , Ticlopidina/farmacologia , Proteínas rap1 de Ligação ao GTP/fisiologia
16.
Chem Biol ; 20(9): 1125-34, 2013 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-23993462

RESUMO

A comprehensive knowledge of the platelet proteome is necessary for understanding thrombosis and for envisioning antiplatelet therapies. To discover other biochemical pathways in human platelets, we screened platelets with a carbamate library designed to interrogate the serine hydrolase subproteome and used competitive activity-based protein profiling to map the targets of active carbamates. We identified an inhibitor that targets arylacetamide deacetylase-like 1 (AADACL1), a lipid deacetylase originally identified in invasive cancers. Using this compound, along with highly selective second-generation inhibitors of AADACL1, metabolomics, and RNA interference, we show that AADACL1 regulates platelet aggregation, thrombus growth, RAP1 and PKC activation, lipid metabolism, and fibrinogen binding to platelets and megakaryocytes. These data provide evidence that AADACL1 regulates platelet and megakaryocyte activation and highlight the value of this chemoproteomic strategy for target discovery in platelets.


Assuntos
Plaquetas/metabolismo , Hidrolases de Éster Carboxílico/metabolismo , Carbamatos/química , Carbamatos/metabolismo , Carbamatos/farmacologia , Hidrolases de Éster Carboxílico/antagonistas & inibidores , Hidrolases de Éster Carboxílico/genética , Linhagem Celular , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Fibrinogênio/metabolismo , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Megacariócitos/citologia , Megacariócitos/efeitos dos fármacos , Megacariócitos/metabolismo , Metabolômica , Ativação Plaquetária/efeitos dos fármacos , Agregação Plaquetária/efeitos dos fármacos , Complexo Glicoproteico GPIIb-IIIa de Plaquetas/metabolismo , Ligação Proteica , Proteína Quinase C/metabolismo , Proteômica , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Esterol Esterase , Proteínas rap1 de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa