Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Biol Chem ; 299(7): 104865, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37268160

RESUMEN

Spleen tyrosine kinase (Syk) is expressed in a variety of hemopoietic cells. Upon phosphorylation of the platelet immunoreceptor-based activation motif of the glycoprotein VI (GPVI)/Fc receptor gamma chain collagen receptor, both the tyrosine phosphorylation and activity of Syk are increased leading to downstream signaling events. Although it has been established that the activity of Syk is regulated by tyrosine phosphorylation, the specific roles of individual phosphorylation sites remain to be elucidated. We observed that Syk Y346 in mouse platelets was still phosphorylated when GPVI-induced Syk activity was inhibited. We then generated Syk Y346F mice and analyzed the effect this mutation exerts on platelet responses. Syk Y346F mice bred normally, and their blood cell count was unaltered. We did observe potentiation of GPVI-induced platelet aggregation and ATP secretion as well as increased phosphorylation of other tyrosines on Syk in the Syk Y346F mouse platelets when compared to WT littermates. This phenotype was specific for GPVI-dependent activation, since it was not seen when AYPGKF, a PAR4 agonist, or 2-MeSADP, a purinergic receptor agonist, was used to activate platelets. Despite a clear effect of Syk Y346F on GPVI-mediated signaling and cellular responses, there was no effect of this mutation on hemostasis as measured by tail-bleeding times, although the time to thrombus formation determined using the ferric chloride injury model was reduced. Thus, our results indicate a significant effect of Syk Y346F on platelet activation and responses in vitro and reveal its complex nature manifesting itself by the diversified translation of platelet activation into physiological responses.


Asunto(s)
Plaquetas , Agregación Plaquetaria , Quinasa Syk , Animales , Ratones , Fosforilación , Activación Plaquetaria , Glicoproteínas de Membrana Plaquetaria/genética , Glicoproteínas de Membrana Plaquetaria/metabolismo , Quinasa Syk/genética , Quinasa Syk/metabolismo , Tirosina
2.
Int J Mol Sci ; 25(1)2023 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-38203381

RESUMEN

Apoptosis signal-regulating kinase 1 (ASK1) is a serine-threonine kinase that is ubiquitously expressed in nucleated cells and is responsible for the activation of multiple mitogen-activated protein kinases (MAPK) to regulate cell stress. Activation of ASK1 via cellular stress leads to activation of downstream signaling components, activation of transcription factors, and proinflammatory cytokine production. ASK1 is also expressed in anucleate platelets and is a key player in platelet activation as it is important for signaling. Interestingly, the mechanism of ASK1 activation is cell type-dependent. In this review we will explore how ASK1 regulates a variety of cellular processes from innate immune function to thrombosis and hemostasis. We will discuss how ASK1 influences FcγRIIA-mediated platelet reactivity and how that reactivity drives platelet clearance. Furthermore, we will explore the role of ASK1 in thromboxane (TxA2) generation, which highlights differences in the way ASK1 functions in mouse and human platelets.


Asunto(s)
MAP Quinasa Quinasa Quinasa 5 , Sepsis , Humanos , Animales , Ratones , Plaquetas , Cinética , Proteínas Quinasas Activadas por Mitógenos
3.
Int J Mol Sci ; 23(19)2022 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-36232816

RESUMEN

Platelets are anucleate cells that mediate hemostasis. This occurs via a primary signal that is reinforced by secreted products such as ADP that bind purinergic receptors (P2Y1 and P2Y12) on the platelet surface. We recently identified a human subject, whom we termed platelet defect subject 25 (PDS25) with a platelet functional disorder associated with the P2Y12 receptor. PDS25 has normal blood cell counts and no history of bleeding diathesis. However, platelets from PDS25 have virtually no response to 2-MeSADP (a stable analogue of ADP). Genetic analysis of P2Y12 from PDS25 revealed a heterozygous mutation of D121N within the DRY motif. Rap1b activity was reduced in platelets from PDS25, while VASP phosphorylation was enhanced, suggesting that signaling from the P2Y12 receptor was interrupted by the heterozygous mutation. To explore this further, we produced knock-in mice that mimic our subject. Bleeding failed to cease in homozygous KI mice during tail bleeding assays, while tail bleeding times did not differ between WT and heterozygous KI mice. Furthermore, occlusions failed to form in most homozygous KI mice following carotid artery injury via FeCl3. These data indicate that the aspartic acid residue found in the DRY motif of P2Y12 is essential for P2Y12 function.


Asunto(s)
Plaquetas/metabolismo , Receptores Purinérgicos P2Y12/metabolismo , Adenosina Difosfato/metabolismo , Animales , Ácido Aspártico/metabolismo , Hemorragia/genética , Hemorragia/metabolismo , Humanos , Ratones , Agregación Plaquetaria , Pruebas de Función Plaquetaria , Antagonistas del Receptor Purinérgico P2Y/farmacología , Receptores Purinérgicos P2Y12/química , Receptores Purinérgicos P2Y12/genética
4.
J Biol Chem ; 298(8): 102189, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35753354

RESUMEN

Immune cells express receptors bearing an immune tyrosine activation motif (ITAM) containing two YXXL motifs or hemITAMs containing only one YXXL motif. Phosphorylation of the ITAM/hemITAM is mediated by Src family kinases allowing for the binding and activation of spleen tyrosine kinase (Syk). It is believed that Syk must be phosphorylated on tyrosine residues for activation, and Tyr342, а conserved tyrosine in the interdomain B region, has been shown to be critical for regulating Syk in FcεR1-activated mast cells. Syk is a key mediator of signaling pathways downstream of several platelet pathways including the ITAM bearing glycoprotein VI (GPVI)/Fc receptor gamma chain collagen receptor and the hemITAM containing C-type lectin-like receptor-2 (CLEC-2). Since platelet activation is a crucial step in both hemostasis and thrombosis, we evaluated the importance of Syk Y342 in these processes by producing an Syk Y342F knock-in mouse. When using a CLEC-2 antibody as an agonist, reduced aggregation and secretion were observed in Syk Y342F mouse platelets when compared with control mouse platelets. Platelet reactivity was also reduced in response to the GPVI agonist collagen-related peptide. Signaling initiated by either GPVI or CLEC-2 was also greatly inhibited, including Syk Y519/520 phosphorylation. Hemostasis, as measured by tail bleeding time, was not altered in Syk Y342F mice, but thrombus formation in response to FeCl3 injury was prolonged in Syk Y342F mice. These data demonstrate that phosphorylation of Y342 on Syk following stimulation of either GPVI or CLEC-2 receptors is important for the ability of Syk to transduce a signal.


Asunto(s)
Glicoproteínas de Membrana Plaquetaria , Quinasa Syk/metabolismo , Tirosina , Animales , Plaquetas/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Ratones , Fosforilación , Glicoproteínas de Membrana Plaquetaria/genética , Glicoproteínas de Membrana Plaquetaria/metabolismo , Receptores Inmunológicos/metabolismo , Quinasa Syk/genética , Tirosina/metabolismo
5.
Res Pract Thromb Haemost ; 6(3): e12710, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-35573643

RESUMEN

Background: C-type lectin receptor family members play a role in many cells including platelets, where they are crucial in the separation of lymphatic and blood vessels during development. The C-type lectin-like receptor 2 (CLEC-2) receptor contains the canonical intracellular hemITAM motif through which it signals to activate Syk. Objectives: One proposed hypothesis for signaling cascade is that Syk bridges two receptors through phosphorylated hemITAM motifs. We demonstrated that the phosphorylated hemITAM stimulates PI3 kinase/Btk pathways to activate Syk. To address this controversy, we used a CLEC-2 selective agonist and studied the role of Btk in platelet activation. Results and Conclusions: Platelet activation and downstream signaling were abolished in murine and human platelets in the presence of the Btk inhibitors ibrutinib or acalabrutinib when a low concentration of a CLEC-2 antibody was used to crosslink CLEC-2 receptors. This inhibition was overcome by increasing concentrations of the CLEC-2 antibody. Similar results were obtained in X-linked immunodeficient mouse platelets, with an inactivating mutation in Btk or in Lyn null platelets. We conclude that at low crosslinking conditions of CLEC-2, Btk plays an important role in the activation of Syk, but at higher crosslinking conditions their role becomes less important and other mechanisms take over to activate Syk.

6.
J Biol Chem ; 296: 100720, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33932405

RESUMEN

Platelets are key mediators of physiological hemostasis and pathological thrombosis, whose function must be carefully balanced by signaling downstream of receptors such as protease-activated receptor (PAR)4. Protein kinase C (PKC) is known to regulate various aspects of platelet function. For instance, PKCδ is known to regulate dense granule secretion, which is important for platelet activation. However, the mechanism by which PKCδ regulates this process as well as other facets of platelet activity is unknown. We speculated that the way PKCδ regulates platelet function may be because of the phosphorylation of tyrosine residues on PKCδ. We investigated phosphorylation of PKCδ following glycoprotein VI-mediated and PAR4-mediated platelet activation and found that Y311 is selectively phosphorylated when PAR4 is activated in human platelets. Therefore, we generated PKCδ Y311F knock-in mice, which are viable and have no gross abnormalities. However, PKCδY311F mice have significantly enhanced tail-bleeding times compared with WT littermate controls, which means hemostasis is interrupted. Furthermore, PKCδY311F mice exhibit longer time to carotid artery occlusion compared with WT control using a ferric chloride in vivo thrombosis model, indicating that the phosphorylation of PKCδ Y311 is prothrombotic. Washed platelets from PKCδY311F mice have reduced reactivity after stimulation with a PAR-4 agonist indicating its importance in platelet signaling. The phenotype observed in Y311F mouse platelets is because of reduced thromboxane generation, as an inhibitor of thromboxane generation equalizes the PKCδY311F platelet response to that of WT. Therefore, phosphorylation of PKCδ on Y311 is important for regulation of platelet function and specifically thromboxane generation, which reinforces platelet activation.


Asunto(s)
Plaquetas/metabolismo , Proteína Quinasa C-delta/química , Proteína Quinasa C-delta/metabolismo , Tromboxanos/biosíntesis , Tirosina/metabolismo , Animales , Humanos , Ratones , Modelos Moleculares , Fosforilación , Conformación Proteica
7.
Circulation ; 142(8): 758-775, 2020 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-32489148

RESUMEN

BACKGROUND: Cardiac rupture is a major lethal complication of acute myocardial infarction (MI). Despite significant advances in reperfusion strategies, mortality from cardiac rupture remains high. Studies suggest that cardiac rupture can be accelerated by thrombolytic therapy, but the relevance of this risk factor remains controversial. METHODS: We analyzed protease-activated receptor 4 (Par4) expression in mouse hearts with MI and investigated the effects of Par4 deletion on cardiac remodeling and function after MI by echocardiography, quantitative immunohistochemistry, and flow cytometry. RESULTS: Par4 mRNA and protein levels were increased in mouse hearts after MI and in isolated cardiomyocytes in response to hypertrophic and inflammatory stimuli. Par4-deficient mice showed less myocyte apoptosis, reduced infarct size, and improved functional recovery after acute MI relative to wild-type (WT). Conversely, Par4-/- mice showed impaired cardiac function, greater rates of myocardial rupture, and increased mortality after chronic MI relative to WT. Pathological evaluation of hearts from Par4-/- mice demonstrated a greater infarct expansion, increased cardiac hemorrhage, and delayed neutrophil accumulation, which resulted in impaired post-MI healing compared with WT. Par4 deficiency also attenuated neutrophil apoptosis in vitro and after MI in vivo and impaired inflammation resolution in infarcted myocardium. Transfer of Par4-/- neutrophils, but not of Par4-/- platelets, in WT recipient mice delayed inflammation resolution, increased cardiac hemorrhage, and enhanced cardiac dysfunction. In parallel, adoptive transfer of WT neutrophils into Par4-/- mice restored inflammation resolution, reduced cardiac rupture incidence, and improved cardiac function after MI. CONCLUSIONS: These findings reveal essential roles of Par4 in neutrophil apoptosis and inflammation resolution during myocardial healing and point to Par4 inhibition as a potential therapy that should be limited to the acute phases of ischemic insult and avoided for long-term treatment after MI.


Asunto(s)
Regulación de la Expresión Génica , Rotura Cardíaca , Infarto del Miocardio , Miocardio/metabolismo , Receptores de Trombina/deficiencia , Animales , Femenino , Rotura Cardíaca/etiología , Rotura Cardíaca/genética , Rotura Cardíaca/metabolismo , Rotura Cardíaca/prevención & control , Inflamación/genética , Inflamación/metabolismo , Inflamación/prevención & control , Masculino , Ratones , Ratones Noqueados , Infarto del Miocardio/clasificación , Infarto del Miocardio/genética , Infarto del Miocardio/metabolismo , Infarto del Miocardio/prevención & control , Receptores de Trombina/biosíntesis
8.
J Biol Chem ; 294(33): 12547-12554, 2019 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-31266805

RESUMEN

Protein tyrosine phosphatase nonreceptor type 7 (PTPN7), also called hematopoietic protein tyrosine phosphatase, controls extracellular signal-regulated protein kinase 1/2 (ERK1/2) and p38 mitogen-activated protein kinase in T lymphocytes. Because ERK1/2 plays an important role in regulating thromboxane A2 (TXA2) generation in platelets, we investigated the function of PTPN7 in these cells. Using immunoblot analysis, we detected PTPN7 in both human and mouse platelets but not in PTPN7-null mice. PTPN7 KO mouse platelets exhibited increased platelet functional responses, including aggregation, dense granule secretion, and TXA2 generation, compared with platelets from WT littermates, upon stimulation with both G protein-coupled receptor (GPCR) and glycoprotein VI (GPVI) agonists. Using the GPCR agonist AYPGKF in the presence of the COX inhibitor indomethacin, we found that PTPN7 KO mouse platelets aggregated and secreted to the same extent as WT platelets, suggesting that elevated TXA2 is responsible for the potentiation of platelet functional responses in PTPN7-KO platelets. Phosphorylation of ERK1/2 was also elevated in PTPN7 KO platelets. Stimulation of platelets with the GPVI agonist collagen-related peptide along with the COX inhibitor indomethacin did not result in phosphorylation of ERK1/2, indicating that GPVI-mediated ERK phosphorylation occurs through TXA2 Although bleeding times did not significantly differ between PTPN7-null and WT mice, time to death was significantly faster in PTPN7-null mice than in WT mice in a pulmonary thromboembolism model. We conclude that PTPN7 regulates platelet functional responses downstream of GPCR agonists, but not GPVI agonists, through inhibition of ERK activation and thromboxane generation.


Asunto(s)
Plaquetas/enzimología , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Tirosina Fosfatasas no Receptoras/metabolismo , Embolia Pulmonar/enzimología , Animales , Plaquetas/patología , Modelos Animales de Enfermedad , Activación Enzimática , Humanos , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/genética , Oligopéptidos/farmacología , Proteínas Tirosina Fosfatasas no Receptoras/genética , Embolia Pulmonar/genética , Embolia Pulmonar/patología
9.
Thromb Haemost ; 119(8): 1321-1331, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31226719

RESUMEN

BACKGROUND AND OBJECTIVE: CD45 is a receptor protein tyrosine phosphatase present on the surface of all hematopoietic cells except for erythrocytes and platelets. Proteomics studies, however, have demonstrated the presence of a CD45 c-terminal catalytic peptide in platelets. Therefore, we investigated the functional role of this truncated isoform of CD45 in platelets, which contains the c-terminal catalytic domain but lacks the extracellular region. METHODS AND RESULTS: We used an antibody specific to the c-terminus of CD45 to confirm the presence of a truncated CD45 isoform in platelets. We also examined ex vivo and in vivo platelet function using CD45 knockout (KO) mice. Aggregation and secretion mediated by the glycoprotein VI (GPVI) receptor was impaired in CD45 KO platelets. Consequently, CD45 KO mice had impaired hemostasis indicated by increased tail bleeding times. Also, using a model of pulmonary embolism we showed that CD45 KO mice had defective in vivo thrombus formation. Next, we investigated whether or not the truncated isoform of CD45 had a role in GPVI signaling. The full-length isoform of CD45 is known to regulate Src family kinase (SFK) activation in lymphocytes. We find a similar role for the truncated isoform of CD45 in platelets. SFK activation was impaired downstream of the GPVI receptor in the CD45 KO murine platelets. Consequently, Syk, PLCγ2, and pleckstrin phosphorylations were also impaired in CD45 KO murine platelets. CONCLUSION: We conclude that the truncated CD45 isoform regulates GPVI-mediated signaling and platelet functional responses by regulating SFK activation.


Asunto(s)
Plaquetas/metabolismo , Antígenos Comunes de Leucocito/metabolismo , Glicoproteínas de Membrana Plaquetaria/metabolismo , Familia-src Quinasas/metabolismo , Animales , Proteínas Sanguíneas/química , Dominio Catalítico , Membrana Celular/metabolismo , Hemostasis , Humanos , Ratones , Ratones Noqueados , Péptidos/química , Fosfoproteínas/química , Fosforilación , Activación Plaquetaria , Unión Proteica , Isoformas de Proteínas , Transducción de Señal , Trombosis/metabolismo
10.
TH Open ; 2(4): e411-e419, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31249969

RESUMEN

Platelet activation is essential for hemostasis. Central to platelet activation are the signals transmitted through surface receptors such as glycoprotein VI, the protease-activated receptors, and C-type lectin-like receptor 2 (CLEC-2). CLEC-2 is a HemITAM (hem-immunoreceptor tyrosine activation motif)-bearing receptor that binds podoplanin and signals through spleen tyrosine kinase (Syk). T-cell ubiquitin ligand-2 (TULA-2) is a protein tyrosine phosphatase that is highly expressed in platelets and targets phosphorylated Y352 of Syk. We wanted to determine whether TULA-2 regulates Syk phosphorylation and activity downstream of CLEC-2. To that end, we used TULA-2 knockout mice and wild-type (WT) littermate controls. We found that TULA-2 deficiency enhances the aggregation and secretion response following stimulation with an excitatory CLEC-2 antibody or the CLEC-2 agonist rhodocytin. Consistently, Syk phosphorylation of Y346 is enhanced, as well as phosphorylation of the downstream signaling molecule PLCγ2, in TULA-2 knockout platelets treated with either CLEC-2 antibody or rhodocytin, compared with WT control platelets. Furthermore, the kinetics of Syk phosphorylation, as well as that of PLCγ2 and SLP-76, is enhanced in TULA-2 knockout platelets treated with 2.5-µg/mL CLEC-2 antibody compared with WT platelets. Similarly, thromboxane production was enhanced, in both amount and kinetics, in TULA-2 -/- platelets treated with 2.5-µg/mL CLEC-2 antibody. TULA-2 acts as a negative regulator of CLEC-2 signaling by dephosphorylating Syk on Y346 and restraining subsequent Syk-mediated signaling.

11.
PLoS One ; 12(8): e0182867, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28783756

RESUMEN

BACKGROUND: It has long been postulated that Protein Kinase C (PKC) is an important regulator of megakaryopoiesis. Recent contributions to the literature have outlined the functions of several individual PKC isoforms with regard to megakaryocyte differentiation and platelet production. However, the exact role of PKCε remains elusive. OBJECTIVE: To delineate the role of PKCε in megakaryopoiesis. APPROACH AND RESULTS: We used a PKCε knockout mouse model to examine the effect of PKCε deficiency on platelet mass, megakaryocyte mass, and bone marrow progenitor cell distribution. We also investigated platelet recovery in PKCε null mice and TPO-mediated signaling in PKCε null megakaryocytes. PKCε null mice have higher platelet counts due to increased platelet production compared to WT littermate controls (p<0.05, n = 8). Furthermore, PKCε null mice have more bone marrow megakaryocyte progenitor cells than WT littermate control mice. Additionally, thrombopoietin-mediated signaling is perturbed in PKCε null mice as Akt and ERK1/2 phosphorylation are enhanced in PKCε null megakaryocytes stimulated with thrombopoietin. Finally, in response to immune-induced thrombocytopenia, PKCε null mice recovered faster and had higher rebound thrombocytosis than WT littermate control mice. CONCLUSIONS: Enhanced platelet recovery could be due to an increase in megakaryocyte progenitor cells found in PKCε null mice as well as enhanced thrombopoietin-mediated signaling observed in PKCε deficient megakaryocytes. These data suggest that PKCε is a negative regulator of megakaryopoiesis.


Asunto(s)
Técnicas de Inactivación de Genes , Proteína Quinasa C-epsilon/deficiencia , Proteína Quinasa C-epsilon/genética , Células Madre/citología , Células Madre/metabolismo , Trombopoyesis , Animales , Células de la Médula Ósea/citología , Diferenciación Celular/efectos de los fármacos , Megacariocitos/citología , Megacariocitos/efectos de los fármacos , Ratones , Recuento de Plaquetas , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Células Madre/efectos de los fármacos , Trombocitopenia/enzimología , Trombocitopenia/inmunología , Trombopoyesis/efectos de los fármacos , Trombopoyesis/genética , Trombopoyetina/farmacología
12.
Int J Mol Sci ; 18(6)2017 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-28598382

RESUMEN

The binding of von Willebrand factor (VWF) to the platelet membrane glycoprotein 1b-IX (GP1b-IX) leads to activation of platelets. GP1b was shown to signal via the FcRγ-ITAM (Fc Receptor γ-Immunoreceptor tyrosine-based activation motif) pathway, activating spleen tyrosine kinase (Syk) and other tyrosine kinases. However, there have been conflicting reports regarding the role of Syk in GP1b signaling. In this study, we sought to resolve these conflicting reports and clarify the role of Syk in VWF-induced platelet activation. The inhibition of Syk with the selective Syk inhibitors, OXSI-2 and PRT-060318, did not inhibit VWF-induced platelet adhesion, agglutination, aggregation, or secretion. In contrast, platelets stimulated with the Glycoprotein VI (GPVI) agonist, collagen-related peptide (CRP), failed to cause any aggregation or secretion in presence of the Syk inhibitors. Furthermore, GP1b-induced platelet signaling was unaffected in the presence of Syk inhibitors, but GPVI-induced signaling was abolished under similar conditions. Thus, we conclude that Syk kinase activity does not play any functional role downstream of GP1b-mediated platelet activation.


Asunto(s)
Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Transducción de Señal , Quinasa Syk/metabolismo , Animales , Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Humanos , Fosforilación , Adhesividad Plaquetaria/genética , Agregación Plaquetaria/genética , Complejo GPIb-IX de Glicoproteína Plaquetaria/genética , Transducción de Señal/efectos de los fármacos , Quinasa Syk/antagonistas & inhibidores , Quinasa Syk/genética , Factor de von Willebrand/metabolismo
13.
Blood ; 125(19): 2995-3005, 2015 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-25824689

RESUMEN

Megakaryocyte maturation and polyploidization are critical for platelet production; abnormalities in these processes are associated with myeloproliferative disorders, including thrombocytopenia. Megakaryocyte maturation signals through cascades that involve p21-activated kinase (Pak) function; however, the specific role for Pak kinases in megakaryocyte biology remains elusive. Here, we identify Pak2 as an essential effector of megakaryocyte maturation, polyploidization, and proplatelet formation. Genetic deletion of Pak2 in murine bone marrow is associated with macrothrombocytopenia, altered megakaryocyte ultrastructure, increased bone marrow megakaryocyte precursors, and an elevation of mature CD41(+) megakaryocytes, as well as an increased number of polyploid cells. In Pak2(-/-) mice, platelet clearance rate was increased, as was production of newly synthesized, reticulated platelets. In vitro, Pak2(-/-) megakaryocytes demonstrate increased polyploidization associated with alterations in ß1-tubulin expression and organization, decreased proplatelet extensions, and reduced phosphorylation of the endomitosis regulators LIM domain kinase 1, cofilin, and Aurora A/B/C. Together, these data establish a novel role for Pak2 as an important regulator of megakaryopoiesis, polyploidization, and cytoskeletal dynamics in developing megakaryocytes.


Asunto(s)
Plaquetas/metabolismo , Citoesqueleto/patología , Megacariocitos/metabolismo , Mitosis/genética , Factor de Transcripción PAX2/fisiología , Trombocitopenia/genética , Trombopoyesis/fisiología , Animales , Plaquetas/patología , Médula Ósea/metabolismo , Médula Ósea/patología , Citoesqueleto/metabolismo , Megacariocitos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Microscopía Fluorescente , Poliploidía , Células Madre/metabolismo , Células Madre/patología , Trombocitopenia/patología
14.
Platelets ; 26(1): 2-9, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-24433221

RESUMEN

Platelets upon activation change their shape, aggregate and secrete alpha and dense granule contents among which ADP acts as a feedback activator. Different Protein Kinase C (PKC) isoforms have specific non-redundant roles in mediating platelet responses including secretion and thrombus formation. Murine platelets lacking specific PKC isoforms have been used to evaluate the isoform specific functions. Novel PKC isoform δ has been shown to play an important role in some pathological processes. Lack of specific inhibitors for PKCδ has restricted analysis of its role in various cells. The current study was carried out to evaluate a novel small molecule PKCδ inhibitor, CGX1037 in platelets. Platelet aggregation, dense granule secretion and western blotting experiments were performed to evaluate CGX1037. In human platelets, CGX1037 inhibited PAR4-mediated phosphorylation on PKD2, a PKCδ-specific substrate. Pre-treatment of human or murine platelets with CGX1037 inhibited PAR4-mediated dense granule secretion whereas it potentiated GPVI-mediated dense granule secretion similar to the responses observed in murine platelets lacking PKCδ· Furthermore, pre-treatment of platelets from PKCδ(-/-) mice with CGX1037 had no significant additive effect on platelet responses suggesting the specificity of CGX1037. Hence, we show that CGX1037 is a selective small molecule inhibitor of PKCδ in platelets.


Asunto(s)
Plaquetas/efectos de los fármacos , Plaquetas/metabolismo , Proteína Quinasa C-delta/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Animales , Humanos , Concentración 50 Inhibidora , Ratones , Ratones Noqueados , Fosforilación/efectos de los fármacos , Agregación Plaquetaria/efectos de los fármacos , Glicoproteínas de Membrana Plaquetaria/metabolismo , Proteína Quinasa C-delta/genética , Proteína Quinasa C-delta/metabolismo , Proteína Quinasa D2 , Proteínas Quinasas/metabolismo , Transporte de Proteínas , Vesículas Secretoras/metabolismo
16.
Arterioscler Thromb Vasc Biol ; 34(12): 2579-85, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25359855

RESUMEN

OBJECTIVE: We previously determined that protein kinase C δ (PKCδ) regulates platelet function. However, the function of PKCδ in megakaryopoiesis is unknown. APPROACH AND RESULTS: Using PKCδ(-/-) and wild-type littermate mice, we found that deficiency of PKCδ caused an increase in white blood cells and platelet counts, as well as in bone marrow and splenic megakaryocytes (P<0.05). Additionally, the megakaryocyte number and DNA content were enhanced in PKCδ(-/-) mouse bone marrow after culturing with exogenous thrombopoietin compared with wild-type (P<0.05). Importantly, thrombopoietin-induced signaling was also altered with PKCδ deletion because both extracellular signal-regulated kinase and Akt308 phosphorylation were heightened in PKCδ(-/-) megakaryocytes compared with wild-type. Finally, PKCδ(-/-) mice recovered faster and had a heightened rebound thrombocytosis after thrombocytopenic challenge. CONCLUSIONS: These data suggest that PKCδ is an important megakaryopoietic protein, which regulates signaling induced by thrombopoietin and represents a potential therapeutic target.


Asunto(s)
Megacariocitos/citología , Megacariocitos/enzimología , Proteína Quinasa C-delta/deficiencia , Trombocitopenia/sangre , Trombocitopenia/enzimología , Trombopoyesis/fisiología , Animales , Células de la Médula Ósea/citología , Quinasas MAP Reguladas por Señal Extracelular/sangre , Recuento de Leucocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Recuento de Plaquetas , Proteína Quinasa C-delta/sangre , Proteína Quinasa C-delta/genética , Proteínas Proto-Oncogénicas c-akt/sangre , ARN Mensajero/sangre , ARN Mensajero/genética , Transducción de Señal , Bazo/citología , Trombocitopenia/inmunología , Trombopoyesis/genética , Trombopoyetina/sangre , Regulación hacia Arriba
17.
Blood ; 119(3): 838-46, 2012 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-22128142

RESUMEN

Megakaryocytes are large, polyploid cells that produce platelets. We have previously reported that calcium- and integrin-binding protein 1 (CIB1) regulates endomitosis in Dami cells. To further characterize the role of CIB1 in megakaryopoiesis, we used a Cib1(-/-) mouse model. Cib1(-/-) mice have more platelets and BM megakaryocytes than wild-type (WT) controls (P < .05). Furthermore, subsequent analysis of megakaryocyte-CFU production revealed an increase with Cib1 deletion compared with WT (P < .05). In addition, BM from Cib1(-/-) mice, cultured with thrombopoietin (TPO) for 24 hours, produced more highly polyploid megakaryocytes than WT BM (P < .05). Subsequent analysis of TPO signaling revealed enhanced Akt and ERK1/2 phosphorylation, whereas FAK(Y925) phosphorylation was reduced in Cib1(-/-) megakaryocytes treated with TPO. Conversely, platelet recovery in Cib1(-/-) mice after platelet depletion was attenuated compared with WT (P < .05). This could be the result of impaired adhesion and migration, as adhesion to fibrinogen and fibronectin and migration toward an SDF-1α gradient were reduced in Cib1(-/-) megakaryocytes compared with WT (P < .05). In addition, Cib1(-/-) megakaryocytes formed fewer proplatelets compared with WT (P < .05), when plated on fibrinogen. These data suggest that CIB1 plays a dual role in megakaryopoiesis, initially by negatively regulating TPO signaling and later by augmenting proplatelet production.


Asunto(s)
Plaquetas/metabolismo , Proteínas de Unión al Calcio/fisiología , Calcio/farmacología , Adhesión Celular/fisiología , Movimiento Celular/fisiología , Megacariocitos/fisiología , Ploidias , Animales , Plaquetas/citología , Western Blotting , Quimiocina CXCL12/metabolismo , Megacariocitos/citología , Ratones , Ratones Noqueados , Fosforilación , Unión Proteica , Transducción de Señal , Trombopoyesis , Trombopoyetina/metabolismo
18.
PLoS One ; 6(1): e14513, 2011 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-21264284

RESUMEN

Endomitosis is a form of mitosis in which both karyokinesis and cytokinesis are interrupted and is a hallmark of megakaryocyte differentiation. Very little is known about how such a dramatic alteration of the cell cycle in a physiological setting is achieved. Thrombopoietin-induced signaling is essential for induction of endomitosis. Here we show that calcium- and integrin-binding protein 1 (CIB1), a known regulator of platelet integrin α(IIb)ß(3) outside-in signaling, regulates endomitosis. We observed that CIB1 expression is increased in primary mouse megakaryocytes compared to mononuclear bone marrow cells as determined by Western blot analysis. Following PMA treatment of Dami cells, a megakaryoblastic cell line, we found that CIB1 protein expression increased concomitant with cell ploidy. Overexpression of CIB1 in Dami cells resulted in multilobated nuclei and led to increased time for a cell to complete cytokinesis as well as increased incidence of furrow regression as observed by time-lapse microscopy. Additionally, we found that surface expression of integrin α(IIb)ß(3,) an important megakaryocyte marker, was enhanced in CIB1 overexpressing cells as determined by flow cytometry. Furthermore, PMA treatment of CIB1 overexpressing cells led to increased ploidy compared to PMA treated control cells. Interestingly, expression of Polo-like kinase 3 (Plk3), an established CIB1-interacting protein and a key regulator of the mitotic process, decreased upon PMA treatment of Dami cells. Furthermore, PMA treatment augmented the interaction between CIB1 and Plk3, which depended on the duration of treatment. These data suggest that CIB1 is involved in regulating endomitosis, perhaps through its interaction with Plk3.


Asunto(s)
Proteínas de Unión al Calcio/fisiología , Endometritis/metabolismo , Megacariocitos/citología , Mitosis , Proteínas Serina-Treonina Quinasas/fisiología , Animales , Línea Celular , Femenino , Expresión Génica , Ratones , Ploidias , Unión Proteica , Acetato de Tetradecanoilforbol/farmacología
19.
Nutr J ; 6: 19, 2007 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-17705825

RESUMEN

BACKGROUND: Prepubescent children may oxidize fatty acids more readily than adults. Therefore, dietary fat needs would be higher for children compared with adults. The dietary fat recommendations are higher for children 4 to 18 yrs (i.e., 25 to 35% of energy) compared with adults (i.e., 20 to 35% of energy). Despite this, many parents and children restrict dietary fat for health reasons. METHODS: This study assessed whether rates of fat oxidation are similar between prepubescent children and adults. Ten children (8.7 +/- 1.4 yr, 33 +/- 13 kg mean +/- SD) in Tanner stage 1 and 10 adults (41.6 +/- 8 yr, 74 +/- 13 kg) were fed a weight maintenance diet for three days to maintain body weight and to establish a consistent background for metabolic rate measurements (all foods provided). Metabolic rate was measured on three separate occasions before and immediately after breakfast and for 9 hrs using a hood system (twice) or a room calorimeter (once) where continuous metabolic measurements were taken. RESULTS: During all three sessions whole body fat oxidation was higher in children (lower RQ) compared to adults (mean RQ= 0.84 +/- .016 for children and 0.87 +/- .02, for adults, p < 0.02). Although, total grams of fat oxidized was similar in children (62.7 +/- 20 g/24 hrs) compared to adults (51.4 +/- 19 g/24 hrs), the grams of fat oxidized relative to calorie expenditure was higher in children (0.047 +/- .01 g/kcal, compared to adults (0.032 +/- .01 p < 0.02). Females oxidized more fat relative to calorie expenditure than males of a similar age. A two way ANOVA showed no interaction between gender and age in terms of fat oxidation. CONCLUSION: These data suggest that fat oxidation relative to total calorie expenditure is higher in prepubescent children than in adults. Consistent with current dietary guidelines, a moderate fat diet is appropriate for children within the context of a diet that meets their energy and nutrient needs.


Asunto(s)
Envejecimiento/metabolismo , Grasas de la Dieta/metabolismo , Metabolismo Energético/fisiología , Adulto , Factores de Edad , Análisis de Varianza , Presión Sanguínea/fisiología , Calorimetría Indirecta , Niño , Dieta/métodos , Femenino , Frecuencia Cardíaca/fisiología , Humanos , Masculino , Persona de Mediana Edad , Oxidación-Reducción , Valores de Referencia , Factores Sexuales , Urea/orina
20.
Am J Physiol Heart Circ Physiol ; 293(4): H2056-63, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17675573

RESUMEN

In adult heart, selective PKCepsilon activation limits ischemia (I)-reperfusion (R) damage and mimics the protection associated with ischemic preconditioning. We sought to determine whether local delivery of PKCepsilon activator peptide psiepsilon-receptor for activated C-kinase (psiepsilon-RACK) is sufficient to produce a similarly protected phenotype in aged hearts. Langendorff-perfused hearts isolated from adult (5 mo; n = 9) and aged (24 mo; n = 9) male Fisher 344 rats were perfused with psiepsilon-RACK conjugated to Tat (500 nM) or Tat only (500 nM) for 10 min before global 31-min ischemia. Western blotting was used to measure mitochondrial targeting of PKCepsilon, PKCdelta, phospho (p)-GSK-3beta (Ser(9)) and GSK-3beta in hearts snap-frozen during I. Recovery of left ventricular developed pressure was significantly improved by psiepsilon-RACK (P < 0.01) and infarct size reduced in 24-mo rats vs. age-matched controls (60% vs. 34%; P < 0.01). Mitochondrial PKCepsilon levels were 30% greater during I with psiepsilon-RACK in aged vs. control rats (P < 0.01). Interestingly, mitochondrial GSK-3beta levels were threefold greater in aged vs. adult rats during I, and psiepsilon-RACK prevented this increase (P < 0.01). Mitochondrial p-GSK-3beta levels were also greater in aged rats after psiepsilon-RACK (P < 0.01), and subsequent inhibition of GSK-3beta with SB-216763 (3 muM) before I/R elicited protection similar to that of psiepsilon-RACK (n = 3/group). Mitochondrial proteomic analysis further identified group differences in the F(1)-ATPase beta-subunit, and coimmunoprecipitation studies revealed a novel interaction with PKCepsilon. F(1)-ATPase-PKCepsilon association was affected by psiepsilon-RACK in adult but not aged rats. Our results provide evidence, for the first time, for PKCepsilon-mediated protection in aged rat heart after I/R and suggest a central role for mitochondrial GSK-3beta but not F(1)-ATPase as a potential target of PKCepsilon to limit I/R damage with aging.


Asunto(s)
Cardiotónicos/farmacología , Glucógeno Sintasa Quinasa 3/metabolismo , Corazón/efectos de los fármacos , Precondicionamiento Isquémico , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Oligopéptidos/farmacología , Proteína Quinasa C-epsilon/metabolismo , ATPasas de Translocación de Protón/metabolismo , Envejecimiento/metabolismo , Animales , Cardiotónicos/administración & dosificación , Cardiotónicos/uso terapéutico , Activación Enzimática/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta , Corazón/fisiopatología , Técnicas In Vitro , Masculino , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/enzimología , Mitocondrias Cardíacas/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Daño por Reperfusión Miocárdica/fisiopatología , Miocardio/enzimología , Miocardio/patología , Oligopéptidos/administración & dosificación , Oligopéptidos/uso terapéutico , Fosforilación , Proteína Quinasa C-delta/metabolismo , Subunidades de Proteína , Proteómica , Ratas , Ratas Sprague-Dawley , Función Ventricular Izquierda/efectos de los fármacos , Presión Ventricular/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...