Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Eur J Neurol ; 28(1): 349-354, 2021 01.
Article in English | MEDLINE | ID: mdl-32961619

ABSTRACT

BACKGROUND AND PURPOSE: When switching between monoamine oxidase type B (MAO-B) inhibitors, a 15-day suspension period is a precautionary measure to avoid a serotonin syndrome and hypertensive crisis. However, this indication results in a major inconvenience for parkinsonian patients because of the worsening of their clinical condition. In routine clinical practice, neurologists often perform a substitution of these two drugs without solution of continuity (i.e. overnight), to avoid worsening of fluctuations and prolonged OFF periods. Therefore, a safety open label study was performed to investigate the possible risks of switching overnight from rasagiline to safinamide. METHODS: The study population included 20 advanced patients with Parkinson's disease on stable treatment with rasagiline and levodopa (alone or in combination with other anti-parkinsonian medication). The possible occurrence of serotonin syndrome and hypertension was monitored through a strict clinical observation and a 24-h Holter recording (ABPM) performed twice, whilst subjects were on rasagiline and immediately after switching to safinamide. RESULTS: No cases of serotonin syndrome or hypertensive crisis occurred during the study. Changes that were not significant occurred in the primary end-point: 24-h mean blood pressure (BP) had a mild +4.4% increase in the ABPM2 versus ABPM1 (P = 0.17), 24-h systolic and diastolic BP values were slightly higher at ABPM2 compared to ABPM1 (respectively +3.3%, P = 0.13; and 5.4%, P = 0.08) and 24-h systolic BP variability was unchanged between the two ABPM evaluations (from 8.6 ± 2.9 to 8.9 ± 1.8; P = 0.27). CONCLUSION: The results of the present study confirm that the overnight switch from rasagiline to safinamide is safe and well tolerated by patients.


Subject(s)
Parkinson Disease , Alanine/analogs & derivatives , Antiparkinson Agents/adverse effects , Benzylamines , Drug Therapy, Combination , Humans , Indans/adverse effects , Levodopa/adverse effects , Monoamine Oxidase Inhibitors/adverse effects , Parkinson Disease/drug therapy
2.
Eur J Phys Rehabil Med ; 51(5): 569-74, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26138088

ABSTRACT

BACKGROUND: The Lee Silverman Voice Treatment (LSVT®) was specifically created and tested to comply with the needs of individuals with Parkinson's disease (PD) and other neurological problems. This is a high effort intensive treatment that aims at increasing vocal intensity through the increase of subglottal air pressure, i.e. respiratory effort, for a better cordal adduction and vibration, following the motto "think loud". AIM: The main goal of this study is to inspect the efficacy of LSVT® treatment in progressive supranuclear palsy (PSP) patients. DESIGN: Longitudinal study. SETTING: Rehabilitative inpatient unit. POPULATION: Sixteen patients with PSP and 23 patients with idiopathic PD as control were enrolled in the study. METHODS: All patients underwent a training consisting in16 sessions of speech therapy following the LSVT® protocol. Initially the two groups of patients had similar voice problems, i.e. low volume and bad articulation of speech. RESULTS: A statistically significant improvement was found among the data collected before and after treatment in the PSP and Parkinson groups. Increase in maximum phonation duration and volume of voice in reading were similar in the two groups. Improvement in quality of voice and articulation were more significant in the PD group as compared to the PSP group. CONCLUSION: These results, along with previous findings, add further support to the generalized therapeutic impact of intensive voice treatment on respiratory and laryngeal functions in individuals with PSP. CLINICAL REHABILITATION IMPACT: The positive results, the absence of dropout and collateral effect following this clinical treatments with LSVT technique encouraged to use this technique in PSP patients.


Subject(s)
Dysarthria/physiopathology , Dysarthria/rehabilitation , Speech Therapy/methods , Supranuclear Palsy, Progressive/physiopathology , Adult , Aged , Aged, 80 and over , Dysarthria/etiology , Female , Humans , Longitudinal Studies , Male , Middle Aged , Parkinson Disease/complications , Parkinson Disease/physiopathology , Phonation , Speech Production Measurement , Supranuclear Palsy, Progressive/complications , Treatment Outcome
3.
J Thromb Haemost ; 11(2): 345-56, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23216754

ABSTRACT

BACKGROUND: The proline-rich tyrosine kinase Pyk2 is a focal adhesion kinase expressed in blood platelets, and is activated downstream of G-protein coupled receptors as well as integrin α2ß1. OBJECTIVE: In this study we have investigated the involvement of Pyk2 in integrin αIIbß3 outside-in signaling in human and murine platelets. METHODS: We analyzed the stimulation of intracellular signaling pathways in platelets from Pyk2 knockout mice adherent to immobilized fibrinogen. RESULTS: Pyk2 was rapidly phosphorylated and activated in human and murine platelets adherent to fibrinogen through integrin αIIbß3. Activation of Pyk2 was Src-dependent, but did not require phospholipase Cγ2 activity. Platelets from Pyk2 knockout mice showed a defective ability to adhere and spread on fibrinogen, in association with a dramatic reduction of phosphatidylinositol 3-kinase (PI3K) activation and Akt phosphorylation. Pharmacological and genetic analysis demonstrated that integrin αIIbß3 engagement selectively stimulated the ß-isoform of PI3K (PI3Kß), and that, as for Pyk2, PI3Kß activation required Src family kinases activity, but not phospholipase Cγ2. In fibrinogen-adherent platelets, both Pyk2 and PI3Kß were necessary for stimulation of the small GTPase Rap1b, a regulator of cell adhesion and spreading. Integrin αIIbß3 engagement triggered the association of the PI3Kß regulatory subunit p85 with the adaptor protein c-Cbl, which was mediated by the p85 SH3 domain, and was independent of c-Cbl tyrosine phosphorylation. However, p85-associated c-Cbl was tyrosine phosphorylated by activated Pyk2 in fibrinogen adherent platelets. CONCLUSIONS: These results identify a novel pathway of integrin αIIbß3 outside-in signaling and recognize the tyrosine kinase Pyk2 as a major regulator of platelet adhesion and spreading on fibrinogen.


Subject(s)
Blood Platelets/enzymology , Focal Adhesion Kinase 2/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Signal Transduction , Animals , Cell Shape , Enzyme Activation , Fibrinogen/metabolism , Focal Adhesion Kinase 1/metabolism , Focal Adhesion Kinase 2/deficiency , Focal Adhesion Kinase 2/genetics , Humans , Integrin alpha2/metabolism , Integrin beta3/metabolism , Mice , Mice, Knockout , Phosphatidylinositol 3-Kinase/genetics , Phosphatidylinositol 3-Kinase/metabolism , Phospholipase C gamma/genetics , Phospholipase C gamma/metabolism , Phosphorylation , Platelet Adhesiveness , Proto-Oncogene Proteins c-abl/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Time Factors , rap GTP-Binding Proteins/metabolism , src-Family Kinases/metabolism
4.
Biochim Biophys Acta ; 1813(3): 500-6, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21167219

ABSTRACT

A balance between the proteolytic processing of amyloid precursor protein APP through the amyloidogenic and the non-amyloidogenic pathways controls the production and release of amyloid ß-protein, whose accumulation in the brain is associated to the onset of Alzheimer Disease. APP is also expressed on circulating platelets. The regulation of APP processing in these cells is poorly understood. In this work we show that platelets store considerable amounts of APP fragments, including sAPPα, that can be released upon stimulation of platelets. Moreover, platelet stimulation also promotes the proteolysis of intact APP expressed on the cell surface. This process is supported by an ADAM metalloproteinase, and causes the release of sAPPα. Processing of intact platelet APP is promoted also by treatment with calmodulin antagonist W7. W7-induced APP proteolysis occurs through the non-amyloidogenic pathway, is mediated by a metalloproteinase, and causes the release of sAPPα. Co-immunoprecipitation and pull-down experiments revealed a physical association between calmodulin and APP. These results document a novel role of calmodulin in the regulation of non-amyloidogenic processing of APP.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Blood Platelets/metabolism , Calmodulin/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyloid beta-Protein Precursor/genetics , Blood Platelets/cytology , Calmodulin/antagonists & inhibitors , Cell Line , Enzyme Inhibitors/pharmacology , Gene Expression , Humans , Platelet Activation , Protein Binding , Sulfonamides/pharmacology
5.
J Thromb Haemost ; 7(7): 1200-6, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19422462

ABSTRACT

BACKGROUND: Platelet adhesion promoted by integrin alpha2beta1 induces integrin alpha(IIb)beta3 activation through the phospholipase C (PLC)-dependent stimulation of the small GTPase Rap1b. OBJECTIVE: To analyze the mechanism of PLC activation downstream of alpha2beta1 that is required for regulation of Rap1b and alpha(IIb)beta3. METHODS: Human and murine platelets were allowed to adhere to immobilized type I monomeric collagen through alpha2beta1. Tyrosine phosphorylation of PLCgamma2, PLC activation, accumulation of GTP-bound Rap1b and fibrinogen binding were measured and compared. RESULTS: Integrin alpha2beta1 recruitment induced an evident PLC activation that was concomitant with robust tyrosine phosphorylation of PLCgamma2, and was suppressed in platelets from PLCgamma2-knockout mice. Moreover, PLCgamma2(-/-) platelets were unable to accumulate active Rap1b and to activate alpha(IIb)beta3 upon adhesion through alpha2beta1. Inhibition of Src kinases completely prevented tyrosine phosphorylation of PLCgamma2 in adherent platelets, but did not affect its activation, and both Rap1b and alpha(IIb)beta3 stimulation occurred normally. Importantly, alpha(IIb)beta3-induced phosphorylation and activation of PLCgamma2, as well as accumulation of active Rap1b, were totally suppressed by Src inhibition. Integrin alpha2beta1 recruitment triggered the Src kinase-independent activation of the small GTPase Rac1, and activation of Rac1 was not required for PLCgamma2 phosphorylation. However, when phosphorylation of PLCgamma2 was blocked by the Src kinase inhibitor PP2, prevention of Rac1 activation significantly reduced PLCgamma2 activation, GTP-Rap1b accumulation, and alpha(IIb)beta3 stimulation. CONCLUSIONS: Src kinases and the Rac GTPases mediate independent pathways for PLCgamma2 activation downstream of alpha2beta1.


Subject(s)
Blood Platelets/enzymology , Integrin alpha2beta1/physiology , Phospholipase C gamma/metabolism , rac GTP-Binding Proteins/metabolism , src-Family Kinases/metabolism , Animals , Cell Adhesion , Electrophoresis, Polyacrylamide Gel , Enzyme Activation , Humans , In Vitro Techniques , Mice , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Tyrosine/metabolism , src-Family Kinases/antagonists & inhibitors
6.
J Thromb Haemost ; 7(3): 478-84, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19067792

ABSTRACT

BACKGROUND: Although mutations of GPIb alpha are among the most frequent causes of inherited platelet disorders, the mechanisms for the onset of thrombocytopenia and platelet macrocytosis are still poorly defined. OBJECTIVE: In this work we analyzed in vitro megakaryocyte differentiation and proplatelet formation in six subjects heterozygous for the Ala156Val mutation in the GPIb alpha (Bolzano mutation). METHODS: Human megakaryocytes were obtained by differentiation of patient cord blood-derived CD34(+) cells and peripheral blood-derived CD45(+) cells. Proplatelet formation was evaluated by phase contrast and fluorescence microscopy. RESULTS: Megakaryocyte differentiation from both cord blood (one patient) and peripheral blood (five patients) was comparable to controls. However, proplatelet formation was reduced by about 50% with respect to controls. An identical defect of proplatelet formation was observed when megakaryocytes were plated on fibrinogen, von Willebrand factor or grown in suspension. Morphological evaluation of proplatelet formation revealed an increased size of proplatelet tips, which was consistent with the increased diameters of patients' blood platelets. Moreover, alpha-tubulin distribution within proplatelets was severely deranged. CONCLUSIONS: Megakaryocytes from patients carrying a Bolzano allele of GPIb alpha display both quantitative and qualitative abnormalities of proplatelet formation in vitro. These results suggest that a defect of platelet formation contributes to macrothrombocytopenia associated to the Bolzano mutation, and indicate a key role for GPIb alpha in proplatelet formation.


Subject(s)
Bernard-Soulier Syndrome/genetics , Blood Platelets/pathology , Megakaryocytes/pathology , Membrane Proteins/genetics , Alleles , Bernard-Soulier Syndrome/pathology , Cell Differentiation , Cell Shape , Heterozygote , Humans , Membrane Glycoproteins , Mutation, Missense , Platelet Glycoprotein GPIb-IX Complex , Thrombocytopenia
7.
J Thromb Haemost ; 6(11): 1900-7, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18752571

ABSTRACT

BACKGROUND: Megakaryocytes release platelets from the tips of cytoplasmic extensions, called proplatelets. In humans, the regulation of this process is still poorly characterized. OBJECTIVE: To analyse the regulation of proplatelet formation by megakaryocyte adhesion to extracellular adhesive proteins through different membrane receptors. METHODS: Human megakaryocytes were obtained by differentiation of cord blood-derived CD34(+) cells, and proplatelet formation was evaluated by phase contrast and fluorescence microscopy. RESULTS: We found that human megakaryocytes extended proplatelets in a time-dependent manner. Adhesion to fibrinogen, fibronectin or von Willebrand factor (VWF) anticipated the development of proplatelets, but dramatically limited both amplitude and duration of the process. Type I, but not type III or type IV, collagen totally suppressed proplatelet extension, and this effect was overcome by the myosin IIA antagonist blebbistatin. Integrin alphaIIbbeta3 was essential for megakaryocyte spreading on fibrinogen or VWF, but was not required for proplatelet formation. In contrast, proplatelet formation was prevented by blockade of GPIb-IX-V, or upon cleavage of GPIbalpha by the metalloproteinase mocarhagin. Membrane-associated VWF was detected exclusively on proplatelet-forming megakaryocytes, but not on round mature cells that do not extend proplatelets. CONCLUSIONS: Our findings show that proplatelet formation in human megakaryocytes undergoes a complex spatio-temporal regulation orchestrated by adhesive proteins, GPIb-IX-V and myosin IIA.


Subject(s)
Blood Platelets/cytology , Blood Proteins/metabolism , Megakaryocytes/cytology , Membrane Glycoproteins/metabolism , Nonmuscle Myosin Type IIA/physiology , Cell Adhesion , Fibrinogen/metabolism , Fibronectins/metabolism , Humans , Kinetics , Megakaryocytes/metabolism , Microscopy , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Platelet Glycoprotein GPIb-IX Complex/metabolism , von Willebrand Factor/metabolism
8.
J Thromb Haemost ; 6(10): 1772-9, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18647220

ABSTRACT

BACKGROUND: The endocannabinoid 2-arachidonoylglycerol (2-AG) is an endogenous lipid that acts through the activation of G-protein-coupled cannabinoid receptors and plays essential roles in many physiological contexts. In the cardiovascular system 2-AG is generated by both activated endothelial cells and platelets, and participates in the regulation of inflammation and thrombosis. Although human platelets actively metabolize endocannabinoids, 2-AG also binds to platelet surface and leads to cell activation. OBJECTIVE: To investigate the biological consequence of 2-AG interactions with human platelets and to clarify the role of cannabinoid receptors. METHODS: Gel-filtered platelets were stimulated with 2-AG in the presence or absence of various inhibitors. Platelet aggregation and secretion were measured in a lumiaggregometer. Calcium ion movements were measured in FURA-2 loaded platelets. Thromboxane A(2) (TxA(2)) generation was evaluated as Thromboxane B(2) accumulation with a commercial EIA assay. RESULTS: 2-AG induced platelet shape change, aggregation and secretion with a dose-dependent mechanism that required engagement of platelet TxA(2) receptors. 2-AG caused also cytosolic calcium increase; however, it was totally dependent on availability of TxA(2). Indeed 2-AG was able to induce a robust generation of TxA(2) through the cyclooxygenase pathway. Treatment of platelets with inhibitors of monoacylglycerol lipase and fatty acid amide hydrolase did not affect the activation induced by 2-AG. Moreover, neither CB(1) and CB(2) proteins nor CB(1)/CB(2) mRNAs were detected in platelets. CONCLUSIONS: 2-AG can be considered a new physiologic platelet agonist able to induce full platelet activation and aggregation with a non-CB(1)/CB(2) receptor-mediated mechanism.


Subject(s)
Arachidonic Acids/pharmacology , Cannabinoid Receptor Modulators/pharmacology , Endocannabinoids , Glycerides/pharmacology , Platelet Activation/drug effects , Receptors, Cannabinoid/physiology , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Blood Platelets/chemistry , Blood Platelets/metabolism , Calcium/metabolism , Cells, Cultured , Humans , Platelet Aggregation , Receptors, Thromboxane A2, Prostaglandin H2/physiology
9.
J Thromb Haemost ; 3(11): 2526-35, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16241951

ABSTRACT

OBJECTIVE: Platelets contain and release some matrix metalloproteinases (MMPs), enzymes involved in the degradation of extracellular matrix, and one of these (MMP-2) exerts a proaggregatory effect. We explored the signal transduction mechanisms activated by MMP-2 in human blood platelets. METHODS AND RESULTS: Recombinant, human MMP-2, added before stimulation with subthreshold doses of different agonists, potentiated platelet activation, calcium influx, IP3 formation, and pleckstrin phosphorylation. Wortmannin and LY29400, two PI3-K inhibitors, suppressed the potentiating effects of MMP-2 and preincubation with MMP-2 enhanced the thrombin-induced association of the p85alpha PI3-K subunit with the cytoskeleton and increased the phosphorylation of PKB. Protein tyrosine kinase inhibitors, MAP kinase inhibitors, PLA2 inhibitors, cyclooxygenase inhibitors and antagonists of the P2Y1 and P2Y12 receptors did not affect the potentiating activity of MMP-2 on platelets. CONCLUSION: Our data show that MMP-2, at a concentration released by activated platelets, facilitates platelet activation acting at the level of a second messenger system common to different agonists and related to the activation of PI3-K. Platelet-released MMP-2 may contribute to platelet activation in vivo.


Subject(s)
Matrix Metalloproteinase 2/pharmacology , Platelet Aggregation/drug effects , Recombinant Proteins/pharmacology , Second Messenger Systems/drug effects , Signal Transduction/drug effects , Androstadienes/pharmacology , Blood Platelets/drug effects , Blood Platelets/metabolism , Calcium/metabolism , Chromones/pharmacology , Drug Synergism , Estrenes/pharmacology , Humans , In Vitro Techniques , Indoles/pharmacology , Inositol 1,4,5-Trisphosphate/metabolism , Matrix Metalloproteinase Inhibitors , Morpholines/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/metabolism , Pyrrolidinones/pharmacology , Recombinant Proteins/antagonists & inhibitors , Thrombin/pharmacology , Tissue Inhibitor of Metalloproteinase-2/pharmacology , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/metabolism , Wortmannin
10.
J Thromb Haemost ; 3(5): 1026-35, 2005 May.
Article in English | MEDLINE | ID: mdl-15869600

ABSTRACT

MYH9-related disease (MYH9-RD) is an autosomal dominant disorder deriving from mutations in the MYH9 gene encoding for the heavy chain of non-muscle myosin IIA, and characterized by thrombocytopenia and giant platelets. Isoform IIA of myosin is the only one expressed in platelets, but the possibility that MYH9 mutations affect the organization of contractile structures in these blood elements has never been investigated. In this work we have analyzed the composition and the agonist-induced reorganization of the platelet cytoskeleton from seven MYH9-RD patients belonging to four different families. We found that an increased amount of myosin was constitutively associated with actin in the cytoskeleton of resting MYH9-RD platelets. Upon platelet stimulation, an impaired increase in the total cytoskeletal proteins was observed. Moreover, selected membrane glycoproteins, tyrosine kinases, and small GTPases failed to interact with the cytoskeleton in agonist-stimulated MYH9-RD platelets. These results demonstrate for the first time that mutations of MYH9 result in an alteration of the composition and agonist-induced reorganization of the platelet cytoskeleton. We suggest that these abnormalities may represent the biochemical basis for the previously reported functional alterations of MYH9-RD platelets, and for the abnormal platelet formation from megakaryocytes, resulting in thrombocytopenia and giant platelets.


Subject(s)
Blood Platelet Disorders/diagnosis , Blood Platelet Disorders/metabolism , Blood Platelets/metabolism , Cytoskeleton/metabolism , Molecular Motor Proteins/metabolism , Molecular Motor Proteins/physiology , Myosin Heavy Chains/metabolism , Myosin Heavy Chains/physiology , Thrombocytopenia/genetics , Adolescent , Adult , Dimerization , Electrophoresis, Polyacrylamide Gel , Family Health , Female , GTP Phosphohydrolases/metabolism , Genes, Dominant , Glycoproteins/metabolism , Humans , Immunoblotting , Male , Megakaryocytes/metabolism , Middle Aged , Mutation , Nonmuscle Myosin Type IIA/chemistry , Polymorphism, Genetic , Signal Transduction
11.
J Thromb Haemost ; 2(12): 2223-30, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15613030

ABSTRACT

The activation of the small GTPase Rap2B in resting and agonist-stimulated human platelets was investigated. Both thrombin, that stimulates heterotrimeric G-protein-coupled receptors, and the GPVI ligand convulxin, that activates a tyrosine-kinase based signaling pathway, were able to induced the rapid and sustained binding of GTP to Rap2B. Similarly, a number of other agonists tested, previously known to activate the highly related protein Rap1B, were also able to stimulate Rap2B. In contrast, platelet antagonists that increase the intracellular concentration of cAMP did not signal to Rap2B. Thrombin- and convulxin-induced activation of Rap2B was not dependent on thromboxane A2, did not require the interaction of the protein with the cytoskeleton, and was not regulated by integrin alphaIIbbeta3-dependent outside-in signaling. When secreted ADP was neutralized, activation of Rap2B induced by thrombin, but not by convulxin, was significantly reduced. ADP itself was found to induce the rapid and sustained binding of GTP to Rap2B, and this effect was predominantly mediated by stimulation of the Gi-coupled P2Y12 receptor. Activation of Rap2B promoted by both thrombin and convulxin was regulated by intracellular Ca2+, while protein kinase C was found to be involved in convulxin- but not in thrombin-induced activation of Rap2B. Moreover, Rap2B activation induced by thrombin, but not by convulxin, was totally dependent on phosphatidylinositol 3-kinase activity. These results demonstrate that the small GTPase Rap2B is involved in platelet activation, and outline some important differences between the regulation of highly related GTPases Rap2B and Rap1B in human platelets.


Subject(s)
Blood Platelets/enzymology , rap GTP-Binding Proteins/metabolism , Adenosine Diphosphate/metabolism , Calcium/metabolism , Chromatography, Gel , Crotalid Venoms/pharmacology , Cyclic AMP/metabolism , Cytoskeleton/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Glutathione Transferase/metabolism , Humans , Lectins, C-Type , Phosphatidylinositol 3-Kinases/metabolism , Platelet Activation , Promoter Regions, Genetic , Protein Kinase C/metabolism , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Thrombin/metabolism , Time Factors
12.
J Biol Chem ; 276(28): 26022-9, 2001 Jul 13.
Article in English | MEDLINE | ID: mdl-11344169

ABSTRACT

Interaction of von Willebrand Factor with glycoprotein Ib-IX-V induces platelet activation through a still poorly defined mechanism. Previous studies have suggested a possible role for the low affinity receptor for immunoglobulin, Fc gamma RIIA, in GPIb-IX-V signaling. Here we show that binding of vWF to platelets induces the tyrosine phosphorylation of Fc gamma RIIA by a Src kinase. Treatment of platelets with the anti-Fc gamma RIIA monoclonal antibody IV.3 specifically inhibits vWF-induced but not thrombin-induced pleckstrin phosphorylation and serotonin secretion. Moreover, vWF fails to induce pleckstrin phosphorylation in mouse platelets, lacking Fc gamma RIIA, and serotonin secretion is impaired. Pleckstrin phosphorylation and serotonin secretion in human platelets stimulated with vWF are blocked by the cyclooxygenase inhibitor acetylsalicylic acid. However, release of arachidonic acid and synthesis of TxA(2) induced by vWF are not affected by the anti-Fc gamma RIIA monoclonal antibody IV.3. Similarly, vWF-induced tyrosine phosphorylation of Fc gamma RIIA, as well as of Syk and PLC gamma 2, occurs normally in aspirinized platelets. Inhibition of the tyrosine kinase Syk by piceatannol does not affect vWF-induced tyrosine phosphorylation of Fc gamma RIIA but prevents phosphorylation of PLC gamma 2. Pleckstrin phosphorylation and platelet secretion induced by vWF, but not by thrombin, are also inhibited by piceatannol. Pleckstrin phosphorylation is also sensitive to the phosphatidylinositol 3-kinase inhibitor wortmannin. These results indicate that PLC gamma 2 plays a central role in platelet activation by vWF and that the stimulation of this enzyme requires coordinated signals through endogenous TxA(2) and Fc gamma RIIA.


Subject(s)
Antigens, CD/physiology , Platelet Activation , Receptors, IgG/physiology , Thromboxane A2/physiology , von Willebrand Factor/physiology , Humans , Signal Transduction
13.
FEBS Lett ; 466(2-3): 355-8, 2000 Jan 28.
Article in English | MEDLINE | ID: mdl-10682859

ABSTRACT

Pretreatment of intact platelets with cytochalasin D prevented actin polymerization and cytoskeleton reorganization induced by thrombin, but did not affect platelet aggregation. Under these conditions, synthesis of phosphatidylinositol 3,4-bisphosphate (PtdIns(3,4)P2) stimulated by thrombin was strongly inhibited, while production of phosphatidic acid was unaffected. The inhibitory effect of cytochalasin D was not observed when platelet aggregation was prevented by the RGDS peptide. We also found that cytochalasin D did not affect PtdIns(3,4)P2 synthesis induced by concanavalin A (ConA), which is known to occur through an aggregation-independent mechanism. Moreover, thrombin, but not ConA, induced the translocation of phosphatidylinositol 3-kinase to the cytoskeleton. This process was equally inhibited by both the RGDS peptide and cytochalasin D. These results demonstrate that the cytoskeleton represents a functional link between thrombin-induced aggregation and synthesis of PtdIns(3,4)P2.


Subject(s)
Blood Platelets/drug effects , Cytoskeleton/drug effects , Phosphatidylinositol Phosphates/chemical synthesis , Platelet Aggregation , Thrombin/pharmacology , Blood Platelets/physiology , Cytochalasin D/pharmacology , Cytoskeleton/physiology , Humans , In Vitro Techniques
14.
Mov Disord ; 15(1): 71-6, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10634244

ABSTRACT

This study was designed to investigate anorectal function in Parkinson's disease and multiple system atrophy (MSA). After a standardized interview, 17 patients with Parkinson's disease (PD) and 16 patients with multiple system atrophy (MSA) underwent anorectal manometry with a continuously perfused multi-lumen catheter, located to record pressures from the anal canal, and a balloon for rectal distension. Data were analyzed by observers blind to the neurologic diagnosis. Disease duration was shorter in the MSA than in the PD group (6+/-4 versus 10+/-5 yrs, p<0.05). Most patients reported a bowel frequency of less than three evacuations per week and some patients had fecal incontinence. Most manometric recordings disclosed an abnormal pattern during straining (a paradoxic contraction or lack of inhibition) in 13 patients with MSA and 11 patients with PD. Mean anal pressures and rectal sensitivity threshold were not significantly higher in the MSA group, whereas the inhibitory anal reflex and rectal compliance thresholds were within the normal range in both groups. Manometric patterns did not differentiate patients with MSA from patients with PD. Most patients in both groups showed an abnormal straining pattern, decreased anal tone, or both dysfunctions. In conclusion, our findings suggest that although bowel and anorectal dysfunctions do not differentiate MSA from PD, both abnormalities occur earlier and develop faster in MSA than in PD.


Subject(s)
Anal Canal/physiopathology , Multiple System Atrophy/physiopathology , Parkinson Disease/physiopathology , Rectum/physiopathology , Aged , Anal Canal/innervation , Ataxia/diagnosis , Ataxia/physiopathology , Autonomic Nervous System/physiopathology , Constipation/diagnosis , Constipation/physiopathology , Fecal Incontinence/diagnosis , Fecal Incontinence/physiopathology , Female , Humans , Male , Manometry , Middle Aged , Multiple System Atrophy/diagnosis , Parkinson Disease/diagnosis , Rectum/innervation
15.
J Cell Biochem ; 75(4): 675-85, 1999 Dec 15.
Article in English | MEDLINE | ID: mdl-10572250

ABSTRACT

The interaction of the low-molecular-weight GTP-binding protein rap2 with the cytoskeleton from thrombin-aggregated platelets was investigated by inducing depolymerization of the actin filaments, followed by in vitro-promoted repolymerization. We found that the association of rap2 with the cytoskeleton was spontaneously restored after one cycle of actin depolymerization and repolymerization. Exogenous rap2, but not unrelated proteins, added to depolymerized actin and solubilized actin-binding proteins, was also specifically incorporated into the in vitro reconstituted cytoskeleton. The incorporation of exogenous rap2 was also observed when the cytoskeleton from resting or thrombin-activated platelets was subjected to actin depolymerization-repolymerization. Moreover, such interaction occurred equally well when exogenous rap2 was loaded with either GDP or GTPgammaS. We also found that polyhistidine-tagged rap2 immobilized on Ni(2+)-Sepharose and loaded with either GDP or GTPgammaS, could specifically bind to cytoskeletal actin. Moreover, when purified monomeric actin was induced to polymerize in vitro in the presence of rap2, the small G-protein specifically associated with the actin filaments. Finally, rap2 loaded with either GDP or GTPgammaS was able to bind to purified F-actin immobilized on a plastic surface. These results demonstrate that rap2 interacts with the platelet cytoskeleton by direct binding to the actin filaments and that this interaction is not regulated by the activation state of the protein.


Subject(s)
Actins/metabolism , Blood Platelets/metabolism , Cytoskeleton/metabolism , rap GTP-Binding Proteins/metabolism , Biotin , Blood Platelets/drug effects , Chymotrypsinogen/metabolism , Electrophoresis, Polyacrylamide Gel , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Guanosine Diphosphate/metabolism , Humans , Immunoblotting , Octoxynol , Thrombin/pharmacology
16.
FEBS Lett ; 455(3): 359-63, 1999 Jul 23.
Article in English | MEDLINE | ID: mdl-10437804

ABSTRACT

CD38 is a multifunctional cell surface ectoenzyme that catalyzes both the synthesis of cyclic ADP-ribose from NAD+ and its hydrolysis to ADP-ribose. In this work, we investigated the metabolism of NADP+ by CD38 expressed on human platelets. Incubation of either platelet membranes or intact cells with NADP+ resulted in the rapid and time-dependent accumulation of ADP-ribose 2'-phosphate that paralleled the consumption of the substrate. However, under the same conditions, synthesis of cyclic ADP-ribose 2'-phosphate was not observed. By immunoprecipitation experiments, we identified CD38 as the enzyme responsible for the observed NADP+ glycohydrolase activity. The lack of detection of cyclic ADP-ribose 2'-phosphate was not due to its rapid hydrolysis, since direct incubation of platelet membranes with cyclic ADP-ribose 2'-phosphate did not result in the formation of ADP-ribose 2'-phosphate. By contrast, the same membrane samples expressed a significant ability to hydrolyze cyclic ADP-ribose to ADP-ribose. The absence of cyclic ADP-ribose 2'-phosphate hydrolase activity was also confirmed using high concentrations of substrate and by analysing both intact Jurkat T-lymphocytes and immunoprecipitated CD38. These results indicate that CD38, which is a multifunctional enzyme towards NAD+, displays exclusively a NADP+ glycohydrolase activity and is unable to catalyze both the synthesis and the hydrolysis of cyclic ADP-ribose 2'-phosphate.


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , Antigens, CD , Antigens, Differentiation/metabolism , Blood Platelets/enzymology , Blood Platelets/immunology , Cyclic ADP-Ribose/analogs & derivatives , NAD+ Nucleosidase/metabolism , NADP/metabolism , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Adenosine Diphosphate Ribose/biosynthesis , Adenosine Diphosphate Ribose/metabolism , Humans , Hydrolysis , In Vitro Techniques , Jurkat Cells , Kinetics , Membrane Glycoproteins , Substrate Specificity , T-Lymphocytes/metabolism
17.
J Biol Chem ; 274(19): 13690-7, 1999 May 07.
Article in English | MEDLINE | ID: mdl-10224142

ABSTRACT

Stimulation of human platelets with von Willebrand factor (vWF) induced the translocation of the small GTPases Rap1B and Rap2B to the cytoskeleton. This effect was specifically prevented by an anti-glycoprotein Ib monoclonal antibody or by the omission of stirring, but was not affected by the peptide RGDS, which antagonizes binding of adhesive proteins to platelet integrins. Association of Rap2B with the cytoskeleton was very rapid, while translocation of Rap1B occurred in a later phase of platelet activation and was totally inhibited by cytochalasin D. vWF also induced the rapid tyrosine phosphorylation of several proteins that was prevented by the tyrosine kinases inhibitor genistein and by cAMP-increasing agents. Under these conditions, also the association of Rap1B and Rap2B with the cytoskeleton was prevented. Translocation of Rap proteins to the cytoskeleton induced by vWF, but not by thrombin, was inhibited by a monoclonal antibody against the FcgammaII receptor. The same antibody inhibited vWF-induced tyrosine phosphorylation of selected substrates with molecular masses of about 75, 95, and 150 kDa. Three of these substrates were identified as the tyrosine kinase pp72(syk), the phospholipase Cgamma2, and the inositol 5-phosphatase SHIP. Our results indicate that translocation of Rap1B and Rap2B to the cytoskeleton is regulated by tyrosine kinases and suggest a novel role for the FcgammaII receptor in the mechanism of platelet activation by vWF.


Subject(s)
Cytoskeleton/metabolism , GTP-Binding Proteins/metabolism , Receptors, IgG/metabolism , Tyrosine/metabolism , von Willebrand Factor/metabolism , Blood Platelets/metabolism , Humans , Kinetics , Phosphorylation , Platelet Activation , Platelet Membrane Glycoproteins/metabolism , rap GTP-Binding Proteins
18.
Thromb Haemost ; 81(1): 124-30, 1999 Jan.
Article in English | MEDLINE | ID: mdl-10348703

ABSTRACT

Tyrosine phosphorylation of the non-receptor tyrosine kinases pp72syk and pp125FAK and of the gamma2 isoform of phospholipase C (PLCgamma2) in human platelets stimulated with the lectin Concanavalin A was investigated. Concanavalin A induced the rapid tyrosine phosphorylation of pp72syk and PLCgamma2 with a similar kinetics, while tyrosine phosphorylation of pp125FAK occurred in a later phase of platelet activation. When compared with other platelet agonists, Concanavalin A revealed to be at least as potent as collagen in inducing tyrosine phosphorylation of PLCgamma2 and pp125FAK, while tyrosine phosphorylation of pp72syk induced by the lectin was much stronger than that induced by thrombin or collagen. Concanavalin A-induced tyrosine phosphorylation of pp72syk, PLCgamma2 and pp125FAK was not dependent on platelet aggregation as it occurred normally even in the absence of sample stirring and when fibrinogen binding to integrin alphaIIb-beta3 was inhibited by the peptide RGDS. Tyrosine phosphorylation of pp72syk, PLCgamma2 and pp125FAK required the binding of the lectin to the platelet surface, but was not observed in platelets treated with succinyl-Concanavalin A, a derivative of the lectin that interacts with the same receptors but does not promote clustering of membrane glycoproteins. Moreover, the aggregation-independent tyrosine phosphorylation of pp125FAK and pp72syk induced by Concanavalin A required the expression of integrin alphaIIb-beta3 on the platelet surface as it was strongly inhibited in platelets from patients affected by Glanzmann thrombasthenia. By contrast, tyrosine phosphorylation of PLCalpha2 occurred normally also in thrombasthenic platelets stimulated with Concanavalin A. These results demonstrate that, even in the absence of aggregation, the clustering of integrin alphaIIb-beta3 induced by Concanavalin A on the platelet surface directly promotes tyrosine phosphorylation of pp72syk and pp125FAK and provide further evidence that the oligomerization of the fibrinogen receptor promoted by its natural ligand during platelet aggregation may be responsible for the tyrosine phosphorylation of these proteins induced by physiological agonists.


Subject(s)
Blood Platelets/physiology , Cell Adhesion Molecules/physiology , Enzyme Precursors/physiology , Platelet Aggregation/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Protein-Tyrosine Kinases/physiology , Signal Transduction/physiology , Concanavalin A/pharmacology , Focal Adhesion Kinase 1 , Focal Adhesion Protein-Tyrosine Kinases , Humans , Intracellular Signaling Peptides and Proteins , Phosphorylation , Platelet Aggregation/drug effects , Signal Transduction/drug effects , Syk Kinase , Tyrosine/metabolism
19.
FEBS Lett ; 431(1): 19-22, 1998 Jul 10.
Article in English | MEDLINE | ID: mdl-9684857

ABSTRACT

Stimulation of human platelets with thrombin caused a 42% inhibition of the ADP-ribosyl cyclase activity of membrane CD38. This effect was mediated by the activation of the platelet thrombin receptor rather than by proteolysis of CD38, and was not due to a different distribution of the synthesised nucleotide or to a reduced accessibility of CD38 to the substrate. The inhibitory effect of thrombin required actin polymerisation and was not observed when interaction of CD38 with the cytoskeleton was prevented by cytochalasin D. Finally, we analysed whether cADPR could play a role as a Ca2+-mobilising agent in human platelets. Using saponin-permeabilised cells, we found that unlike IP3, cADPR did not induce any release of Ca2+ from intracellular stores. These results indicate that the enzymatic activity of membrane CD38 can be modulated by platelet activation, and that the function of this glycoprotein is probably not related to Ca2+ mobilisation.


Subject(s)
Antigens, CD , Antigens, Differentiation/metabolism , Blood Platelets/enzymology , Cytoskeleton/physiology , NAD+ Nucleosidase/metabolism , Platelet Activation , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Calcium/metabolism , Collagen/pharmacology , Humans , In Vitro Techniques , Inositol 1,4,5-Trisphosphate/metabolism , Membrane Glycoproteins , Thrombin/pharmacology
20.
FEBS Lett ; 428(3): 200-4, 1998 May 29.
Article in English | MEDLINE | ID: mdl-9654134

ABSTRACT

The effect of platelet stimulation on the subcellular localization of CD38, a membrane glycoprotein that catalyses the synthesis of cyclic ADP-ribose from beta-NAD+ was investigated. Treatment of human platelets with thrombin caused the association of about 40% of the total ADP-ribosyl cyclase activity with the cytoskeleton, through the translocation of the CD38 molecule from the Triton X-100-soluble to the insoluble fraction. The interaction of CD38 with the cytoskeleton was a specific and reversible process, mediated by the binding to the actin-rich filaments and was inhibited by treatment of platelets with cytochalasin D. This event was regulated by integrin alphaIIb beta3 and platelet aggregation as it was prevented by the inhibition of fibrinogen binding and was not observed in platelets from a patient affected by Glanzmann thrombasthenia. These results demonstrate that the subcellular localization of CD38 can be influenced by platelet stimulation with physiological agonists, and that membrane CD38 can interact with intracellular proteins.


Subject(s)
Adenosine Diphosphate Ribose/analogs & derivatives , Antigens, Differentiation/blood , Blood Platelets/physiology , Cytoskeleton/physiology , NAD+ Nucleosidase/blood , Thrombin/pharmacology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Actins/blood , Adenosine Diphosphate Ribose/biosynthesis , Adenosine Diphosphate Ribose/blood , Antigens, CD/blood , Antigens, CD/drug effects , Antigens, Differentiation/drug effects , Blood Platelets/drug effects , Collagen/pharmacology , Cyclic ADP-Ribose , Cytoskeleton/drug effects , Humans , In Vitro Techniques , Membrane Glycoproteins , Multienzyme Complexes/blood , Multienzyme Complexes/drug effects , NAD+ Nucleosidase/drug effects , Oligopeptides/pharmacology , Platelet Aggregation/drug effects , Protein Binding
SELECTION OF CITATIONS
SEARCH DETAIL
...