Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
J Am Soc Mass Spectrom ; 29(3): 455-462, 2018 03.
Article in English | MEDLINE | ID: mdl-29313205

ABSTRACT

Sulfotyrosine and phosphotyrosine are two post-translational modifications present in higher eukaryotes. A simple and direct mass spectrometry method to distinguish between these modifications is crucial to advance our understanding of the sulfoproteome. While sulfation and phosphorylation are nominally isobaric, the accurate mass of the sulfuryl moiety is 9.6 mDa less than the phosphoryl moiety. Based on this difference, we have used an Orbitrap Fusion Lumos mass spectrometer to characterize, resolve, and distinguish between sulfotyrosine and phosphotyrosine modifications using a set of model peptides. Multiple fragmentation techniques, namely HCD, CID, ETD, ETciD, and EThcD, have been used to compare the different fragmentation behaviors between peptides modified with these species. Sulfotyrosine undergoes neutral loss using HCD and CID, but the sulfuryl moiety is largely stable under ETD. In contrast, phosphotyrosine is stable during fragmentation using all these methods. This differential stability provides a mechanism to distinguish sulfopeptides from phosphopeptides. Based on the rigorous characterization presented herein, this work serves as a model for accurate identification of phosphotyrosine and, more challenging, sulfotyrosine, in complex proteomic samples. Graphical Abstract ᅟ.


Subject(s)
Peptides/chemistry , Phosphotyrosine/analysis , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Tyrosine/analogs & derivatives , Amino Acid Sequence , Humans , Peptides/blood , Phosphotyrosine/blood , Tyrosine/analysis , Tyrosine/blood
2.
Fertil Steril ; 94(5): 1616-21, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20153468

ABSTRACT

OBJECTIVE: To investigate the biochemical parameters of the erythrocyte response to diamide-induced oxidative stress, alone or as adjuncts to serum values of CA-125 and human epididymal secretory protein E4 (HE4), in the diagnosis and study of endometriosis. SETTING: University of Padova. DESIGN: Prospective study. PATIENT(S): Forty-five patients of reproductive age undergoing laparoscopy. INTERVENTION(S): All women were studied for endometriotic foci during laparoscopic surgery. Forty-one had laparoscopically and histologically confirmed endometriosis, and four did not. Twenty women with confirmed endometriosis were reassessed 1-4 months later. MAIN OUTCOME MEASURE(S): CA-125 and HE4 and two new parameters evaluated in erythrocytes after diamide-induced stress, that is, band 3 tyrosine phosphorylation (Tyr-P) level and decrease in total glutathione content (ΔGSH), were assessed in all patients. RESULT(S): In association with serum CA-125 levels but not with HE4, diamide-related erythrocyte band 3 Tyr-P and ΔGSH were significantly higher in patients with endometriosis and were able to discriminate with high sensitivity and specificity between patients before and after surgery. CONCLUSION(S): Endometriosis is associated with an increase in systemic oxidative stress, affecting the antioxidative defenses of circulating erythrocytes. All related implications, including evaluation of other oxidative stress-related changes, warrant further study.


Subject(s)
Anion Exchange Protein 1, Erythrocyte , CA-125 Antigen/blood , Endometriosis/blood , Epididymal Secretory Proteins/metabolism , Erythrocytes/metabolism , Glutathione/blood , Phosphotyrosine/blood , Uterine Diseases/blood , Adult , Biomarkers/blood , Case-Control Studies , Endometriosis/diagnosis , Female , Humans , Oxidative Stress/physiology , Prospective Studies , Sensitivity and Specificity , Uterine Diseases/diagnosis , beta-Defensins
3.
Arch Biochem Biophys ; 484(2): 173-82, 2009 Apr 15.
Article in English | MEDLINE | ID: mdl-18996080

ABSTRACT

Peroxynitrite crosses the red blood cell (RBC) membrane and reacts with hemoglobin (Hb) producing mainly metHb, which is reduced back to ferrousHb by NADH- and NADPH-dependent reductases. Peroxynitrite also induces band 3 (B3) tyrosine phosphorylation, a signaling pathway believed to activate glucose metabolism. This study was aimed to decipher the relationship between these two peroxynitrite-dependent processes. Peroxynitrite induced a burst of the hexose monophosphate shunt (HMS), revealed by NMR studies, and a burst of the glycolytic pathway, measured by lactate production. The HMS plays a prominent role in membrane signaling, as demonstrated by B3 phosphotyrosine inhibition by the glycolytic pathway inhibitor 2-deoxy-glucose (2DG) and activation by dehydroepiandrosterone (DHEA), an inhibitor of HMS. Peroxynitrite-induced B3 tyrosine phosphorylation was paralleled by the inhibition of membrane-associated phosphotyrosine phosphatase (PTP) activity, which was protected by 2DG but not DHEA. Interestingly, heme poisoning with CO inhibited peroxynitrite-dependent Hb oxidation and lactate production but did not affect PTP down regulation. These results suggest two distinct and concurrent effects of peroxynitrite: one mediated by Hb which, likely in its oxidized state, binds more strongly to B3, and another mediated by PTP-dependent B3 phosphorylation. Both effects are directed towards a surge in glucose utilization.


Subject(s)
Anion Exchange Protein 1, Erythrocyte/metabolism , Erythrocytes/metabolism , Oxyhemoglobins/metabolism , Peroxynitrous Acid/pharmacology , Phosphotyrosine/blood , Anion Exchange Protein 1, Erythrocyte/drug effects , Carbon Dioxide/pharmacology , Cyclooxygenase 1/blood , Erythrocyte Membrane/drug effects , Erythrocyte Membrane/metabolism , Erythrocytes/drug effects , Humans , Lipids/blood , Lipoproteins/blood , Lipoxygenase/blood , Nitric Oxide/pharmacology
4.
Ann Hematol ; 88(1): 29-36, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18696070

ABSTRACT

Assay of phosphotyrosine levels using flow cytometry has been used to identify patients with chronic myelogenous leukemia positive for the Bcr-Abl fusion gene. We hypothesized that clinical monitoring could identify treatment response through reductions in intragranulocyte phosphotyrosine. Initially, we studied cell lines FDC-P1 and HL60 (Bcr-Abl-negative) and FDrv210 and K562 (Bcr-Abl-positive) with our technique. A fluorescein isothiocyanate-conjugated monoclonal antibody was used along with fluorescence-conjugated microspheres for reference (ratio of sample fluorescence: bead fluorescence = relative fluorescence unit [RFU]). Samples from 20 controls and 32 patients undergoing treatment were analyzed using the same method. Bcr-Abl status for each patient was confirmed using fluorescent in situ hybridization or polymerase chain reaction gene amplification (PCR). Testing of cell lines consistently produced expected results. Patient values were found to be significantly higher than control values (P < 0.001) and values for patients with advanced disease were significantly higher than for patients with chronic-phase disease (P < 0.05). Results of clinical monitoring were consistent with results from PCR. Two patients who received allogeneic stem cell transplantation had molecular remission confirmed by PCR and had a marked decrease in RFU value (from 62 to five and from 131 to 23). No such fluorescence change was observed in patients who achieved clinical remission. Flow cytometric analysis of phosphotyrosine levels is a reliable and convenient adjuvant technique for diagnosis of Bcr-Abl-positive leukemias and shows promise for serial evaluation of patients undergoing treatment.


Subject(s)
Biomarkers/blood , Flow Cytometry/methods , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/blood , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/diagnosis , Phosphotyrosine/blood , Humans , In Situ Hybridization, Fluorescence , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/physiopathology , Polymerase Chain Reaction , Prognosis , Sensitivity and Specificity
5.
Platelets ; 18(1): 35-42, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17365852

ABSTRACT

TGF-beta1 plays a main role in tissue repair by regulating extracellular matrix production and tissue granulation. Platelets are one of the main sources of this cytokine in the circulation. The aim of this study was to evaluate the presence of the TGF-beta receptors on platelets, the effect of TGF-beta1 on platelet aggregation and the underlying intracellular mechanisms. TGF-beta receptors on platelets were studied by flow cytometry and their mRNA by PCR. Platelet aggregation was assessed by turbidimetric methods and intracellular pathways by Western blot. TGF-beta receptor type II and mRNA codifying for TbetaRI and TbetaRII were found in platelets. We demonstrated that TGF-beta1 did not trigger platelet aggregation by itself but had a modulating effect on ADP-induced platelet aggregation. Either inhibition or increase in platelet aggregation, depending on the exposure time to TGF-beta1 and the ADP concentration used, were shown. We found that platelets possess Smad2 protein and that its phosphorylation state is increased after exposure to TGF-beta1. Besides, TGF-beta1 modified the pattern of ADP-induced tyrosine phosphorylation. Increased phosphorylation levels of 64-, 80- and 125-kDa proteins during short time incubation with TGF-beta1 and increased phosphorylation of 64- and 125-kDa proteins after longer incubation were observed. The modulating effect of TGF-beta1 on platelet aggregation could play a role during pathological states in which circulating TGF-beta1 levels are increased and intravascular platelet activation is present, such as myeloproliferative disorders. In vascular injury, in which platelet activation followed by granule release generates high local ADP concentrations, it could function as a physiological mechanism of platelet activation control.


Subject(s)
Activin Receptors, Type I/blood , Platelet Aggregation/drug effects , Receptors, Transforming Growth Factor beta/blood , Smad2 Protein/blood , Transforming Growth Factor beta1/physiology , Activin Receptors, Type I/genetics , Adenosine Diphosphate/pharmacology , Blood Proteins/metabolism , Drug Synergism , Humans , Molecular Weight , Phosphoproteins/blood , Phosphorylation , Phosphotyrosine/blood , Platelet Activation/drug effects , Platelet Activation/physiology , Platelet Aggregation/physiology , Protein Processing, Post-Translational , Protein Serine-Threonine Kinases , RNA, Messenger/blood , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Time Factors , Transforming Growth Factor beta1/blood , Transforming Growth Factor beta1/pharmacology
6.
Clin Biochem ; 38(5): 457-64, 2005 May.
Article in English | MEDLINE | ID: mdl-15820777

ABSTRACT

OBJECTIVES: Magnesium sulfate has been recognized as a neuroprotective agent against hypoxia-ischemia, mainly by the protection from the excitotoxicity associated with increased glutamate concentration. However, the mechanism of MgSO4 action is not fully understood and is considerably controversial. DESIGN AND METHODS: During the 2 first hours of life, the asphyxiated full-term newborns were treated intravenously with one dose of MgSO4 250 mg/kg body weight. At birth, after 6 and 48 h of life the activity of ATP-dependent enzymes in erythrocyte membranes: Mg2+-ATPase, Ca2+-ATPase, protein kinases A and C, were determined. Using monoclonal antibodies, the band 3 and its phosphotyrosine level were also assayed. RESULTS: The time-dependent decrease of Ca2+-ATPase activity was detected in untreated newborns, whereas MgSO4 prevented this reduction. After 48 h, protein kinases activities differed in MgSO4-treated and untreated groups. Magnesium therapy increased the amount of band 3 and diminished proteolytic degradation of this protein. CONCLUSION: Our results demonstrated, for the first time, that magnesium sulfate treatment significantly altered the activities of some important enzymes in erythrocyte membrane from asphyxiated newborns. It also reduced the post-asphyxial damages of membrane compounds. These data may partly explain the molecular mechanisms of MgSO4 action in asphyxiated newborns.


Subject(s)
Asphyxia Neonatorum/drug therapy , Erythrocyte Membrane/drug effects , Magnesium Sulfate/therapeutic use , Neuroprotective Agents/therapeutic use , Anion Exchange Protein 1, Erythrocyte/analysis , Asphyxia Neonatorum/enzymology , Ca(2+) Mg(2+)-ATPase/blood , Calcium-Transporting ATPases/blood , Cyclic AMP-Dependent Protein Kinases/blood , Erythrocyte Membrane/enzymology , Humans , Infant, Newborn , Magnesium/blood , Phosphotyrosine/blood , Protein Kinase C/blood
7.
J Ren Nutr ; 15(1): 8-12, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15647999

ABSTRACT

The bioincompatibility of dialytic systems along with the loss of antioxidant substances via the dialysis may contribute to peripheral blood mononuclear cell (PBMC) activation and the production of inflammatory mediators, such as cytokines, oxygen radicals, and complement fragments, that may sustain a state of chronic microinflammation responsible for the pathogenesis of a variety of diseases, including atherosclerosis, anemia, and malnutrition. Moreover, during hemodialysis (HD), oxidative stress may influence several intracellular signaling enzymes, including some stress-activated kinases, such as jun-N-terminal kinase (JNK), potentially leading to PBMC activation and proinflammatory cytokine production. Recent reports suggest that L-carnitine may play an important role in balancing antioxidative systems. Therefore, we sought to evaluate the effect of L-carnitine supplementation on the PBMC responses to oxidative stress induced by different HD membranes. We observed in PBMC from cellulosic (C)-treated patients an increase in the amount of intracellular tyrosine-phosphorylated proteins and a striking activation of JNK, as compared with synthetic (S)-treated patients. On the contrary, 3 months of L-carnitine supplementation significantly lowered intracellular levels of phosphorylated proteins and JNK activity in PBMC from C-treated patients. In addition, after 180 minutes of HD, a significant decrease in global plasma antioxidant capacity was found, particularly in C-treated patients, whereas L-carnitine supplementation improved plasma antioxidant capacity levels in these patients. These observations were also confirmed by in vitro experiments, showing the ability of L-carnitine to reduce the JNK activation in normal PBMC exposed to different amounts of hydrogen peroxide. In conclusion, the uremic milieu is characterized by an enhanced inflammatory response and an increased oxidant load, affecting lipids, carbohydrates, and proteins. Regular L-carnitine supplementation in HD patients can improve cellular defense against chronic inflammation and oxidative stress, most likely by modulating the specific signal transduction cascade activated by an overproduction of proinflammatory cytokines and oxidative stress.


Subject(s)
Carnitine/physiology , Inflammation/etiology , Renal Dialysis/adverse effects , Atherosclerosis/etiology , Carnitine/administration & dosage , Chemokine CCL2/physiology , Humans , Interleukin-6/physiology , JNK Mitogen-Activated Protein Kinases/blood , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/complications , Kidney Failure, Chronic/therapy , Leukocytes, Mononuclear/chemistry , Leukocytes, Mononuclear/enzymology , Leukocytes, Mononuclear/physiology , Phosphoproteins/blood , Phosphotyrosine/blood , Signal Transduction
8.
Anticancer Res ; 23(2A): 991-7, 2003.
Article in English | MEDLINE | ID: mdl-12820337

ABSTRACT

INTRODUCTION: Tyrosine kinase signal transduction pathways are a focus of interest for therapeutic interventions. The oncoprotein HER-2/neu shows tyrosine kinase activity leading to phosphorylation and activation of numerous second-messenger systems. One target of phosphorylation processes is assumed to be the tumor type M2 isoenzyme of pyruvate kinase (TuM2-PK) which has been shown to be elevated in metastatic breast cancer. MATERIALS AND METHODS: We measured the plasma levels of HER-2/neu, TuM2-PK and tyrosine-phosphorylated TuM2-PK (p-TuM2-PK) in 69 patients (pts) with breast cancer and correlated these parameters to each other and to the classical tumor marker CA 27.29. The samples were measured with ELISA assays while CA 27.29 was determined with an automated chemiluminescence assay. For analysis, we formed 5 subgroups according to the plasma HER-2/neu levels (group 1: < 15 ng/ml, n = 28; group 2: 15 < or = x < 50 ng/ml, n = 21; group 3: 50 < or = x < 100 ng/ml, n = 9; group 4: 100 < or = x < 500 ng/ml, n = 7; group 5: > or = 500 ng/ml, n = 4). RESULTS: From the HER-2/neu group 1 to group 5, there was a statistically significant increase of CA 27.29 from 35.8 U/ml to 1095.8 U/ml (p < 0.001). There was also a trend for increasing TuM2-PK levels with increasing HER-2/neu levels (p = 0.126). From the lowest extinction (0.088) to the highest extinction result (2.167) of p-TuM2-PK we found a 25-fold increase, which was reproducible in spiking and dilution experiments proving that TuM2-PK is phosphorylated at tyrosine residues to a certain extent. However, there was no correlation between plasma HER-2/neu and p-TuM2-PK levels. CONCLUSION: TuM2-PK is phosphorylated at tyrosine residues in breast cancer patients. Using the shed antigen of HER-2/neu in plasma as a surrogate marker, we did not find any evidence that this phosphorylation is initiated by the oncoprotein HER-2/neu.


Subject(s)
Breast Neoplasms/pathology , Phosphotyrosine/blood , Pyruvate Kinase/blood , Receptor, ErbB-2/blood , Adult , Aged , Antigens, Tumor-Associated, Carbohydrate/blood , Biomarkers, Tumor/blood , Breast Neoplasms/blood , Female , Humans , Isoenzymes/blood , Middle Aged , Neoplasm Staging , Protein-Tyrosine Kinases/blood , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis
9.
Platelets ; 13(8): 437-42, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12487776

ABSTRACT

The effects of Mg2+ and EDTA on collagen-induced platelet dense granule secretion and protein tyrosine phosphorylation were studied in the presence and absence of various inhibitors of autocrine agonists that are released from secretory granules. Addition of EDTA to gel-filtered platelets in Tyrode's solution caused a decrease in pH of 0.7-0.9 pH units, due to chelation of Mg2+, and a marked inhibition of cATP+ADP secretion induced by collagen (25 (micro)g/ml). Lowering pH of the platelet suspension to the same extent by HCl had no effect on secretion which also was the same in the absence of exogenous Mg2+ as in its presence. Similarly, secretion induced by collagen per se, i.e., isolated from autocrine agonists by inhibitors, which was about 50% less than with autocrine agonists, was not affected by the presence or absence of exogenous Mg2+. The level of tyrosine phosphorylation of several proteins in resting platelets was higher in the absence of Mg2+ than in its presence, but the onset of collagen-induced phosphorylation and dephosphorylation was more rapid in the absence of Mg2+. Tyrosine phosphorylation of p38 was specifically enhanced in the presence of inhibitors of autocrine phosphorylation and even more enhanced in the absence of Mg2+. EDTA inhibited the protein phosphorylation to the same extent in the presence as in the absence of exogenous Mg2+. These results show that EDTA inhibits collagen-induced dense granule secretion neither through chelation of Mg2+ nor lowering of pH, and thus probably through other effects on the platelet by the EDTA2- species that are reflected in protein tyrosine phosphorylation signalling.


Subject(s)
Adenosine Diphosphate/blood , Adenosine Triphosphate/blood , Blood Platelets/physiology , Collagen/pharmacology , Edetic Acid/pharmacology , Hydrogen-Ion Concentration , Magnesium/blood , Blood Platelets/drug effects , Chelating Agents/pharmacology , Collagen/antagonists & inhibitors , Humans , In Vitro Techniques , Kinetics , Phosphorylation , Phosphotyrosine/blood
10.
Am J Physiol Cell Physiol ; 280(5): C1045-56, 2001 May.
Article in English | MEDLINE | ID: mdl-11287316

ABSTRACT

Activated neutrophils display an array of physiological responses, including initiation of the oxidative burst, phagocytosis, and cell migration, that are associated with cellular adhesion. Under conditions that lead to cellular adhesion, we observed rapid tyrosine phosphorylation of an intracellular protein with an approximate relative molecular mass of 92 kDa (p92). Phosphorylation of p92 was inducible when Mac-1 was activated by phorbol 12-myristate 13-acetate, the beta(2)-specific activating antibody CBR LFA-1/2, or interleukin-8 (77 amino acids). In addition, tyrosine phosphorylation of p92 was dependent on engagement of Mac-1 with ligand. Several observations suggest that this event may be an important step in the signaling pathway initiated by Mac-1 binding. p92 phosphorylation was specifically blocked with antibodies to CD11b, the alpha-subunit of Mac-1, and was rapidly reversible on disengagement of the integrin ligand interaction. Integrin-stimulated phosphorylation of p92 created binding sites that were recognized in vitro by the SH2 domains of c-CrkII and Src. Our observations suggest that neutrophil adhesion mediated through the binding of the beta(2)-integrin Mac-1 initiates a signaling cascade that involves the activation of protein tyrosine kinases and leads to the regulation of protein-protein interactions via SH2 domains, a key process shared with growth factor signaling pathways.


Subject(s)
Cell Adhesion/physiology , Macrophage-1 Antigen/physiology , Neutrophil Activation/physiology , Neutrophils/physiology , Antibodies, Monoclonal/pharmacology , Cytochalasin B/pharmacology , Enzyme Inhibitors/pharmacology , Fibrinogen/physiology , Genistein/pharmacology , Glutathione Transferase/chemistry , Glutathione Transferase/metabolism , Humans , In Vitro Techniques , Interleukin-8/pharmacology , Kinetics , Macrophage-1 Antigen/drug effects , Macrophage-1 Antigen/immunology , Neutrophil Activation/drug effects , Neutrophils/drug effects , Phosphorylation , Phosphotyrosine/blood , Protein Kinase Inhibitors , Protein Kinases/metabolism , Recombinant Fusion Proteins/metabolism , Staurosporine/pharmacology , Tetradecanoylphorbol Acetate/pharmacology , Vanadates/pharmacology , src Homology Domains
11.
J Biol Chem ; 276(24): 20882-9, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11287424

ABSTRACT

Aggretin, a potent platelet activator, was isolated from Calloselasma rhodostoma venom, and 30-amino acid N-terminal sequences of both subunits were determined. Aggretin belongs to the heterodimeric snake C-type lectin family and is thought to activate platelets by binding to platelet glycoprotein alpha(2)beta(1). We now show that binding to glycoprotein (GP) Ib is also required. Aggretin-induced platelet activation was inhibited by a monoclonal antibody to GPIb as well as by antibodies to alpha(2)beta(1). Binding of both of these platelet receptors to aggretin was confirmed by affinity chromatography. No binding of other major platelet membrane glycoproteins, in particular GPVI, to aggretin was detected. Aggretin also activates platelets from Fc receptor gamma chain (Fcgamma)-deficient mice to a greater extent than those from normal control mice, showing that it does not use the GPVI/Fcgamma pathway. Platelets from Fcgamma-deficient mice expressed fibrinogen receptors normally in response to collagen, although they did not aggregate, indicating that these platelets may partly compensate via other receptors including alpha(2)beta(1) or GPIb for the lack of the Fcgamma pathway. Signaling by aggretin involves a dose-dependent lag phase followed by rapid tyrosine phosphorylation of a number of proteins. Among these are p72(SYK), p125(FAK), and PLCgamma2, whereas, in comparison with collagen and convulxin, the Fcgamma subunit neither is phosphorylated nor coprecipitates with p72(SYK). This supports an independent, GPIb- and integrin-based pathway for activation of p72(SYK) not involving the Fcgamma receptor.


Subject(s)
Blood Platelets/physiology , Enzyme Precursors/blood , Integrins/blood , Integrins/physiology , Isoenzymes/blood , Lectins, C-Type , Lectins/pharmacology , Platelet Activation/physiology , Platelet Glycoprotein GPIb-IX Complex/physiology , Protein-Tyrosine Kinases/blood , Type C Phospholipases/blood , Viper Venoms/chemistry , Viper Venoms/pharmacology , Agkistrodon , Amino Acid Sequence , Animals , Blood Platelets/drug effects , Chromatography, Affinity , Collagen/pharmacology , Crotalid Venoms/pharmacology , Humans , In Vitro Techniques , Intracellular Signaling Peptides and Proteins , Lectins/chemistry , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Phospholipase C gamma , Phosphorylation , Phosphotyrosine/blood , Platelet Activation/drug effects , Platelet Glycoprotein GPIb-IX Complex/drug effects , Protein Subunits , Receptors, Collagen , Sequence Alignment , Sequence Homology, Amino Acid , Syk Kinase , Viper Venoms/isolation & purification
12.
J Biol Chem ; 276(24): 21039-45, 2001 Jun 15.
Article in English | MEDLINE | ID: mdl-11274196

ABSTRACT

Cellular adhesion molecules such as E-selectin function to recruit leukocytes into the inflammatory lesions of diseases such as rheumatoid arthritis (RA) and atherosclerosis. Monocytes are the key components of the cellular infiltrates present in these disorders. We hypothesized that soluble E-selectin (sE-selectin) might mediate the chemotaxis of monocytes. In this report, we show that sE-selectin induced normal human peripheral blood monocyte migration in the nanomolar range in a concentration-dependent manner. Neutralization studies using RA human joint synovial fluids and anti-E-selectin antibody showed a mean 31% reduction in RA synovial fluid-mediated monocyte chemotaxis (p < 0.05), indicating that sE-selectin is a major monocyte recruiter in RA. Next, we investigated the role of tyrosine phosphorylation pathways in sE-selectin-induced monocyte chemotaxis. Human peripheral blood monocytes stimulated with sE-selectin showed a time-dependent increase in the tyrosine phosphorylation of a broad range of cellular proteins, predominantly in the molecular size range of Src family kinases (50-60 kDa) and mitogen-activated protein kinases (MAPKs). Western blot analysis of Src family kinases showed a time-dependent increase in Src, Hck, and Lyn phosphorylation. The pretreatment of monocytes with the Src inhibitor AG1879: 4-amino-5-(4-chlorophenyl)-7-(t-butyl)pyrazolol[3,4-d]pyrimidine (PP2) prior to stimulation with sE-selectin markedly inhibited Hck and Lyn phosphorylation, whereas the phosphorylation of Src was partially inhibited. In addition, the sE-selectin stimulation of monocytes resulted in the increased phosphorylation of extracellular signal-related kinase (ERK1/2) and p38 MAPK. The pretreatment of monocytes with PP2 showed 89 and 83% inhibition of ERK1/2 and p38 MAPK phosphorylation, respectively. sE-selectin also showed a time-dependent activation of Ras kinase. Furthermore, the pretreatment of monocytes with PP2 completely inhibited sE-selectin-mediated monocyte chemotaxis. Taken together, our data demonstrate a novel function for sE-selectin as a monocyte chemotactic agent and suggest that sE-selectin might be mediating its biological functions through the Src-MAPK pathway.


Subject(s)
Chemotaxis, Leukocyte/physiology , E-Selectin/pharmacology , Leukocytes, Mononuclear/physiology , Monocytes/physiology , src-Family Kinases/blood , Chemotaxis, Leukocyte/drug effects , E-Selectin/immunology , Enzyme Activation , Humans , Immunoglobulin G/pharmacology , In Vitro Techniques , Kinetics , Leukocytes, Mononuclear/drug effects , Mitogen-Activated Protein Kinases/blood , Monocytes/drug effects , Phosphorylation , Phosphotyrosine/blood , Recombinant Proteins/pharmacology , p38 Mitogen-Activated Protein Kinases
13.
Electrophoresis ; 21(13): 2622-36, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10949139

ABSTRACT

Different search programs were compared to judge their particular efficiency in protein identification. We established a human blood platelet protein map and identified tyrosine-phosphorylated proteins. The cytosolic fraction of human blood platelets was separated by two-dimensional polyacrylamide gel electrophoresis (2-D PAGE) and phosphorylated proteins were detected by Western blotting using anti-phosphotyrosine antibodies. Visualized protein spots were excised, digested with trypsin and analyzed by matrix assisted laser desorption/ionization-time of flight-mass spectrometry (MALDI-TOF-MS). The obtained mass fingerprint data sets have been analyzed using ProFound, MS-Fit and Mascot. For those protein spots with no significant search results MALDI post source decay (PSD) spectra have been acquired on the same sample. For automatic interpretation of these fragment ion spectra, the SEQUEST and Mascot algorithm were applied. Another approach for the identification of phosphorylated proteins is immunoprecipitation using an anti-phosphotyrosine antibody. A method for immunoprecipitation of tyrosine-phosphorylated peptides was optimized.


Subject(s)
Blood Platelets/chemistry , Blood Protein Electrophoresis/methods , Blood Proteins/isolation & purification , Electrophoresis, Gel, Two-Dimensional/methods , Phosphoproteins/blood , Phosphotyrosine/blood , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Adult , Algorithms , Blotting, Western , Cytosol/chemistry , Databases, Factual , Humans , Isoelectric Focusing , Peptide Fragments/analysis , Peptide Fragments/chemical synthesis , Peptide Mapping , Phosphorylation , Protein Processing, Post-Translational , Software
14.
J Cell Physiol ; 183(3): 314-20, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10797305

ABSTRACT

In this study, we investigated the activation of a new member of the focal adhesion kinase family of tyrosine kinases, the proline-rich tyrosine kinase, or PYK2, in platelets. We show that PYK2 is tyrosine phosphorylated and its activity is increased during early stages of platelet aggregation. This activation coincided with increased association of phosphatidylinositol (PI) 3-kinase and PYK2, as determined by both anti-PI 3-kinase and anti-PYK2 immunoprecipitates. However, under basal conditions, some association of PYK2 and PI 3-kinase was consistently observed, even though little or no tyrosine phosphorylated PYK2 could be detected. In addition, both increased PI 3-kinase activity and increased PYK2 activity could be detected in immunoprecipitates following thrombin stimulation. All of these events were unaffected by blocking platelet aggregation with arginine-glycine-aspartate-serine (RGDS) peptide, which interferes with binding of the platelet integrin alpha(IIb)beta(3) to fibrinogen. Neither was the activation of the PYK2 kinase activity affected by blocking PI 3-kinase activity. These results support a model in which PYK2 is associated with PI 3-kinase in unstimulated platelets and following activation of platelets, there is an increase in tyrosine phosphorylation of PYK2, increased PYK2 activity, and increased association of PYK2 with PI 3-kinase, which may contribute to the increase in PI 3-kinase activity. All of these were found to be early events independent of subsequent platelet aggregation.


Subject(s)
Blood Platelets/physiology , Phosphatidylinositol 3-Kinases/blood , Platelet Aggregation , Protein-Tyrosine Kinases/blood , Thrombin/physiology , Blood Platelets/drug effects , Blood Platelets/enzymology , Enzyme Activation , Focal Adhesion Kinase 2 , Humans , In Vitro Techniques , Oligopeptides/pharmacology , Phosphotyrosine/blood , Platelet Aggregation Inhibitors/pharmacology , Thrombin/pharmacology
15.
Blood ; 95(9): 2947-53, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10779444

ABSTRACT

Severe congenital neutropenia (SCN) or Kostmann syndrome is a disorder of myelopoiesis characterized by a maturation arrest at the stage of promyelocytes or myelocytes in bone marrow and absolute neutrophil counts less than 200/microL in peripheral blood. Treatment of these patients with granulocyte colony-stimulating factor (G-CSF) leads to a significant increase in circulating neutrophils and a reduction in infection-related events in more than 95% of the patients. To date, little is known regarding the underlying pathomechanism of SCN. G-CSF-induced neutrophils of patients with SCN are functionally defective (eg, chemotaxis, superoxide anion generation, Ca(++ )mobilization). Two guanosine triphosphatases (GTPases), Rac2 and RhoA, were described to be involved in many neutrophil functions. The expression of these GTPases and their regulation in patients' neutrophils were of interest. This study determined that the guanosine diphosphate (GDP)-dissociation inhibitor RhoGDI is overexpressed at the protein level in patients' neutrophils and that overexpression is a result of G-CSF treatment. RhoA and LyGDI are expressed at similar levels, whereas Rac2 shows a decreased expression. In addition, association of Rac2 and RhoGDI or LyGDI is abrogated or not detectable based on the low Rac2 expression in patients' neutrophils. (Blood. 2000;95:2947-2953)


Subject(s)
Guanine Nucleotide Dissociation Inhibitors/genetics , Neutropenia/blood , Neutropenia/genetics , Neutrophils/metabolism , Proteins/genetics , rac GTP-Binding Proteins/genetics , rhoA GTP-Binding Protein/genetics , Guanine Nucleotide Dissociation Inhibitors/blood , Humans , Neutropenia/congenital , Phosphotyrosine/blood , Proteins/metabolism , Reference Values , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Proteins , rac GTP-Binding Proteins/blood , rho GTP-Binding Proteins/blood , rho GTP-Binding Proteins/genetics , rho Guanine Nucleotide Dissociation Inhibitor alpha , rho Guanine Nucleotide Dissociation Inhibitor beta , rho-Specific Guanine Nucleotide Dissociation Inhibitors , rhoA GTP-Binding Protein/blood , RAC2 GTP-Binding Protein
16.
Biochem J ; 347 Pt 1: 183-92, 2000 Apr 01.
Article in English | MEDLINE | ID: mdl-10727417

ABSTRACT

We have investigated the mechanism of Ca(2+) entry into fura-2-loaded human platelets by preventing the prenylation of proteins such as small GTP-binding proteins. The farnesylcysteine analogues farnesylthioacetic acid (FTA) and N-acetyl-S-geranylgeranyl-L-cysteine (AGGC), which are inhibitors of the methylation of prenylated and geranylgeranylated proteins respectively, significantly decreased thrombin-evoked increases in intracellular free Ca(2+) concentration ([Ca(2+)](i)) in the presence, but not in the absence, of external Ca(2+), suggesting a relatively selective inhibition of Ca(2+) entry over internal release. Both these compounds and N-acetyl-S-farnesyl-L-cysteine, which had similar effects to those of FTA, also decreased Ca(2+) entry evoked by the depletion of intracellular Ca(2+) stores with thapsigargin. The inactive control N-acetyl-S-geranyl-L-cysteine was without effect. Patulin, an inhibitor of prenylation that is inert with respect to methyltransferases, also decreased store-regulated Ca(2+) entry. Cytochalasin D, an inhibitor of actin polymerization, significantly decreased store-regulated Ca(2+) entry in a time-dependent manner. Both cytochalasin D and the farnesylcysteine analogues FTA and AGGC inhibited actin polymerization; however, when evoking the same extent of decrease in actin filament formation, FTA and AGGC showed greater inhibitory effects on Ca(2+) entry, indicating a cytoskeleton-independent component in the regulation of Ca(2+) entry by small GTP-binding-protein. These findings suggest that prenylated proteins such as small GTP-binding proteins are involved in store-regulated Ca(2+) entry through actin cytoskeleton-dependent and cytoskeleton-independent mechanisms in human platelets.


Subject(s)
Actins/blood , Blood Platelets/physiology , Calcium/blood , Cysteine/analogs & derivatives , Cytoskeleton/physiology , Diterpenes/pharmacology , Farnesol/analogs & derivatives , Monomeric GTP-Binding Proteins/blood , Actins/drug effects , Blood Platelets/drug effects , Blood Platelets/ultrastructure , Cysteine/pharmacology , Cytochalasin D/pharmacology , Cytoskeleton/drug effects , Farnesol/pharmacology , Humans , In Vitro Techniques , Kinetics , Phosphotyrosine/blood , Protein Methyltransferases/antagonists & inhibitors , Protein Prenylation/drug effects , Thapsigargin/pharmacology , Thrombin/pharmacology
17.
Blood ; 95(4): 1199-206, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10666191

ABSTRACT

Previously, we demonstrated that B-chronic lymphocytic leukemia (B-CLL) cells could be divided into 2 groups depending on the expression of CD38 by the malignant cells. The 2 groups differed in their signal-transducing capacities initiated by cross-linking of surface IgM; only in CD38-positive cells was an efficient signal delivered, invariably resulting in cell apoptosis. In this study, we investigated the effect of surface IgD cross-linking in 10 patients with CD38-positive B-CLL. Exposure of the malignant cells to goat antihuman delta-chain antibodies (Gadelta-ab) caused [Ca(++)]i mobilization and tyrosine kinase phosphorylation in a manner not different from that observed after goat antihuman mu-chain antibody (Gamu-ab) treatment in vitro. However, Gadelta-ab-treated cells failed to undergo apoptosis and instead displayed prolonged survival in culture and differentiated into plasma cells when rIL2 was concomitantly present. Cross-linking of surface IgD failed to induce proliferation of the malignant cells in vitro. Moreover, treatment with Gadelta-ab did not prevent apoptosis of B-CLL cells induced by Gamu-ab. Collectively, these experiments demonstrated that IgM and IgD expressed by the same cell may deliver opposite signals under particular circumstances and provide some clues for the understanding of the pathophysiology of B-CLL. (Blood. 2000;95:1199-1206)


Subject(s)
Antigens, Differentiation/blood , B-Lymphocytes/immunology , Immunoglobulin D/blood , Immunoglobulin M/blood , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , NAD+ Nucleosidase/blood , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Antibodies/pharmacology , Antigens, CD/blood , Apoptosis , B-Lymphocytes/drug effects , B-Lymphocytes/pathology , Calcium/blood , Cell Cycle , Cell Differentiation , Cross-Linking Reagents , Flow Cytometry , Humans , Immunoglobulin gamma-Chains/blood , Lymphocyte Activation , Membrane Glycoproteins , Necrosis , Phosphotyrosine/blood , Recombinant Proteins/pharmacology , Signal Transduction , Tumor Cells, Cultured
18.
Arch Biochem Biophys ; 374(2): 356-62, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10666318

ABSTRACT

Platelet activation by collagen depends principally on two receptors, alpha(2)beta(1) integrin (GPIa-IIa) and GPVI. During this activation, the nonreceptor protein tyrosine kinase pp72(syk) is rapidly phosphorylated, but the precise contribution of alpha(2)beta(1) integrin and GPVI to signaling for this phosphorylation is not clear. We have recently found that proteolysis of platelet alpha(2)beta(1) integrin by the snake venom metalloproteinase, jararhagin, results in inhibition of collagen-induced platelet aggregation and pp72(syk) phosphorylation. In order to verify whether the treatment of platelets with jararhagin had any effect on GPVI signaling, in this study we stimulated platelets treated with either jararhagin or anti-alpha(2)beta(1) antibody with two GPVI agonists, an antibody to GPVI and convulxin. Platelet shape change and phosphorylation of pp72(syk) by both GPVI agonists was preserved, as was the structure and function of GPVI shown by (125)I-labeled convulxin binding to immunoprecipitated GPVI from jararhagin-treated platelets. In contrast, defective platelet aggregation in response to GPVI agonists occurred in both jararhagin-treated and alpha(2)beta(1)-blocked platelets. This apparent cosignaling role of alpha(2)beta(1) integrin for platelet aggregation suggests the possibility of a topographical association of this integrin with GPVI. We found that both platelet alpha(2)beta(1) integrin and GPVI coimmunoprecipitated with alpha(IIb)beta(3) integrin. Since platelet aggregation requires activation of alpha(IIb)beta(3) integrin, defective aggregation in the absence of alpha(2)beta(1) suggests that this receptor may provide a signaling link between GPVI and alpha(IIb)beta(3). Our study therefore demonstrates that platelet signaling leading to pp72(syk) phosphorylation initiated with GPVI engagement by either convulxin or GPVI antibody does not depend on alpha(2)beta(1) integrin. However, alpha(IIb)beta(3) integrin may, in this model, require functional alpha(2)beta(1) integrin for its activation.


Subject(s)
Blood Platelets/physiology , Integrins/physiology , Lectins, C-Type , Platelet Aggregation , Platelet Membrane Glycoproteins/physiology , Antibodies/pharmacology , Blood Platelets/cytology , Blood Platelets/drug effects , Cell Size/drug effects , Collagen/pharmacology , Crotalid Venoms/pharmacology , Enzyme Precursors/blood , Humans , In Vitro Techniques , Integrins/blood , Intracellular Signaling Peptides and Proteins , Metalloendopeptidases/pharmacology , Phosphotyrosine/blood , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Protein-Tyrosine Kinases/blood , Receptors, Collagen , Syk Kinase , Bothrops jararaca Venom
19.
J Biol Chem ; 274(31): 22072-80, 1999 Jul 30.
Article in English | MEDLINE | ID: mdl-10419535

ABSTRACT

Neutrophil-mediated organ damage is a common feature of many disease states. We previously demonstrated that resuscitation with hypertonic salt solutions prevented the endotoxin-induced leukosequestration and consequent lung injury, and this effect was partially attributed to an altered surface expression of adhesion molecules, CD11b and L-selectin. In this study we investigated the mechanisms whereby osmotic stress evokes L-selectin shedding. The metalloprotease inhibitor RO 31-9790 prevented the osmotic down-regulation of L-selectin, indicating that this process was catalyzed by the same "sheddase" responsible for L-selectin cleavage induced by diverse inflammatory stimuli. The trigger for hypertonic shedding was cell shrinkage and not increased osmolarity, ionic strength, or intracellular pH. Volume reduction caused robust tyrosine phosphorylation and its inhibition by genistein and erbstatin abrogated shedding. Shrinkage stimulated tyrosine kinases Hck, Syk, and Pyk2, but prevention of their activation by the Src-family inhibitor PP1 failed to affect the L-selectin response. Hypertonicity elicited the Src family-independent activation of p38, and the inhibition of this kinase by SB203580 strongly reduced shedding. p38 was also essential for the N-formyl-methionyl-leucyl-phenylalanine- and lipopolysaccharide-induced shedding but not the phorbol ester-induced shedding. Thus, cell volume regulates L-selectin surface expression in a p38-mediated, metalloprotease-dependent manner. Moreover, p38 has a central role in shedding induced by many inflammatory mediators.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/blood , Hydroxamic Acids , L-Selectin/blood , Mitogen-Activated Protein Kinases , Neutrophils/cytology , Neutrophils/physiology , Cell Size/drug effects , Cytosol/physiology , Enzyme Inhibitors/pharmacology , Genistein/pharmacology , Humans , Hydrogen-Ion Concentration , Hypertonic Solutions , Imidazoles/pharmacology , In Vitro Techniques , L-Selectin/genetics , Macrophage-1 Antigen/blood , Metalloendopeptidases/antagonists & inhibitors , Neutrophils/drug effects , Phosphotyrosine/blood , Protease Inhibitors/pharmacology , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases
20.
Biochem Biophys Res Commun ; 261(1): 58-63, 1999 Jul 22.
Article in English | MEDLINE | ID: mdl-10405323

ABSTRACT

Thrombopoietin (TPO) plays a crucial role in megakaryocyte differentiation and platelet production. c-Mpl, a receptor for TPO, is also expressed in terminally differentiated platelets. We investigated the effects of TPO on activation of p38 mitogen-activated protein kinase in human platelets. Thrombin, a thrombin receptor agonist peptide, a thromboxane A(2) analogue, collagen, crosslinking the glycoprotein VI, ADP, and epinephrine, but not phorbol 12, 13-dibutyrate activated p38. TPO did not activate p38 by itself, whereas TPO pretreatment potentiated the agonist-induced activation of p38. TPO did not promote phosphorylation of Hsp27 and cytosolic phospholipase A(2) by itself, but enhanced thrombin-induced phosphorylation of them. The specific p38 inhibitor SB203580 strongly inhibited such phosphorylation. Thus, TPO possesses the priming effect on p38 activation in human platelets and could affect platelet functions through the p38 pathway.


Subject(s)
Blood Platelets/enzymology , Calcium-Calmodulin-Dependent Protein Kinases/blood , Heat-Shock Proteins , Mitogen-Activated Protein Kinases , Receptors, Thrombin/agonists , Thrombopoietin/pharmacology , Blood Platelets/drug effects , Calcium-Calmodulin-Dependent Protein Kinases/antagonists & inhibitors , Drug Synergism , Enzyme Activation/drug effects , HSP27 Heat-Shock Proteins , Humans , Imidazoles/pharmacology , Molecular Chaperones , Neoplasm Proteins/blood , Phospholipases A/blood , Phosphorylation/drug effects , Phosphotyrosine/blood , Pyridines/pharmacology , Receptors, Thrombin/physiology , Thrombin/agonists , Thrombin/antagonists & inhibitors , Thrombin/pharmacology , p38 Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL
...