Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 138
Filter
1.
Inverse Probl Imaging (Springfield) ; 15(5): 1135-1169, 2021 Oct.
Article in English | MEDLINE | ID: mdl-35173824

ABSTRACT

The first numerical implementation of a t exp method in 3D using simulated electrode data is presented. Results are compared to Calderón's method as well as more common TV and smoothness regularization-based methods. The t exp method for EIT is based on tailor-made non-linear Fourier transforms involving the measured current and voltage data. Low-pass filtering in the non-linear Fourier domain is used to stabilize the reconstruction process. In 2D, t exp methods have shown great promise for providing robust real-time absolute and time-difference conductivity reconstructions but have yet to be used on practical electrode data in 3D, until now. Results are presented for simulated data for conductivity and permittivity with disjoint non-radially symmetric targets on spherical domains and noisy voltage data. The 3D t exp and Calderón methods are demonstrated to provide comparable quality to their 2D counterparts, and hold promise for real-time reconstructions due to their fast, non-optimized, computational cost.

2.
J Thromb Haemost ; 16(12): 2501-2514, 2018 12.
Article in English | MEDLINE | ID: mdl-30347494

ABSTRACT

Essentials The rs773902 SNP results in differences in platelet protease-activated receptor (PAR4) function. The functional consequences of rs773902 were analyzed in human platelets and stroke patients. rs773902 affects thrombin-induced platelet function, PAR4 desensitization, stroke association. Enhanced PAR4 Thr120 effects on platelet function are blocked by ticagrelor. SUMMARY: Background F2RL3 encodes protease-activated receptor (PAR) 4 and harbors an A/G single-nucleotide polymorphism (SNP) (rs773902) with racially dimorphic allelic frequencies. This SNP mediates an alanine to threonine substitution at residue 120 that alters platelet PAR4 activation by the artificial PAR4-activation peptide (PAR4-AP) AYPGKF. Objectives To determine the functional effects of rs773902 on stimulation by a physiological agonist, thrombin, and on antiplatelet antagonist activity. Methods Healthy human donors were screened and genotyped for rs773902. Platelet function in response to thrombin was assessed without and with antiplatelet antagonists. The association of rs773902 alleles with stroke was assessed in the Stroke Genetics Network study. Results As compared with rs773902 GG donors, platelets from rs773902 AA donors had increased aggregation in response to subnanomolar concentrations of thrombin, increased granule secretion, and decreased sensitivity to PAR4 desensitization. In the presence of PAR1 blockade, this genotype effect was abolished by higher concentrations of or longer exposure to thrombin. We were unable to detect a genotype effect on thrombin-induced PAR4 cleavage, dimerization, and lipid raft localization; however, rs773902 AA platelets required a three-fold higher level of PAR4-AP for receptor desensitization. Ticagrelor, but not vorapaxar, abolished the PAR4 variant effect on thrombin-induced platelet aggregation. A significant association of modest effect was detected between the rs773902 A allele and stroke. Conclusion The F2RL3 rs773902 SNP alters platelet reactivity to thrombin; the allelic effect requires P2Y12 , and is not affected by gender. Ticagrelor blocks the enhanced reactivity of rs773902 A platelets. PAR4 encoded by the rs773902 A allele is relatively resistant to desensitization and may contribute to stroke risk.


Subject(s)
Blood Platelets/drug effects , Pharmacogenomic Variants , Platelet Aggregation Inhibitors/pharmacology , Polymorphism, Single Nucleotide , Purinergic P2Y Receptor Antagonists/pharmacology , Receptors, Purinergic P2Y12/drug effects , Receptors, Thrombin/agonists , Receptors, Thrombin/genetics , Thrombin/pharmacology , Ticagrelor/pharmacology , Adult , Animals , Blood Platelets/metabolism , COS Cells , Chlorocebus aethiops , Drug Interactions , Female , HEK293 Cells , Humans , Male , Middle Aged , Platelet Aggregation/drug effects , Receptors, Purinergic P2Y12/metabolism , Receptors, Thrombin/metabolism , Risk Factors , Stroke/blood , Stroke/genetics , Young Adult
3.
Clin Pharmacol Ther ; 99(1): 72-81, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26501993

ABSTRACT

Platelet activation and thrombus formation play a central role in ischemic vascular disease. Thrombin, an especially potent physiologic agonist mediating in vivo activation of platelets, acts via a unique family of G-protein-coupled receptors called protease-activated receptors (PARs) with a broad tissue expression. This review focuses on current antiplatelet therapies as well as innovative approaches to targeting PARs in patients with atherothrombotic vascular disease.


Subject(s)
Blood Platelets/drug effects , Platelet Aggregation Inhibitors/therapeutic use , Protease Inhibitors/pharmacology , Protease Inhibitors/therapeutic use , Receptors, Thrombin/antagonists & inhibitors , Vascular Diseases/drug therapy , Drugs, Investigational/pharmacology , Drugs, Investigational/therapeutic use , Humans , Models, Biological , Platelet Aggregation Inhibitors/pharmacology
4.
J Thromb Haemost ; 11 Suppl 1: 340-50, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23809137

ABSTRACT

Recent work by the Encyclopedia of DNA Elements project showed that non-protein-coding RNAs account for an unexpectedly large proportion of the human genome. Among these non-coding RNAs are microRNAs (miRNAs), which are small RNA molecules that modulate protein expression by degrading mRNA or repressing mRNA translation. MiRNAs have been shown to play important roles in hematopoiesis including embryonic stem cell differentiation, erythropoiesis, granulocytopoiesis/monocytopoiesis, lymphopoiesis, and megakaryocytopoiesis. Additionally, disordered miRNA biogenesis and quantitative or qualitative alterations in miRNAs and their targets are associated with hematological pathologies. Platelets contain machinery to process pre-miRNAs into mature miRNAs, and specific platelet miRNA levels have been found to correlate with platelet reactivity. This review summarizes the current state of knowledge of miRNAs in megakaryocytes and platelets, and the exciting possibilities for future megakaryocyte-platelet transcriptome research.


Subject(s)
Blood Platelets/cytology , MicroRNAs/metabolism , Platelet Activation , Blood Platelets/metabolism , Humans
5.
J Thromb Haemost ; 8(11): 2377-84, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20723028

ABSTRACT

BACKGROUND: Contradictory results have been published on the effects of T13254C (rs1613662), which distinguishes the two major isoforms of GP6, the gene encoding the platelet receptor glycoprotein VI, on platelet function and the risk of cardiovascular disease. METHODS: We performed a population-based case-control study, the Study of Myocardial Infarctions in Leiden, among 547 male patients with a first myocardial infarction (MI) and 646 control subjects, as well as a prospective cohort study in which the same MI patients were followed for recurrent events (fatal and non-fatal MI and unstable angina) and mortality (median follow-up of 12 years). P-selectin expression by platelets induced by crosslinked collagen-related peptide (CRP-XL) was measured by whole blood flow cytometry in 274 MI patients. RESULTS: T13254C was not associated with a first MI, but seemed to be associated with a reduced incidence of recurrent events [per-allele hazard ratio 0.77, 95% confidence interval (CI) 0.56-1.06] and mortality (hazard ratio 0.57, 95% CI 0.37-0.89). Pooling with the Heart and Estrogen/Progestin Replacement Study revealed hazard ratios of 0.81 (95% CI 0.66-0.99) and 0.73 (95% CI 0.55-0.96). The minor C-allele was also strongly associated with a reduced percentage of P-selectin-expressing platelets. The reduction per C-allele was 23% (95% CI 18-28%). In an independent study of 219 healthy volunteers, the per-allele reduction of CRP-XL-induced aggregation was 10% (95% CI 2-18%). CONCLUSION: The minor allele of GP6 T13254C that reduced platelet activation and aggregation also seemed to be associated with a reduced incidence of recurrent cardiovascular events and mortality, but was not associated with first MI.


Subject(s)
Blood Platelets/cytology , Cardiovascular Diseases/genetics , Platelet Activation/genetics , Platelet Membrane Glycoproteins/genetics , Aged , Alleles , Cardiovascular Diseases/epidemiology , Case-Control Studies , Cohort Studies , Female , Humans , Incidence , Male , Middle Aged , P-Selectin/blood , Polymorphism, Genetic , Proportional Hazards Models , Protein Isoforms , Recurrence
6.
J Thromb Haemost ; 8(2): 369-78, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19943878

ABSTRACT

BACKGROUND: Variation in platelet reactivity contributes to disorders of hemostasis and thrombosis, but the molecular mechanisms are not well understood. OBJECTIVES: To discover associations between interindividual platelet variability and the responsible platelet genes, and to begin to define the molecular mechanisms altering platelet gene expression. SUBJECTS/METHODS: Two hundred and eighty-eight healthy subjects were phenotyped for platelet responsiveness. Platelet RNA from subjects demonstrating hyperreactivity (n=18) and hyporeactivity (n=11) was used to screen the human transcriptome. RESULTS: Distinctly different mRNA profiles were observed between subjects with differing platelet reactivity. Increased levels of mRNA for VAMP8/endobrevin, a critical v-SNARE involved in platelet granule secretion, were associated with platelet hyperreactivity (Q=0.0275). Validation studies of microarray results showed 4.8-fold higher mean VAMP8 mRNA levels in hyperreactive than hyporeactive platelets (P=0.0023). VAMP8 protein levels varied 13-fold among platelets from these normal subjects, and were 2.5-fold higher in hyperreactive platelets (P=0.05). Among our cohort of 288 subjects, a VAMP8 single-nucleotide polymorphism (rs1010) was associated with platelet reactivity in an age-dependent manner (P<0.003). MicroRNA-96 was predicted to bind to the 3'-untranslated regionof VAMP8 mRNA and was detected in platelets. Overexpression of microRNA-96 in VAMP8-expressing cell lines caused a dose-dependent decrease in VAMP8 protein and mRNA, suggesting a role in VAMP8 mRNA degradation. CONCLUSIONS: These findings support a role for VAMP8/endobrevin in the heterogeneity of platelet reactivity, and suggest a role for microRNA-96 in the regulation of VAMP8 expression.


Subject(s)
Blood Platelets/metabolism , MicroRNAs/blood , Platelet Aggregation/genetics , Polymorphism, Single Nucleotide , R-SNARE Proteins/genetics , 3' Untranslated Regions , Adult , Age Factors , Binding Sites , Epinephrine , Female , Gene Expression Profiling/methods , Genotype , HCT116 Cells , Humans , Male , Oligonucleotide Array Sequence Analysis , Phenotype , R-SNARE Proteins/blood , RNA, Messenger/blood , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Up-Regulation , Young Adult
7.
Pediatr Blood Cancer ; 51(1): 86-92, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18253960

ABSTRACT

BACKGROUND: Although "aspirin resistance" (AR-inadequate platelet inhibition as suggested by in vitro testing of aspirin-treated patients) has been widely studied in adults and linked to increased risk of adverse outcomes, its prevalence and clinical significance are largely unknown in children. PROCEDURE: To determine AR prevalence in children and its relationship to assay methodology, we undertook a cross-sectional study of 44 children (1-17 years, 24 male) on aspirin for various indications and considered three published definitions of AR in adults: platelet aggregation >/=70% to 10 microM adenosine diphosphate and >/=20% to 0.5 mg/ml arachidonic acid (AA), normal PFA-100(R) closure time and elevated urinary 11-dehydro thromboxane B(2) (11dhTxB(2)) concentration. RESULTS: Six subjects exhibited AR according to at least one of the criteria (5 by PFA-100(R), 1 by aggregometry and 11dhTxB(2) criteria); nearly all subjects had low levels of 11dhTxB(2) compared with controls. Subjects studied prior to therapy showed pronounced changes in AR parameters after aspirin dosing (e.g., mean aggregation to AA decreased from 82% to 6%, P < 0.001), confirming an aspirin effect. Subjects with AR did not differ from aspirin responsive subjects in terms of age, race, platelet count, or aspirin dose, indication or therapy duration. There was minimal correlation between assays. CONCLUSIONS: In this initial prevalence study of a clinically diverse group of pediatric patients, frequencies of AR were assay-dependent; however, the prevalence of true AR is likely low in children (2.3%; 95% CI 0.1-10.7%), in agreement with adult studies. To better define the clinical relevance of AR in children, multicenter, prospective cohort studies are imperative.


Subject(s)
Aspirin , Drug Resistance , Adolescent , Aspirin/pharmacology , Aspirin/therapeutic use , Child , Child, Preschool , Cross-Sectional Studies , Epidemiologic Measurements , Female , Humans , Infant , Male , Platelet Aggregation/drug effects , Prevalence , Risk Factors
9.
J Thromb Haemost ; 5(8): 1617-23, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17663734

ABSTRACT

BACKGROUND: Variations in platelet function among individuals may be related to differences in platelet-related genes. The major goal of our study was to estimate the contribution of inheritance to the variability in platelet function in unaffected individuals from white and African American families with premature coronary artery disease. METHODS: Platelet reactivity, in the absence of antiplatelet agents, was assessed by in vitro aggregation and the platelet function analyzer closure time. Heritability was estimated using a variance components model. RESULTS: Both white (n = 687) and African American (n = 321) subjects exhibited moderate to strong heritability (h(2)) for epinephrine- and adenosine diphosphate-induced aggregation (0.36-0.42 for white and >0.71 for African American subjects), but heritability for collagen-induced platelet aggregation in platelet-rich plasma was prominent only in African American subjects. Platelet lag phase after collagen stimulation was heritable in both groups (0.47-0.50). A limited genotype analysis demonstrated that the C825T polymorphism of GNB3 was associated with the platelet aggregation response to 2 muM epinephrine, but the effect differed by race. CONCLUSIONS: Considering the few and modest genetic effects reported to affect platelet function, our findings suggest the likely existence of undiscovered important genes that modify platelet reactivity, some of which affect multiple aspects of platelet biology.


Subject(s)
Blood Platelets/physiology , Coronary Artery Disease/blood , Adult , Coronary Artery Disease/complications , Coronary Artery Disease/ethnology , Family Health , Female , Fibrinogen/metabolism , Genotype , Humans , Male , Middle Aged , Platelet Aggregation , Polymorphism, Genetic , Thrombosis/complications , Thrombosis/diagnosis , Thromboxane B2/blood , von Willebrand Factor/metabolism
10.
J Thromb Haemost ; 4(9): 2043-50, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16961612

ABSTRACT

BACKGROUND: Although platelet hyperreactivity constitutes an important cardiovascular risk factor, standardized methods for its measurement are lacking. We recently reported that aggregometry using a submaximal concentration of epinephrine identifies individuals with in vitro platelet hyperreactivity; this hyperreactivity was reproducible on multiple occasions over long periods of time. OBJECTIVE AND METHODS: To better understand this aberrant reactivity, we studied in a large group of subjects (n = 386) the relationship between healthy individuals' platelet reactivity to epinephrine and their platelet phenotype as measured by other functional assays. RESULTS: Subjects with hyperreactivity to epinephrine were more likely to exhibit hyperfunction in each major aspect of platelet activity, including adhesion (response to low-dose ristocetin; P < 0.001), activation (surface P-selectin expression and PAC-1 binding after stimulation; P

Subject(s)
Blood Platelet Disorders/genetics , GTP-Binding Protein beta Subunits/genetics , Platelet Activation/genetics , Platelet Activation/physiology , Adult , Epinephrine/pharmacology , Female , Genotype , Humans , Male , Middle Aged , Platelet Adhesiveness/drug effects , Platelet Adhesiveness/genetics , Platelet Aggregation/drug effects , Platelet Aggregation/genetics , Polymorphism, Genetic
12.
J Thromb Haemost ; 1(2): 355-62, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12871511

ABSTRACT

The His131Arg polymorphism of platelet FcgammaRIIA affects the binding affinity of certain IgG subclasses. The Arg131 allele has been associated with (auto)immune thrombocytopenia and heparin-induced thrombocytopenia in some studies. Because FcgammaRIIA can transmit platelet activation signals, we studied platelet responsiveness from 73 healthy donors to determine if this polymorphism modulated platelet function. Platelet function was studied by agonist and shear-induced activation, and standard aggregation. FcgammaRIIA was genotyped by allele-specific PCR. Compared with His131, the Arg131 allele was associated with significantly greater binding of activation-dependent antibodies. This effect was most prominent for the receptor-induced binding site (RIBS) antibodies F26 (P < 0.0001) and RIBS1 (P = 0.0057), and the ligand-induced binding site antibody LIBS1 (P = 0.0367). Unexpectedly, Arg131-positive platelets did not show greater fibrinogen binding, platelet aggregation or shear-induced platelet activation. We considered whether enhanced Fc binding and FcgammaRIIA cross-linking were responsible for those discrepancies. The increased binding of the two RIBS antibodies to the Arg131 isoform was abolished by blocking FcgammaRIIA, and the FcgammaRIIA genotype effect on F26 IgG binding was lost when F26 F(ab')2 fragments were used. Furthermore, intact F26 and RIBS1 IgG directly and specifically induced P-selectin expression, and this effect was greatest in Arg131-positive platelets. We concluded that (a) the His131Arg polymorphism of FcgammaRIIA does not affect intrinsic platelet reactivity; (b) RIBS antibodies are able to cross-link FcgammaRIIA and activate platelets, and this activation has a modest effect on Arg131 platelets; and (c) flow cytometric based platelet assays may need to compensate for this FcgammaRIIA His131Arg effect on platelet activation.


Subject(s)
Antigens, CD/genetics , Antigens, CD/physiology , Blood Platelets/immunology , Blood Platelets/physiology , Polymorphism, Genetic , Receptors, IgG/genetics , Receptors, IgG/physiology , Adult , Antibodies, Monoclonal/pharmacology , Base Sequence , DNA/genetics , Female , Fibrinogen/immunology , Fibrinogen/physiology , Genotype , Humans , Immunoglobulin G/metabolism , In Vitro Techniques , Male , Platelet Activation/genetics , Platelet Activation/immunology , Platelet Activation/physiology , Platelet Aggregation/genetics , Platelet Aggregation/immunology , Platelet Aggregation/physiology
13.
Am J Cardiol ; 88(4): 347-52, 2001 Aug 15.
Article in English | MEDLINE | ID: mdl-11545752

ABSTRACT

Chromosome 17q21-23 harbors genes for platelet glycoprotein IIIa and angiotensin-converting enzyme (ACE), which are polymorphic for alleles Pl(A2) and ACE "D." These alleles have been independently and often associated with ischemic coronary artery disease (CAD). We sought to determine if the Pl(A2) and ACE D polymorphisms were risk factors for recurrent coronary events. In the Cholesterol And Recurrent Events (CARE) trial, 4,159 men and women with documented myocardial infarction (MI) were randomized to receive either placebo or pravastatin, and were followed prospectively for 5 years. Pl(A) and ACE genotypes were determined in 767 patients: 385 cases who had experienced a recurrent primary event (death due to coronary disease or nonfatal MI), and 382 age- and gender-matched controls. In patients receiving placebo, the Pl(A1,A2) genotype conferred a relative risk (RR) of 1.38 (confidence intervals [CI] 1.04 to 1.83; p = 0.028; adjusted RR = 1.32, CI = 0.99 to 1.76; p = 0.058]) for the primary end point. Compared with the placebo group, pravastatin reduced the excess RR of coronary disease death and recurrent MI in the Pl(A1,A2) patient population by 31% (p = 0.06). The ACE D allele appeared to have modestly additive effects on the Pl(A1,A2) risk. Among the Pl(A1,A2) patients, pravastatin had little effect on the risk of recurrent events with the ACE II genotype, but reduced the adjusted RR from 1.42 (placebo) to 0.58 for ACE ID patients, and from 1.56 (placebo) to 0.83 for ACE DD. The Pl(A1,A2) genotype was associated with an excess of recurrent coronary events in patients after MI who did not receive pravastatin, and the ACE D allele added to this risk. These data suggest that it would be important to perform a larger study to address the potential role of these genotypes in therapeutic decision making.


Subject(s)
Myocardial Infarction/genetics , Peptidyl-Dipeptidase A/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Aged , Alleles , Anticholesteremic Agents/therapeutic use , Female , Gene Deletion , Genotype , Humans , Male , Middle Aged , Multicenter Studies as Topic , Polymorphism, Genetic , Pravastatin/therapeutic use , Protein Isoforms , Randomized Controlled Trials as Topic , Recurrence , Risk Factors
14.
Transplantation ; 72(2): 233-7, 2001 Jul 27.
Article in English | MEDLINE | ID: mdl-11477344

ABSTRACT

BACKGROUND: Acute cellular rejection in cardiac allografts is a major cause of graft loss, and is associated with activation of the coagulation system. We investigated whether plasma markers of coagulation predict the presence of allograft rejection. METHODS: A total of 132 blood specimens and endomyocardial biopsies were collected from 35 patients, between February of 1997 and May of 1998. We measured plasma prothrombin fragment 1.2 (PF1.2) and p-selectin, fibrinogen, thrombomodulin, and d-dimer. Biopsies were graded according to the International Society of Heart and Lung Transplantation system, with a range of 0 to 4. Grades 0 and 1A were grouped as "no rejection," and the higher grades as "rejection." Linear and logistic regression, accounting for longitudinal data, were the principal analytic tools. RESULTS: p-Selectin level increased progressively with increasing rejection grade (P<0.001). With multivariate analysis, both p-selectin and prothrombin fragment levels significantly predicted rejection. p-Selectin levels were predictive of prothrombin fragment levels (P<0.0001) but not of d-dimer, fibrinogen, or thrombomodulin levels. This model allowed correct prediction of rejection, based on p-selectin and prothrombin fragment values, up to 85% of the time. Dichotomizing patients by a p-selectin level of 65 ng/ml resulted in an odds of rejection of 21.4 [95% C.I. 7.1-64.7] for the patients in the high- compared with the lower risk group. CONCLUSIONS: In heart transplant recipients, p-selectin levels and PF 1.2 levels are highly predictive of organ rejection. The elevation of PF 1.2 suggests that there is systemic generation of thrombin generation. These markers may be useful for noninvasively monitoring patients for organ rejection or for after response to treatment.


Subject(s)
Blood Coagulation , Graft Rejection/epidemiology , Heart Transplantation/physiology , P-Selectin/blood , Peptide Fragments/analysis , Prothrombin/analysis , Biomarkers/blood , Graft Rejection/blood , Heart Transplantation/immunology , Humans , Predictive Value of Tests , Probability , Prognosis , Regression Analysis , Thrombomodulin/analysis , Time Factors
15.
Exp Biol Med (Maywood) ; 226(5): 409-19, 2001 May.
Article in English | MEDLINE | ID: mdl-11393168

ABSTRACT

Coronary artery disease is a leading cause of death worldwide and the largest killer of men and women in the United States. The pathophysiology of myocardial infarction is multifactorial, and numerous physiologic systems converge to dictate the formation of the two fundamental lesions, thrombosis and atherosclerosis. In this review we address genetic aspects of arterial thrombosis and the key thrombotic factors that have been associated with the increased risk for its development. Specifically, we consider components of coagulation, fibrinolysis, and platelet adhesive receptors, and we review the genetic epidemiology and in vitro laboratory data regarding their risk for the acute coronary syndromes. In combination with traditional risk factor assessment, in the near future these inherited markers can be used to manage patients with vascular disease through a better utilization of invasive or expensive diagnostic testing, as well as pharmacologic intervention.


Subject(s)
Coronary Thrombosis/genetics , Blood Coagulation Factors/genetics , Coronary Thrombosis/blood , Coronary Thrombosis/etiology , Female , Humans , Inflammation/etiology , Male , Mutation , Myocardial Infarction/etiology , Platelet Membrane Glycoproteins/genetics , Polymorphism, Genetic , Risk Factors
16.
Mol Cell Biol ; 21(9): 3192-205, 2001 May.
Article in English | MEDLINE | ID: mdl-11287623

ABSTRACT

Alterations in the expression of integrin receptors for extracellular matrix (ECM) proteins are strongly associated with the acquisition of invasive and/or metastatic properties by human cancer cells. Despite this, comparatively little is known of the biochemical mechanisms that regulate the expression of integrin genes in cells. Here we demonstrate that the Ras-activated Raf-MEK-extracellular signal-regulated kinase (ERK) signaling pathway can specifically control the expression of individual integrin subunits in a variety of human and mouse cell lines. Pharmacological inhibition of MEK1 in a number of human melanoma and pancreatic carcinoma cell lines led to reduced cell surface expression of alpha6- and beta3-integrin. Consistent with this, conditional activation of the Raf-MEK-ERK pathway in NIH 3T3 cells led to a 5 to 20-fold induction of cell surface alpha6- and beta3-integrin expression. Induced beta3-integrin was expressed on the cell surface as a heterodimer with alphav-integrin; however, the overall level of alphav-integrin expression was not altered by Ras or Raf. Raf-induced beta3-integrin was observed in primary and established mouse fibroblast lines and in mouse and human endothelial cells. Consistent with previous reports of the ability of the Raf-MEK-ERK signaling pathway to induce beta3-integrin gene transcription in human K-562 erythroleukemia cells, Raf activation in NIH 3T3 cells led to elevated beta3-integrin mRNA. However, unlike immediate-early Raf targets such as heparin binding epidermal growth factor and Mdm2, beta3-integrin mRNA was induced by Raf in a manner that was cycloheximide sensitive. Surprisingly, activation of the Raf-MEK-ERK signaling pathway by growth factors and mitogens had little or no effect on beta3-integrin expression, suggesting that the expression of this gene requires sustained activation of this signaling pathway. In addition, despite the robust induction of cell surface alphavbeta3-integrin expression by Raf in NIH 3T3 cells, such cells display decreased spreading and adhesion, with a loss of focal adhesions and actin stress fibers. These data suggest that oncogene-induced alterations in integrin gene expression may participate in the changes in cell adhesion and migration that accompany the process of oncogenic transformation.


Subject(s)
Antigens, CD/genetics , Gene Expression Regulation , MAP Kinase Signaling System , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Platelet Membrane Glycoproteins/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-raf/metabolism , ras Proteins/metabolism , 3T3 Cells , Animals , Cell Membrane/metabolism , Dimerization , Enzyme Activation , Humans , Integrin beta3 , K562 Cells , MAP Kinase Kinase 1 , Mice , Mitogen-Activated Protein Kinase Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-raf/genetics , Receptors, Vitronectin
17.
Circ Res ; 88(4): 438-42, 2001 Mar 02.
Article in English | MEDLINE | ID: mdl-11230112

ABSTRACT

Platelets play an important role in the coronary thrombus formation that leads to myocardial ischemia and infarction. Gender differences in the development of coronary heart disease and its outcomes are partly regulated by estrogen and its receptors, but the roles of the latter in thrombogenicity are less well-defined. We previously demonstrated the presence of estrogen receptor (ER) beta in cells of the megakaryocytic lineage. In this study, we characterize human platelet ERbeta and its expression using biochemical and molecular biological techniques. Western immunoblotting showed that platelet ERbeta migrated with an apparent molecular mass approximately 3.7 kDa larger than ERbeta in a variety of cell lines (including those of prostate and breast origin). A rigorous investigation of platelet ERbeta mRNA by reverse transcriptase-polymerase chain reaction revealed normal transcripts and a single alternately spliced mRNA. However, this variant form was smaller, lacking exon 2, and could not account for the larger protein size seen in platelets. Treatment of ERbeta with N-glycosidase F, which removes core carbohydrate residues, caused a more rapid migration through polyacrylamide gels but had no effect on ERbeta from human cell lines. We conclude that the larger form of ERbeta in human platelets is not attributable to alternate mRNA splicing but primarily to tissue-specific glycosylation.


Subject(s)
Blood Platelets/chemistry , Receptors, Estrogen/blood , Adult , Animals , CHO Cells , Coronary Artery Disease/etiology , Cricetinae , Electrophoresis, Polyacrylamide Gel , Estrogen Receptor beta , Female , Glycosylation , Humans , Male , Middle Aged , RNA Splicing , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured
18.
Arch Pathol Lab Med ; 125(1): 112-5, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11151063

ABSTRACT

BACKGROUND: Although estrogen has been shown to contribute to retardation of the development of coronary heart disease in premenopausal women, the efficacy of hormone replacement therapy for coronary heart disease prevention in women with established coronary heart disease remains controversial. Hence, additional research is needed to clarify the effects of hormone replacement therapy on the cardiovascular and clotting systems. We investigated the effect of estrogen on platelet aggregation induced by standard agonists (epinephrine and adenosine diphosphate), with and without the platelet antagonist aspirin. Furthermore, we analyzed our data according to the presence or absence of a prevalent polymorphism of the glycoprotein (GP) IIIa subunit of the platelet fibrinogen receptor GPIIb-IIIa, PlA2. METHODS AND RESULTS: The effect of estrogen on aggregation of platelets was studied in healthy men (n = 20, 10 PlA1/A1 and 10 PlA1/A2) and premenopausal healthy women (n = 10, 5 PlA1/A1 and 5 PlA1/A2). The PlA1/A1 and PlA1/A2 individuals were matched for age and race. Platelet response to agonists was investigated in the presence of (1) estrogen (10(-11) to 10(-8) mol/L), (2) aspirin (0.056 to 56 micromol/L), (3) estrogen plus aspirin, and (4) estrogen plus ICI 182 780 (ICI, 10(-9) mol/L, an inhibitor of the estrogen receptor). We found that physiologic concentrations of estrogen strongly and significantly inhibited the aggregation of PlA1/A2 platelets (P<.005 for epinephrine and P<.05 for adenosine diphosphate, induced aggregation, respectively) in both men and women. Concentrations of estrogen that were 1000-fold greater were required to observe the same level of inhibition with PlA1/A1 platelets. In the presence of aspirin, estrogen failed to provide additional inhibitory effect on aggregation of both PlA1/A1 and PlA1/A2 platelets. The estrogen-specific inhibitor ICI blocked the effect of estrogen on aggregation, suggesting that this effect is mediated by the estrogen receptor. CONCLUSIONS: Estrogen inhibits the aggregation of platelets, but such inhibition is highly dependent on the presence or absence of the PlA2 polymorphism of GPIIIa. However, in the presence of aspirin, the inhibitory effect of estrogen on aggregation was no longer detectable.


Subject(s)
Estradiol/analogs & derivatives , Estradiol/pharmacology , Platelet Aggregation/drug effects , Platelet Aggregation/genetics , Platelet Glycoprotein GPIIb-IIIa Complex/genetics , Polymorphism, Genetic , Adult , Coronary Disease/prevention & control , Epinephrine/pharmacology , Estrogen Replacement Therapy , Female , Fulvestrant , Humans , In Vitro Techniques , Male , Platelet Aggregation/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Receptors, Estrogen/antagonists & inhibitors
19.
Curr Opin Hematol ; 7(5): 284-9, 2000 Sep.
Article in English | MEDLINE | ID: mdl-10961578

ABSTRACT

Both blood platelets and genetics contribute to the development of acute ischemic arterial diseases. A careful analysis of the various clinical association studies supports a modest increased risk for coronary artery disease events in carriers of the PIA2 polymorphism of GPIIIa. Investigations with both platelets and stable cells lines have shown the PIA2 polymorphism is prothrombotic. Only a handful of studies have been performed for platelet GPla (integrin alpha2) and GPIb-IX-V, but there is support for the 807 T/C polymorphism of GPIa and the met145 and VNTR B/C genotype of GPIbalpha as risk factors in younger age groups. And isolated reports suggest other platelet polymorphisms (GPIIb, FcgammaRIIa, P-selectin, alpha2 adrenergic receptor, transforming growth factor [TGF]beta) are risk factors for arterial disease or produce a prothrombotic phenotype. Platelet glycoprotein polymorphisms should be added to the list of genetic risk factors for arterial thrombosis, particularly in younger patients and women.


Subject(s)
Platelet Membrane Glycoproteins/genetics , Thrombosis/genetics , Humans , Platelet Membrane Glycoproteins/adverse effects , Platelet Membrane Glycoproteins/physiology , Polymorphism, Genetic , Risk Factors , Thrombosis/etiology
20.
Blood ; 96(5): 1789-97, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10961878

ABSTRACT

Accumulating evidence suggests that successful metastatic spread may depend on the ability of tumor cells to undergo extensive interactions with platelets. However, the mechanisms mediating tumor cell adhesion to platelets under conditions of flow remain largely unknown. Therefore, this study was designed to analyze the ability of 3 human colon carcinoma cell lines (LS174T, COLO205, and HCT-8) to bind to surface-anchored platelets under flow and to identify the receptors involved in these processes. Immobilized platelets support LS174T cell adhesion at wall shear stresses up to 1.4 dyn/cm(2). Our data suggest that platelets primarily recruit LS174T cells through a 2-step, sequential process of adhesive interactions that shares common features but is distinct from that elaborated for neutrophils. Platelet P-selectin mediates LS174T cell tethering and rolling in a PSGL-1- and CD24-independent manner. Moreover, platelet alpha(IIb)beta(3)-integrins appear to be capable of directly capturing LS174T cells from the fluid stream, and also convert instantaneously transient tethers initiated by P-selectin into stable adhesion. This step is at least partially mediated by von Willebrand factor, but not fibrinogen or fibronectin, that bridges platelet alpha(IIb)beta(3) with a yet unidentified receptor on the LS174T cell surface via an RGD-dependent mechanism. The sequential engagement of platelet P-selectin and alpha(IIb)beta(3) is also requisite for the optimal adhesion of COLO205. Furthermore, HCT-8 cells, which fail to interact with P-selectin, tether minimally to surface-anchored platelets under flow, despite their extensive adhesive interactions under static conditions. This cascade of events depicts an efficacious process for colon carcinoma arrest at sites of vascular injury. (Blood. 2000;96:1789-1797)


Subject(s)
Blood Platelets/cytology , Cell Adhesion , Cell Movement , Biomechanical Phenomena , Blood Platelets/metabolism , Colonic Neoplasms , Flow Cytometry , Humans , Membrane Glycoproteins/metabolism , Oligosaccharides/metabolism , P-Selectin/metabolism , Platelet Adhesiveness , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Protein Binding , Sialyl Lewis X Antigen , Tumor Cells, Cultured/cytology , Tumor Cells, Cultured/metabolism , von Willebrand Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...