Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
medRxiv ; 2023 Oct 26.
Article in English | MEDLINE | ID: mdl-37961335

ABSTRACT

Background: CYP2C19 loss-of-function (LOF) alleles decrease the antiplatelet effect of clopidogrel following percutaneous coronary intervention (PCI) in patients presenting with acute coronary syndrome (ACS). The impact of genotype in stable ischemic heart disease (SIHD) is unclear. Objectives: Determine the association of CYP2C19 genotype with major adverse cardiac events (MACE) after PCI for ACS or SIHD. Methods: Million Veterans Program (MVP) participants age <65 years with a PCI documented in the VA Clinical Assessment, Reporting and Tracking (CART) Program between 1/1/2009 to 9/30/2017, treated with clopidogrel were included. Time to MACE defined as the composite of all-cause death, stroke or myocardial infarction within 12 months following PCI. Results: Among 4,461 Veterans (mean age 59.1 ± 5.1 years, 18% Black); 44% had ACS, 56% had SIHD and 29% carried a CYP2C19 LOF allele. 301 patients (6.7%) experienced MACE while being treated with clopidogrel, 155 (7.9%) in the ACS group and 146 (5.9%) in the SIHD group. Overall, MACE was not significantly different between LOF carriers vs. noncarriers (adjusted hazard ratio [HR] 1.18, confidence interval [95%CI] 0.97-1.45, p=0.096). Among patients presenting with ACS, MACE risk in LOF carriers versus non-carriers was numerically higher (HR 1.30, 95%CI 0.98-1.73, p=0.067). There was no difference in MACE risk in patients with SIHD (HR 1.09, 95%CI 0.82-1.44; p=0.565). Conclusions: CYP2C19 LOF carriers presenting with ACS treated with clopidogrel following PCI experienced a numerically greater elevated risk of MACE events. CYP2C19 LOF genotype is not associated with MACE among patients presenting with SIHD.

2.
Biology (Basel) ; 11(5)2022 Apr 29.
Article in English | MEDLINE | ID: mdl-35625411

ABSTRACT

Neuregulin-1ß (NRG-1ß) is a growth and differentiation factor with pleiotropic systemic effects. Because NRG-1ß has therapeutic potential for heart failure and has known growth effects in skeletal muscle, we hypothesized that it might affect heart failure-associated cachexia, a severe co-morbidity characterized by a loss of muscle mass. We therefore assessed NRG-1ß's effect on intercostal skeletal muscle gene expression in a swine model of heart failure using recombinant glial growth factor 2 (USAN-cimaglermin alfa), a version of NRG-1ß that has been tested in humans with systolic heart failure. Animals received one of two intravenous doses (0.67 or 2 mg/kg) of NRG-1ß bi-weekly for 4 weeks, beginning one week after infarct. Based on paired-end RNA sequencing, NRG-1ß treatment altered the intercostal muscle gene expression of 581 transcripts, including genes required for myofiber growth, maintenance and survival, such as MYH3, MYHC, MYL6B, KY and HES1. Importantly, NRG-1ß altered the directionality of at least 85 genes associated with cachexia, including myostatin, which negatively regulates myoblast differentiation by down-regulating MyoD expression. Consistent with this, MyoD was increased in NRG-1ß-treated animals. In vitro experiments with myoblast cell lines confirmed that NRG-1ß induces ERBB-dependent differentiation. These findings suggest a NRG-1ß-mediated anti-atrophic, anti-cachexia effect that may provide additional benefits to this potential therapy in heart failure.

3.
Clin Pharmacol Ther ; 108(5): 1067-1077, 2020 11.
Article in English | MEDLINE | ID: mdl-32472697

ABSTRACT

Antiplatelet response to clopidogrel shows wide variation, and poor response is correlated with adverse clinical outcomes. CYP2C19 loss-of-function alleles play an important role in this response, but account for only a small proportion of variability in response to clopidogrel. An aim of the International Clopidogrel Pharmacogenomics Consortium (ICPC) is to identify other genetic determinants of clopidogrel pharmacodynamics and clinical response. A genomewide association study (GWAS) was performed using DNA from 2,750 European ancestry individuals, using adenosine diphosphate-induced platelet reactivity and major cardiovascular and cerebrovascular events as outcome parameters. GWAS for platelet reactivity revealed a strong signal for CYP2C19*2 (P value = 1.67e-33). After correction for CYP2C19*2 no other single-nucleotide polymorphism reached genomewide significance. GWAS for a combined clinical end point of cardiovascular death, myocardial infarction, or stroke (5.0% event rate), or a combined end point of cardiovascular death or myocardial infarction (4.7% event rate) showed no significant results, although in coronary artery disease, percutaneous coronary intervention, and acute coronary syndrome subgroups, mutations in SCOS5P1, CDC42BPA, and CTRAC1 showed genomewide significance (lowest P values: 1.07e-09, 4.53e-08, and 2.60e-10, respectively). CYP2C19*2 is the strongest genetic determinant of on-clopidogrel platelet reactivity. We identified three novel associations in clinical outcome subgroups, suggestive for each of these outcomes.


Subject(s)
Blood Platelets/drug effects , Cardiovascular Diseases/prevention & control , Clopidogrel/therapeutic use , Coronary Artery Disease/therapy , Cytochrome P-450 CYP2C19/genetics , Percutaneous Coronary Intervention , Pharmacogenomic Variants , Platelet Aggregation Inhibitors/therapeutic use , Polymorphism, Single Nucleotide , Aged , Blood Platelets/metabolism , Cardiovascular Diseases/blood , Cardiovascular Diseases/genetics , Cardiovascular Diseases/mortality , Clopidogrel/adverse effects , Coronary Artery Disease/mortality , Cytochrome P-450 CYP2C19/metabolism , Female , Genome-Wide Association Study , Humans , Male , Middle Aged , Percutaneous Coronary Intervention/adverse effects , Percutaneous Coronary Intervention/mortality , Pharmacogenetics , Platelet Aggregation Inhibitors/adverse effects , Risk Assessment , Risk Factors , Treatment Outcome
4.
Eur Heart J Cardiovasc Pharmacother ; 6(4): 203-210, 2020 07 01.
Article in English | MEDLINE | ID: mdl-31504375

ABSTRACT

AIMS: Clopidogrel is prescribed for the prevention of atherothrombotic events. While investigations have identified genetic determinants of inter-individual variability in on-treatment platelet inhibition (e.g. CYP2C19*2), evidence that these variants have clinical utility to predict major adverse cardiovascular events (CVEs) remains controversial. METHODS AND RESULTS: We assessed the impact of 31 candidate gene polymorphisms on adenosine diphosphate (ADP)-stimulated platelet reactivity in 3391 clopidogrel-treated coronary artery disease patients of the International Clopidogrel Pharmacogenomics Consortium (ICPC). The influence of these polymorphisms on CVEs was tested in 2134 ICPC patients (N = 129 events) in whom clinical event data were available. Several variants were associated with on-treatment ADP-stimulated platelet reactivity (CYP2C19*2, P = 8.8 × 10-54; CES1 G143E, P = 1.3 × 10-16; CYP2C19*17, P = 9.5 × 10-10; CYP2B6 1294 + 53 C > T, P = 3.0 × 10-4; CYP2B6 516 G > T, P = 1.0 × 10-3; CYP2C9*2, P = 1.2 × 10-3; and CYP2C9*3, P = 1.5 × 10-3). While no individual variant was associated with CVEs, generation of a pharmacogenomic polygenic response score (PgxRS) revealed that patients who carried a greater number of alleles that associated with increased on-treatment platelet reactivity were more likely to experience CVEs (ß = 0.17, SE 0.06, P = 0.01) and cardiovascular-related death (ß = 0.43, SE 0.16, P = 0.007). Patients who carried eight or more risk alleles were significantly more likely to experience CVEs [odds ratio (OR) = 1.78, 95% confidence interval (CI) 1.14-2.76, P = 0.01] and cardiovascular death (OR = 4.39, 95% CI 1.35-14.27, P = 0.01) compared to patients who carried six or fewer of these alleles. CONCLUSION: Several polymorphisms impact clopidogrel response and PgxRS is a predictor of cardiovascular outcomes. Additional investigations that identify novel determinants of clopidogrel response and validating polygenic models may facilitate future precision medicine strategies.


Subject(s)
Clopidogrel/therapeutic use , Coronary Artery Disease/therapy , Decision Support Techniques , Percutaneous Coronary Intervention , Pharmacogenomic Variants , Platelet Aggregation Inhibitors/therapeutic use , Platelet Aggregation/drug effects , Polymorphism, Single Nucleotide , Aged , Brain Ischemia/mortality , Brain Ischemia/prevention & control , Clopidogrel/adverse effects , Coronary Artery Disease/blood , Coronary Artery Disease/diagnosis , Coronary Artery Disease/mortality , Coronary Thrombosis/mortality , Coronary Thrombosis/prevention & control , Europe , Female , Humans , Male , Middle Aged , Myocardial Infarction/mortality , Myocardial Infarction/prevention & control , Percutaneous Coronary Intervention/adverse effects , Percutaneous Coronary Intervention/instrumentation , Percutaneous Coronary Intervention/mortality , Platelet Aggregation/genetics , Platelet Aggregation Inhibitors/adverse effects , Predictive Value of Tests , Risk Assessment , Risk Factors , Stents , Stroke/mortality , Stroke/prevention & control , Treatment Outcome
5.
Am Heart J ; 198: 152-159, 2018 04.
Article in English | MEDLINE | ID: mdl-29653637

ABSTRACT

RATIONALE: The P2Y12 receptor inhibitor clopidogrel is widely used in patients with acute coronary syndrome, percutaneous coronary intervention, or ischemic stroke. Platelet inhibition by clopidogrel shows wide interpatient variability, and high on-treatment platelet reactivity is a risk factor for atherothrombotic events, particularly in high-risk populations. CYP2C19 polymorphism plays an important role in this variability, but heritability estimates suggest that additional genetic variants remain unidentified. The aim of the International Clopidogrel Pharmacogenomics Consortium (ICPC) is to identify genetic determinants of clopidogrel pharmacodynamics and clinical response. STUDY DESIGN: Based on the data published on www.clinicaltrials.gov, clopidogrel intervention studies containing genetic and platelet function data were identified for participation. Lead investigators were invited to share DNA samples, platelet function test results, patient characteristics, and cardiovascular outcomes to perform candidate gene and genome-wide studies. RESULTS: In total, 17 study sites from 13 countries participate in the ICPC, contributing individual patient data from 8,829 patients. Available adenosine diphosphate-stimulated platelet function tests included vasodilator-stimulated phosphoprotein assay, light transmittance aggregometry, and the VerifyNow P2Y12 assay. A proof-of-principle analysis based on genotype data provided by each group showed a strong and consistent association between CYP2C19*2 and platelet reactivity (P value=5.1 × 10-40). CONCLUSION: The ICPC aims to identify new loci influencing clopidogrel efficacy by using state-of-the-art genetic approaches in a large cohort of clopidogrel-treated patients to better understand the genetic basis of on-treatment response variability.


Subject(s)
Acute Coronary Syndrome/drug therapy , Clopidogrel/therapeutic use , Genome-Wide Association Study , Molecular Targeted Therapy/methods , Receptors, Purinergic P2Y12/genetics , Acute Coronary Syndrome/diagnosis , Acute Coronary Syndrome/mortality , Aged , Female , Genetic Association Studies , Humans , Internationality , Male , Middle Aged , Pharmacogenetics , Prognosis , Receptors, Purinergic P2Y12/drug effects , Risk Assessment , Survival Rate , Treatment Outcome
6.
J Pharmacol Exp Ther ; 365(2): 219-225, 2018 05.
Article in English | MEDLINE | ID: mdl-29491039

ABSTRACT

Ser54 of Gsα binds guanine nucleotide and Mg2+ as part of a conserved sequence motif in GTP binding proteins. Mutating the homologous residue in small and heterotrimeric G proteins generates dominant-negative proteins, but by protein-specific mechanisms. For αi/o, this results from persistent binding of α to ßγ, whereas for small GTP binding proteins and αs this results from persistent binding to guanine nucleotide exchange factor or receptor. This work examined the role of ßγ interactions in mediating the properties of the Ser54-like mutants of Gα subunits. Unexpectedly, WT-αs or N54-αs coexpressed with α1B-adrenergic receptor in human embryonic kidney 293 cells decreased receptor stimulation of IP3 production by a cAMP-independent mechanism, but WT-αs was more effective than the mutant. One explanation for this result would be that αs, like Ser47 αi/o, blocks receptor activation by sequestering ßγ; implying that N54-αS has reduced affinity for ßγ since it was less effective at blocking IP3 production. This possibility was more directly supported by the observation that WT-αs was more effective than the mutant in inhibiting ßγ activation of phospholipase Cß2. Further, in vitro synthesized N54-αs bound biotinylated-ßγ with lower apparent affinity than did WT-αs The Cys54 mutation also decreased ßγ binding but less effectively than N54-αs Substitution of the conserved Ser in αo with Cys or Asn increased ßγ binding, with the Cys mutant being more effective. This suggests that Ser54 of αs is involved in coupling changes in nucleotide binding with altered subunit interactions, and has important implications for how receptors activate G proteins.


Subject(s)
GTP-Binding Protein alpha Subunits, Gs/chemistry , GTP-Binding Protein alpha Subunits, Gs/metabolism , Mutation , Protein Multimerization , Protein Subunits/metabolism , Conserved Sequence , GTP-Binding Protein alpha Subunits, Gs/genetics , HEK293 Cells , Humans , Phospholipase C beta/metabolism , Protein Binding/genetics , Protein Structure, Quaternary , Receptors, Adrenergic, alpha-1/metabolism , Signal Transduction
7.
Toxicol Appl Pharmacol ; 332: 92-99, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28780372

ABSTRACT

Neuregulin-1ß is a member of the neuregulin family of growth factors and is critically important for normal development and functioning of the heart and brain. A recombinant version of neuregulin-1ß, cimaglermin alfa (also known as glial growth factor 2 or GGF2) is being investigated as a possible therapy for heart failure. Previous studies suggest that neuregulin-1ß stimulation of skeletal muscle increases glucose uptake and, specifically, sufficient doses of cimaglermin alfa acutely produce hypoglycemia in pigs. Since acute hypoglycemia could be a safety concern, blood glucose changes in the above pig study were further investigated. In addition, basal glucose and glucose disposal were investigated in mice. Finally, as part of standard clinical chemistry profiling in a single ascending-dose human safety study, blood glucose levels were evaluated in patients with heart failure after cimaglermin alfa treatment. A single intravenous injection of cimaglermin alfa at doses of 0.8mg/kg and 2.6mg/kg in mice resulted in a transient reduction of blood glucose concentrations of approximately 20% and 34%, respectively, at 2h after the treatment compared to pre-treatment levels. Similar results were observed in diabetic mice. Treatment with cimaglermin alfa also increased blood glucose disposal following oral challenge in mice. However, no significant alterations in blood glucose concentrations were found in human heart failure patients at 0.5 and 2h after treatment with cimaglermin alfa over an equivalent human dose range, based on body surface area. Taken together, these data indicate strong species differences in blood glucose handling after cimaglermin alfa treatment, and particularly do not indicate that this phenomenon should affect human subjects.


Subject(s)
Blood Glucose/metabolism , Heart Failure/blood , Neuregulin-1/pharmacology , Adolescent , Adult , Aged , Animals , Diabetes Mellitus, Experimental/drug therapy , Dose-Response Relationship, Drug , Female , Humans , Insulin/blood , Male , Mice , Mice, Inbred C57BL , Middle Aged , Models, Animal , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Species Specificity , Swine , Young Adult
8.
AMIA Jt Summits Transl Sci Proc ; 2017: 185-192, 2017.
Article in English | MEDLINE | ID: mdl-28815128

ABSTRACT

Electronic health records (EHRs) linked with biobanks have been recognized as valuable data sources for pharmacogenomic studies, which require identification of patients with certain adverse drug reactions (ADRs) from a large population. Since manual chart review is costly and time-consuming, automatic methods to accurately identify patients with ADRs have been called for. In this study, we developed and compared different informatics approaches to identify ADRs from EHRs, using clopidogrel-induced bleeding as our case study. Three different types of methods were investigated: 1) rule-based methods; 2) machine learning-based methods; and 3) scoring function-based methods. Our results show that both machine learning and scoring methods are effective and the scoring method can achieve a high precision with a reasonable recall. We also analyzed the contributions of different types of features and found that the temporality information between clopidogrel and bleeding events, as well as textual evidence from physicians' assertion of the adverse events are helpful. We believe that our findings are valuable in advancing EHR-based pharmacogenomic studies.

9.
J Thromb Thrombolysis ; 41(4): 656-62, 2016 May.
Article in English | MEDLINE | ID: mdl-26446588

ABSTRACT

Abnormal platelet reactivity is associated with recurrent ischemia and bleeding following percutaneous coronary intervention (PCI). Protease-activated receptor-1 (PAR1), encoded by F2R, is a high affinity thrombin receptor on platelets and the target of the antiplatelet drug vorapaxar. The intronic single nucleotide polymorphism F2R IVS-14 A/T affects PAR1 receptor density and function. We hypothesized that carriers of the T allele, who have been shown to have decreased platelet reactivity, would be at lower risk for thrombotic events, but higher risk for bleeding following PCI. Using BioVU, the Vanderbilt DNA repository linked to the electronic medical record, we studied 660 patients who underwent PCI for unstable or stable coronary artery disease. Primary outcome measures were major adverse cardiovascular events (MACE, composite of revascularization, MI, stroke, death) and bleeding (assessed by Bleeding Academic Research Consortium scale) over 24 months. The minor allele (T) frequency was 14.8 %. There were no genotypic differences in the frequency of MACE (33.7, 28.8, and 31.6 % for A/A, A/T, and T/T respectively, P = 0.50) or bleeding (15.7, 14.7, and 18.8 % for A/A, A/T, and T/T respectively, P = 0.90). In a Cox regression model, fully adjusted for age, race, sex, BMI, and smoking status, carrying a T allele was not associated with MACE (HR 1.19, 95 % CI 0.89-1.59, P = 0.23) or bleeding (HR 0.73, 95 % CI 0.37-1.4, P = 0.34). In conclusion, in our population, F2R IVS-14 PAR1 variability does not affect risk of MACE or bleeding following PCI.


Subject(s)
Coronary Artery Disease/genetics , Coronary Artery Disease/surgery , Percutaneous Coronary Intervention/adverse effects , Polymorphism, Genetic , Postoperative Hemorrhage/genetics , Receptor, PAR-1/genetics , Aged , Aged, 80 and over , Coronary Artery Disease/mortality , Female , Humans , Male , Middle Aged , Myocardial Infarction/etiology , Myocardial Infarction/genetics , Myocardial Infarction/mortality , Postoperative Hemorrhage/etiology , Postoperative Hemorrhage/mortality , Stroke/etiology , Stroke/genetics , Stroke/mortality
10.
J Am Heart Assoc ; 3(5): e000773, 2014 Oct 23.
Article in English | MEDLINE | ID: mdl-25341890

ABSTRACT

BACKGROUND: Neuregulin-1ß (NRG-1ß) is a growth factor critical for cardiac development and repair with therapeutic potential for heart failure. We previously showed that the glial growth factor 2 (GGF2) isoform of NRG-1ß improves cardiac function in rodents after myocardial infarction (MI), but its efficacy in a large animal model of cardiac injury has not been examined. We therefore sought to examine the effects of GGF2 on ventricular remodeling, cardiac function, and global transcription in post-MI swine, as well as potential mechanisms for anti-remodeling effects. METHODS AND RESULTS: MI was induced in anesthetized swine (n=23) by intracoronary balloon occlusion. At 1 week post-MI, survivors (n=13) received GGF2 treatment (intravenous, biweekly for 4 weeks; n=8) or were untreated (n=5). At 5 weeks post-MI, fractional shortening was higher (32.8% versus 25.3%, P=0.019), and left ventricular (LV) end-diastolic dimension lower (4.5 versus 5.3 cm, P=0.003) in GGF2-treated animals. Treatment altered expression of 528 genes, as measured by microarrays, including collagens, basal lamina components, and matricellular proteins. GGF2-treated pigs exhibited improvements in LV cardiomyocyte mitochondria and intercalated disk structures and showed less fibrosis, altered matrix structure, and fewer myofibroblasts (myoFbs), based on trichrome staining, electron microscopy, and immunostaining. In vitro experiments with isolated murine and rat cardiac fibroblasts demonstrate that NRG-1ß reduces myoFbs, and suppresses TGFß-induced phospho-SMAD3 as well as αSMA expression. CONCLUSIONS: These results suggest that GGF2/NRG-1ß prevents adverse remodeling after injury in part via anti-fibrotic effects in the heart.


Subject(s)
Heart Failure/drug therapy , Myocardium/pathology , Neuregulin-1/pharmacology , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Actins/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Dose-Response Relationship, Drug , Fibrosis , Gene Expression Regulation/drug effects , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Male , Mice, Inbred C57BL , Myocardial Contraction/drug effects , Myocardium/metabolism , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Myofibroblasts/pathology , Phosphorylation , Rats, Sprague-Dawley , Smad3 Protein/metabolism , Swine , Time Factors , Transcription, Genetic/drug effects , Ventricular Remodeling/genetics
11.
J Pharmacol Exp Ther ; 351(1): 33-43, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25052834

ABSTRACT

Although resistance to the P2Y12 antagonist clopidogrel is linked to altered drug metabolism, some studies suggest that these pharmacokinetic abnormalities only partially account for drug resistance. To circumvent pharmacokinetic complications and target P2Y12 receptor function we applied the direct P2Y12 antagonist 2-methylthio-AMP (2-methylthioadenosine 5'-monophosphate triethylammonium salt) to purified platelets ex vivo. Platelets were purified from healthy and type 2 diabetes mellitus (T2DM) patients and stimulated with thrombin or the selective protease-activated receptor agonists, protease-activated receptor 1-activating peptide (PAR1-AP), or PAR4-AP. Platelet activation as measured by αIIbß3 activation, and P-selectin expression was monitored in 141 subjects. Our results demonstrate that, compared with healthy subjects, platelets from diabetic patients are resistant to inhibition by 2-methylthio-AMP, demonstrating P2Y12 pharmacodynamic defects among diabetic patients. Inhibition of thrombin-mediated αIIbß3 activation by 2-methylthio-AMP was lower in diabetic platelets versus healthy platelets. Subgroup analysis revealed a racial difference in the resistance to 2-methylthio-AMP. We found no resistance in platelets from diabetic African Americans; they were inhibited by 2-methylthio-AMP equally as well as platelets from healthy African Americans. In contrast, platelets from Caucasian patients with diabetes were resistant to P2Y12 antagonism compared with healthy Caucasians. Multivariable analysis demonstrated that other variables, such as obesity, age, or gender, could not account for the differential resistance to 2-methylthio-AMP among races. These results suggest that in addition to altered drug metabolism, P2Y12 receptor function itself is altered in the Caucasian diabetic population. The racial difference in platelet function in T2DM is a novel finding, which may lead to differences in treatment as well as new targets for antiplatelet therapy.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/pharmacology , Blood Platelets/drug effects , Diabetes Mellitus, Type 2/ethnology , Drug Resistance , Purinergic P2Y Receptor Antagonists/pharmacology , Adenosine Monophosphate/pharmacokinetics , Adult , Black or African American , Diabetes Mellitus, Type 2/metabolism , Female , Humans , Male , White People
12.
J Thromb Thrombolysis ; 35(2): 209-22, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23054462

ABSTRACT

Bivalirudin is a direct thrombin inhibitor that is increasingly used in percutaneous coronary intervention (PCI) and has been previously shown to lack inherent platelet activation. Thrombin works through activation of protease activated receptor-1 (PAR1) and PAR4 on human platelets to initiate signaling cascades leading to platelet aggregation. Despite the increasing usage of bivalirudin, the effects on platelet function have not been well defined. Bivalirudin exposure during PCI was therefore assessed for its potential short-term effects on washed platelet function through PAR1 and PAR4. Bivalirudin significantly inhibited low-dose thrombin-mediated platelet aggregation, dense granule secretion, integrin αIIbß3 activation and Rap1 activation and high dose thrombin-mediated dense granule secretion and Rap1 activation. Exposure to bivalirudin did not alter PAR1 or 4 agonist peptide (PAR1-AP or PAR4-AP) induced aggregation, dense granule secretion, integrin glycoprotein IIbIIIa activation or Rap1 activation. However, exposure to bivalirudin significantly potentiated surface expression of P-selectin following stimulation with high dose thrombin and PAR1-AP, and both low and high dose PAR4-AP. Hence, our data are the first to show that exposure to bivalirudin increased P-selectin expression with certain conditions demonstrating that bivalirudin can increase inherent platelet activity.


Subject(s)
Antithrombins/pharmacology , Hirudins/pharmacology , Peptide Fragments/pharmacology , Percutaneous Coronary Intervention , Platelet Activation/drug effects , Receptor, PAR-1/physiology , Receptors, Thrombin/metabolism , Aged , Antithrombins/adverse effects , Dose-Response Relationship, Drug , Female , Hirudins/adverse effects , Humans , Male , Middle Aged , P-Selectin/biosynthesis , Peptide Fragments/adverse effects , Percutaneous Coronary Intervention/trends , Platelet Activation/physiology , Recombinant Proteins/adverse effects , Recombinant Proteins/pharmacology , Treatment Outcome
13.
J Invasive Cardiol ; 24(11): 599-604, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23117316

ABSTRACT

OBJECTIVE: To determine whether radial artery access is associated with a reduction in fluoroscopy time, procedure time, and other procedural variables over a 27-month period during which the radial artery approach was incorporated in a single academic Medical Center. BACKGROUND: Although previous studies have demonstrated a relationship between increased volume and decreased procedural time, no studies have looked at the integration of radial access over time. METHODS: Data were collected from consecutive patients who presented to the Vanderbilt University Medical Center cardiac catheterization laboratory from January 1, 2009 to April 1, 2011. Patients who underwent radial access diagnostic catheterization with and without percutaneous coronary intervention were included in this study. A total of 1112 diagnostic cardiac catheterizations through the radial access site were analyzed. High-volume, intermediate-volume, and low-volume operators were grouped based on the percentage of procedures performed through a radial approach. RESULTS: From 2009 to 2011, there was a significant decrease in fluoroscopy time in all operator groups for diagnostic catheterization (P=.035). The high-volume operator group had 1.88 and 3.66 minute reductions in fluoroscopy time compared to the intermediate- and low-volume operator groups, respectively (both P<.001). Likewise, the intermediate-volume operator group had a 1.77 minute improvement compared to the low-volume operator group, but this did not reach statistical significance (P=.102). The improvement in fluoroscopy time and other procedure-related parameters was seen after approximately 25 cases with further improvement after 75 cases. CONCLUSIONS: The incorporation of the radial access approach in the cardiac catheterization laboratory led to a decrease in fluoroscopy time for each operator and operator group over the last 3 years. Our data demonstrated that higher-volume radial operators have better procedure, room, and fluoroscopy times when compared to intermediate- and low-volume operators. However, lower-volume operators have a reduction in procedure-related parameters with increased radial cases. Number of procedures needed to become sufficient was demonstrated in the current study.


Subject(s)
Cardiac Catheterization/methods , Learning Curve , Radial Artery , Specialization , Aged , Fluoroscopy , Hospitals, High-Volume/statistics & numerical data , Hospitals, Low-Volume/statistics & numerical data , Humans , Middle Aged , Retrospective Studies , Time Factors
14.
J Am Coll Cardiol ; 53(3): 232-41, 2009 Jan 20.
Article in English | MEDLINE | ID: mdl-19147039

ABSTRACT

OBJECTIVES: This study sought to report our experience with a routine completion angiogram after coronary artery bypass surgery (CABG) and simultaneous (1-stop) percutaneous coronary intervention (PCI) at the time of CABG performed in the hybrid catheterization laboratory/operating room. BACKGROUND: The value of a routine completion angiogram after CABG and 1-stop hybrid CABG/PCI remains unresolved. METHODS: Between April 2005 and July 2007, 366 consecutive patients underwent CABG surgery, with (n = 112) or without (n = 254) concomitant 1-stop PCI (hybrid), all with completion angiography before chest closure. Among the 112 1-stop hybrid CABG/PCI patients, 67 (60%) underwent a planned hybrid procedure based on pre-operative assessment, whereas 45 (40%) underwent open-chest PCI (unplanned hybrid) based on intraoperative findings. RESULTS: Among the 796 CABG grafts (345 left internal mammary artery, 12 right internal mammary artery/radial, and 439 veins), 97 (12%) angiographic defects were identified. Defects were repaired with either a minor adjustment of the graft (n = 22, 2.8%), with intraoperative open-chest PCI (unplanned hybrid, n = 48, 6%) or with traditional surgical revision (n = 27, 3.4%). Hybrid patients had clinical outcomes similar to standard CABG patients. CONCLUSIONS: Routine completion angiography detected 12% of grafts with important angiographic defects. One-stop hybrid coronary revascularization is reasonable, safe, and feasible. Combining the tools of the catheterization laboratory and operating room greatly enhances the options available to the surgeon and cardiologist for patients with complex coronary artery disease.


Subject(s)
Angioplasty, Balloon, Coronary/methods , Coronary Angiography/methods , Coronary Artery Bypass/methods , Coronary Disease/diagnostic imaging , Coronary Disease/surgery , Operating Rooms , Adult , Aged , Aged, 80 and over , Angioplasty, Balloon, Coronary/adverse effects , Cardiac Catheterization/methods , Cohort Studies , Combined Modality Therapy , Coronary Artery Bypass/adverse effects , Coronary Disease/therapy , Female , Follow-Up Studies , Humans , Intraoperative Care/methods , Male , Middle Aged , Probability , Radiography, Interventional , Retrospective Studies , Risk Assessment , Sensitivity and Specificity , Statistics, Nonparametric , Stents , Treatment Outcome , Vascular Patency
16.
Blood ; 107(7): 2736-44, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16332977

ABSTRACT

Thrombin-mediated endothelial-cell release of von Willebrand factor (VWF) and P-selectin functionally links protease-activated receptors (PARs) to thrombosis and inflammation. VWF release can be stimulated by both Ca2+ and cAMP, and, although both VWF and P-selectin are found in Weibel-Palade bodies (WPBs), we found that their release could be differentially regulated. In these studies, human umbilical vein endothelial cells stimulated with cAMP or PAR2-AP led to a delayed release of VWF and significantly less P-selectin release compared with histamine, thrombin, or PAR1-AP. Dose-response studies revealed that PAR2-AP was significantly less efficacious in promoting the release of P-selectin compared with VWF. PAR2-AP-induced robust stimulation of intracellular Ca2+ coupled with a significantly greater inhibitory effect of calcium chelation on release of VWF compared with cell-surface expression of P-selectin, suggests an additional Ca2+-independent pathway involved in release of P-selectin. PAR2-AP failed to increase global cAMP levels; however, inhibition of protein kinase A led to a significant attenuation of PAR2-AP-mediated release of VWF. Confocal microscopy studies revealed that PAR2 and forskolin caused preferential release of a population of Weibel-Palade bodies (WPBs) consisting of only VWF. Thus, WPBs are pharmacologically and morphologically heterogeneous, and distinct granule populations are susceptible to differential regulation.


Subject(s)
Calcium Signaling/physiology , Cyclic AMP/physiology , Endothelium, Vascular/physiology , P-Selectin/metabolism , Peptide Hydrolases/metabolism , Receptor, PAR-1/metabolism , Receptor, PAR-2/metabolism , Receptors, Cell Surface/physiology , Signal Transduction/physiology , von Willebrand Factor/metabolism , Cell Line , Endothelium, Vascular/cytology , Exocytosis , Humans , Kinetics , Protein Transport , Thrombin/pharmacology , Umbilical Veins
17.
Biochemistry ; 44(35): 11882-90, 2005 Sep 06.
Article in English | MEDLINE | ID: mdl-16128590

ABSTRACT

The Gbeta and Ggamma subunit of the heterotrimeric G proteins form a functional dimer that is stable once assembled in vivo or in vitro. The requirements, mechanism, and specificity of dimer formation are still incompletely understood, but represent important biochemical processes involved in the specificity of cellular signaling through G proteins. Here, seven Gbeta and 12 FLAG-epitope-tagged Ggamma subunits were separately synthesized in vitro using a rabbit reticulocyte lysate expression system. The translation products were combined and dimers isolated by immunoprecipitation. Gbeta1 and Gbeta4 formed dimers with all Ggamma subunit isoforms, generally with Gbeta/Ggamma stoichiometries between 0.2:1 and 0.5:1. Gbeta5, Gbeta5L, and Gbeta3s did not form significant amounts of dimer with any of the gamma subunit isoforms. Gbeta2 and Gbeta3 formed dimers with selected Ggamma isoforms to levels intermediate between that of Gbeta1/Gbeta4 and Gbeta3s/Gbeta5/Gbeta5L. We also expressed selected Gbetagamma in HEK293 cells and measured PLCbeta2 activity. Gbetagamma dimer-dependent increases in IP3 production were seen with most Gbeta1, Gbeta2, and Gbeta5 combinations, indicating functional dimer expression in intact cells. These results define the complete set of G protein betagamma dimers that are formed using a single biochemical assay method and suggest that there are Gbeta isoform-specific factors in rabbit reticulocyte lysates that determine the efficacy of Gbetagamma dimer formation.


Subject(s)
GTP-Binding Protein beta Subunits/chemistry , GTP-Binding Protein beta Subunits/metabolism , GTP-Binding Protein gamma Subunits/chemistry , GTP-Binding Protein gamma Subunits/metabolism , Heterotrimeric GTP-Binding Proteins/biosynthesis , Animals , Cells, Cultured , Dimerization , Epitopes/metabolism , Humans , Isoenzymes/metabolism , Oligopeptides , Peptides/metabolism , Phospholipase C beta , Rabbits , Reticulocytes/metabolism , Type C Phospholipases/metabolism
18.
J Biol Chem ; 280(23): 22172-80, 2005 Jun 10.
Article in English | MEDLINE | ID: mdl-15817447

ABSTRACT

Thrombospondin-1 (THBS1) is a large extracellular matrix glycoprotein that affects vasculature systems such as platelet activation, angiogenesis, and wound healing. Increases in THBS1 expression have been liked to disease states including tumor progression, atherosclerosis, and arthritis. The present study focuses on the effects of thrombin activation of the G-protein-coupled, protease-activated receptor-1 (PAR-1) on THBS1 gene expression in the microvascular endothelium. Thrombin-induced changes in gene expression were characterized by microarray analysis of approximately 11,000 different human genes in human microvascular endothelial cells (HMEC-1). Thrombin induced the expression of a set of at least 65 genes including THBS1. Changes in THBS1 mRNA correlated with an increase in the extracellular THBS1 protein concentration. The PAR-1-specific agonist peptide (TFLLRNK-PDK) mimicked thrombin stimulation of THBS1 expression, suggesting that thrombin signaling is through PAR-1. Further studies showed THBS1 expression was sensitive to pertussis toxin and protein kinase C inhibition indicating G(i/o)- and G(q)-mediated pathways. THBS1 up-regulation was also confirmed in human umbilical vein endothelial cells stimulated with thrombin. Analysis of the promoter region of THBS1 and other genes of similar expression profile identified from the microarray predicted an EBOX/EGRF transcription model. Expression of members of each family, MYC and EGR1, respectively, correlated with THBS1 expression. These results suggest thrombin formed at sites of vascular injury increases THBS1 expression into the extracellular matrix via activation of a PAR-1, G(i/o), G(q), EBOX/EGRF-signaling cascade, elucidating regulatory points that may play a role in increased THBS1 expression in disease states.


Subject(s)
Endothelium, Vascular/cytology , Gene Expression Regulation , Microcirculation/metabolism , Thrombin/chemistry , Thrombospondin 1/biosynthesis , Adenosine Diphosphate/chemistry , Algorithms , Amides/pharmacology , Apoptosis , Cells, Cultured , Cluster Analysis , Culture Media , DNA Primers/chemistry , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Electric Impedance , Endothelium, Vascular/metabolism , Enzyme-Linked Immunosorbent Assay , Extracellular Matrix/metabolism , GTP-Binding Protein alpha Subunits, Gi-Go/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Indoles/pharmacology , Maleimides/pharmacology , Models, Biological , Nucleic Acid Hybridization , Oligonucleotide Array Sequence Analysis , Peptides/chemistry , Pertussis Toxin/pharmacology , Promoter Regions, Genetic , Protein Binding , Pyridines/pharmacology , RNA/metabolism , Receptor, PAR-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Thrombin/metabolism , Time Factors , Umbilical Veins/cytology , Up-Regulation
19.
J Biol Chem ; 279(35): 36601-7, 2004 Aug 27.
Article in English | MEDLINE | ID: mdl-15234971

ABSTRACT

A Ser to Asn mutation at position 54 of the alpha subunit of G(s) (designated N54-alpha(s)) was characterized after transient expression of it with various components of the receptor-adenylyl cyclase pathway in COS-1, COS-7, and HEK 293 cells. Previous studies of the N54-alpha(s) mutant revealed that it has a conditional dominant negative phenotype that prevents hormone-stimulated increases in cAMP without interfering with the regulation of basal cAMP levels (Cleator, J. H., Mehta, N. D., Kurtz, D. K., Hildebrandt, J. D. (1999) FEBS Lett. 243, 205-208). Experiments reported here were conducted to localize the mechanism of the dominant negative effect of the mutant. Competition studies conducted with activated alpha(s)* (Q212L) showed that the N54 mutant did not work down-stream by blocking the interaction of endogenous alpha(s) with adenylyl cyclase. The co-expression of wild type or N54-alpha(s) along with the thyroid-stimulating hormone (TSH) receptor and adenylyl cyclase isotypes differing with respect to betagamma stimulation (AC II or AC III) revealed that the phenotype of the mutant is not dependent upon the presence of adenylyl cyclase isoforms regulated by betagamma. These studies ruled out a downstream site of action of the mutant. To investigate an upstream site of action, N54-alpha(s) was co-expressed with either the TSH receptor that activates both alpha(s) and alpha(q) or with the alpha(1B)-adrenergic receptor that activates only alpha(q). N54-alpha(s) failed to inhibit alpha(1B)-adrenergic receptor stimulation of inositol 1,4,5-trisphosphate production but did inhibit TSH stimulation of inositol 1,4,5-trisphosphate. These results show that G(s) and G(q) compete for activation by the TSH receptor. They also indicate that the N54 protein has a dominant negative phenotype by blocking upstream receptor interactions with normal G proteins. This phenotype is different from that seen in analogous mutants of other G protein alpha subunits and suggests that either regulation or protein-protein interactions differ among G protein alpha subunits.


Subject(s)
Cyclic AMP/metabolism , GTP-Binding Protein alpha Subunits, Gs/genetics , Receptors, Thyrotropin/metabolism , Adenylyl Cyclases/metabolism , Animals , Binding, Competitive , COS Cells , Cell Line , DNA, Complementary/metabolism , Dimerization , Dose-Response Relationship, Drug , GTP-Binding Protein alpha Subunits, Gs/metabolism , Genes, Dominant , Genetic Vectors , Humans , Immunoblotting , Inositol 1,4,5-Trisphosphate/metabolism , Inositol Phosphates/metabolism , Mutation , Phenotype , Protein Isoforms , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL