Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Circ Genom Precis Med ; 11(4): e002178, 2018 04.
Article in English | MEDLINE | ID: mdl-29654098

ABSTRACT

The National Institutes of Health have made substantial investments in genomic studies and technologies to identify DNA sequence variants associated with human disease phenotypes. The National Heart, Lung, and Blood Institute has been at the forefront of these commitments to ascertain genetic variation associated with heart, lung, blood, and sleep diseases and related clinical traits. Genome-wide association studies, exome- and genome-sequencing studies, and exome-genotyping studies of the National Heart, Lung, and Blood Institute-funded epidemiological and clinical case-control studies are identifying large numbers of genetic variants associated with heart, lung, blood, and sleep phenotypes. However, investigators face challenges in identification of genomic variants that are functionally disruptive among the myriad of computationally implicated variants. Studies to define mechanisms of genetic disruption encoded by computationally identified genomic variants require reproducible, adaptable, and inexpensive methods to screen candidate variant and gene function. High-throughput strategies will permit a tiered variant discovery and genetic mechanism approach that begins with rapid functional screening of a large number of computationally implicated variants and genes for discovery of those that merit mechanistic investigation. As such, improved variant-to-gene and gene-to-function screens-and adequate support for such studies-are critical to accelerating the translation of genomic findings. In this White Paper, we outline the variety of novel technologies, assays, and model systems that are making such screens faster, cheaper, and more accurate, referencing published work and ongoing work supported by the National Heart, Lung, and Blood Institute's R21/R33 Functional Assays to Screen Genomic Hits program. We discuss priorities that can accelerate the impressive but incomplete progress represented by big data genomic research.


Subject(s)
Genetic Variation , Genomics/methods , High-Throughput Nucleotide Sequencing/methods , Animals , Cells, Cultured , Diffusion of Innovation , Forecasting , Genetic Predisposition to Disease , Genome-Wide Association Study , Genomics/trends , High-Throughput Nucleotide Sequencing/trends , Humans , Models, Animal , National Heart, Lung, and Blood Institute (U.S.) , Phenotype , Reproducibility of Results , Risk Factors , United States , Workflow
2.
Am Heart J ; 166(2): 199-207.e15, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23895801

ABSTRACT

BACKGROUND: Inflammation plays a fundamental role in atherothrombosis. Yet, whether direct inhibition of inflammation will reduce the occurrence of adverse cardiovascular outcomes is not known. DESIGN: The Cardiovascular Inflammation Reduction Trial (CIRT) (ClinicalTrials.govNCT01594333) will randomly allocate 7,000 patients with prior myocardial infarction (MI) and either type 2 diabetes or the metabolic syndrome to low-dose methotrexate (target dose 15-20 mg/wk) or placebo over an average follow-up period of 3 to 5 years. Low-dose methotrexate is a commonly used anti-inflammatory regimen for the treatment of rheumatoid arthritis and lacks significant effects on lipid levels, blood pressure, or platelet function. Both observational and mechanistic studies suggest that low-dose methotrexate has clinically relevant antiatherothrombotic effects. The CIRT primary end point is a composite of nonfatal MI, nonfatal stroke, and cardiovascular death. Secondary end points are all-cause mortality, coronary revascularization plus the primary end point, hospitalization for congestive heart failure plus the primary end point, all-cause mortality plus coronary revascularization plus congestive heart failure plus the primary end point, incident type 2 diabetes, and net clinical benefit or harm. CIRT will use standardized central methodology designed to ensure consistent performance of all dose adjustments and safety interventions at each clinical site in a manner that protects the blinding to treatment but maintains safety for enrolled participants. SUMMARY: CIRT aims to test the inflammatory hypothesis of atherothrombosis in patients with prior MI and either type 2 diabetes or metabolic syndrome, conditions associated with persistent inflammation. If low-dose methotrexate reduces cardiovascular events, CIRT would provide a novel therapeutic approach for the secondary prevention of heart attack, stroke, and cardiovascular death.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Diabetes Mellitus, Type 2/complications , Inflammation/drug therapy , Metabolic Syndrome/complications , Methotrexate/therapeutic use , Myocardial Infarction/drug therapy , Algorithms , Anti-Inflammatory Agents/administration & dosage , Atherosclerosis , Cardiovascular Diseases/mortality , Cardiovascular Diseases/prevention & control , Humans , Inflammation/complications , Methotrexate/administration & dosage , Myocardial Infarction/complications , Research Design
5.
Microbiology (Reading) ; 156(Pt 12): 3660-3668, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20705662

ABSTRACT

Since the late 1980s, a worldwide increase of severe Streptococcus pyogenes infections has been associated with strains of the M1 serotype, strains which all secrete the streptococcal inhibitor of complement-mediated lysis (SIC). Previous work has shown that SIC blocks complement-mediated haemolysis, inhibits the activity of antibacterial peptides and has affinity for the human plasma proteins clusterin and histidine-rich glycoprotein; the latter is a member of the cystatin protein family. The present work demonstrates that SIC binds to cystatin C, high-molecular-mass kininogen (HK) and low-molecular-mass kininogen, which are additional members of this protein family. The binding sites in HK are located in the cystatin-like domain D3 and the endothelial cell-binding domain D5. Immobilization of HK to cellular structures plays a central role in activation of the human contact system. SIC was found to inhibit the binding of HK to endothelial cells, and to reduce contact activation as measured by prolonged blood clotting time and impaired release of bradykinin. These results suggest that SIC modifies host defence systems, which may contribute to the virulence of S. pyogenes strains of the M1 serotype.


Subject(s)
Bacterial Proteins/immunology , Streptococcal Infections/microbiology , Streptococcus pyogenes/immunology , Streptococcus pyogenes/pathogenicity , Bacterial Proteins/genetics , Complement System Proteins , Cystatin C/immunology , Host-Pathogen Interactions , Humans , Kininogen, High-Molecular-Weight/immunology , Protein Binding , Streptococcal Infections/immunology , Streptococcus pyogenes/genetics , Virulence
7.
FEBS Lett ; 579(1): 25-9, 2005 Jan 03.
Article in English | MEDLINE | ID: mdl-15620686

ABSTRACT

The angiotensin converting enzyme breakdown product of bradykinin, bradykinin 1-5 (RPPGF), inhibits thrombin-induced human or mouse platelet aggregation. RPPGF binds to the exodomain of human protease-activated receptor 1 (PAR1). Studies determined if RPPGF also binds to the exodomain of human PAR4. RPPGF binds to a peptide of the thrombin cleavage site on PAR4. Recombinant wild-type and mutated exodomain of human PAR4 was prepared. The N-terminal arginine on RPPGF binds to the P2 position or proline46 on PAR4 to block thrombin cleavage. These data indicate that RPPGF influences thrombin activity by binding to the thrombin cleavage site on both PAR4 and PAR1.


Subject(s)
Bradykinin/chemistry , Bradykinin/metabolism , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Receptors, Thrombin/chemistry , Receptors, Thrombin/metabolism , Binding, Competitive , Biotin/chemistry , Humans , Protein Interaction Mapping , Protein Structure, Tertiary/genetics , Receptors, Thrombin/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thrombin/antagonists & inhibitors
8.
J Pharmacol Exp Ther ; 311(2): 492-501, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15210836

ABSTRACT

Thrombin and protease-activated receptor 1 (PAR1) activation antagonists were prepared based upon the peptide RPPGF, the angiotensin-converting enzyme breakdown product of bradykinin. A library of 72 peptides consisting of d and/or synthetic amino acids was designed with various substitutions in positions 1 to 5 in Arg-Pro-Pro-Gly-Phe (RPPGF). Two compounds, rOicPGF (TH146) and betaAK2K-4(rOicPGF) (MAP4-TH146), were characterized further. TH146 or MAP4-TH146 completely inhibits threshold gamma-thrombin-induced platelet aggregation at a concentration of 142 +/- 0.05 or 19 +/- 0.06 microM, respectively. TH146 completely inhibits threshold alpha-thrombin-induced washed platelet aggregation at 444 +/- 0.04 microM. TH146 or MAP4-TH146 blocks 2 nM alpha-thrombin-induced fibroblast calcium mobilization with an IC(50) value of 110 or 18 microM, respectively. Furthermore, significant prolongation of the activated partial thromboplastin time, prothrombin time, or thrombin clotting time occurs at 31, 62, or 7.8 microM TH146 and 0.4, 6.25, or 1.56 microM MAP4-TH146, respectively. TH146 and MAP4-TH146 inhibit both alpha-thrombin with a K(i) value of 97 and 49 microM, respectively, and factor VIIa with a K(i) value of 44 and 5 microM, respectively. Both TH146 and MAP4-TH146 specifically bind to the exodomain of recombinant PAR1. MAP4-TH146 (200 microM) completely blocks thrombocytin, a PAR1-activating snake venom protease, without inhibiting the enzyme's active site. TH146 inhibits gamma-thrombin-induced aggregation of mouse platelets, prolongs mouse bleeding times, and delays the time to mouse carotid artery thrombosis. TH146 and MAP4-TH146 inhibit human and mouse platelet aggregation and mouse thrombosis. Analogs of RPPGF are model compounds to develop PAR1 activation antagonists as well as direct inhibitors to thrombin and factor VIIa.


Subject(s)
Bradykinin/analogs & derivatives , Bradykinin/pharmacology , Factor VIIa/antagonists & inhibitors , Peptide Fragments/pharmacology , Receptor, PAR-1/antagonists & inhibitors , Thrombin/antagonists & inhibitors , Animals , Bleeding Time , Blood Pressure/drug effects , Bradykinin/blood , Bradykinin/therapeutic use , Disease Models, Animal , Humans , Mice , Peptide Fragments/blood , Peptide Fragments/therapeutic use , Peptides/pharmacology , Platelet Aggregation/drug effects , Thrombosis/blood , Thrombosis/prevention & control
9.
Am J Physiol Heart Circ Physiol ; 285(1): H183-93, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12598231

ABSTRACT

Investigations determined the mechanism(s) by which Arg-Pro-Pro-Gly-Phe (RPPGF) inhibits thrombin-induced platelet activation. High concentrations of RPPGF inhibit thrombin-induced coagulant activity. RPPGF binds to the active site of thrombin by forming a parallel beta-strand with Ser214-Gly216 and interacts with His57, Asp189, and Ser195 of the catalytic triad. RPPGF competitively inhibits alpha-thrombin from hydrolyzing Sar-Pro-Arg-paranitroanilide with a Ki = 1.75 +/- 0.03 mM. Other mechanisms were sought to explain why RPPGF inhibits thrombin activation of platelets at concentrations below that which inhibits its active site. Soluble RPPGF blocks biotinylated NATLDPRSFLLR of the thrombin cleavage site on protease-activated receptor (PAR)1 from binding to the peptide RPPGC (IC50 = 20 microM). The soluble recombinant extracellular domain of PAR1 (rPAR1EC) blocks biotinylated RPPGF binding to rPAR1EC (IC50 = 50 microM) bound to microtiter plates, but rPAR1EC deletion mutants missing the sequence LDPR or PRSF do not. RPPGF and related forms prevent the thrombin-like enzyme thrombocytin from proteolyzing rPAR1EC at concentrations that do not block thrombocytin's active site. These studies indicate that RPPGF is a bifunctional inhibitor of thrombin: it binds to PAR1 to prevent thrombin cleavage at Arg41 and interacts with the active site of alpha-thrombin.


Subject(s)
Bradykinin/pharmacology , Hemostatics/antagonists & inhibitors , Peptide Fragments/pharmacology , Thrombin/antagonists & inhibitors , Anticoagulants/pharmacology , Blood Platelets/drug effects , Crotalid Venoms/metabolism , DNA, Complementary/biosynthesis , DNA, Complementary/genetics , Humans , In Vitro Techniques , Kinetics , Models, Molecular , Molecular Conformation , Mutagenesis, Site-Directed , Peptides/pharmacology , Protein Binding , Receptor, PAR-1 , Receptors, Thrombin/drug effects , Receptors, Thrombin/metabolism , Serine Endopeptidases/metabolism
10.
Cardiovasc Res ; 53(4): 984-92, 2002 Mar.
Article in English | MEDLINE | ID: mdl-11922908

ABSTRACT

OBJECTIVE: Thrombin activates platelets and contributes to the occlusion of arteries following thrombolytic therapy or angioplasty. Thrombostatin (RPPGF), the angiotensin converting enzyme degradation product of bradykinin, inhibits alpha-thrombin induced platelet activation. We hypothesized that thrombostatin prevents platelet aggregation and adhesion after balloon angioplasty (BA). METHODS: Platelet-rich plasma (PRP) was obtained from 22 Beagle dogs before sacrifice and 10% of the PRP was labeled with 111In. Carotid arteries were then removed from each dog and mounted in a dual perfusion chamber and intimal injury was performed with BA. 111In-PRP with or without thrombostatin or aspirin alone was perfused through the arteries for 60 min. During perfusion, platelet volume was measured using a Coulter counter and a laser-light scattering technique. Platelet adhesion to arteries was measured by radioactivity count. RESULTS: Arterial injury alone compared to non-injury increased platelet volume in the circuit by 1.4 times (x) (P<0.05) using a Coulter counter or 1.8x (P<0.05) using laser-light scattering and increased platelet adhesion by 2.3x (P<0.01). When compared to BA injury alone, the addition of thrombostatin reduced platelet volume by 1.8x (P<0.03) as measured by Coulter counter or 1.9x (P<0.01) by laser-light scattering and platelet adhesion by 4.2x (P<0.05). Compared to BA injury alone, aspirin reduced platelet volume by 1.2x (P<0.01) as assessed by Coulter counter or 1.5x (P<0.03) using laser-light scattering and platelet adhesion by 1.8x (P<0.02). CONCLUSION: Thrombostatin or aspirin independently decreases evidence of platelet activation in the canine carotid artery model of BA injury.


Subject(s)
Bradykinin/metabolism , Bradykinin/pharmacology , Carotid Artery Injuries/pathology , Catheterization/adverse effects , Peptide Fragments/pharmacology , Platelet Activation/drug effects , Animals , Aspirin/pharmacology , Blood Platelets/drug effects , Blood Platelets/pathology , Carotid Arteries/ultrastructure , Carotid Artery Injuries/etiology , Carotid Stenosis/therapy , Cell Size/drug effects , Disease Models, Animal , Dogs , Lasers , Microscopy, Electron, Scanning , P-Selectin/metabolism , Platelet Aggregation , Platelet Aggregation Inhibitors/pharmacology , Scattering, Radiation
SELECTION OF CITATIONS
SEARCH DETAIL
...