98 results on '"Ware JA"'
Search Results
2. Commentary on: 'Influence of OATP1B1 Function on the Disposition of Sorafenib-beta-D-Glucuronide'
- Author
-
Morrissey, KM, Benet, LZ, and Ware, JA
- Published
- 2017
- Full Text
- View/download PDF
3. Platelet activation and subsequent inhibition by plasmin and recombinant tissue-type plasminogen activator
- Author
-
Penny, WF, primary and Ware, JA, additional
- Published
- 1992
- Full Text
- View/download PDF
4. Platelet activation by a synthetic hydrophobic polymer, polymethylmethacrylate
- Author
-
Ware, JA, primary, Kang, J, additional, DeCenzo, MT, additional, Smith, M, additional, Watkins, SC, additional, Slayter, HS, additional, and Saitoh, M, additional
- Published
- 1991
- Full Text
- View/download PDF
5. Endothelial cell activation in patients with decompensated heart failure.
- Author
-
Colombo PC, Banchs JE, Celaj S, Talreja A, Lachmann J, Malla S, DuBois NB, Ashton AW, Latif F, Jorde UP, Ware JA, and LeJemtel TH
- Published
- 2005
6. Abnormalities of cytoplasmic Ca2+ in platelets from patients with uremia
- Author
-
Ware, JA, Clark, BA, Smith, M, and Salzman, EW
- Abstract
Uremic patients have a hemorrhagic tendency, often associated with prolonged bleeding times and decreased platelet function in vitro. Whether these defects result from abnormalities in plasma factors affecting platelet activity, platelet surface receptors, intracellular platelet mediators, or other aspects of platelet behavior is unknown. To examine the possibility that the abnormality in platelet function may result from aberrations in Ca2+ homeostasis, blood was obtained from 29 patients with severe uremia. The platelets were washed, loaded with the Ca2+ -sensitive probes indo-1 and aequorin, gel-filtered, and resuspended in either plasma or buffer. Of the 29 patients, seven had template bleeding times prolonged to 11 minutes or more, but platelet aggregation in plasma was not consistently impaired in these patients. However, in aequorin-loaded platelets from the patients with long bleeding times, the highest elevation of cytoplasmic calcium [( Ca2+]i) in response to the Ca2+ ionophore A23187, arachidonate, adenosine diphosphate (ADP), or epinephrine was lower than that seen in platelets from both uremic patients with less prolonged bleeding times and normal volunteers. The reduced [Ca2+]i response was associated with decreased aggregation of gel-filtered platelets suspended in buffer. Suspending washed aequorin-loaded uremic platelets in normal plasma for 20 minutes did not reverse the decreased agonist-induced rise in [Ca2+]i; platelets from a normal donor resuspended in uremic plasma aggregated and produced a normal increase in [Ca2+]i in response to agonists. We conclude that the platelet defect seen in some patients with uremia is associated with a decreased rise in platelet [Ca2+]i after stimulation and that this is a manifestation of an intrinsic platelet defect.
- Published
- 1989
- Full Text
- View/download PDF
7. Activation of protein kinase C in platelets by epinephrine and A23187: correlation with fibrinogen binding
- Author
-
Saitoh, M, Salzman, EW, Smith, M, and Ware, JA
- Abstract
Activation of protein kinase C (PKC), as revealed by phosphorylation of a 47 kd protein (p47), occurs in platelets stimulated by some agonists (eg, thrombin or phorbol esters). It is not known if activation of PKC occurs with pairs of agonists, such as epinephrine and A23187, that do not individually phosphorylate p47, nor is it known what role the concentration of cytoplasmic Ca++ ([Ca++]i) plays in these events. We stimulated aequorin-loaded platelets with subaggregating concentrations of epinephrine and A23187, neither of which by itself phosphorylated p47. The combination of agonists resulted in p47 phosphorylation, an increase in platelet-bound fibrinogen, and aggregation, but only if the concentration of each agonist was sufficient to increase [Ca++]i if it was added separately. Subaggregating concentrations of A23187 alone released platelet fibrinogen and increased platelet membrane binding of [3H]-phorbol dibutyrate, but these were not enhanced by epinephrine. Epinephrine and A23187 did not increase production of diacylglycerol. Thus, epinephrine and A23187 together activate PKC by a mechanism that does not require phospholipase C or enhanced binding of PKC to the plasma membrane; PKC activation in turn is correlated with enhanced platelet fibrinogen binding and aggregation. These events require an initial elevation of [Ca++]i above a threshold.
- Published
- 1989
- Full Text
- View/download PDF
8. Changes in von Willebrand factor during cardiac surgery: effect of desmopressin acetate
- Author
-
Weinstein, M, Ware, JA, Troll, J, and Salzman, E
- Abstract
Patients who receive desmopressin acetate (dDAVP) after cardiopulmonary bypass bleed less during operation and in the first 24 hours after operation than do patients who receive a placebo. To study the mechanism of improved hemostasis in bypass patients, we examined the relationship between von Willebrand factor (vWF) and blood loss in 70 cardiopulmonary bypass patients, one-half of whom received desmopressin intraoperatively. vWF concentration and multimeric composition were analyzed before and after bypass, after drug treatment, and 24 hours after operation. Before operation, patients with valvular disease had lower percentages of vWF high-mol-wt multimers (HMWMs) than did healthy subjects or patients with coronary artery disease, but subsequent blood loss, vWF activity, and bleeding times were not related to this finding. Irrespective of drug treatment, patients who had low preoperative vWF and who had a net loss of the protein during bypass bled more after bypass than did similar patients who had a net increase of vWF during bypass. HMWMs rose to above normal levels after bypass regardless of desmopressin infusion. Differences in the concentration of vWF between desmopressin and placebo patients after receipt of the drug, although small, were better correlated with reduced blood loss than were differences in HMWM distribution. We conclude that the beneficial effect of desmopressin on hemostasis following cardiopulmonary bypass cannot be attributed to a drug-induced change in HMWM distribution but may be related to an increase in overall vWF concentration.
- Published
- 1988
- Full Text
- View/download PDF
9. Platelet aggregation by fibrinogen polymers crosslinked across the E domain
- Author
-
McManama, G, Lindon, JN, Kloczewiak, M, Smith, MA, Ware, JA, Hawiger, J, Merrill, EW, and Salzman, EW
- Abstract
There is evidence that platelet interactions with artificial surfaces are mediated by plasma proteins, especially fibrinogen, adsorbed on the surfaces. Multiple site interactions between fibrinogen molecules adsorbed in high concentration and receptors in the unactivated platelet may be sufficient for platelet adhesion and subsequent activation. To examine this hypothesis, we prepared soluble polymers of fibrinogen. Polymers produced by interaction of fibrinogen with Fab'2 fragments of antibodies against fibrinogen's E (central) domain (Fg- Fab'2(E] induced, in gel-filtered platelets, aggregation and serotonin release, which were blocked by monoclonal antibodies against the GPIIb/IIIa complex, by Fab fragments against the D domain, and by metabolic inhibitors; aggregation was attenuated but not abolished by enzymatic removal of ADP (with CP/CPK) or by blockage of ADP binding sites (with FSBA), and when secretion was inhibited by aspirin. Fg- Fab'2(E) also induced a dose-dependent elevation in cytoplasmic Ca2+ (measured by Aequorin luminescence) which was attenuated by CP/CPK and by FSBA, and was eliminated by metabolic inhibitors and by anti- IIb/IIIa antibody. Fibrinogen complexes crosslinked with dimethylsuberimidate or Factor XIII neither aggregated gel-filtered platelets nor inhibited platelet aggregation by ADP and fibrinogen, probably because of inaccessibility of lysine residues in the D (terminal) domain of fibrinogen, which are thought to be required for platelet binding. Thus, soluble complexes of fibrinogen having multiple available platelet receptor recognition sites activate gel-filtered platelets and may provide a useful model for platelet-surface interactions mediated by adsorbed fibrinogen.
- Published
- 1986
- Full Text
- View/download PDF
10. An assessment of functioning and non-functioning distractors in multiple-choice questions: a descriptive analysis
- Author
-
Mohammed Ahmed M, Ware James, and Tarrant Marie
- Subjects
Special aspects of education ,LC8-6691 ,Medicine - Abstract
Abstract Background Four- or five-option multiple choice questions (MCQs) are the standard in health-science disciplines, both on certification-level examinations and on in-house developed tests. Previous research has shown, however, that few MCQs have three or four functioning distractors. The purpose of this study was to investigate non-functioning distractors in teacher-developed tests in one nursing program in an English-language university in Hong Kong. Methods Using item-analysis data, we assessed the proportion of non-functioning distractors on a sample of seven test papers administered to undergraduate nursing students. A total of 514 items were reviewed, including 2056 options (1542 distractors and 514 correct responses). Non-functioning options were defined as ones that were chosen by fewer than 5% of examinees and those with a positive option discrimination statistic. Results The proportion of items containing 0, 1, 2, and 3 functioning distractors was 12.3%, 34.8%, 39.1%, and 13.8% respectively. Overall, items contained an average of 1.54 (SD = 0.88) functioning distractors. Only 52.2% (n = 805) of all distractors were functioning effectively and 10.2% (n = 158) had a choice frequency of 0. Items with more functioning distractors were more difficult and more discriminating. Conclusion The low frequency of items with three functioning distractors in the four-option items in this study suggests that teachers have difficulty developing plausible distractors for most MCQs. Test items should consist of as many options as is feasible given the item content and the number of plausible distractors; in most cases this would be three. Item analysis results can be used to identify and remove non-functioning distractors from MCQs that have been used in previous tests.
- Published
- 2009
- Full Text
- View/download PDF
11. Safety and efficacy of bivalirudin with and without glycoprotein IIb/IIIa inhibitors in patients with acute coronary syndromes undergoing percutaneous coronary intervention 1-year results from the ACUITY (Acute Catheterization and Urgent Intervention Triage strategY) trial.
- Author
-
White HD, Ohman EM, Lincoff AM, Bertrand ME, Colombo A, McLaurin BT, Cox DA, Pocock SJ, Ware JA, Manoukian SV, Lansky AJ, Mehran R, Moses JW, Stone GW, White, Harvey D, Ohman, E Magnus, Lincoff, A Michael, Bertrand, Michel E, Colombo, Antonio, and McLaurin, Brent T
- Abstract
Objectives: This study was designed to determine the impact of bivalirudin on 1-year outcomes in acute coronary syndrome (ACS) patients undergoing percutaneous coronary intervention (PCI).Background: The ACUITY (Acute Catheterization and Urgent Intervention Triage strategY) trial demonstrated that in moderate- and high-risk ACS patients undergoing PCI, bivalirudin alone compared to unfractionated heparin (UFH) or enoxaparin plus a glycoprotein (GP) IIb/IIIa inhibitor resulted in less major bleeding and similar ischemic outcomes at 30 days. The impact of bivalirudin on 1-year outcomes in ACS patients undergoing PCI is unknown.Methods: In the ACUITY trial, 13,819 patients were enrolled, and 7,789 (56.4%) patients had PCI. Composite ischemia (death, myocardial infarction, or unplanned revascularization) and mortality at 1 year were assessed.Results: Among patients undergoing PCI, 2,561, 2,609, and 2,619 were randomized to UFH or enoxaparin plus a GP IIb/IIIa inhibitor, bivalirudin plus a GP IIb/IIIa inhibitor, and bivalirudin monotherapy, respectively. At 1 year, there were no differences in composite ischemia (17.8% vs. 19.4% vs. 19.2%, p = NS) or mortality (3.2% vs. 3.3% vs. 3.1%, p = NS) among the 3 groups, respectively.Conclusions: Bivalirudin compared with UFH or enoxaparin plus a GP IIb/IIIa inhibitor results in similar rates of composite ischemia and mortality at 1 year in moderate- and high-risk ACS patients undergoing PCI. [ABSTRACT FROM AUTHOR]- Published
- 2008
- Full Text
- View/download PDF
12. Bioavailability of acalabrutinib suspension delivered via nasogastric tube in the presence or absence of a proton pump inhibitor in healthy subjects.
- Author
-
Sharma S, Pepin X, Cheung J, Zheng L, Wei H, Townsley D, Han D, Majewski M, Ware JA, Mann J, Munugalavadla V, Sheridan L, Patel P, Gupta A, and Tomkinson H
- Subjects
- Adult, Benzamides, Biological Availability, Cross-Over Studies, Healthy Volunteers, Humans, Pyrazines, Suspensions, Critical Illness, Proton Pump Inhibitors adverse effects, Proton Pump Inhibitors pharmacokinetics
- Abstract
Aims: Acalabrutinib, a selective Bruton tyrosine kinase inhibitor, is approved for the treatment of mantle cell lymphoma and chronic lymphocytic leukaemia. Many critically ill patients are unable to swallow and need oral medications to be delivered via a nasogastric (NG) tube. Furthermore, critically ill patients are typically administered proton-pump inhibitors (PPIs) to prevent stress ulcers. Concomitant administration with PPIs reduces acalabrutinib exposure and is not currently recommended. To evaluate acalabrutinib in subjects co-administered with PPIs who require NG delivery, a phase 1, open-label, randomized, crossover, single-dose study was conducted in healthy subjects., Methods: The study assessed the relative bioavailability of an acalabrutinib suspension-in regular, degassed Coca-Cola-administered via NG tube (Acala-NG) versus the pharmacokinetics (PK) of an acalabrutinib capsule administered orally with water. In addition, the PPI effect was evaluated by comparing the PK following Acala-NG in the presence or absence of rabeprazole., Results: Exposure of acalabrutinib and its active metabolite (ACP-5862) were comparable following administration of Acala-NG versus the oral capsule (Geo mean ratio, % ref [90% confidence interval, CI]: acalabrutinib AUC
inf : 103 [93-113]; Cmax : 144 [120-173]). In addition, exposure was similar following administration of Acala-NG with and without a PPI (Geo mean ratio, % ref [90% CI]: acalabrutinib AUCinf : 105 [79-138]; Cmax : 95 [66-137]). No safety or tolerability concerns were observed, and all adverse events were mild and resolved without treatment., Conclusions: Acala-NG with or without a PPI is safe and well-tolerated without impeding bioavailability., (© 2022 British Pharmacological Society.)- Published
- 2022
- Full Text
- View/download PDF
13. Quantitative systems pharmacology model-based investigation of adverse gastrointestinal events associated with prolonged treatment with PI3-kinase inhibitors.
- Author
-
Gadkar K, Friedrich C, Hurez V, Ruiz ML, Dickmann L, Kumar Jolly M, Schutt L, Jin J, Ware JA, and Ramanujan S
- Subjects
- Diarrhea chemically induced, Humans, Network Pharmacology, Phosphoinositide-3 Kinase Inhibitors, Protein Isoforms, Colitis chemically induced, Phosphatidylinositol 3-Kinases
- Abstract
Several PI3K inhibitors are in clinical development for the treatment of various forms of cancers, including pan-PI3K inhibitors targeting all four PI3K isoforms (α, β, γ, and δ), and isoform-selective inhibitors. Diarrhea and immune-mediated colitis are among the adverse events observed with PI3K inhibition which limits the maximal tolerated dose. A quantitative systems pharmacology model was developed to investigate PI3K-inhibitor-induced colitis. The effects of individual PI3K isoforms on relevant cellular pathways were incorporated into a mechanistic representation of mucosal inflammation. A virtual clinical population captures the observed clinical variability in the onset timing and rates of diarrhea and colitis for seven clinically tested PI3K inhibitors. Model-based analysis suggests that colitis development is governed by both the inhibition of PI3Kδ, which drives T cell differentiation and proliferation, and PI3Kα, which regulates epithelial barrier integrity. Specifically, when PI3Kα is inhibited below a given threshold, epithelial barrier dysfunction precipitates an exaggerated T effector response due to PI3Kδ-inhibition, leading to risk of diarrhea and colitis. This synergy explains why the lowest diarrhea and colitis rates are seen with the weakest PI3Kδ inhibition (alpelisib), and higher rates are seen with strong PI3Kδ inhibition if PI3Kα is even mildly inhibited (e.g., idelalisib), whereas strong PI3Kδ inhibition in the absence of PI3Kα inhibition does not result in high colitis rates (umbralisib). Thus, the model-based analysis suggests that PI3Kα and δ inhibition play unique but synergistic roles in driving colitis. Finally, we explore if and how dose-regimen might influence colitis rates for molecules that inhibit both PI3Kα and PI3Kδ., (© 2021 Genentech, Inc. CPT: Pharmacometrics & Systems Pharmacology published by Wiley Periodicals LLC on behalf of American Society for Clinical Pharmacology and Therapeutics.)
- Published
- 2022
- Full Text
- View/download PDF
14. Intrinsic and Extrinsic Pharmacokinetic Variability of Small Molecule Targeted Cancer Therapy.
- Author
-
Reyner E, Lum B, Jing J, Kagedal M, Ware JA, and Dickmann LJ
- Subjects
- Administration, Oral, Adult, Age Factors, Antineoplastic Agents administration & dosage, Area Under Curve, Biological Variation, Population, Body Mass Index, Drug Approval, Europe, Female, Healthy Volunteers, Humans, Male, Middle Aged, Molecular Targeted Therapy methods, Neoplasms blood, United States, United States Food and Drug Administration, Antineoplastic Agents pharmacokinetics, Neoplasms drug therapy
- Abstract
Pharmacokinetic (PK) variability in cancer clinical trials may be due to heterogeneous populations and identifying sources of variability is important. Use of healthy subjects in clinical pharmacology studies together with detailed knowledge of the characteristics of patients with cancer can allow for quick identification and quantification of factors affecting PK variability. PK data and sources of variability of 40 marketed molecularly targeted oncology therapeutics were compiled from regulatory approval documents covering an 18-year period (1999-2017). Variability in PK parameters was compared and contributors to variability were identified. The results show that PK variability was ~ 16% higher for peak plasma concentration (C
max ) and area under the concentration time curve (AUC) in patients with cancer compared with healthy subjects. Several factors were identified as major contributors to variability including hepatic/renal impairment and cytochrome P450 inhibition/induction. Lower PK variability in healthy subjects may represent an opportunity to perform rapid and robust pharmacological and PK assessments to inform subsequent studies in the development of new cancer therapies., (© 2019 Genentech Inc. Clinical and Translational Science published by Wiley Periodicals, Inc. on behalf of the American Society for Clinical Pharmacology and Therapeutics.)- Published
- 2020
- Full Text
- View/download PDF
15. Investigation of the absolute bioavailability and human mass balance of navoximod, a novel IDO1 inhibitor.
- Author
-
Ma S, Suchomel J, Yanez E, Yost E, Liang X, Zhu R, Le H, Siebers N, Joas L, Morley R, Royer-Joo S, Pirzkall A, Salphati L, Ware JA, and Morrissey KM
- Subjects
- Administration, Intravenous, Administration, Oral, Adult, Biological Availability, Cross-Over Studies, Female, Healthy Volunteers, Humans, Imidazoles administration & dosage, Indoles administration & dosage, Intestinal Elimination, Male, Metabolic Clearance Rate, Middle Aged, Neoplasms drug therapy, Neoplasms immunology, Renal Elimination, Tumor Escape drug effects, Young Adult, Imidazoles pharmacokinetics, Indoleamine-Pyrrole 2,3,-Dioxygenase antagonists & inhibitors, Indoles pharmacokinetics
- Abstract
Aims: Navoximod (GDC-0919, NLG-919) is a small molecule inhibitor of indoleamine-2,3-dioxygenase 1 (IDO1), developed to treat the acquired immune tolerance associated with cancer. The primary objectives of this study were to assess navoximod's absolute bioavailability (aBA), determine the mass balance and routes of elimination of [
14 C]-navoximod, and characterize navoximod's metabolite profile., Methods: A phase 1, open-label, two-part study was conducted in healthy volunteers. In Part 1 (aBA), subjects (n = 16) were randomized to receive oral (200 mg tablet) or intravenous (5 mg solution) navoximod in a crossover design with a 5-day washout. In Part 2 (mass balance), subjects (n = 8) were administered [14 C]-navoximod (200 mg/600 μCi) as an oral solution., Results: The aBA of navoximod was estimated to be 55.5%, with a geometric mean (%CV) plasma clearance and volume of distribution of 62.0 L/h (21.0%) and 1120 L (28.4%), respectively. Mean recovery of total radioactivity was 87.8%, with 80.4% detected in urine and the remainder (7.4%) in faeces. Navoximod was extensively metabolized, with unchanged navoximod representing 5.45% of the dose recovered in the urine and faeces. Glucuronidation was identified as the primary route of metabolism, with the major glucuronide metabolite, M28, accounting for 57.5% of the total drug-derived exposure and 59.7% of the administered dose recovered in urine., Conclusions: Navoximod was well tolerated, quickly absorbed and showed moderate bioavailability, with minimal recovery of the dose as unchanged parent in the urine and faeces. Metabolism was identified as the primary route of clearance and navoximod glucuronide (M28) was the most abundant metabolite in circulation with all other metabolites accounting for <10% of drug-related exposure., (© 2019 Genentech Inc. British Journal of Clinical Pharmacology published by John Wiley & Sons Ltd on behalf of British Pharmacological Society.)- Published
- 2019
- Full Text
- View/download PDF
16. A Phase I Dose-Escalation Study of the Safety and Pharmacokinetics of Pictilisib in Combination with Erlotinib in Patients with Advanced Solid Tumors.
- Author
-
Leong S, Moss RA, Bowles DW, Ware JA, Zhou J, Spoerke JM, Lackner MR, Shankar G, Schutzman JL, van der Noll R, Voest EE, and Schellens JHM
- Subjects
- Adult, Aged, Antineoplastic Combined Chemotherapy Protocols administration & dosage, Cell Proliferation genetics, Dose-Response Relationship, Drug, ErbB Receptors antagonists & inhibitors, Erlotinib Hydrochloride adverse effects, Erlotinib Hydrochloride pharmacokinetics, Female, Humans, Indazoles adverse effects, Indazoles pharmacokinetics, Male, Maximum Tolerated Dose, Middle Aged, Neoplasm Staging, Neoplasms genetics, Neoplasms pathology, Phosphatidylinositol 3-Kinases genetics, Phosphoinositide-3 Kinase Inhibitors, Protein Kinase Inhibitors adverse effects, Protein Kinase Inhibitors pharmacokinetics, Sulfonamides adverse effects, Sulfonamides pharmacokinetics, ErbB Receptors genetics, Erlotinib Hydrochloride administration & dosage, Indazoles administration & dosage, Neoplasms drug therapy, Protein Kinase Inhibitors administration & dosage, Sulfonamides administration & dosage
- Abstract
Background: Epidermal growth factor receptor (EGFR) and phosphatidylinositol 3-kinase (PI3K) are involved in the proliferation and survival of many cancer types. Enhanced antitumor activity may be achieved through combined inhibition of these pathways. We report results for pictilisib (GDC-0941, a class I pan-PI3K inhibitor) plus erlotinib (an EGFR tyrosine kinase inhibitor) in patients with advanced solid tumors., Materials and Methods: A 3 + 3 dose-escalation study was carried out at a starting daily dose of 60 mg pictilisib on days 1-21 of a 28-day cycle and 150 mg erlotinib from day 2 of cycle 1. The primary objectives of the study were to assess safety and tolerability, identify dose-limiting toxicities (DLTs), estimate the maximum tolerated dose, and identify the recommended phase II dose (RP2D). Evaluation of a dose-expansion cohort at the RP2D was performed., Results: Fifty-seven patients were treated in the study. All patients experienced at least one adverse event (AE). Grade ≥3 AEs, serious AEs, and deaths were reported in 38 (66.7%), 19 (33.3%), and 4 (7.0%) patients, respectively. DLTs occurred in nine patients across eight cohorts and the RP2D was determined to be 340 mg pictilisib on a "5 days on, 2 days off" schedule plus 100 mg erlotinib. Two patients (3.5%) experienced partial response and 19 (33.3%) had stable disease., Conclusion: Combining pictilisib with erlotinib in patients with advanced solid tumors is feasible; however, antitumor activity is limited. Additional studies may identify patients likely to benefit from combined inhibition of EGFR and PI3K pathways., Implications for Practice: Combining drugs targeting different signaling pathways in cancer growth and survival could overcome drug resistance and improve antitumor activity. In this first-in-human study for the combination, addition of the PI3K inhibitor pictilisib to the EGFR tyrosine kinase inhibitor erlotinib resulted in toxicity that led to dose and schedule modifications to identify a tolerable recommended phase II dose of 340 mg pictilisib on a "5 days on, 2 days off" schedule plus 100 mg erlotinib daily. The limited antitumor activity observed, however, suggests that additional studies are needed to identify patients most likely to benefit from combined EGFR and PI3K inhibition., Competing Interests: Disclosures of potential conflicts of interest may be found at the end of this article., (© AlphaMed Press 2017.)
- Published
- 2017
- Full Text
- View/download PDF
17. Combining "Bottom-up" and "Top-down" Approaches to Assess the Impact of Food and Gastric pH on Pictilisib (GDC-0941) Pharmacokinetics.
- Author
-
Lu T, Fraczkiewicz G, Salphati L, Budha N, Dalziel G, Smelick GS, Morrissey KM, Davis JD, Jin JY, and Ware JA
- Subjects
- Administration, Oral, Biological Availability, Computer Simulation, Cross-Over Studies, Diet, High-Fat, Female, Healthy Volunteers, Humans, Hydrogen-Ion Concentration, Male, Models, Biological, Random Allocation, Anti-Ulcer Agents administration & dosage, Indazoles administration & dosage, Indazoles pharmacokinetics, Intestines chemistry, Rabeprazole administration & dosage, Sulfonamides administration & dosage, Sulfonamides pharmacokinetics
- Abstract
Pictilisib, a weakly basic compound, is an orally administered, potent, and selective pan-inhibitor of phosphatidylinositol 3-kinases for oncology indications. To investigate the significance of high-fat food and gastric pH on pictilisib pharmacokinetics (PK) and enable label recommendations, a dedicated clinical study was conducted in healthy volunteers, whereby both top-down (population PK, PopPK) and bottom-up (physiologically based PK, PBPK) approaches were applied to enhance confidence of recommendation and facilitate the clinical development through scenario simulations. The PopPK model identified food (for absorption rate constant (K
a )) and proton pump inhibitors (PPI, for relative bioavailability (Frel ) and Ka ) as significant covariates. Food and PPI also impacted the variability of Frel . The PBPK model accounted for the supersaturation tendency of pictilisib, and gastric emptying physiology successfully predicted the food and PPI effect on pictilisib absorption. Our research highlights the importance of applying both quantitative approaches to address critical drug development questions., (© 2017 The Authors CPT: Pharmacometrics & Systems Pharmacology published by Wiley Periodicals, Inc. on behalf of American Society for Clinical Pharmacology and Therapeutics.)- Published
- 2017
- Full Text
- View/download PDF
18. Phase I Study of Apitolisib (GDC-0980), Dual Phosphatidylinositol-3-Kinase and Mammalian Target of Rapamycin Kinase Inhibitor, in Patients with Advanced Solid Tumors.
- Author
-
Dolly SO, Wagner AJ, Bendell JC, Kindler HL, Krug LM, Seiwert TY, Zauderer MG, Lolkema MP, Apt D, Yeh RF, Fredrickson JO, Spoerke JM, Koeppen H, Ware JA, Lauchle JO, Burris HA 3rd, and de Bono JS
- Subjects
- Antineoplastic Agents adverse effects, Antineoplastic Agents pharmacokinetics, Bridged Bicyclo Compounds, Heterocyclic adverse effects, Bridged Bicyclo Compounds, Heterocyclic pharmacokinetics, Female, Humans, Male, Middle Aged, Protein Kinase Inhibitors adverse effects, Protein Kinase Inhibitors pharmacokinetics, Pyrimidines adverse effects, Pyrimidines pharmacokinetics, Antineoplastic Agents therapeutic use, Bridged Bicyclo Compounds, Heterocyclic therapeutic use, Class I Phosphatidylinositol 3-Kinases antagonists & inhibitors, Neoplasms drug therapy, Protein Kinase Inhibitors therapeutic use, Pyrimidines therapeutic use, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Purpose: This first-in-human phase I trial assessed the safety, tolerability, and preliminary antitumor activity of apitolisib (GDC-0980), a dual inhibitor of class I PI3K, and mTOR kinases., Experimental Design: Once-daily oral apitolisib was administered to patients with solid tumors for days 1 to 21 or 1 to 28 of 28-day cycles. Pharmacokinetic and pharmacodynamic parameters were assessed., Results: Overall, 120 patients were treated at doses between 2 and 70 mg. The commonest ≥G3 toxicities related to apitolisib at the recommended phase 2 dose (RP2D) at 40 mg once daily included hyperglycemia (18%), rash (14%), liver dysfunction (12%), diarrhea (10%), pneumonitis (8%), mucosal inflammation (6%), and fatigue (4%). Dose-limiting toxicities (1 patient each) were G4 fasting hyperglycemia at 40 mg (21/28 schedule) and G3 maculopapular rash and G3 fasting hyperglycemia at 70 mg (21/28 schedule). The pharmacokinetic profile was dose-proportional. Phosphorylated serine-473 AKT levels were suppressed by ≥90% in platelet-rich plasma within 4 hours at the MTD (50 mg). Pharmacodynamic decreases in fluorodeoxyglucose positron emission tomography uptake of >25% occurred in 66% (21/32) of patients dosed at 40 mg once daily. Evidence of single-agent activity included 10 RECIST partial responses (PR; confirmed for peritoneal mesothelioma, PIK3CA mutant head-and-neck cancer, and three pleural mesotheliomas)., Conclusions: Apitolisib exhibited dose-proportional pharmacokinetics with target modulation at doses ≥16 mg. The RP2D was 40 mg once-daily 28/28 schedule; severe on-target toxicities were apparent at ≥40 mg, particularly pneumonitis. Apitolisib was reasonably tolerated at 30 mg, the selected dose for pleural mesothelioma patients given limited respiratory reserve. Modest but durable antitumor activity was demonstrated. Clin Cancer Res; 22(12); 2874-84. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
19. Breast cancer resistance protein (ABCG2) in clinical pharmacokinetics and drug interactions: practical recommendations for clinical victim and perpetrator drug-drug interaction study design.
- Author
-
Lee CA, O'Connor MA, Ritchie TK, Galetin A, Cook JA, Ragueneau-Majlessi I, Ellens H, Feng B, Taub ME, Paine MF, Polli JW, Ware JA, and Zamek-Gliszczynski MJ
- Subjects
- ATP Binding Cassette Transporter, Subfamily G, Member 2, ATP-Binding Cassette Transporters genetics, Clinical Trials as Topic, Drug Resistance, Multiple, Drug-Related Side Effects and Adverse Reactions genetics, Humans, Neoplasm Proteins genetics, Polymorphism, Single Nucleotide, Practice Guidelines as Topic, Research Design, Substrate Specificity, ATP-Binding Cassette Transporters antagonists & inhibitors, Drug Interactions, Drug-Related Side Effects and Adverse Reactions metabolism, Neoplasm Proteins antagonists & inhibitors, Pharmaceutical Preparations metabolism, Pharmacokinetics
- Abstract
Breast cancer resistance protein (BCRP; ABCG2) limits intestinal absorption of low-permeability substrate drugs and mediates biliary excretion of drugs and metabolites. Based on clinical evidence of BCRP-mediated drug-drug interactions (DDIs) and the c.421C>A functional polymorphism affecting drug efficacy and safety, both the US Food and Drug Administration and European Medicines Agency recommend preclinical evaluation and, when appropriate, clinical assessment of BCRP-mediated DDIs. Although many BCRP substrates and inhibitors have been identified in vitro, clinical translation has been confounded by overlap with other transporters and metabolic enzymes. Regulatory recommendations for BCRP-mediated clinical DDI studies are challenging, as consensus is lacking on the choice of the most robust and specific human BCRP substrates and inhibitors and optimal study design. This review proposes a path forward based on a comprehensive analysis of available data. Oral sulfasalazine (1000 mg, immediate-release tablet) is the best available clinical substrate for intestinal BCRP, oral rosuvastatin (20 mg) for both intestinal and hepatic BCRP, and intravenous rosuvastatin (4 mg) for hepatic BCRP. Oral curcumin (2000 mg) and lapatinib (250 mg) are the best available clinical BCRP inhibitors. To interrogate the worst-case clinical BCRP DDI scenario, study subjects harboring the BCRP c.421C/C reference genotype are recommended. In addition, if sulfasalazine is selected as the substrate, subjects having the rapid acetylator phenotype are recommended. In the case of rosuvastatin, subjects with the organic anion-transporting polypeptide 1B1 c.521T/T genotype are recommended, together with monitoring of rosuvastatin's cholesterol-lowering effect at baseline and DDI phase. A proof-of-concept clinical study is being planned by a collaborative consortium to evaluate the proposed BCRP DDI study design., (Copyright © 2015 by The American Society for Pharmacology and Experimental Therapeutics.)
- Published
- 2015
- Full Text
- View/download PDF
20. First-in-human phase I study of pictilisib (GDC-0941), a potent pan-class I phosphatidylinositol-3-kinase (PI3K) inhibitor, in patients with advanced solid tumors.
- Author
-
Sarker D, Ang JE, Baird R, Kristeleit R, Shah K, Moreno V, Clarke PA, Raynaud FI, Levy G, Ware JA, Mazina K, Lin R, Wu J, Fredrickson J, Spoerke JM, Lackner MR, Yan Y, Friedman LS, Kaye SB, Derynck MK, Workman P, and de Bono JS
- Subjects
- Administration, Oral, Adult, Aged, Dose-Response Relationship, Drug, Drug-Related Side Effects and Adverse Reactions pathology, Female, Humans, Indazoles blood, Male, Maximum Tolerated Dose, Middle Aged, Neoplasms blood, Neoplasms genetics, Neoplasms pathology, Phosphatidylinositol 3-Kinases genetics, Protein Kinase Inhibitors blood, Protein Kinase Inhibitors pharmacokinetics, Proto-Oncogene Proteins B-raf genetics, Sulfonamides blood, Indazoles administration & dosage, Neoplasms drug therapy, Phosphoinositide-3 Kinase Inhibitors, Protein Kinase Inhibitors administration & dosage, Sulfonamides administration & dosage
- Abstract
Purpose: This first-in-human dose-escalation trial evaluated the safety, tolerability, maximal-tolerated dose (MTD), dose-limiting toxicities (DLT), pharmacokinetics, pharmacodynamics, and preliminary clinical activity of pictilisib (GDC-0941), an oral, potent, and selective inhibitor of the class I phosphatidylinositol-3-kinases (PI3K)., Patients and Methods: Sixty patients with solid tumors received pictilisib at 14 dose levels from 15 to 450 mg once-daily, initially on days 1 to 21 every 28 days and later, using continuous dosing for selected dose levels. Pharmacodynamic studies incorporated (18)F-FDG-PET, and assessment of phosphorylated AKT and S6 ribosomal protein in platelet-rich plasma (PRP) and tumor tissue., Results: Pictilisib was well tolerated. The most common toxicities were grade 1-2 nausea, rash, and fatigue, whereas the DLT was grade 3 maculopapular rash (450 mg, 2 of 3 patients; 330 mg, 1 of 7 patients). The pharmacokinetic profile was dose-proportional and supported once-daily dosing. Levels of phosphorylated serine-473 AKT were suppressed >90% in PRP at 3 hours after dose at the MTD and in tumor at pictilisib doses associated with AUC >20 h·μmol/L. Significant increase in plasma insulin and glucose levels, and >25% decrease in (18)F-FDG uptake by PET in 7 of 32 evaluable patients confirmed target modulation. A patient with V600E BRAF-mutant melanoma and another with platinum-refractory epithelial ovarian cancer exhibiting PTEN loss and PIK3CA amplification demonstrated partial response by RECIST and GCIG-CA125 criteria, respectively., Conclusion: Pictilisib was safely administered with a dose-proportional pharmacokinetic profile, on-target pharmacodynamic activity at dose levels ≥100 mg and signs of antitumor activity. The recommended phase II dose was continuous dosing at 330 mg once-daily., (©2014 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
21. The use of betaine HCl to enhance dasatinib absorption in healthy volunteers with rabeprazole-induced hypochlorhydria.
- Author
-
Yago MR, Frymoyer A, Benet LZ, Smelick GS, Frassetto LA, Ding X, Dean B, Salphati L, Budha N, Jin JY, Dresser MJ, and Ware JA
- Subjects
- Achlorhydria chemically induced, Adult, Antineoplastic Agents administration & dosage, Antineoplastic Agents blood, Area Under Curve, Betaine administration & dosage, Cross-Over Studies, Dasatinib, Drug Interactions, Female, Gastric Acid chemistry, Healthy Volunteers, Humans, Hydrogen-Ion Concentration, Male, Middle Aged, Proton Pump Inhibitors blood, Proton Pump Inhibitors pharmacokinetics, Pyrimidines administration & dosage, Pyrimidines blood, Rabeprazole blood, Rabeprazole pharmacokinetics, Thiazoles administration & dosage, Thiazoles blood, Young Adult, Absorption, Physiological drug effects, Achlorhydria metabolism, Antineoplastic Agents pharmacokinetics, Betaine pharmacology, Proton Pump Inhibitors pharmacology, Pyrimidines pharmacokinetics, Rabeprazole pharmacology, Thiazoles pharmacokinetics
- Abstract
Many orally administered, small-molecule, targeted anticancer drugs, such as dasatinib, exhibit pH-dependent solubility and reduced drug exposure when given with acid-reducing agents. We previously demonstrated that betaine hydrochloride (BHCl) can transiently re-acidify gastric pH in healthy volunteers with drug-induced hypochlorhydria. In this randomized, single-dose, three-way crossover study, healthy volunteers received dasatinib (100 mg) alone, after pretreatment with rabeprazole, and with 1500 mg BHCl after rabeprazole pretreatment, to determine if BHCl can enhance dasatinib absorption in hypochlorhydric conditions. Rabeprazole (20 mg b.i.d.) significantly reduced dasatinib Cmax and AUC0-∞ by 92 and 78%, respectively. However, coadministration of BHCl significantly increased dasatinib Cmax and AUC0-∞ by 15- and 6.7-fold, restoring them to 105 and 121%, respectively, of the control (dasatinib alone). Therefore, BHCl reversed the impact of hypochlorhydria on dasatinib drug exposure and may be an effective strategy to mitigate potential drug-drug interactions for drugs that exhibit pH-dependent solubility and are administered orally under hypochlorhydric conditions.
- Published
- 2014
- Full Text
- View/download PDF
22. Bridging the gap between preclinical and clinical studies using pharmacokinetic-pharmacodynamic modeling: an analysis of GDC-0973, a MEK inhibitor.
- Author
-
Wong H, Vernillet L, Peterson A, Ware JA, Lee L, Martini JF, Yu P, Li C, Del Rosario G, Choo EF, Hoeflich KP, Shi Y, Aftab BT, Aoyama R, Lam ST, Belvin M, and Prescott J
- Subjects
- Animals, Antineoplastic Agents therapeutic use, Azetidines pharmacokinetics, Cell Line, Tumor, Humans, Mice, Mice, Nude, Mitogen-Activated Protein Kinase Kinases pharmacokinetics, Mitogen-Activated Protein Kinase Kinases pharmacology, Models, Biological, Piperidines pharmacokinetics, Protein Kinase Inhibitors pharmacokinetics, Protein Kinase Inhibitors pharmacology, Xenograft Model Antitumor Assays, Antineoplastic Agents pharmacokinetics, Antineoplastic Agents pharmacology, Azetidines pharmacology, Mitogen-Activated Protein Kinase Kinases antagonists & inhibitors, Piperidines pharmacology
- Abstract
Purpose: GDC-0973 is a potent and selective mitogen-activated protein (MAP)/extracellular signal-regulated kinase (ERK) kinase (MEK) inhibitor. Pharmacokinetic-pharmacodynamic (PK-PD) modeling was used to relate GDC-0973 plasma and tumor concentrations, tumor pharmacodynamics and antitumor efficacy to establish pharmacokinetic endpoints and predict active doses in the clinic., Experimental Design: A PK-PD model was used to characterize GDC-0973 tumor disposition and in vivo potency in WM-266-4 xenograft mice. Simulations were conducted using the PK-PD model along with human pharmacokinetics to identify a target plasma concentration and predict active doses. In vivo potency and antitumor efficacy were characterized in A375 melanoma xenograft mice, and a population-based integrated PK-PD-efficacy model was used to relate tumor pharmacodynamics (%pERK decrease) to antitumor activity., Results: GDC-0973 showed a sustained tumor pharmacodynamic response due to longer residence in tumor than in plasma. Following single doses of GDC-0973, estimated in vivo IC(50) values of %pERK decrease based on tumor concentrations in xenograft mice were 0.78 (WM-266-4) and 0.52 μmol/L (A375). Following multiple doses of GDC-0973, the estimated in vivo IC(50) value in WM-266-4 increased (3.89 μmol/L). Human simulations predicted a minimum target plasma concentration of 83 nmol/L and an active dose range of 28 to 112 mg. The steep relationship between tumor pharmacodynamics (%pERK decrease) and antitumor efficacy suggests a pathway modulation threshold beyond which antitumor efficacy switches on., Conclusions: Clinical observations of %pERK decrease and antitumor activity were consistent with model predictions. This article illustrates how PK-PD modeling can improve the translation of preclinical data to humans by providing a means to integrate preclinical and early clinical data.
- Published
- 2012
- Full Text
- View/download PDF
23. Thromboxane A2 receptor signaling inhibits vascular endothelial growth factor-induced endothelial cell differentiation and migration.
- Author
-
Ashton AW and Ware JA
- Subjects
- Animals, Capillaries cytology, Cell Movement drug effects, Endothelial Cells cytology, Endothelial Cells metabolism, Endothelium, Vascular cytology, Fatty Acids, Unsaturated, Focal Adhesions drug effects, Humans, Hydrazines pharmacology, Neovascularization, Physiologic drug effects, Nitric Oxide metabolism, Nitric Oxide Donors pharmacology, Phosphorylation drug effects, Protein Isoforms antagonists & inhibitors, Protein Isoforms drug effects, Protein Isoforms physiology, Protein Kinases genetics, Protein Kinases physiology, Protein Processing, Post-Translational drug effects, Rats, Receptors, Thromboxane A2, Prostaglandin H2 antagonists & inhibitors, Receptors, Thromboxane A2, Prostaglandin H2 drug effects, Recombinant Proteins pharmacology, Signal Transduction drug effects, Transfection, Umbilical Veins, 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid pharmacology, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Endothelial Cells drug effects, Endothelium, Vascular drug effects, Receptors, Thromboxane A2, Prostaglandin H2 physiology, Vascular Endothelial Growth Factor A pharmacology
- Abstract
Vascular endothelial growth factor (VEGF) is an important patho-physiological mediator of angiogenesis. VEGF-induced endothelial cell (EC) migration and angiogenesis often occur in complicated environments containing multiple agents capable of modifying the response. Thromboxane (TX) A2 is released from multiple cell types and is a prime mediator of pathogenesis of many vascular diseases. Human EC express both TXA2 receptor (TP) isoforms; however, the effects of individual TP isoforms on VEGF-induced EC migration and angogenesis are unknown. We report here that the TXA2 mimetic [1S-(1alpha, 2beta(5Z), 3alpha(1E, 3R), 4alpha]-7-[3-(3-hydroxy-4-(4'-iodophenoxy)-1-butenyl)-7-oxab icyclo-[2.2.1]heptan-2yl]-5'-heptenoic acid (IBOP) (100 nmol/L) is a potent antagonist (IC50 30 nmol/L) of VEGF-induced EC migration and differentiation. TPbeta, but not TPalpha, expression is required for the inhibition of VEGF-induced migration and angiogenesis. IBOP costimulation suppressed nitric oxide (NO) release from VEGF-treated EC through decreased activation of Akt, eNOS, and PDK1. TPbeta costimulation also ablated the increase in focal adhesion formation in response to VEGF. This mechanism was characterized by decreased recruitment of focal adhesion kinase (FAK) and vinculin to the alpha(v)beta3 integrin and reduced FAK and Src activation in response to VEGF. Addition of NO donors together with transfection of a constitutively active Src construct could circumvent the blockade of VEGF-induced migration by TP; however, neither intervention alone was sufficient. Thus, TP stimulation appears to limit angiogenesis, at least in part, by inhibiting the pro-angiogenic cytokine VEGF. These data further support a role for antagonism of TP activation in enhancing the angiogenic response in tissues exposed to elevated TXA2 levels in which revascularization is important.
- Published
- 2004
- Full Text
- View/download PDF
24. Myopathy related to administration of a cationic amphiphilic drug and the use of multidose drug distribution analysis to predict its occurrence.
- Author
-
Vonderfecht SL, Stone ML, Eversole RR, Yancey MF, Schuette MR, Duncan BA, and Ware JA
- Subjects
- Animals, Autoradiography, Dopamine Antagonists blood, Dose-Response Relationship, Drug, Drug Evaluation, Preclinical, Lipidoses chemically induced, Male, Microscopy, Electron, Muscle, Skeletal drug effects, Muscle, Skeletal ultrastructure, Phospholipids analysis, Rats, Rats, Sprague-Dawley, Tissue Distribution, Dopamine Antagonists pharmacokinetics, Dopamine Antagonists toxicity, Muscle, Skeletal pathology, Muscular Diseases chemically induced
- Abstract
Many cationic amphiphilic (phospholipidosis-inducing) drugs (CADs) accumulate in tissues following repeated dosing in preclinical models, and this is sometimes associated with dose-limiting toxicities. Plasma drug levels cannot be used to estimate tissue accumulation of CADs since it occurs in tissues despite stabilization of plasma levels. Severe myopathy was found in skeletal muscles of rats during the initial safety evaluation of a dopamine D3 receptor antagonist, PNU-177864, and was associated with phospholipidosis in numerous tissues. The myopathy was observed only when plasma levels of PNU-177864 remained essentially constant throughout the 24-hour dosing period. A repeat dose drug distribution study using whole body autoradiography demonstrated that drug-related material did not accumulate in skeletal muscle or other tissues following repeated doses at levels considered within the therapeutic range and showing toxicokinetic profiles acceptable for further development. These observations provided support for the continued development of and longer-term toxicity studies with this candidate compound.
- Published
- 2004
- Full Text
- View/download PDF
25. Thromboxane A2 receptor agonists antagonize the proangiogenic effects of fibroblast growth factor-2: role of receptor internalization, thrombospondin-1, and alpha(v)beta3.
- Author
-
Ashton AW, Cheng Y, Helisch A, and Ware JA
- Subjects
- Cell Cycle physiology, Cell Movement drug effects, Endothelial Cells drug effects, Endothelium, Vascular cytology, Endothelium, Vascular metabolism, Humans, Hydrazines pharmacology, Inflammation metabolism, Integrin alphaVbeta3 physiology, Ischemia metabolism, Ligands, Protein Isoforms agonists, Protein Isoforms chemistry, Receptor, Fibroblast Growth Factor, Type 1, Receptors, Thromboxane A2, Prostaglandin H2 antagonists & inhibitors, Receptors, Thromboxane A2, Prostaglandin H2 chemistry, Thrombospondin 1 metabolism, Thrombospondin 1 pharmacology, Thromboxane A2 physiology, Transcription, Genetic, Tumor Suppressor Protein p53 physiology, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Endocytosis drug effects, Fatty Acids, Unsaturated pharmacology, Fibroblast Growth Factor 2 antagonists & inhibitors, Neovascularization, Physiologic drug effects, Receptor Protein-Tyrosine Kinases physiology, Receptors, Fibroblast Growth Factor physiology, Receptors, Thromboxane A2, Prostaglandin H2 agonists
- Abstract
Thromboxane (TX) A2 is released from multiple cell types and is a prime mediator of the pathogenesis of many vascular events, including angiogenesis. Endothelial cells express TXA2 receptors (TP) but the effects of TP stimulation on angiogenesis remain controversial. In this study, we show that stimulation of endothelial cell TP impairs ligand-induced FGF receptor internalization and consequently abrogates FGF-2-induced endothelial cell migration in vitro and angiogenesis in vivo. Prevention of FGF-2-induced angiogenesis was associated with expression of the TPbeta isoform. The deficit in FGFR1 internalization was mediated through activation of TPbeta preventing the FGF-2-mediated decrease in p53 expression, thus enhancing thrombospondin-1 (TSP-1) release from EC and reducing FGFR1 internalization. Once released TSP-1 interacted with the alpha(v)beta3 integrin on the EC surface. On stimulation, FGFR1 and alpha(v)beta3 were found to associate in a complex. We determined that complex formation was important for receptor internalization as conditions that inhibit FGFR1 internalization, such as inappropriate ligation of alpha(v)beta3 by either TSP-1 or a neutralizing antibody, disrupted the complex. These results establish a novel role for isoform specific regulation of angiogenesis by TP, provide the first functional significance for the existence of two TP isoforms in humans, and clarify the mechanism by which TP signaling regulates FGFR1 kinetics and signaling.
- Published
- 2004
- Full Text
- View/download PDF
26. Inhibition of tumor necrosis factor alpha-mediated NFkappaB activation and leukocyte adhesion, with enhanced endothelial apoptosis, by G protein-linked receptor (TP) ligands.
- Author
-
Ashton AW, Ware GM, Kaul DK, and Ware JA
- Subjects
- Antigens, CD metabolism, Caspases metabolism, Cell Adhesion drug effects, Cell Adhesion immunology, Cells, Cultured, GTP-Binding Proteins metabolism, Heptanoic Acids pharmacology, Humans, Interleukin-1 pharmacology, Ligands, Receptor Cross-Talk drug effects, Receptor Cross-Talk physiology, Receptors, Cell Surface metabolism, Receptors, Tumor Necrosis Factor metabolism, Receptors, Tumor Necrosis Factor, Type I, Receptors, Tumor Necrosis Factor, Type II, Signal Transduction drug effects, Signal Transduction physiology, Thromboxane A2 metabolism, Umbilical Veins cytology, Antineoplastic Agents pharmacology, Apoptosis physiology, Endothelium, Vascular cytology, Leukocytes cytology, NF-kappa B metabolism, Tumor Necrosis Factor-alpha pharmacology
- Abstract
Tumor necrosis factor (TNF) alpha is a critical mediator of inflammation; however, TNFalpha is rarely released alone and the "cross-talk" between different classes of inflammatory mediators is largely unexplored. Thromboxane A(2) (TXA(2)) is released during I/R injury and exerts its effects via a G protein-linked receptor (TP). In this study, we found that TXA(2) mimetics stimulate leukocyte adhesion molecule (LAM) expression on endothelium via TPbeta. The potential interaction between TXA(2) and TNFalpha in altering endothelial survival and LAM expression was examined. IBOP, a TXA(2) mimetic, attenuated TNFalpha-induced LAM expression in vitro, in a concentration-dependent manner, by preventing TNFalpha-enhanced gene expression, and also reduced TNFalpha-induced leukocyte adhesion to endothelium both in vitro and in vivo. IBOP abrogated TNFalpha-induced NFkappaB activation in endothelial cells, as determined by reduced IkappaB phosphorylation and NFkappaB nuclear translocation, by inhibiting the assembly of signaling intermediates with the intracellular domain of TNF receptors 1 and 2 in response to TNFalpha. This inhibition resulted from the Galpha(q)-mediated enhancement of STAT1 activation and was reversed by anti-STAT1 antisense oligonucleotides. TNFalpha-mediated TNFR1-FADD association and caspase 8 activation were not inhibited by IBOP co-stimulation, however, resulting in a 2.6-fold increase in endothelial cell apoptosis. By stimulating the vessel wall and inducing endothelial cell apoptosis, TXA(2), in combination with TNFalpha, may hamper the angiogenic response during inflammation or ischemia, thus reducing revascularization and tissue viability.
- Published
- 2003
- Full Text
- View/download PDF
27. Long-term effects of surgical angiogenic therapy with fibroblast growth factor 2 protein.
- Author
-
Ruel M, Laham RJ, Parker JA, Post MJ, Ware JA, Simons M, and Sellke FW
- Subjects
- Double-Blind Method, Female, Follow-Up Studies, Heart diagnostic imaging, Humans, Male, Middle Aged, Technetium Tc 99m Sestamibi, Thallium Radioisotopes, Time Factors, Tomography, Emission-Computed, Single-Photon, Coronary Artery Bypass, Fibroblast Growth Factor 2 therapeutic use
- Abstract
Objective: The long-term effects of surgical fibroblast growth factor 2 therapy are examined., Methods: In a randomized, double-blind study, fibroblast growth factor 2 (10 microg or 100 microg) or placebo (n = 8 each) was delivered in the ungraftable myocardial territory of patients concomitantly undergoing coronary artery bypass grafting. Patients were followed up to 32.2 +/- 6.8 months postoperatively with clinical assessment and nuclear perfusion imaging., Results: Baseline patient characteristics were similar between the 3 groups. There were 2 late deaths, one of pancreatic cancer and one of undetermined cause (both in the 100-microg fibroblast growth factor 2 group). Two patients (both in the control group) underwent a total of 6 repeat cardiac catheterizations for recurrent coronary events. Mean Canadian Cardiovascular Society angina class improved at late follow-up from baseline in all groups (P < or = .02); however, patients treated with either dose of fibroblast growth factor 2 had significantly more freedom from angina recurrence than those treated with placebo (P =.03). Late nuclear perfusion scans revealed a persistent reversible or a new, fixed perfusion defect in the ungraftable territory of 4 of 5 patients who received placebo versus only 1 of 9 patients treated with fibroblast growth factor 2 (P =.02). The overall sum of left ventricular stress perfusion defect scores was also lower in fibroblast growth factor 2-treated patients than in control subjects (1.3 +/- 1.4 vs 3.9 +/- 2.1, respectively; P =.04). A trend toward a higher late left ventricular ejection fraction was noted in fibroblast growth factor 2-treated patients (55.1% +/- 14.6% vs 44.3% +/- 6.5%, fibroblast growth factor 2-treated patients versus control subjects; P =.12)., Conclusions: These data suggest that surgical angiogenic therapy with sustained-release fibroblast growth factor 2 may result in a prolonged myocardial revascularization effect that could translate into clinical benefit.
- Published
- 2002
- Full Text
- View/download PDF
28. Protein kinase C (PKC) delta regulates PKCalpha activity in a Syndecan-4-dependent manner.
- Author
-
Murakami M, Horowitz A, Tang S, Ware JA, and Simons M
- Subjects
- Amino Acid Sequence, Animals, Molecular Sequence Data, Phosphorylation, Protein Kinase C-alpha, Protein Kinase C-delta, Rats, Syndecan-4, Isoenzymes metabolism, Membrane Glycoproteins metabolism, Protein Kinase C metabolism, Proteoglycans metabolism
- Abstract
The phosphorylation state of Ser(183) in the cytoplasmic tail of syndecan-4 determines the binding affinity of the cytoplasmic tail to phosphatidylinositol 4,5-bisphosphate (PIP(2)), the capacity of the tail to multimerize, and its ability to activate protein kinase C (PKC) alpha. We sought to identify the kinase responsible for this phosphorylation and to determine its downstream effects on PKCalpha activity and on endothelial cell function. Among several PKC isoenzymes tested, only PKCalpha and -delta were able to specifically phosphorylate Ser(183) in vitro. However, studies in cultured endothelial cells showed that the phosphorylation level of syndecan-4 was significantly reduced in endothelial cells expressing a dominant negative (DN) PKCdelta but not a DN PKCalpha mutant. Syndecan-4/PIP(2)-dependent PKCalpha activity was significantly increased in PKCdelta DN cells, while PKCdelta overexpression was accompanied by decreased PKCalpha activity. PKCdelta-overexpressing cells exhibited a significantly lower proliferation rate and an impaired tube formation in response to FGF2, which were mirrored by similar observations in PKCalpha DN endothelial cells. These findings suggest that PKCdelta is the kinase responsible for syndecan-4 phosphorylation, which, in turn, attenuates the cellular response to FGF2 by reducing PKCalpha activity. The reduced PKCalpha activity then leads to impaired endothelial cell function. We conclude that PKCdelta regulates PKCalpha activity in a syndecan-4-dependent manner.
- Published
- 2002
- Full Text
- View/download PDF
29. Inhibition of protein kinase Calpha prevents endothelial cell migration and vascular tube formation in vitro and myocardial neovascularization in vivo.
- Author
-
Wang A, Nomura M, Patan S, and Ware JA
- Subjects
- Animals, Cell Adhesion drug effects, Cell Division drug effects, Cells, Cultured, Coronary Vessels drug effects, Coronary Vessels metabolism, Endothelial Growth Factors pharmacology, Endothelium, Vascular cytology, Endothelium, Vascular drug effects, Gene Expression drug effects, Humans, Isoenzymes genetics, Isoenzymes metabolism, Ligation, Lymphokines pharmacology, MAP Kinase Kinase Kinase 3, MAP Kinase Kinase Kinases metabolism, Male, Mice, Mice, Inbred C57BL, Mitogen-Activated Protein Kinase 3, Mitogen-Activated Protein Kinases metabolism, Myocardial Infarction drug therapy, Myocardial Infarction pathology, Myocardial Revascularization, Neovascularization, Physiologic drug effects, Oligonucleotides, Antisense pharmacology, Organ Specificity, Phosphorylation drug effects, Protein Kinase C genetics, Protein Kinase C metabolism, Protein Kinase C-alpha, Signal Transduction drug effects, Signal Transduction physiology, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Vitronectin metabolism, Cell Movement drug effects, Endothelium, Vascular metabolism, Isoenzymes antagonists & inhibitors, Myocardial Infarction metabolism, Neovascularization, Physiologic physiology, Protein Kinase C antagonists & inhibitors
- Abstract
Although protein kinase C (PKC) activation is required for endothelial cell (EC) growth, migration, adhesion, and vessel formation, the role of individual PKC isoenzymes in these events is not defined. Because PKCalpha has been previously linked with enhanced EC migration and response to angiogenic growth factors, we characterized a specific phosphorothioate-modified 21-mer antisense PKCalpha (AS-PKCalpha). AS-PKCalpha (500 nmol/L) prevented the expression of PKCalpha protein by 90% in human ECs and did not reduce the expression of any other PKC isoenzyme. AS-PKCalpha reduced human EC migration by 64% compared with its control oligonucleotide in a "scratch" wounding assay, and AS-PKCalpha reduced human EC adhesion to the extracellular matrix protein vitronectin by 18%. Phosphorylation of mitogen-activated protein kinase (extracellular signal-regulated kinase 1/2) induced by vascular endothelial growth factor was inhibited by 30% in human ECs transfected with AS-PKCalpha. Compared with control, AS-PKCalpha also reduced the number of EC tubes formed in a 3D type I collagen gel assay by 37.5%. Finally, using an osmotic minipump, we infused AS-PKCalpha into mice in which myocardial infarction was induced by coronary ligation and found that the oligonucleotide was primarily taken up by intramyocardial blood vessels. Compared with the results with control oligonucleotide, AS-PKCalpha oligonucleotide inhibited the number of anti-PKCalpha-stained blood vessels by 48% and reduced the total vessel number by 72% as well. In conclusion, the expression of PKCalpha is required for full EC migration, adhesion to vitronectin, vascular endothelial growth factor-induced extracellular signal-regulated kinase activation, and tube formation and is likely to be of importance in myocardial angiogenesis in vivo after ischemia.
- Published
- 2002
- Full Text
- View/download PDF
30. Biopsy coupled to quantitative immunofluorescence: a new method to study the human vascular endothelium.
- Author
-
Colombo PC, Ashton AW, Celaj S, Talreja A, Banchs JE, Dubois NB, Marinaccio M, Malla S, Lachmann J, Ware JA, and Le Jemtel TH
- Subjects
- Aged, Arteries, Biopsy, Cells, Cultured, Chronic Disease, Cyclooxygenase 2, Fluorescent Antibody Technique, Humans, Isoenzymes metabolism, Male, Membrane Proteins, Middle Aged, NF-kappa B metabolism, Nitric Oxide biosynthesis, Nitric Oxide Synthase metabolism, Nitric Oxide Synthase Type III, Prostaglandin-Endoperoxide Synthases metabolism, Reference Values, Reproducibility of Results, Tyrosine metabolism, Veins, Cardiac Output, Low metabolism, Endothelium, Vascular metabolism, Endothelium, Vascular pathology, Tyrosine analogs & derivatives
- Abstract
Limited availability of endothelial tissue is a major constraint when investigating the cellular mechanisms of endothelial dysfunction in patients with metabolic and cardiovascular diseases. We propose a novel approach that combines collection of 200-1,000 endothelial cells from a superficial forearm vein or the radial artery, with reliable measurements of protein expression by quantitative immunofluorescence analysis. This method was validated against immunoblot analysis in cultured endothelial cells. Levels of vascular endothelial cell activation, oxidative stress, and nitric oxide synthase expression were measured and compared in five patients with severe chronic heart failure and in four healthy age-matched subjects. In summary, vascular endothelial biopsy coupled with measurement of protein expression by quantitative immunofluorescence analysis provides a novel approach to the study of the vascular endothelium in humans.
- Published
- 2002
- Full Text
- View/download PDF
31. Differential effects of protein kinase C on human vascular smooth muscle cell proliferation and migration.
- Author
-
Itoh H, Yamamura S, Ware JA, Zhuang S, Mii S, Liu B, and Kent KC
- Subjects
- Cell Division drug effects, Cell Movement drug effects, Cells, Cultured, Chemotaxis drug effects, Chemotaxis physiology, Cyclic AMP physiology, Cyclic AMP-Dependent Protein Kinases physiology, Humans, Isoenzymes metabolism, Muscle, Smooth, Vascular drug effects, Muscle, Smooth, Vascular enzymology, Protein Kinase C metabolism, Muscle, Smooth, Vascular cytology, Muscle, Smooth, Vascular physiology, Protein Kinase C pharmacology
- Abstract
Vascular smooth muscle cell (SMC) migration and proliferation contribute to intimal hyperplasia, and protein kinase C (PKC) may be required for both events. In this report, we investigated the role of PKC in proliferation and migration of SMC derived from the human saphenous vein. Activation of PKC by phorbol-12,13-dibutyrate (PDBu) or (-)-indolactam [(-)-ILV] increases SMC proliferation. Downregulation of PKC activity by prolonged incubation with phorbol ester or inhibition of PKC with chelerythrine in SMC diminished agonist-stimulated proliferation. In contrast, stimulation of PKC with PDBu or (-)-ILV inhibited basal and agonist-induced SMC chemotaxis. Moreover, downregulation of PKC or inhibition with chelerythrine accentuated migration. We postulated that the inhibitory effect of PKC on SMC chemotaxis was mediated through cAMP-dependent protein kinase (protein kinase A, PKA). In support of this hypothesis, we found that activation of PKC in SMC stimulated PKA activity. The cAMP agonist forskolin significantly inhibited SMC chemotaxis. Furthermore, the inhibitory effect of PKC on SMC chemotaxis was completely reversed by cAMP or PKA inhibitors. In search of the PKC isotype(s) underlying these differential effects of PKC in SMC, we identified eight isotypes expressed in human SMC. Only PKC-alpha, -beta I, -delta, and -epsilon were eliminated by downregulation, suggesting that one or more of these four enzymes facilitate the observed phorbol ester-dependent effects of PKC in SMC. In summary, we found that PKC activation enhances proliferation but inhibits migration of human vascular SMC. These differential effect of PKC on vascular cells appears to be mediated through PKC-alpha, -beta I, -delta, and/or -epsilon.
- Published
- 2001
- Full Text
- View/download PDF
32. Too many vessels? Not enough? The wrong kind? The VEGF debate continues.
- Author
-
Ware JA
- Subjects
- Animals, Arteriosclerosis etiology, Clinical Trials as Topic, Endothelial Growth Factors adverse effects, Endothelial Growth Factors physiology, Humans, Lymphokines adverse effects, Lymphokines physiology, Myocardial Ischemia drug therapy, Neovascularization, Pathologic, Safety, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Endothelial Growth Factors pharmacology, Lymphokines pharmacology, Neovascularization, Physiologic drug effects
- Published
- 2001
- Full Text
- View/download PDF
33. Reversal of angiogenesis in vitro, induction of apoptosis, and inhibition of AKT phosphorylation in endothelial cells by thromboxane A(2).
- Author
-
Gao Y, Yokota R, Tang S, Ashton AW, and Ware JA
- Subjects
- Adenylyl Cyclase Inhibitors, Adenylyl Cyclases metabolism, Bridged Bicyclo Compounds, Heterocyclic, Cell Survival, Clone Cells, Colforsin pharmacology, Cyclic AMP metabolism, Endothelium, Vascular cytology, Enzyme Activation, Fatty Acids, Unsaturated, Humans, Hydrazines pharmacology, Isoquinolines pharmacology, Phosphorylation drug effects, Protein Isoforms antagonists & inhibitors, Protein Isoforms biosynthesis, Protein Serine-Threonine Kinases antagonists & inhibitors, Protein Serine-Threonine Kinases metabolism, Proto-Oncogene Proteins c-akt, Receptors, Thromboxane antagonists & inhibitors, Receptors, Thromboxane biosynthesis, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction, Thromboxane A2 antagonists & inhibitors, Time Factors, Angiogenesis Inhibitors pharmacology, Apoptosis, Endothelium, Vascular drug effects, Proto-Oncogene Proteins, Sulfonamides, Thromboxane A2 pharmacology
- Abstract
Thromboxane A(2) (TxA(2)) causes platelet aggregation, vasoconstriction, and inhibition of endothelial cell (EC) migration and prevents vascular tube formation via its specific receptors (TP), of which there are two isoforms (TPalpha and TPbeta), both expressed in human ECs. In this study, we demonstrate that the TxA(2) mimetic IBOP increases apoptosis of human ECs and inhibits the phosphorylation of Akt kinase, an intracellular mediator required for cell survival. Treatment with IBOP destroyed EC networks formed on a basement membrane matrix in vitro. To distinguish the role of the TP isoforms, each isoform was expressed in TP-null ECs to create TPalpha and TPbeta ECs. IBOP induced apoptosis and inhibited phosphorylation of Akt kinase in both TPalpha and TPbeta. IBOP increased cAMP levels in TPalpha but not in TPbeta. Apoptosis induced by IBOP in TPalpha was not affected by either the adenylyl cyclase activator forskolin or the protein kinase A inhibitor 14-22 amide or H-89, whereas that in TPbeta was suppressed by forskolin and enhanced by the protein kinase A inhibitor 14-22 amide or H-89, suggesting that the TP isoforms differ in their signal pathways in mediating apoptosis. In conclusion, apoptosis may be the mechanism by which TxA(2)-mediated destruction of vascular structures in ECs occurs; although both TP isoforms induce apoptosis, possibly via inhibiting Akt phosphorylation, the signaling differs in each isoform, in that activation of the adenylyl cyclase pathway prevents apoptosis caused by TPbeta, but not by TPalpha, stimulation.
- Published
- 2000
- Full Text
- View/download PDF
34. A pair of ACEs, for openers?
- Author
-
Sibinga NE and Ware JA
- Subjects
- Angiotensin-Converting Enzyme Inhibitors, Angiotensins metabolism, Humans, Peptidyl-Dipeptidase A metabolism, Receptors, Angiotensin metabolism, Renin-Angiotensin System physiology
- Published
- 2000
- Full Text
- View/download PDF
35. Inhibition of endothelial cell migration, intercellular communication, and vascular tube formation by thromboxane A(2).
- Author
-
Ashton AW, Yokota R, John G, Zhao S, Suadicani SO, Spray DC, and Ware JA
- Subjects
- Calcium metabolism, Capillaries drug effects, Cells, Cultured, Connexin 43 metabolism, Cytoskeleton drug effects, Cytoskeleton ultrastructure, Endothelium, Vascular cytology, Endothelium, Vascular drug effects, Gap Junctions drug effects, Gap Junctions physiology, Humans, Integrins analysis, Umbilical Veins, Wound Healing drug effects, Bridged Bicyclo Compounds, Heterocyclic pharmacology, Capillaries physiology, Cell Communication drug effects, Chemotaxis drug effects, Endothelium, Vascular physiology, Fatty Acids, Unsaturated pharmacology, Neovascularization, Physiologic drug effects, Thromboxane A2 pharmacology
- Abstract
The eicosanoid thromboxane A(2) (TXA(2)) is released by activated platelets, monocytes, and the vessel wall and interacts with high affinity receptors expressed in several tissues including endothelium. Whether TXA(2) might alter endothelial migration and tube formation, two determinants of angiogenesis, is unknown. Thus, we investigated the effect of the TXA(2) mimetic [1S-(1alpha, 2beta(5Z),3alpha(1E,3R), 4alpha]-7-[3-(3-hydroxy-4-(4'-iodophenoxy)-1-butenyl)-7-o xab icyclo- [2.2.1]heptan-2-yl]-5'-heptenoic acid (IBOP) on human endothelial cell (HEC) migration and angiogenesis in vitro. IBOP stimulation inhibited HEC migration by 50% and in vitro capillary formation by 75%. These effects of IBOP were time- and concentration-dependent with an IC(50) of 25 nM. IBOP did not affect integrin expression or cytoskeletal morphology of HEC. Since gap junction-mediated intercellular communication increases in migrating HEC, we determined whether IBOP might inhibit coupling or connexin expression in HEC. IBOP reduced the passage of microinjected dyes between HEC by 50%, and the effects of IBOP on migration and tube formation were mimicked by the gap junction inhibitor 18beta-glycyrrhetinic acid (1 microM) with a similar time course and efficacy. IBOP (24 h) did not affect the expression or phosphorylation of connexin 43 in whole HEC lysates. Immunohistologic examination of HEC suggested that IBOP may impair functional coupling by altering the cellular distribution of gap junctions, leading to increased connexin 43 internalization. Thus, this finding that TXA(2) mimetics can prevent HEC migration and tube formation, possibly by impairing intercellular communication, suggests that antagonizing TXA(2) signaling might enhance vascularization of ischemic tissue.
- Published
- 1999
- Full Text
- View/download PDF
36. Enhancement of endothelial cell migration and in vitro tube formation by TAP20, a novel beta 5 integrin-modulating, PKC theta-dependent protein.
- Author
-
Tang S, Gao Y, and Ware JA
- Subjects
- Amino Acid Sequence, Animals, Base Sequence, Cell Adhesion physiology, Cell Division physiology, Cloning, Molecular, Cytoplasm chemistry, Endothelium, Vascular cytology, Endothelium, Vascular physiology, Humans, Integrins metabolism, Membrane Glycoproteins genetics, Molecular Sequence Data, Neovascularization, Physiologic physiology, Nuclear Proteins, Protein Kinase C-theta, Proteins chemistry, Rats, Receptors, Vitronectin metabolism, Tumor Cells, Cultured, Zinc Fingers, Cell Movement physiology, Integrin beta Chains, Integrins physiology, Isoenzymes physiology, Membrane Glycoproteins physiology, Protein Kinase C physiology
- Abstract
Migration, proliferation, and tube formation of endothelial cells are regulated by a protein kinase C isoenzyme PKCtheta. A full-length cDNA encoding a novel 20-kD protein, whose expression was PKCtheta-dependent, was identified in endothelial cells, cloned, characterized, and designated as theta-associated protein (TAP) 20. Overexpression of TAP20 decreased cell adhesion and enhanced migration on vitronectin and tube formation in three-dimensional culture. An antiintegrin alphavbeta5 antibody prevented these TAP20 effects. Overexpression of TAP20 also decreased focal adhesion formation in alphavbeta3-deficient cells. The interaction between TAP20 and beta5 integrin cytoplasmic domain was demonstrated by protein coprecipitation and immunoblotting. Thus, the discovery of TAP20, which interacts with integrin beta5 and modulates cell adhesion, migration, and tube formation, further defines a possible pathway to angiogenesis dependent on PKCtheta.
- Published
- 1999
- Full Text
- View/download PDF
37. Downregulation of protein kinase cdelta activity enhances endothelial cell adaptation to hypoxia.
- Author
-
Shizukuda Y, Helisch A, Yokota R, and Ware JA
- Subjects
- Apoptosis physiology, Cell Division physiology, Down-Regulation, Endothelium, Vascular cytology, Enzyme Inhibitors pharmacology, Humans, Nitric Oxide Synthase antagonists & inhibitors, Protein Kinase C-delta, omega-N-Methylarginine pharmacology, Adaptation, Physiological, Endothelium, Vascular metabolism, Hypoxia metabolism, Isoenzymes metabolism, Protein Kinase C metabolism
- Abstract
Background: Although protein kinase C (PKC) has been implicated in ischemic cell death, the role of individual PKC isoenzymes in the response of endothelial cells (ECs) to hypoxia is unknown., Methods and Results: To test the effect of hypoxia on the activity of individual PKC isoenzymes, human ECs were exposed to 95% N(2) with 5% CO(2) for 24 hours. This severe hypoxia reduced PKCdelta specific activity in both human umbilical vein ECs (HUVECs) and a HUVEC-derived EC line (ECVs) significantly (80.5+/-5.7% and 55.5+/-8. 6% of normoxia controls, respectively); the activities of PKCalpha and PKCepsilon were unchanged. The protein levels of PKCalpha, PKCdelta, and PKCepsilon were unchanged by hypoxia. To determine whether PKCdelta downregulation by hypoxia was linked to EC function, ECVs in which PKCdelta was stably overexpressed (PKCdelta-ECs) were exposed to hypoxia. A significant increase in cell death was observed in PKCdelta-ECs compared with controls (5.8+/-0.6% versus 2. 3+/-0.4% at 24 hours, 13.2+/-1.2% versus 4.1+/-0.4% at 48 hours, P<0. 05) during hypoxia. Neither the DNA laddering assay nor TUNEL staining revealed an increase in apoptosis of PKCdelta-ECs exposed to hypoxia, suggesting a hypoxia-induced increase in nonapoptotic cell death of PKCdelta-ECs. Inhibition of NO synthase with N(G)-monomethyl-L-arginine (L-NMMA) affected neither the decline in PKCdelta activity nor the EC death induced by hypoxia., Conclusions: PKCdelta activity is decreased by hypoxia by a mechanism that does not involve NO synthase; this downregulation appears to enhance EC survival during hypoxia by decreasing nonapoptotic cell death.
- Published
- 1999
- Full Text
- View/download PDF
38. Local perivascular delivery of basic fibroblast growth factor in patients undergoing coronary bypass surgery: results of a phase I randomized, double-blind, placebo-controlled trial.
- Author
-
Laham RJ, Sellke FW, Edelman ER, Pearlman JD, Ware JA, Brown DL, Gold JP, and Simons M
- Subjects
- Alginates, Coronary Vessels, Delayed-Action Preparations, Double-Blind Method, Drug Carriers, Drug Compounding, Drug Implants, Female, Follow-Up Studies, Glucuronic Acid, Heparin, Hexuronic Acids, Humans, Male, Middle Aged, Patient Selection, Placebos, Recombinant Proteins administration & dosage, Coronary Artery Bypass, Fibroblast Growth Factor 2 administration & dosage
- Abstract
Background: Angiogenesis is a promising treatment strategy for patients who are not candidates for standard revascularization, because it promotes the growth of new blood vessels in ischemic myocardium., Methods and Results: We conducted a randomized, double-blind, placebo-controlled study of basic fibroblast growth factor (bFGF; 10 or 100 microg versus placebo) delivered via sustained-release heparin-alginate microcapsules implanted in ischemic and viable but ungraftable myocardial territories in patients undergoing CABG. Twenty-four patients were randomized to 10 microg of bFGF (n=8), 100 microg of bFGF (n=8), or placebo (n=8), in addition to undergoing CABG. There were 2 operative deaths and 3 Q-wave myocardial infarctions. There were no treatment-related adverse events, and there was no rise in serum bFGF levels. Clinical follow-up was available for all patients (16.0+/-6.8 months). Three control patients had recurrent angina, 2 of whom required repeat revascularization. One patient in the 10-microg bFGF group had angina, whereas all patients in the 100-microg bFGF group remained angina-free. Stress nuclear perfusion imaging at baseline and 3 months after CABG showed a trend toward worsening of the defect size in the placebo group (20.7+/-3.7% to 23.8+/-5.7%, P=0.06), no significant change in the 10-microg bFGF group, and significant improvement in the 100-microg bFGF group (19.2+/-5.0% to 9.1+/-5.9%, P=0.01). Magnetic resonance assessment of the target ischemic zone in a subset of patients showed a trend toward a reduction in the target ischemic area in the 100-microg bFGF group (10.7+/-3.9% to 3. 7+/-6.3%, P=0.06)., Conclusions: This study of bFGF in patients undergoing CABG demonstrates the safety and feasibility of this mode of therapy in patients with viable myocardium that cannot be adequately revascularized.
- Published
- 1999
- Full Text
- View/download PDF
39. Vascular endothelial growth factor-induced endothelial cell migration and proliferation depend on a nitric oxide-mediated decrease in protein kinase Cdelta activity.
- Author
-
Shizukuda Y, Tang S, Yokota R, and Ware JA
- Subjects
- Cell Division drug effects, Cell Movement drug effects, Cells, Cultured, Endothelium, Vascular cytology, Endothelium, Vascular drug effects, Fibroblast Growth Factor 2 pharmacology, Humans, Isoenzymes genetics, Isoenzymes metabolism, Nitric Oxide metabolism, Nitric Oxide Donors pharmacology, Nitric Oxide Synthase antagonists & inhibitors, Nitroprusside pharmacology, Protein Kinase C genetics, Protein Kinase C metabolism, Protein Kinase C-delta, RNA, Messenger metabolism, Receptor Protein-Tyrosine Kinases metabolism, Receptors, Growth Factor metabolism, Receptors, Vascular Endothelial Growth Factor, Tetradecanoylphorbol Acetate pharmacology, Time Factors, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, Endothelial Growth Factors pharmacology, Endothelium, Vascular physiology, Isoenzymes antagonists & inhibitors, Lymphokines pharmacology, Nitric Oxide physiology, Protein Kinase C antagonists & inhibitors
- Abstract
Vascular endothelial growth factor (VEGF) promotes angiogenesis and endothelial cell (EC) migration and proliferation by affecting intracellular mediators, only some of which are known, distal to its receptors. Protein kinase C (PKC) participates in the function of VEGF, but the role of individual PKC isoenzymes is unknown. In this study, we tested the importance of the activity of specific PKC isoenzymes in human EC migration and proliferation in response to VEGF. PKCdelta specific activity was depressed by the addition of VEGF (by 41+/-8% [P<0.05] at 24 hours) in human umbilical vein ECs (HUVECs) and in a HUVEC-derived EC line, ECV, without changing the total amount of either protein or mRNA encoding PKCdelta. Neither basic fibroblast growth factor (FGF-2) nor serum altered PKCdelta specific activity. The VEGF-induced decrease of PKCdelta activity, which began at 8 hours after stimulation, was strongly blocked by pretreatment with the nitric oxide (NO) synthase inhibitor N(G)-monomethyl-L-arginine in HUVECs; NO release peaked within 2 hours after stimulation. An exogenous NO donor, sodium nitroprusside, also decreased PKCdelta activity. The inhibition by N(G)-monomethyl-L-arginine of VEGF-induced HUVEC migration and proliferation, but not that induced by FGF-2 or serum, suggested that the decrease in PKCdelta via NO pathway is required for VEGF-induced EC migration and proliferation. Overexpression of PKCdelta in ECV cells specifically prevented EC response to VEGF but not to FGF-2 or serum. Thus, we conclude that suppression of PKCdelta activity via a NO synthase mechanism is required for VEGF-induced EC migration and proliferation, but not for that induced by FGF-2 or serum.
- Published
- 1999
- Full Text
- View/download PDF
40. Protein kinase Cdelta inhibition of S-phase transition in capillary endothelial cells involves the cyclin-dependent kinase inhibitor p27(Kip1).
- Author
-
Ashton AW, Watanabe G, Albanese C, Harrington EO, Ware JA, and Pestell RG
- Subjects
- Animals, Cells, Cultured, Culture Media, Serum-Free, Cyclin-Dependent Kinase Inhibitor p27, Cyclin-Dependent Kinases antagonists & inhibitors, Genes, Tumor Suppressor, Protein Kinase C-delta, Rats, Cell Cycle Proteins, Endothelium, Vascular cytology, Endothelium, Vascular metabolism, Isoenzymes metabolism, Microtubule-Associated Proteins metabolism, Protein Kinase C metabolism, S Phase, Tumor Suppressor Proteins
- Abstract
Distinct protein kinase C (PKC) isoforms differentially regulate cellular proliferation in rat microvascular endothelial cells (EC). Overexpression of PKCalpha has little effect on proliferation, whereas PKCdelta slows endothelial cell proliferation and induces S-phase arrest. Analyses were performed on EC overexpressing PKCalpha (PKCalphaEC) or PKCdelta (PKCdeltaEC) to determine the role of specific cell cycle regulatory proteins in the PKCdelta-induced cell cycle arrest. Serum-induced stimulation of cyclins D1, E, and A-associated kinase activity was delayed by 12 h in the PKCdeltaEC line in association with S-phase arrest. However, the protein levels for cyclins D1, E, and A were similar. Nuclear accumulation of cyclin D1 protein in response to serum was also delayed in PKCdeltaEC. In the PKCdeltaEC line, serum induced p27(Kip1) but not p16(Ink4a) or p21(Cip1). Serum did not affect p27(Kip1) levels in the control vascular endothelial cell line. Immunoprecipitation-Western blotting analysis of p27(Kip1) showed serum stimulation of the vascular endothelial cell line resulted in increased amounts of cyclin D1 bound to p27(Kip1). In the PKCdeltaEC line, serum did not increase the amount of cyclin D1 bound to p27(Kip1). Transfection of full-length p27(Kip1) antisense into the PCKdeltaEC line reversed the S-phase arrest and resulted in normal cell cycle progression, suggesting a critical role for p27(Kip1) in the PKCdelta-mediated S-phase arrest.
- Published
- 1999
- Full Text
- View/download PDF
41. Angiogenesis activators and inhibitors differentially regulate caveolin-1 expression and caveolae formation in vascular endothelial cells. Angiogenesis inhibitors block vascular endothelial growth factor-induced down-regulation of caveolin-1.
- Author
-
Liu J, Razani B, Tang S, Terman BI, Ware JA, and Lisanti MP
- Subjects
- Calcium-Calmodulin-Dependent Protein Kinases metabolism, Caveolin 1, Cell Division drug effects, Cell Line, Down-Regulation drug effects, Fibroblast Growth Factor 2 pharmacology, Hepatocyte Growth Factor pharmacology, Humans, Proto-Oncogene Proteins metabolism, Receptor Protein-Tyrosine Kinases metabolism, Receptors, Growth Factor metabolism, Receptors, Vascular Endothelial Growth Factor, Signal Transduction drug effects, Vascular Endothelial Growth Factor A, Vascular Endothelial Growth Factors, ets-Domain Protein Elk-1, Caveolins, DNA-Binding Proteins, Endothelial Growth Factors pharmacology, Endothelium, Vascular drug effects, Lymphokines pharmacology, Membrane Proteins metabolism, Neovascularization, Physiologic drug effects, Transcription Factors
- Abstract
Angiogenesis is the process by which new blood vessels are formed via proliferation of vascular endothelial cells. A variety of angiogenesis inhibitors that antagonize the effects of vascular endothelial growth factor (VEGF) and basic fibroblast growth factor (bFGF) have recently been identified. However, the mechanism by which these diverse angiogenesis inhibitors exert their common effects remains largely unknown. Caveolin-1 and -2 are known to be highly expressed in vascular endothelial cells both in vitro and in vivo. Here, we examine the potential role of caveolins in the angiogenic response. For this purpose, we used the well established human umbilical vein endothelial cell line, ECV 304. Treatment of ECV 304 cells with known angiogenic growth factors (VEGF, bFGF, or hepatocyte growth factor/scatter factor), resulted in a dramatic reduction in the expression of caveolin-1. This down-regulation event was selective for caveolin-1, as caveolin-2 levels remained constant under these conditions of growth factor stimulation. VEGF-induced down-regulation of caveolin-1 expression also resulted in the morphological loss of cell surface caveolae organelles as seen by transmission electron microscopy. A variety of well characterized angiogenesis inhibitors (including angiostatin, fumagillin, 2-methoxy estradiol, transforming growth factor-beta, and thalidomide) effectively blocked VEGF-induced down-regulation of caveolin-1 as seen by immunoblotting and immunofluorescence microscopy. However, treatment with angiogenesis inhibitors alone did not significantly affect the expression of caveolin-1. PD98059, a specific inhibitor of mitogen-activated protein kinase and a known angiogenesis inhibitor, also blocked the observed VEGF-induced down-regulation of caveolin-1. Furthermore, we show that caveolin-1 can function as a negative regulator of VEGF-R (KDR) signal transduction in vivo. Thus, down-regulation of caveolin-1 may be an important step along the pathway toward endothelial cell proliferation.
- Published
- 1999
- Full Text
- View/download PDF
42. Myb-dependent regulation of thrombospondin 2 expression. Role of mRNA stability.
- Author
-
Bein K, Ware JA, and Simons M
- Subjects
- 3T3 Cells, Animals, Base Sequence, Cloning, Molecular, DNA Primers, Mice, Proto-Oncogene Proteins c-myb, RNA, Messenger genetics, Transcription, Genetic, Gene Expression Regulation, Proto-Oncogene Proteins metabolism, Thrombospondins genetics, Trans-Activators metabolism
- Abstract
The nuclear transcription factor c-Myb, which is highly expressed in hematopoietic cells, has been shown to be functional in NIH 3T3 cells: cells that do not possess detectable levels of c-Myb. To identify endogenous target genes of c-Myb in fibroblasts, RNA isolated from NIH 3T3 cells stably transfected with a full-length or a dominant negative c-myb construct (GREMyb and GREMEn, respectively) was subjected to differential display analysis. 5'-Rapid amplification of cDNA ends of a selected band, sequencing, and a nucleotide homology search led to the identification of thrombospondin 2 (TSP 2) as the gene product repressed in GREMyb and induced in GREMEn cells. The pattern of TSP 2 expression during the cell cycle was consistent with c-myb-dependent regulation. The possibility that the identified transcript was TSP 1, a homologous product known to be repressed by v-Src, c-Jun, and v-Myc, was ruled out by using a TSP 2-specific DNA probe and by showing a distinct pattern of regulation of TSP 1 and TSP 2 expression. Nuclear run-on and TSP 2 promoter-reporter (chloramphenicol acetyltransferase) assays showed similar transcriptional levels in GREMyb and NIH 3T3 cells. However, mRNA stability studies showed a much shorter TSP 2 mRNA half-life in GREMyb compared with wild type NIH 3T3 cells, suggesting that c-myb affects TSP 2 expression via a post-transcriptional mechanism. The implications of a protooncogene-mediated suppression of TSP expression are discussed.
- Published
- 1998
- Full Text
- View/download PDF
43. Requirement for protein kinase C theta for cell cycle progression and formation of actin stress fibers and filopodia in vascular endothelial cells.
- Author
-
Tang S, Morgan KG, Parker C, and Ware JA
- Subjects
- Animals, Blotting, Western, Capillaries cytology, Cell Division, Enzyme Activation, Humans, Microscopy, Fluorescence, Protein Kinase C-theta, Rats, Wound Healing, Actins metabolism, Cell Cycle, Endothelium, Vascular metabolism, Isoenzymes metabolism, Protein Kinase C metabolism, Zinc Fingers
- Abstract
Activation of the protein kinase C (PKC) family with phorbol esters induces endothelial proliferation and angiogenesis, but which of the events that constitute angiogenesis are affected by individual members of the PKC family is unknown. In rat capillary endothelial (RCE) cells, serum stimulation increased expression of a single PKC isoenzyme, PKCtheta, and its translocation to the periphery. Conditional overexpression of a dominant-negative mutant of PKCtheta markedly inhibited RCE proliferation, as well as closure of a "wound" by RCE migration and formation of capillary rings and tubules in vitro. PKCtheta inhibition delayed the endothelial cell cycle at the G2/M phase and prevented formation of actin stress fibers and filopodia but not lamellipodia. The defect in cell morphology and wound closure in PKCtheta-kn cells was reversed by overexpressing kinase-active PKCtheta, indicating that these RCE functions depend upon PKCtheta substrates. Thus, PKCtheta is required for multiple processes essential for angiogenesis and wound repair, including endothelial mitosis, maintenance of a normal actin cytoskeleton, and formation of an enclosed tube.
- Published
- 1997
- Full Text
- View/download PDF
44. Differential desensitization of thromboxane A2 receptor subtypes.
- Author
-
Yukawa M, Yokota R, Eberhardt RT, von Andrian L, and Ware JA
- Subjects
- 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid, Animals, CHO Cells, Calcium metabolism, Colforsin pharmacology, Cricetinae, Cyclic AMP-Dependent Protein Kinases metabolism, Enzyme Activation, Enzyme Inhibitors pharmacology, Naphthalenes pharmacology, Prostaglandin Endoperoxides, Synthetic pharmacology, Protein Kinase C antagonists & inhibitors, Protein Kinase C metabolism, Receptors, Thromboxane agonists, Tetradecanoylphorbol Acetate pharmacology, Thromboxane A2 analogs & derivatives, Thromboxane A2 pharmacology, Receptors, Thromboxane metabolism
- Abstract
Two subtypes of the thromboxane A2 (TxA2) receptor (TxA2R-E and TxA2R-P), which differ in their alternatively spliced cytoplasmic tails, have been identified. The initial concentration of the TxA2 mimetic IBOP required to reduce peak intracellular Ca2+ concentration ([Ca2+]i) induced by a second addition of IBOP (100 nmol/L) was similar (IC50 for TxA2R-E and TxA2R-P, 0.46 +/- 0.16 and 0.40 +/- 0.07 nmol/L) in fibroblasts overexpressing either the TxA2R-E or -P subtype. Although the number of TxA2 binding sites decreased in TxA2R-P cells after prolonged stimulation with a TxA2 mimetic, those in the TxA2R-E cells increased markedly. To determine whether the mechanism for desensitization differs between subtypes, the effect of activation of protein kinase C (PKC) or cAMP-dependent kinase on TxA2-induced [Ca2+]i mobilization was measured. Forskolin reduced the IBOP-induced peak [Ca2+]i in neither TxA2R-E nor TxA2R-P cells; however, treatment with phorbol esters (IC50, 0.57 +/- 0.70 nmol/L) strongly prevented IBOP-mediated [Ca2+]i rise in TxA2R-E but not in TxA2R-P cells. Desensitization of TxA2R-E by phorbol esters was prevented by the PKC inhibitor calphostin C or by downregulation of PKC-alpha. Thus, the response of TxA2R-E to prolonged stimulation differs from that of TxA2R-P in both the regulation of the number of binding sites and the mechanism for desensitization; agonists that activate PKC-alpha might interfere with TxA2R-E-mediated signaling.
- Published
- 1997
- Full Text
- View/download PDF
45. Enhancement of migration by protein kinase Calpha and inhibition of proliferation and cell cycle progression by protein kinase Cdelta in capillary endothelial cells.
- Author
-
Harrington EO, Löffler J, Nelson PR, Kent KC, Simons M, and Ware JA
- Subjects
- Animals, Cell Division genetics, Cell Line, Endothelium, Vascular enzymology, Gene Transfer Techniques, Protein Kinase C-alpha, Protein Kinase C-delta, Rats, Cell Cycle genetics, Cell Movement genetics, Endothelium, Vascular cytology, Gene Expression Regulation, Enzymologic, Isoenzymes genetics, Protein Kinase C genetics
- Abstract
Activation of protein kinase C (PKC) induces angiogenesis, migration, and proliferation of endothelial cells (EC), but can also prevent growth factor-induced EC proliferation. To determine whether these disparate effects are mediated by substrates of individual PKC isoenzymes, PKCalpha and PKCdelta were overexpressed in rat microvascular EC. Basal and stimulated migration were enhanced in PKCalpha EC compared with either PKCdelta or control EC. Serum-induced growth of PKCdelta EC was decreased, while that of PKCalpha cells was similar to control EC. Phorbol ester markedly inhibited PKCdelta EC growth but enhanced growth of PKCalpha and control EC. To determine possible causes for this altered proliferation, the effect of PKCdelta on adhesion, mitogen-activated protein kinase activity, and cell cycle progression was measured. Adherence of PKCdelta EC to vitronectin was significantly enhanced. Serum-induced extracellular signal-regulated kinase-2 activity was increased equally in both PKCalpha and PKCdelta EC above that of control, while extracellular signal-regulated kinase-1 activity was similar in all EC. Cell cycle analysis suggested that PKCdelta EC entered S phase inappropriately and were delayed in passage through S phase. Thus, PKCalpha may mediate some proangiogenic effects of PKC activation; conversely, PKCdelta may direct antiangiogenic aspects of overall PKC activation, including slowing of the cell cycle progression.
- Published
- 1997
- Full Text
- View/download PDF
46. Angiogenesis in ischemic heart disease.
- Author
-
Ware JA and Simons M
- Subjects
- Animals, Humans, Myocardial Ischemia physiopathology, Coronary Vessels, Myocardial Ischemia pathology, Neovascularization, Pathologic
- Published
- 1997
- Full Text
- View/download PDF
47. Food for starving hearts.
- Author
-
Simons M and Ware JA
- Subjects
- Animals, Fibroblast Growth Factor 5, Fibroblast Growth Factors genetics, Humans, Recombinant Proteins therapeutic use, Swine, Fibroblast Growth Factors therapeutic use, Genetic Therapy methods, Myocardial Ischemia therapy, Neovascularization, Physiologic drug effects
- Published
- 1996
- Full Text
- View/download PDF
48. Selective inhibition of thrombin receptor-mediated Ca2+ entry by protein kinase C beta.
- Author
-
Xu Y and Ware JA
- Subjects
- Base Sequence, Cell Line, DNA, Antisense pharmacology, Fluorescent Dyes, Fura-2, Humans, Isoenzymes antagonists & inhibitors, Isoenzymes biosynthesis, Isoenzymes metabolism, Kinetics, Megakaryocytes, Molecular Sequence Data, Mutagenesis, Site-Directed, Oligodeoxyribonucleotides, Protein Kinase C antagonists & inhibitors, Protein Kinase C biosynthesis, Receptors, Thrombin antagonists & inhibitors, Receptors, Thrombin biosynthesis, Recombinant Proteins antagonists & inhibitors, Recombinant Proteins biosynthesis, Recombinant Proteins metabolism, Spectrometry, Fluorescence, Transfection, Calcium metabolism, Protein Kinase C metabolism, Receptors, Thrombin physiology
- Abstract
Thrombin initiates many physiological processes in platelets and other megakaryocyte-lineage cells by interacting with surface receptors and generating rises in cytoplasmic Ca2+; these rises result from both Ca2+ release from intracellular stores and receptor-mediated Ca2+ entry. Regulators that limit Ca2+ entry after its initiation by thrombin have not been identified. In this study, prevention of expression of a single protein kinase C isoenzyme (PKC beta) by antisense cDNA overexpressed in HEL cells, a human megakaryoblastic cell line that expresses thrombin receptors, promotes thrombin receptor-mediated Ca2+ entry without altering thrombin-induced intracellular release of Ca2+. The cytoplasmic Ca2+ rise initiated by endoperoxide analogs was not affected by inhibiting PKC beta. Overexpression of a cDNA encoding wild-type PKC beta mutated to prevent recognition by the antisense cDNA abolished the enhancement of Ca2+ influx following thrombin. Thus, PKC beta appears to be a specific negative regulator of thrombin receptor-mediated Ca2+ entry.
- Published
- 1995
- Full Text
- View/download PDF
49. Requirement for protein kinase C activation in basic fibroblast growth factor-induced human endothelial cell proliferation.
- Author
-
Kent KC, Mii S, Harrington EO, Chang JD, Mallette S, and Ware JA
- Subjects
- Alkaloids, Benzophenanthridines, Calcium metabolism, Cell Division, Cells, Cultured, Endothelium metabolism, Enzyme Activation, Enzyme Inhibitors pharmacology, Fibroblast Growth Factor 2 pharmacology, Fibroblast Growth Factor 2 physiology, Humans, Immunoblotting, Neovascularization, Pathologic, Phenanthridines pharmacology, Phorbol Esters pharmacology, Polymerase Chain Reaction, Protein Kinase C antagonists & inhibitors, Protein Kinase C drug effects, RNA analysis, Signal Transduction, Endothelium cytology, Endothelium enzymology, Isoenzymes metabolism, Protein Kinase C metabolism
- Abstract
The intracellular signaling mechanisms that mediate basic fibroblast growth factor (bFGF)-induced angiogenesis have not been fully identified. In particular, whether activation of the intracellular enzyme protein kinase C (PKC) is necessary or sufficient for bFGF-induced mitogenesis of human endothelial cells is not clear. Accordingly, the effect of bFGF stimulation on the Ca2+ increase and PKC activity of normal human endothelial cells (HEC) was studied, as was the effect of inhibition of PKC and the distribution of PKC isoenzymes in these cells. The addition of bFGF to cultured HEC increased overall PKC activity in the absence of an increase in intracellular Ca2+ and markedly stimulated their proliferation, as did the addition of PKC-activating phorbol esters. bFGF-induced proliferation was prevented by the PKC inhibitors chelerythrine and H-7 and by downregulation of PKC after prolonged incubation with phorbol esters. In contrast, these inhibitors did not prevent HEC proliferation induced by epidermal growth factor. Because of the failure of bFGF to increase Ca2+, we determined whether bFGF-induced proliferation could be mediated by novel or atypical PKC isoenzymes (which are not regulated by Ca2+). Investigation of the isoenzyme distribution of confluent and subconfluent HEC by immunoblotting, Northern transfer analysis, and polymerase chain reaction of reverse-transcribed RNA revealed the presence of several novel and atypical isoenzymes (PKC-delta, -eta, -theta, and -zeta) as well as small amounts of the conventional (Ca(2+)-regulated) isoenzymes PKC-alpha and -beta. Activation of PKC by bFGF, in the absence of an increase in intracellular Ca2+, suggests that one or more of these Ca(2+)-independent PKC isoenzymes are both necessary and sufficient for HEC proliferation after bFGF.
- Published
- 1995
- Full Text
- View/download PDF
50. Alternative splicing produces a divergent cytoplasmic tail in the human endothelial thromboxane A2 receptor.
- Author
-
Raychowdhury MK, Yukawa M, Collins LJ, McGrail SH, Kent KC, and Ware JA
- Subjects
- Alternative Splicing, Amino Acid Sequence, Animals, Base Sequence, Humans, Molecular Sequence Data, Endothelium, Vascular metabolism, Receptors, Thromboxane genetics, Thromboxane A2 metabolism
- Published
- 1995
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.