Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Molecules ; 21(4): 440, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-27534722

ABSTRACT

Isosorbide-2-carbamates-5-aryl esters are highly potent and very selective butyrylcholinesterase inhibitors. The objective of the present work was to address the hypothesis that the isosorbide-aryl-5-ester group could be replaced with an antioxidant functionality while maintaining inhibitor effects and selectivity. We successfully incorporated ferulic acid or lipoic acid groups producing potent selective inhibitors of butyrylcholinesterase (BuChE). The hybrid compounds were non-toxic to the murine hippocampal cell line HT-22 and lipoate esters were neuroprotective at 10 and 25 µM when the cells were challenged with glutamate (5 mM) in a similar manner to the positive control quercetin. The benzyl carbamate 7a was a potent inhibitor of BuChE (IC50 150 nM) and it was effective in reducing glutamate toxicity to neuronal cells at >5 µM. Representative compounds exhibited an antioxidant effect in the oxygen radical absorbance capacity assay as the lipoate 7d was not active, whereas the ferulate 8a showed a weak, but significant, activity with 0.635 ± 0.020 Trolox Equivalent.


Subject(s)
Antioxidants/pharmacology , Carbamates/chemical synthesis , Carbamates/pharmacology , Cholinesterase Inhibitors/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/enzymology , Animals , Antioxidants/chemical synthesis , Antioxidants/chemistry , Butyrylcholinesterase , Carbamates/chemistry , Cell Line , Cholinesterase Inhibitors/chemical synthesis , Cholinesterase Inhibitors/chemistry , Esters , Hippocampus/cytology , Hippocampus/drug effects , Mice , Molecular Structure , Neurons/cytology , Neurons/drug effects , Structure-Activity Relationship
2.
Atherosclerosis ; 221(2): 478-83, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22326030

ABSTRACT

OBJECTIVES: To evaluate the pharmacokinetics and effects of the first immediate-release (IR) niacin-aspirin prodrug (ST0702) on lipid, prostaglandin and thromboxane levels in non-human primates (NHPs). METHODS: We compared 28 mg/kg crystalline IR niacin, equimolar doses of crystalline IR ST0702 and control on low density lipoprotein cholesterol (LDL-C), apolipoprotein B (ApoB) and triglycerides (Tg) in NHPs (6 per group) over 48 h (daily oral gavage). In addition, we compared IR niacin and ST0702 effects on prostaglandin (PG)D(2), ex vivo thromboxane B(2) (TXB(2)) levels and plasma pharmacokinetics. RESULTS: ST0702 is metabolised in vivo to aspirin, niacin and salicylic acid with T(max) values of 30, 45 and 95 min respectively using a non-compartmental model. ST0702 resulted in 38% and 40% reductions in LDL-C and ApoB levels compared to control over the 48 h period (p = 0.027 and p = 0.012 respectively). Corresponding values were 32% and 25% for niacin (both p = NS vs control). ST0702, but not niacin, decreased Tg levels (p = 0.017 for between group difference). Post prandial glycaemia was attenuated vs baseline in the ST0702 group only. Ex vivo serum TXB(2) generation was suppressed at 15 min and complete suppression of TXB(2) was sustained at 24h (p<0.01 vs niacin). ST0702 suppressed PGD(2) exposure eightfold (p = 0.012) compared to niacin over the first 24h. CONCLUSIONS: This two-dose study in NHPs suggests that ST0702 is more effective than IR niacin on lipid profiles, while suppressing TXB(2) and PGD(2) increases and prevents post-prandial glycaemia. ST0702 shows promise as a new IR therapeutic option for niacin.


Subject(s)
Aspirin/analogs & derivatives , Aspirin/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Hypolipidemic Agents/pharmacology , Isosorbide/analogs & derivatives , Lipid Metabolism/drug effects , Lipids/blood , Niacin/analogs & derivatives , Niacin/pharmacology , Prodrugs/pharmacology , Prostaglandin D2/blood , Salicylates/pharmacology , Animals , Apolipoproteins B/blood , Aspirin/blood , Aspirin/pharmacokinetics , Blood Glucose/drug effects , Chemistry, Pharmaceutical , Cholesterol, LDL/blood , Cyclooxygenase Inhibitors/blood , Cyclooxygenase Inhibitors/pharmacokinetics , Hyperglycemia/blood , Hyperglycemia/prevention & control , Hypolipidemic Agents/blood , Hypolipidemic Agents/pharmacokinetics , Isosorbide/blood , Isosorbide/pharmacokinetics , Isosorbide/pharmacology , Macaca fascicularis , Models, Biological , Niacin/blood , Niacin/pharmacokinetics , Postprandial Period , Prodrugs/pharmacokinetics , Salicylates/blood , Salicylates/pharmacokinetics , Thromboxane B2/blood , Triglycerides/blood
3.
J Med Chem ; 55(5): 2154-62, 2012 Mar 08.
Article in English | MEDLINE | ID: mdl-22248361

ABSTRACT

We describe a new type of barbiturate-based matrix metalloproteinase (MMP) inhibitor incorporating a nitric oxide (NO) donor/mimetic group (series 1). The compounds were designed to inhibit MMP at enzyme level and to attenuate MMP-9 secretion arising from inflammatory signaling. To detect effects related to the nitrate, we prepared and studied an analogous series of barbiturate C5-alkyl alcohols that were unable to release NO (series 2). Both series inhibited recombinant human MMP-2/9 activity with nanomolar potency. Series 1 consistently inhibited the secretion of MMP-9 from TNFα/IL1ß stimulated Caco-2 cells at 10 µM, which could be attributed to NO related effects because the non-nitrate panel did not affect enzyme levels. Several compounds from series 1 (10 µM) inhibited tumor cell invasion but none from the non-nitrate panel did. The work shows that MMP-inhibitory barbiturates are suitable scaffolds for hybrid design, targeting additional facets of MMP pathophysiology, with potential to improve risk-benefit ratios.


Subject(s)
Barbiturates/chemical synthesis , Matrix Metalloproteinase Inhibitors , Nitric Oxide Donors/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Barbiturates/chemistry , Barbiturates/pharmacology , Cell Line , Cell Movement/drug effects , Cell Survival/drug effects , Collagen , Drug Combinations , Drug Design , Humans , Interleukin-1beta/pharmacology , Laminin , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Models, Molecular , Molecular Mimicry , Neoplasm Invasiveness , Nitric Oxide Donors/chemistry , Nitric Oxide Donors/pharmacology , Proteoglycans , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/pharmacology
4.
Br J Pharmacol ; 166(3): 938-49, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22122360

ABSTRACT

BACKGROUND AND PURPOSE: Tumour cell-induced platelet aggregation (TCIPA) facilitates cancer cell invasion, angiogenesis and the formation of metastatic foci. TCIPA can be modulated by pharmacological inhibitors of MMP-2 and ADP; however, the COX inhibitor aspirin did not prevent TCIPA. In this study, we have tested the pharmacological effects of a new group of isosorbide-based aspirin prodrugs on TCIPA. EXPERIMENTAL APPROACH: TCIPA was induced in human platelets by mixing with human adenocarcinoma or fibrosarcoma cells under no flow and flow conditions. The release of gelatinases and P-selectin expression during TCIPA were studied by zymography and flow cytometry respectively. KEY RESULTS: Tumour cells caused platelet aggregation. This aggregation resulted in the release of MMP-2 and a significant up-regulation of P-selectin on platelets, indicative of platelet activation. Pharmacological modulation of TCIPA revealed that ST0702, one of the aspirin prodrugs, down-regulated TCIPA while aspirin was ineffective. The deacetylated metabolite of ST0702, 5-nicotinate salicylate (ST0702 salicylate), down-regulated both ADP-stimulated platelet aggregation and TCIPA. CONCLUSIONS AND IMPLICATIONS: Our results show that ST0702 was an effective inhibitor of TCIPA in vitro. Its deacetylated metabolite may contribute to the effects of ST0702 by inhibiting ADP-mediated TCIPA.


Subject(s)
Aspirin/analogs & derivatives , Blood Platelets/drug effects , Cell Communication/drug effects , Isosorbide/analogs & derivatives , Niacin/analogs & derivatives , Platelet Aggregation/drug effects , Prodrugs/pharmacology , Aspirin/pharmacology , Blood Platelets/cytology , Cell Line, Tumor , Chromatography, High Pressure Liquid , Drug Stability , Flow Cytometry , Humans , Isosorbide/pharmacology , Microscopy, Phase-Contrast , Niacin/pharmacology , Physostigmine/pharmacology
5.
J Pharm Pharmacol ; 64(1): 77-89, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22150675

ABSTRACT

OBJECTIVES: Aspirin is the mainstay of anti-platelet therapy in the secondary prevention of cardiovascular disease. However, problems with aspirin safety and resistance demand clinical strategies based on multiple pharmacological approaches. Prodrugs of aspirin may offer beneficial effects in terms of gastro-intestinal safety and multiple pharmacological approaches. However, the pharmacological profile of aspirin prodrugs in human platelets has not been completed yet. We aimed to compare the effects of aspirin and prodrugs of aspirin (1-5) on human platelet aggregation stimulated by ADP and collagen and associated receptor expression (GPIIb/IIIa and P-selectin) in platelet-rich plasma (PRP) and washed platelets (WP). METHODS: As aspirin is released from prodrugs following esterase hydrolysis we studied the expression and activity of butyrylcholineterase (BuChE) and carboxyesterase (CE) in plasma and platelets. The mechanism of prodrug-induced platelet aggregation inhibition was explored by studying the effects of plasma and purified human BuChE on aggregation. Finally, the relative contribution of nitric oxide (NO) bioactivity to nitrate-containing prodrugs of aspirin-induced inhibition of aggregation was determined using 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ,) a selective inhibitor of the soluble guanylyl cyclase. KEY FINDINGS: ST0702, 2, a nicotinic acid-aspirin codrug was equipotent with aspirin with respect to inhibition of collagen-induced platelet aggregation. Compound 4, a NO releasing aspirin was the most potent inhibitor of ADP-induced platelet aggregation, an effect partially reversed by ODQ. The platelet inhibitory effects of aspirin prodrugs were time-dependent as the maximal inhibitory effects against collagen-induced aggregation were achieved by aspirin at 2 min, 1 at 5 min and ST0702 at 15 min. The aspirin prodrugs were significantly less potent in WP than in PRP and the reverse was true of aspirin. In the presence of complete BuChE inhibition in PRP, there was almost complete loss of aspirin prodrug, but not aspirin anti-aggregatory activity. Interestingly, CE activity was observed in WP and platelet lysate with pNPA substrate. Accordingly, 1 and ST0702 retained 50% and 100% anti-aggregatory activity at maximal concentrations in WP, which was attenuated in the presence of esterase inhibitor phenylmethylsulphonyl fluoride. CONCLUSIONS: The inhibitory effect of aspirin prodrugs in PRP is due to prodrug activation by BuChE. In contrast, the platelet-inhibitory effects of aspirin prodrugs in WP may be mediated through the activity of platelet CE. Compound 4, a NO-containing aspirin prodrug, may exert dual inhibitory effects in platelets. Thus, aspirin prodrugs effectively inhibit human platelet aggregation and as such may be an alternative to conventional aspirin.


Subject(s)
Aspirin/pharmacology , Blood Platelets/drug effects , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Prodrugs/pharmacology , Blood Platelets/enzymology , Butyrylcholinesterase/metabolism , Carboxylesterase/metabolism , Flow Cytometry , Humans , Immunoblotting , Nitrates/pharmacology , Platelet Count
6.
J Med Chem ; 52(21): 6588-98, 2009 Nov 12.
Article in English | MEDLINE | ID: mdl-19821574

ABSTRACT

Aspirin prodrugs and related nitric oxide releasing compounds hold significant therapeutic promise, but they are hard to design because aspirin esterification renders its acetate group very susceptible to plasma esterase mediated hydrolysis. Isosorbide-2-aspirinate-5-salicylate is a true aspirin prodrug in human blood because it can be effectively hydrolyzed to aspirin upon interaction with plasma BuChE. We show that the identity of the remote 5-ester dictates whether aspirin is among the products of plasma-mediated hydrolysis. By observing the requirements for aspirin release from an initial panel of isosorbide-based esters, we were able to introduce nitroxymethyl groups at the 5-position while maintaining ability to release aspirin. Several of these compounds are potent inhibitors of platelet aggregation. The design of these compounds will allow better exploration of cross-talk between COX inhibition and nitric oxide release and potentially lead to the development of selective COX-1 acetylating drugs without gastric toxicity.


Subject(s)
Aspirin/analogs & derivatives , Aspirin/chemical synthesis , Isosorbide/analogs & derivatives , Isosorbide/chemical synthesis , Nitrates/chemical synthesis , Nitric Oxide Donors/chemical synthesis , Platelet Aggregation Inhibitors/chemical synthesis , Prodrugs/chemical synthesis , Aspirin/blood , Aspirin/pharmacology , Butyrylcholinesterase/blood , Esters , Humans , Hydrolysis , In Vitro Techniques , Isosorbide/pharmacology , Models, Molecular , Nitrates/pharmacology , Nitric Oxide Donors/blood , Nitric Oxide Donors/pharmacology , Platelet Aggregation/drug effects , Platelet Aggregation Inhibitors/blood , Platelet Aggregation Inhibitors/pharmacology , Prodrugs/pharmacology , Structure-Activity Relationship
7.
J Biol Chem ; 284(9): 5869-75, 2009 Feb 27.
Article in English | MEDLINE | ID: mdl-19129181

ABSTRACT

Platelet factor 4 (PF4) is an abundant platelet alpha-granule chemokine released following platelet activation. PF4 interacts with thrombomodulin and the gamma-carboxyglutamic acid (Gla) domain of protein C, thereby enhancing activated protein C (APC) generation by the thrombin-thrombomodulin complex. However, the protein C Gla domain not only mediates protein C activation in vivo, but also plays a critical role in modulating the diverse functional properties of APC once generated. In this study we demonstrate that PF4 significantly inhibits APC anti-coagulant activity. PF4 inhibited both protein S-dependent APC anticoagulant function in plasma and protein S-dependent factor Va (FVa) proteolysis 3- to 5-fold, demonstrating that PF4 impairs protein S cofactor enhancement of APC anticoagulant function. Using recombinant factor Va variants FVa-R506Q/R679Q and FVa-R306Q/R679Q, PF4 was shown to impair APC proteolysis of FVa at position Arg(306) by 3-fold both in the presence and absence of protein S. These data suggest that PF4 contributes to the poorly understood APC resistance phenotype associated with activated platelets. Finally, despite PF4 binding to the APC Gla domain, we show that APC in the presence of PF4 retains its ability to initiate PAR-1-mediated cytoprotective signaling. In summary, we propose that PF4 acts as a critical regulator of APC generation, but also differentially targets APC toward cytoprotective, rather than anticoagulant function at sites of vascular injury with concurrent platelet activation.


Subject(s)
Anticoagulants/pharmacology , Platelet Factor 4/metabolism , Protein C/metabolism , Apoptosis/physiology , Cells, Cultured , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Factor Va/metabolism , Hexadimethrine Bromide/pharmacology , Humans , Platelet Factor 4/genetics , Protamines/pharmacology , Protein C/antagonists & inhibitors , Protein S/metabolism , Thrombin/metabolism
8.
J Biol Chem ; 283(45): 30531-9, 2008 Nov 07.
Article in English | MEDLINE | ID: mdl-18779332

ABSTRACT

Activated protein C (APC) plays a critical anticoagulant role in vivo by inactivating procoagulant factor Va and factor VIIIa and thus down-regulating thrombin generation. In addition, APC bound to the endothelial cell protein C receptor can initiate protease-activated receptor-1 (PAR-1)-mediated cytoprotective signaling. Protein S constitutes a critical cofactor for the anticoagulant function of APC but is not known to be involved in regulating APC-mediated protective PAR-1 signaling. In this study we utilized a site-directed mutagenesis strategy to characterize a putative protein S binding region within the APC Gla domain. Three single amino acid substitutions within the APC Gla domain (D35T, D36A, and A39V) were found to mildly impair protein S-dependent anticoagulant activity (<2-fold) but retained entirely normal cytoprotective activity. However, a single amino acid substitution (L38D) ablated the ability of protein S to function as a cofactor for this APC variant. Consequently, in assays of protein S-dependent factor Va proteolysis using purified proteins or in the plasma milieu, APC-L38D variant exhibited minimal residual anticoagulant activity compared with wild type APC. Despite the location of Leu-38 in the Gla domain, APC-L38D interacted normally with endothelial cell protein C receptor and retained its ability to trigger PAR-1 mediated cytoprotective signaling in a manner indistinguishable from that of wild type APC. Consequently, elimination of protein S cofactor enhancement of APC anticoagulant function represents a novel and effective strategy by which to separate the anticoagulant and cytoprotective functions of APC for potential therapeutic gain.


Subject(s)
Coenzymes/metabolism , Endothelial Cells/metabolism , Protein C/metabolism , Protein S/metabolism , Receptor, PAR-1/metabolism , Signal Transduction/physiology , Amino Acid Substitution , Antigens, CD/genetics , Antigens, CD/metabolism , Binding Sites/genetics , Cell Line , Coenzymes/genetics , Endothelial Cells/cytology , Endothelial Protein C Receptor , Factor VIIIa/genetics , Factor VIIIa/metabolism , Factor Va/genetics , Factor Va/metabolism , Humans , Mutagenesis, Site-Directed , Peptide Mapping/methods , Protein C/genetics , Protein S/genetics , Receptor, PAR-1/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
9.
Metabolism ; 52(9): 1102-12, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14506614

ABSTRACT

Alcoholic myopathy is characterized by muscle weakness and difficulties in gait and locomotion. It is one of the most prevalent skeletal muscle disorders in the Western hemisphere, affecting between 40% and 60% of all chronic alcohol misusers. However, the pathogenic mechanisms are unknown, although recent studies have suggested that membrane defects occur as a consequence of chronic alcohol exposure. It was our hypothesis that alcohol ingestion perturbs membrane-located proteins associated with intracellular signalling and contractility, in particular those relating to calcium homeostasis. To test this, we fed male Wistar rats nutritionally complete liquid diets containing ethanol as 35% of total dietary energy. Controls were pair-fed identical amounts of the same diet in which ethanol was replaced by isocaloric glucose. At the end of 6 weeks, rats were killed and skeletal muscles dissected. These were used to determine important ion-regulatory skeletal muscle proteins including sarcalumenin (SAR), sarcoplasmic-endoplasmic reticulum Ca(2+)-adenosine triphosphatase (ATPase) (SERCA1), the junctional face protein of 90 kd (90-JFP), alpha(1)- and alpha(2)-dihydropyridine receptor (alpha(1)-DHPR and alpha(2)-DHPR), and calsequestrin (CSQ) by immunoblotting. The relative abundance of microsomal proteins was determined by immunoblotting using the enhanced chemiluminescence (ECL) technique. The data showed that alcohol-feeding significantly reduced gastrocnemius and hind limb muscle weights (P <.05 in both instances). Concomitant changes included increases in the relative amounts of SERCA1 (P <.05) and Ca(2+)-ATPase activity (P <.025). However, there were no statistically significant changes in either SAR, 90-JFP, alpha(1)-DHPR or alpha(2)-DHPR (P >.2 in all instances). Reductions in CSQ were of marginal significance (P =.0950). We conclude that upregulation of SERCA1 protein and Ca(2+)-ATPase activity may be an adaptive mechanism and/or a contributory process in the pathology of alcohol-induced muscle disease.


Subject(s)
Calcium-Transporting ATPases/metabolism , Calcium/metabolism , Ethanol/toxicity , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Alcoholism/complications , Animals , Body Weight/drug effects , Calcium Channels, L-Type/analysis , Calsequestrin/metabolism , Diet , Glucose/administration & dosage , Immunoblotting , Luminescent Measurements , Male , Membrane Proteins/metabolism , Muscle, Skeletal/anatomy & histology , Muscular Diseases/chemically induced , Organ Size/drug effects , Rats , Rats, Wistar , Sarcoplasmic Reticulum Calcium-Transporting ATPases
SELECTION OF CITATIONS
SEARCH DETAIL