Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Br J Pharmacol ; 167(1): 80-94, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22519452

ABSTRACT

BACKGROUND AND PURPOSE: Recently, a small molecule (Q94) was reported to selectively block PAR(1) /Gα(q) interaction and signalling. Here, we describe the pharmacological properties of Q94 and two analogues that share its benzimidazole scaffold (Q109, Q89). Q109 presents a modest variation from Q94 in the substituent group at the 2-position, while Q89 has quite different groups at the 1- and 2-positions. EXPERIMENTAL APPROACH: Using human microvascular endothelial cells, we examined intracellular Ca(2+) mobilization and inositol 1,4,5-trisphosphate accumulation as well as isoprenaline- or forskolin-stimulated cAMP production in response to thrombin. KEY RESULTS: Q89 (10 µM) produced a leftward shift in the thrombin-mediated intracellular Ca(2+) mobilization concentration-response curve while having no effect on the E(max) . Both Q94 (10 µM) and Q109 (10 µM) reduced intracellular Ca(2+) mobilization, leading to a decrease in E(max) and an increase in EC(50) values. Experiments utilizing receptor-specific activating peptides confirmed that Q94 and Q109 were selective for PAR(1) as they did not alter the Ca(2+) response mediated by a PAR(2) activating peptide. Consistent with our Ca(2+) results, micromolar concentrations of either Q94 or Q109 significantly reduced thrombin-induced inositol 1,4,5-trisphosphate production. Neither Q94 nor Q109 diminished the inhibitory effects of thrombin on cAMP production, indicating they inhibit signalling selectively through the G(q) pathway. Our results also suggest the 1,2-disubstituted benzimidazole derivatives act as 'allosteric agonists' of PAR(1) . CONCLUSIONS AND IMPLICATIONS: The Q94 and Q109 benzimidazole derivatives represent a novel scaffold for the development of new PAR(1) inhibitors and provide a starting point to develop dual signalling pathway-selective positive/negative modulators of PAR(1) .


Subject(s)
Benzimidazoles/pharmacology , Receptor, PAR-1/metabolism , Calcium/metabolism , Cell Line , Colforsin/pharmacology , Cyclic AMP/metabolism , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Isoproterenol/pharmacology , Receptor, PAR-1/agonists , Signal Transduction/drug effects
2.
Curr Top Med Chem ; 12(4): 312-20, 2012.
Article in English | MEDLINE | ID: mdl-22204489

ABSTRACT

Indolylglyoxylamides are a class of distinctive benzodiazepine receptor ligands, proposed in the mid-eighties as open analogues of -carbolines. Thorough and long-lasting studies of their structure-activity relationships led to the development of a great deal of derivatives, to satisfy increasingly structural and pharmacophoric requirements of the benzodiazepine binding site in the central nervous system. Efforts to pre-organize their flexible structure in the three-dimensional shape adopted when bound to the receptor led to the identification of two novel classes of rigid ligands, characterized by planar tricyclic heteroaromatic cores: the [1,2,4]triazino[4,3-a]benzimidazol-4(10H)-one and the [1,2,3]triazolo[1,2-a][1,2,4]benzotriazin-1,5(6H)-dione. The present review focuses on these selected classes of ligands, whose rational development, in terms of chemical structures and structure-activity relationships, will be fully discussed.


Subject(s)
Amides/chemistry , Anti-Anxiety Agents/chemistry , Glyoxylates/chemistry , Hypnotics and Sedatives/chemistry , Indoles/chemistry , Receptors, GABA-A/metabolism , Amides/pharmacology , Animals , Anti-Anxiety Agents/pharmacology , Binding Sites , Brain/drug effects , Brain/metabolism , GABA-A Receptor Agonists/chemistry , GABA-A Receptor Agonists/pharmacology , GABA-A Receptor Antagonists/chemistry , GABA-A Receptor Antagonists/pharmacology , Glyoxylates/pharmacology , Humans , Hypnotics and Sedatives/pharmacology , Indoles/pharmacology , Ligands , Protein Binding , Protein Subunits/agonists , Protein Subunits/antagonists & inhibitors , Protein Subunits/metabolism , Structure-Activity Relationship , Triazines/chemistry , Triazines/pharmacology , Triazoles/chemistry , Triazoles/pharmacology
3.
Curr Med Chem ; 17(35): 4270-90, 2010.
Article in English | MEDLINE | ID: mdl-20939813

ABSTRACT

DNA topoisomerases (topos) are essential enzymes that regulate the topological state of DNA during cellular processes such as replication, transcription, recombination, and chromatin remodeling. Topoisomerase I (Topo I) is a ubiquitous nuclear enzyme which catalyzes the relaxation of superhelical DNA generating a transient single strand nick in the duplex, through cycles of cleavage and religation. Topoisomerase II (Topo II) mediates the ATP-dependent induction of coordinated nicks in both strands of the DNA duplex, followed by crossing of another double strand DNA through the transiently broken duplex. Although the biological functions of Topoisomerases are important for ensuing genomic integrity, the ability to interfere with enzymes or generate enzyme-mediated damage is an effective strategy for cancer therapy and, in this connection, DNA topos (I and II) proved to be the excellent targets of clinically significant classes of anticancer drugs. Actually, specific Topo I and Topo II inhibitors reversibly trap the enzyme-DNA complexes, thus converting Topos into physiological poisons, able to produce permanent DNA damage, which triggers cell death. Given that both enzymes are good targets, it would be desirable to jointly inhibit them, but use-limiting toxicity of sequential or simultaneous combinations of topo I and II poisons include severe to life-threatening neutropenia and anemia. Furthermore, the emergence of resistance phenomena to topo I inhibitors is often accompanied by a concomitant rise in the level of topo II expression and viceversa, leading to the failure of clinical therapies. In this regard, a single compound able to inhibit both Topo I and II may present the advantage of improving antitopoisomerase activity, with reduced toxic side effects, with respect to the combination of two inhibitors. Due to the high interest in such compounds, this review represents an update of previous works dealing with the development of dual Topo I and II inhibitors as novel anti-cancer agents. The newly collected derivatives have been described focusing attention on their chemical structures and their biological profiles.


Subject(s)
Antineoplastic Agents/pharmacology , DNA Topoisomerases, Type II/metabolism , DNA Topoisomerases, Type I/metabolism , Topoisomerase I Inhibitors/pharmacology , Topoisomerase II Inhibitors/pharmacology , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , DNA/chemistry , DNA/metabolism , Humans , Structure-Activity Relationship , Topoisomerase I Inhibitors/chemical synthesis , Topoisomerase I Inhibitors/chemistry , Topoisomerase II Inhibitors/chemical synthesis , Topoisomerase II Inhibitors/chemistry
4.
Curr Top Med Chem ; 10(10): 976-92, 2010.
Article in English | MEDLINE | ID: mdl-20370657

ABSTRACT

Adenosine is a ubiquitous homeostatic substance which exerts its action by triggering four different cell membrane G protein-coupled receptors, classified as A(1), A(2A), A(2B), and A(3). Being widely distributed and deeply involved in several physiological functions, as well as pathological disorders, these receptors represent an excellent drug target and the development of specific ligands has been tested as a promising therapeutic concept. Among the obtainable ligands, allosteric modulators offer higher advantages with respect to classical orthosteric compounds, as they possible to achieve greater selectivity and better modulatory control at disease mediating receptors. Actually, synergizing with adenosine bound to the primary binding site, these compounds may modify receptor functions through interaction with an additional binding site. As a consequence, their actions depend directly on the release of the endogenous agonist. A number of compound have been developed as effective allosteric modulators. Most of them target adenosine A(1) and A(3) receptor subtypes as, to date, little or no research attempt have been made to improve the field of A(2A) and A(2B) ligands. This review updates literature on the allosteric modulators that has appeared in the last few years, focusing its attention on medicinal chemistry, in terms of chemical structure and structure-activity relationships. This will provide new perspectives on existing data and an exciting starting point for the development of novel and more effective modulators.


Subject(s)
Adenosine/pharmacology , Receptors, Purinergic P1/metabolism , Adenosine/analogs & derivatives , Adenosine/chemistry , Allosteric Regulation/drug effects , Animals , Humans , Ligands , Purinergic P1 Receptor Agonists/metabolism , Structure-Activity Relationship
5.
Curr Med Chem ; 16(26): 3359-80, 2009.
Article in English | MEDLINE | ID: mdl-19548867

ABSTRACT

The Translocator protein (TSPO), formerly known as the peripheral-type benzodiazepine receptor, is an 18 kDa mitochondrial protein primarily involved in steroid biosynthesis in both peripheral and glial cells. It has been extensively reported that TSPO regulates the rate-limiting translocation of cholesterol from the outer to the inner mitochondrial membrane before its transformation by cytochrome P450(scc) into pregnenolone, which is further converted into an array of different steroids. In the brain, neurosteroids such as allopregnanolone and pregnenolone, acting as positive modulators of gamma-aminobutyric type A (GABA(A)) receptors, exert anxiolytic activity. Specific ligands targeting TSPO increase neurosteroid production and for this reason they have been suggested to play an important role in anxiety modulation. Unlike benzodiazepines (Bzs), which represent the most common anti-anxiety drugs administered around the world, selective TSPO ligands have shown anxiolytic effects in animal models without any of the side effects associated with Bzs. Therefore, specific TSPO ligands that are able to promote neurosteroidogenesis may represent the future of therapeutic treatment of anxiety disorders. Furthermore, TSPO expression levels are altered in several different psychiatric disorders in which anxiety is the main symptom. This article reviews the primary and patent literature over the last decade concerning the development of novel TSPO ligands that have resulted effective in various models of anxiety, taking into special consideration their structure-activity relationships.


Subject(s)
Anxiety Disorders/drug therapy , Ligands , Receptors, GABA/metabolism , Animals , Anti-Anxiety Agents/chemistry , Anti-Anxiety Agents/pharmacology , Anxiety Disorders/diagnosis , Benzodiazepines/chemistry , Benzodiazepines/pharmacology , Humans , Indoleacetic Acids/chemistry , Indoleacetic Acids/pharmacology , Neurotransmitter Agents/chemistry , Neurotransmitter Agents/pharmacology , Oxazines/chemistry , Oxazines/pharmacology , Receptors, GABA-A/metabolism , Structure-Activity Relationship
6.
Curr Med Chem ; 14(25): 2680-701, 2007.
Article in English | MEDLINE | ID: mdl-17979718

ABSTRACT

The gamma-aminobutyric acid type A (GABA(A)) receptors are the major inhibitory neuronal receptors in the mammalian brain. Their activation by GABA opens the intrinsic ion channel, enabling chloride flux into the cell with subsequent hyperpolarization. Several GABA(A) receptor subunit isoforms have been cloned, the major isoform containing alpha, beta, and gamma subunits, and a regional heterogeneity associated with distinct physiological effects has been suggested. As a variety of allosteric ligands can modulate GABA-gated conductance changes through binding to distinct sites, the development of subtype-selective ligands may lead to the selective treatment of GABA system-associated pathology. In particular, the best characterized binding site is the benzodiazepine site (BzR), localized at the alpha/gamma subunit interface, in which the alpha subunit is the main determinant of BzR ligand action selectivity. The alpha1-containing BzR have been proposed to be responsible for the sedative action; the alpha2 and/or the alpha3 subtypes have been suggested to mediate the anxiolytic activity and the myorelaxation effects, and the alpha5 subtype has been associated with cognition processes. The discovery of alpha-selective subtype ligands may help in the specific treatment of anxiety, sleep disorders, convulsions and memory deficits with fewer side effects. Selectivity may be achieved by two approaches: selective affinity or selective efficacy. Selective affinity needs a compound to bind with a higher affinity to one receptor subtype compared with another, whereas subtype-selective efficacy relies on a compound binding to all subtypes, but having different efficacies at various subtypes. The status of BzR ligands, subdivided on the basis of their main chemical structural features, is reviewed in relation to structure-activity relationships which determine their affinity or efficacy selectivity for a certain BzR subtype.


Subject(s)
Receptors, GABA-A/drug effects , Benzodiazepines/metabolism , Carbolines/metabolism , Flavones/metabolism , Indoles/metabolism , Pyrazoles/metabolism , Pyridazines/metabolism , Pyridones/metabolism , Receptors, GABA-A/metabolism , Thiophenes/metabolism , Triazoles/metabolism
7.
J Med Chem ; 44(25): 4359-69, 2001 Dec 06.
Article in English | MEDLINE | ID: mdl-11728182

ABSTRACT

Acetic acid derivatives of [1,2,4]triazino[4,3-a]benzimidazole (TBI) were synthesized and tested in vitro and in vivo as a novel class of aldose reductase (ALR2) inhibitors. Compound 3, (10-benzyl[1,2,4]triazino[4,3-a]benzimidazol-3,4(10H)-dion-2-yl)acetic acid, displayed the highest inhibitory activity (IC(50) = 0.36 microM) and was found to be effective in preventing cataract development in severely galactosemic rats when administered as an eyedrop solution. All the compounds investigated were selective for ALR2, since none of them inhibited appreciably aldehyde reductase, sorbitol dehydrogenase, or glutathione reductase. The activity of 3 was lowered by inserting various substituents on the pendant phenyl ring, by shifting the acetic acid moiety from the 2 to the 3 position of the TBI nucleus, or by cleaving the TBI system to yield benzimidazolylidenehydrazines as open-chain analogues. A three-dimensional model of human ALR2 was built, taking into account the conformational changes induced by the binding of inhibitors such as zopolrestat, to simulate the docking of 3 into the enzyme active site. The theoretical binding mode of 3 was fully consistent with the structure-activity relationships in the TBI series and will guide the design of novel ALR2 inhibitors.


Subject(s)
Acetates/chemical synthesis , Aldehyde Reductase/antagonists & inhibitors , Benzimidazoles/chemical synthesis , Enzyme Inhibitors/chemical synthesis , Triazines/chemical synthesis , Acetates/chemistry , Acetates/pharmacology , Animals , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Binding Sites , Cataract/etiology , Cataract/prevention & control , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Galactosemias/complications , Humans , Models, Molecular , Ophthalmic Solutions , Protein Binding , Rats , Stereoisomerism , Structure-Activity Relationship , Triazines/chemistry , Triazines/pharmacology
8.
J Med Chem ; 44(3): 316-27, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11462973

ABSTRACT

Radioligand binding assays using bovine cortical membrane preparations and biochemical in vitro studies revealed that various 3-aryl[1,2,4]triazino[4,3-a]benzimidazol-4(10H)-one (ATBI) derivatives, previously reported by us as ligands of the central benzodiazepine receptor (BzR) (Primofiore, G.; et al. J. Med. Chem. 2000, 43, 96-102), behaved as antagonists at the A1 adenosine receptor (A1AR). Alkylation of the nitrogen at position 10 of the triazinobenzimidazole nucleus conferred selectivity for the A1AR vs the BzR. The most potent ligand of the ATBI series (10-methyl-3-phenyl[1,2,4]triazino[4,3-a]benzimidazol-4(10H)-one 12) displayed a Ki value of 63 nM at the A1AR without binding appreciably to the adenosine A2A and A3 nor to the benzodiazepine receptor. Pharmacophore-based modeling studies in which 12 was compared against a set of well-established A1AR antagonists suggested that three hydrogen bonding sites (HB1 acceptor, HB2 and HB3 donors) and three lipophilic pockets (L1, L2, and L3) might be available to antagonists within the A1AR binding cleft. According to the proposed pharmacophore scheme, the lead compound 12 engages interactions with the HB2 site (via the N2 nitrogen) as well as with the L2 and L3 sites (through the pendant and the fused benzene rings). The results of these studies prompted the replacement of the methyl with more lipophilic groups at the 10-position (to fill the putative L1 lipophilic pocket) as a strategy to improve A1AR affinity. Among the new compounds synthesized and tested, the 3,10-diphenyl[1,2,4]triazino[4,3-a]benzimidazol-4(10H)-one (23) was characterized by a Ki value of 18 nM which represents a 3.5-fold gain of A1AR affinity compared with the lead 12. A rhodopsin-based model of the bovine adenosine A1AR was built to highlight the binding mode of 23 and two well-known A1AR antagonists (III and VII) and to guide future lead optimization projects. In our docking simulations, 23 receives a hydrogen bond (via the N1 nitrogen) from the side chain of Asn247 (corresponding to the HB1 and HB2 sites) and fills the L1, L2, and L3 lipophilic pockets with the 10-phenyl, 3-phenyl, and fused benzene rings, respectively.


Subject(s)
Benzimidazoles/chemical synthesis , Purinergic P1 Receptor Antagonists , Amino Acid Sequence , Animals , Benzimidazoles/chemistry , Benzimidazoles/metabolism , Brain/metabolism , Cattle , In Vitro Techniques , Ligands , Models, Molecular , Molecular Sequence Data , Radioligand Assay , Receptors, Purinergic P1/metabolism
9.
J Med Chem ; 44(14): 2286-97, 2001 Jul 05.
Article in English | MEDLINE | ID: mdl-11428922

ABSTRACT

A series of N-(arylalkyl)indol-3-ylglyoxylylamides (4-8) was synthesized as ligands of the benzodiazepine receptor (BzR) and tested for their ability to displace [(3)H]flumazenil from bovine brain membranes. The new compounds, bearing a branched (4) or a geometrically constrained benzyl/phenylethyl amide side chain (5-8), represent the continuation of our research on N-benzylindol-3-ylglyoxylylamides 1 (Da Settimo et al., 1996), N'-phenylindol-3-ylglyoxylohydrazides 2 (Da Settimo et al., 1998), and N-(indol-3-ylglyoxylyl)alanine derivatives 3 (Primofiore et al., 1989). A few indoles belonging to the previously investigated benzylamides 1 and phenylhydrazides 2 were synthesized and tested to enrich the SARs in these two series. The affinities and the GABA ratios of selected compounds for clonal mammalian alpha(1)beta(2)gamma(2), alpha(3)beta(2)gamma(2), and alpha(5)beta(3)gamma(2) BzR subtypes were also determined. It was hypothesized that the reduced flexibility of indoles 4-8 would both facilitate the mapping of the BzR binding cleft and increase the chances of conferring selectivity for the considered receptor subtypes. In the series of indoles 4, the introduction of a methyl group on the benzylic carbon with the R configuration improved affinity of the 5-substituted (5-Cl and 5-NO(2)) derivatives, whereas it was detrimental for their 5-unsubtituted (5-H) counterparts. All S enantiomers were less potent than the R ones. Replacement of the methyl with hydrophilic substituents on the benzylic carbon lowered affinity. The isoindolinylamide side chain was tolerated if the 5-position was unsubstituted (K(i) of 5a = 123 nM), otherwise affinity was abolished (5b, c). All the 2-indanylamides 6 and (S)-1-indanylamides 8 were devoid of any appreciable affinity. The 5-Cl and 5-NO(2) (R)-1-indanylamides 7b (K(i) 80 nM) and 7c (K(i) 28 nM) were the most potent among the indoles 5-8 geometrically constrained about the side chain. The 5-H (R)-1-indanylamide 7a displayed a lower affinity (K(i) 675 nM). The SARs developed from the new compounds, together with those collected from our previous studies, confirmed the hypothesis of different binding modes for 5-substituted and 5-unsubstituted indoles, suggesting that the shape of the lipophilic pocket L(1) (notation in accordance with Cook's BzR topological model) is asymmetric and highlighted the stereoelectronic and conformational properties of the amide side chain required for high potency. Several of the new indoles showed selectivity for the alpha(1)beta(2)gamma(2) subtype compared with the alpha(3)beta(2)gamma(2) and alpha(5)beta(3)gamma(2) subtypes (e.g.: 4t and 7c bind to these three BzR isoforms with K(i) values of 14 nM, 283 nM, 239 nM, and 9 nM, 1960 nM, 95 nM, respectively). The GABA ratios close to unity exhibited by all the tested compounds on each BzR subtype were predictive of an efficacy profile typical of antagonists.


Subject(s)
Glyoxylates/chemical synthesis , Indoles/chemical synthesis , Receptors, GABA-A/metabolism , Amides/chemical synthesis , Amides/chemistry , Amides/metabolism , Animals , Brain/metabolism , Cattle , Glyoxylates/chemistry , Glyoxylates/metabolism , In Vitro Techniques , Indoles/chemistry , Indoles/metabolism , Ligands , Models, Molecular , Radioligand Assay , Structure-Activity Relationship
10.
Ann Anat ; 182(4): 311-8, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10932321

ABSTRACT

In this study we measured the ability of three newly-synthesized N-arylalkylindol-3-ylglyoxylylamide derivatives, which have recently been characterized as partial agonists at central benzodiazepine binding sites, to prevent the rat cardiac mitochondrial alterations resulting from acute loud noise exposure. In particular, we evaluated the effects of these new compounds on the ultrastructural damage induced by noise stress on the right atrium and ventricle after 6 and 12 hr of loud noise exposure. In parallel experiments, we measured the affinity of these compounds for peripheral benzodiazepine binding sites. Following a single injection of the test products, we observed a cardioprotective effect which was more marked after 6 hr compared with 12 hr of noise exposure. Confirming our recent data showing that full agonists at benzodiazepine receptors produce cardioprotection, we demonstrate in this study that partial agonists, like indolylglyoxylylamides, can also produce a cardioprotective effect. Based on their greater affinity in binding studies, the protective activity seems to be related more to their action at central than at peripheral benzodiazepine receptors.


Subject(s)
Benzodiazepinones/pharmacology , Isoquinolines/pharmacology , Mitochondria, Heart/pathology , Myocardium/pathology , Noise , Receptors, GABA-A/metabolism , Stress, Psychological/pathology , Animals , Benzodiazepinones/pharmacokinetics , Binding, Competitive , Cardiotonic Agents/pharmacokinetics , Cardiotonic Agents/pharmacology , Heart Atria , Heart Ventricles , Isoquinolines/pharmacokinetics , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Myocardium/ultrastructure , Rats , Rats, Wistar , Stress, Psychological/metabolism , Time Factors
11.
J Med Chem ; 43(1): 96-102, 2000 Jan 13.
Article in English | MEDLINE | ID: mdl-10633040

ABSTRACT

A series of 3-substituted [1,2,4]triazino[4,3-c]benzimidazoles V were prepared and tested at the central benzodiazepine receptor (BzR). These compounds were designed as rigid analogues of the previously described N-benzylindolylglyoxylylamide derivatives IV. The title compounds V showed an affinity which depended directly on the presence of the N(10)-H group and an aromatic ring at position 3. Some of them elicited a 2- or 3-fold higher affinity with respect to that of the indolylglyoxylylamide derivatives IV (R = H). The GABA ratio and [(35)S]-tert-butylcyclophosphorothionate binding data revealed an efficacy profile of partial inverse agonists/antagonists for compounds 1c,e,f,j,k, and of a partial agonist for 2c. This last compound proved to be effective in antagonizing pentylenetetrazole-induced seizures in mice. Attempts were made to interpret the structure-affinity relationships of compounds V in the light of possible tautomeric equilibria involving the ligands.


Subject(s)
Benzimidazoles/chemical synthesis , Receptors, GABA-A/drug effects , Animals , Anticonvulsants/chemical synthesis , Anticonvulsants/pharmacology , Benzimidazoles/pharmacology , Brain/metabolism , Cattle , Convulsants/chemical synthesis , Convulsants/pharmacology , Diazepam/antagonists & inhibitors , Diazepam/pharmacology , Flumazenil/pharmacology , GABA Modulators/pharmacology , In Vitro Techniques , Ligands , Membranes/drug effects , Membranes/metabolism , Mice , Models, Molecular , Radioligand Assay
SELECTION OF CITATIONS
SEARCH DETAIL
...