Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 185: 114451, 2021 03.
Article in English | MEDLINE | ID: mdl-33545115

ABSTRACT

The secretin receptor (SCTR) is a prototypic Class B1 G protein-coupled receptor (GPCR) that represents a key target for the development of therapeutics for the treatment of cardiovascular, gastrointestinal, and metabolic disorders. However, no non-peptidic molecules targeting this receptor have yet been disclosed. Using a high-throughput screening campaign directed at SCTR to identify small molecule modulators, we have identified three structurally related scaffolds positively modulating SCTRs. Here we outline a comprehensive study comprising a structure-activity series based on commercially available analogs of the three hit scaffold sets A (2-sulfonyl pyrimidines), B (2-mercapto pyrimidines) and C (2-amino pyrimidines), which revealed determinants of activity, cooperativity and specificity. Structural optimization of original hits resulted in analog B2, which substantially enhances signaling of truncated secretin peptides and prolongs residence time of labeled secretin up to 13-fold in a dose-dependent manner. Furthermore, we found that investigated compounds display structural similarity to positive allosteric modulators (PAMs) active at the glucagon-like peptide-1 receptor (GLP-1R), and we were able to confirm cross-recognition of that receptor by a subset of analogs. Studies using SCTR and GLP-1R mutants revealed that scaffold A, but not B and C, likely acts via two distinct mechanisms, one of which constitutes covalent modification of Cys-347GLP-1R known from GLP-1R-selective modulators. The scaffolds identified in this study might not only serve as novel pharmacologic tools to decipher SCTR- or GLP-1R-specific signaling pathways, but also as structural leads to elucidate allosteric binding sites facilitating the future development of orally available therapeutic approaches targeting these receptors.


Subject(s)
Drug Discovery/methods , Pyrimidines/chemistry , Pyrimidines/metabolism , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/metabolism , Receptors, Gastrointestinal Hormone/chemistry , Receptors, Gastrointestinal Hormone/metabolism , Allosteric Regulation/drug effects , Allosteric Regulation/physiology , Amino Acid Sequence , Animals , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Protein Binding/physiology , Pyrimidines/pharmacology , Rats , Structure-Activity Relationship
2.
SLAS Discov ; 26(1): 1-16, 2021 01.
Article in English | MEDLINE | ID: mdl-32749201

ABSTRACT

The secretin receptor (SCTR), a prototypical class B G protein-coupled receptor (GPCR), exerts its effects mainly by activating Gαs proteins upon binding of its endogenous peptide ligand secretin. SCTRs can be found in a variety of tissues and organs across species, including the pancreas, stomach, liver, heart, lung, colon, kidney, and brain. Beyond that, modulation of SCTR-mediated signaling has therapeutic potential for the treatment of multiple diseases, such as heart failure, obesity, and diabetes. However, no ligands other than secretin and its peptide analogs have been described to regulate SCTRs, probably due to inherent challenges in family B GPCR drug discovery. Here we report creation of a testing funnel that allowed targeted detection of SCTR small-molecule activators. Pursuing the strategy to identify positive allosteric modulators (PAMs), we established a unique primary screening assay employing a mixture of three orthosteric stimulators that was compared in a screening campaign testing 12,000 small-molecule compounds. Beyond that, we developed a comprehensive set of secondary assays, such as a radiolabel-free target engagement assay and a NanoBiT (NanoLuc Binary Technology)-based approach to detect ß-arrestin-2 recruitment, all feasible in a high-throughput environment as well as capable of profiling ligands and hits regarding their effect on binding and receptor function. This combination of methods enabled the discovery of five promising scaffolds, four of which have been validated and further characterized with respect to their allosteric activities. We propose that our results may serve as starting points for developing the first in vivo active small molecules targeting SCTRs.


Subject(s)
Drug Development/methods , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Receptors, Gastrointestinal Hormone/antagonists & inhibitors , Receptors, Gastrointestinal Hormone/chemistry , Animals , Biobehavioral Sciences , CHO Cells , Calcium/metabolism , Carrier Proteins , Cricetulus , Cyclic AMP/metabolism , Gene Expression , Genes, Reporter , HEK293 Cells , High-Throughput Screening Assays/methods , Humans , Ligands , Peptides/chemistry , Peptides/pharmacology , Protein Binding , Protein Conformation , Structure-Activity Relationship
3.
Nat Chem Biol ; 13(5): 486-493, 2017 05.
Article in English | MEDLINE | ID: mdl-28244987

ABSTRACT

The proteasome is a vital cellular machine that maintains protein homeostasis, which is of particular importance in multiple myeloma and possibly other cancers. Targeting of proteasome 20S peptidase activity with bortezomib and carfilzomib has been widely used to treat myeloma. However, not all patients respond to these compounds, and those who do eventually suffer relapse. Therefore, there is an urgent and unmet need to develop new drugs that target proteostasis through different mechanisms. We identified quinoline-8-thiol (8TQ) as a first-in-class inhibitor of the proteasome 19S subunit Rpn11. A derivative of 8TQ, capzimin, shows >5-fold selectivity for Rpn11 over the related JAMM proteases and >2 logs selectivity over several other metalloenzymes. Capzimin stabilized proteasome substrates, induced an unfolded protein response, and blocked proliferation of cancer cells, including those resistant to bortezomib. Proteomic analysis revealed that capzimin stabilized a subset of polyubiquitinated substrates. Identification of capzimin offers an alternative path to develop proteasome inhibitors for cancer therapy.


Subject(s)
Proteasome Inhibitors/pharmacology , Quinolines/pharmacology , Trans-Activators/antagonists & inhibitors , Dose-Response Relationship, Drug , Humans , Molecular Structure , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/chemistry , Quinolines/chemistry , Structure-Activity Relationship , Trans-Activators/metabolism
4.
Bioorg Med Chem Lett ; 24(3): 1000-1004, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24412070

ABSTRACT

Alkaline phosphatase (AP) isozymes are present in a wide range of species from bacteria to man and are capable of dephosphorylation and transphosphorylation of a wide spectrum of substrates in vitro. In humans, four AP isozymes have been identified-one tissue-nonspecific (TNAP) and three tissue-specific-named according to the tissue of their predominant expression: intestinal (IAP), placental (PLAP) and germ cell (GCAP) APs. Modulation of activity of the different AP isozymes may have therapeutic implications in distinct diseases and cellular processes. For instance, changes in the level of IAP activity can affect gut mucosa tolerance to microbial invasion due to the ability of IAP to detoxify bacterial endotoxins, alter the absorption of fatty acids and affect ectopurinergic regulation of duodenal bicarbonate secretion. To identify isozyme selective modulators of the human and mouse IAPs, we developed a series of murine duodenal IAP (Akp3-encoded dIAP isozyme), human IAP (hIAP), PLAP, and TNAP assays. High throughput screening and subsequent SAR efforts generated a potent inhibitor of dIAP, ML260, with specificity for the Akp3-, compared to the Akp5- and Akp6-encoded mouse isozymes.


Subject(s)
Acetanilides/chemistry , Acetanilides/pharmacology , Alkaline Phosphatase/antagonists & inhibitors , Sulfonamides/chemistry , Sulfonamides/pharmacology , Acetanilides/isolation & purification , Animals , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/isolation & purification , Enzyme Inhibitors/pharmacology , Humans , Mice , Protein Isoforms/chemistry , Sulfonamides/isolation & purification
5.
ACS Chem Biol ; 7(2): 367-77, 2012 Feb 17.
Article in English | MEDLINE | ID: mdl-22070201

ABSTRACT

The hematopoietic protein tyrosine phosphatase (HePTP) is implicated in the development of blood cancers through its ability to negatively regulate the mitogen-activated protein kinases (MAPKs) ERK1/2 and p38. Small-molecule modulators of HePTP activity may become valuable in treating hematopoietic malignancies such as T cell acute lymphoblastic leukemia (T-ALL) and acute myelogenous leukemia (AML). Moreover, such compounds will further elucidate the regulation of MAPKs in hematopoietic cells. Although transient activation of MAPKs is crucial for growth and proliferation, prolonged activation of these important signaling molecules induces differentiation, cell cycle arrest, cell senescence, and apoptosis. Specific HePTP inhibitors may promote the latter and thereby may halt the growth of cancer cells. Here, we report the development of a small molecule that augments ERK1/2 and p38 activation in human T cells, specifically by inhibiting HePTP. Structure-activity relationship analysis, in silico docking studies, and mutagenesis experiments reveal how the inhibitor achieves selectivity for HePTP over related phosphatases by interacting with unique amino acid residues in the periphery of the highly conserved catalytic pocket. Importantly, we utilize this compound to show that pharmacological inhibition of HePTP not only augments but also prolongs activation of ERK1/2 and, especially, p38. Moreover, we present similar effects in leukocytes from mice intraperitoneally injected with the inhibitor at doses as low as 3 mg/kg. Our results warrant future studies with this probe compound that may establish HePTP as a new drug target for acute leukemic conditions.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Protein Tyrosine Phosphatases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Line , Enzyme Activation/drug effects , Humans , Mice , Mice, Inbred C57BL , Models, Molecular , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/metabolism , Structure-Activity Relationship , T-Lymphocytes/drug effects , T-Lymphocytes/enzymology
6.
J Biomol Screen ; 17(3): 350-60, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22156224

ABSTRACT

Overexpression of the anti-apoptotic Bcl-2 family proteins occurs commonly in human cancers. Bfl-1 is highly expressed in some types of malignant cells, contributing significantly to tumor cell survival and chemoresistance. Therefore, it would be desirable to have chemical antagonists of Bfl-1. To this end, we devised a fluorescence polarization assay (FPA) using Bfl-1 protein and fluorescein-conjugated Bid BH3 peptide, which was employed for high-throughput screening of chemical libraries. Approximately 66 000 compounds were screened for the ability to inhibit BH3 peptide binding to Bfl-1, yielding 14 reproducible hits with ≥50% displacement. After dose-response analysis and confirmation using a secondary assay based on time-resolved fluorescence resonance energy transfer (TR-FRET), two groups of Bfl-1-specific inhibitors were identified, including chloromaleimide and sulfonylpyrimidine series compounds. FPAs generated for each of the six anti-apoptotic Bcl-2 proteins demonstrated selective binding of both classes of compounds to Bfl-1. Analogs of the sulfonylpyrimidine series were synthesized and compared with the original hit for Bfl-1 binding by both FPAs and TR-FRET assays. The resulting structure-activity relation analysis led to the chemical probe compound CID-2980973 (ML042). Collectively, these findings demonstrate the feasibility of using the HTS assay for discovery of selective chemical inhibitors of Bfl-1.


Subject(s)
Drug Evaluation, Preclinical/methods , High-Throughput Screening Assays/methods , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/metabolism , Small Molecule Libraries/analysis , Fluorescence , Fluorescence Polarization/methods , Fluorescence Resonance Energy Transfer/methods , HeLa Cells , Humans , Maleimides/metabolism , Maleimides/pharmacology , Minor Histocompatibility Antigens , Neoplasms/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Pyrimidines/metabolism , Pyrimidines/pharmacology
7.
ACS Med Chem Lett ; 2(10): 780-785, 2011 Oct 13.
Article in English | MEDLINE | ID: mdl-22003428

ABSTRACT

NOD1 (nucleotide-binding oligomerization domain 1) protein is a member of the NLR (NACHT and leucine rich repeat domain containing proteins) protein family, which plays a key role in innate immunity as a sensor of specific microbial components derived from bacterial peptidoglycans and induction of inflammatory responses. Mutations in NOD proteins have been associated with various inflammatory diseases that affect NF-κB (nuclear factor κB) activity, a major signaling pathway involved in apoptosis, inflammation, and immune response. A luciferase-based reporter gene assay was utilized in a high-throughput screening program conducted under the NIH-sponsored Molecular Libraries Probe Production Center Network program to identify the active scaffolds. Herein, we report the chemical synthesis, structure-activity relationship studies, downstream counterscreens, secondary assay data, and pharmacological profiling of the 2-aminobenzimidazole lead (compound 1c, ML130) as a potent and selective inhibitor of NOD1-induced NF-κB activation.

8.
Chem Biol ; 18(7): 825-32, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21802003

ABSTRACT

NLR family proteins play important roles in innate immune response. NOD1 (NLRC1) activates various signaling pathways including NF-κB in response to bacterial ligands. Hereditary polymorphisms in the NOD1 gene are associated with asthma, inflammatory bowel disease, and other disorders. Using a high throughput screening (HTS) assay measuring NOD1-induced NF-κB reporter gene activity, followed by multiple downstream counter screens that eliminated compounds impacting other NF-κB effectors, 2-aminobenzimidazole compounds were identified that selectively inhibit NOD1. Mechanistic studies of a prototypical compound, Nodinitib-1 (ML130; CID-1088438), suggest that these small molecules cause conformational changes of NOD1 in vitro and alter NOD1 subcellular targeting in cells. Altogether, this inaugural class of inhibitors provides chemical probes for interrogating mechanisms regulating NOD1 activity and tools for exploring the roles of NOD1 in various infectious and inflammatory diseases.


Subject(s)
Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Nod1 Signaling Adaptor Protein/antagonists & inhibitors , Signal Transduction/drug effects , Cell Line , Cells, Cultured , Dendritic Cells/drug effects , Drug Evaluation, Preclinical , Genes, Reporter/drug effects , High-Throughput Screening Assays , Humans , NF-kappa B/genetics , Nod1 Signaling Adaptor Protein/immunology
9.
J Med Chem ; 54(10): 3661-8, 2011 May 26.
Article in English | MEDLINE | ID: mdl-21539312

ABSTRACT

We report the discovery and validation of a series of benzoisothiazolones as potent inhibitors of phosphomannose isomerase (PMI), an enzyme that converts mannose-6-phosphate (Man-6-P) into fructose-6-phosphate (Fru-6-P) and, more importantly, competes with phosphomannomutase 2 (PMM2) for Man-6-P, diverting this substrate from critical protein glycosylation events. In congenital disorder of glycosylation type Ia, PMM2 activity is compromised; thus, PMI inhibition is a potential strategy for the development of therapeutics. High-throughput screening (HTS) and subsequent chemical optimization led to the identification of a novel class of benzoisothiazolones as potent PMI inhibitors having little or no PMM2 inhibition. Two complementary synthetic routes were developed, enabling the critical structural requirements for activity to be determined, and the compounds were subsequently profiled in biochemical and cellular assays to assess efficacy. The most promising compounds were also profiled for bioavailability parameters, including metabolic stability, plasma stability, and permeability. The pharmacokinetic profile of a representative of this series (compound 19; ML089) was also assessed, demonstrating the potential of this series for in vivo efficacy when dosed orally in disease models.


Subject(s)
Congenital Disorders of Glycosylation/diagnosis , Mannose-6-Phosphate Isomerase/antagonists & inhibitors , Thiazoles/chemistry , Administration, Oral , Chemistry, Pharmaceutical/methods , Combinatorial Chemistry Techniques/methods , Congenital Disorders of Glycosylation/genetics , Drug Design , Humans , Inhibitory Concentration 50 , Models, Chemical , Permeability , Structure-Activity Relationship
10.
Bioorg Med Chem Lett ; 20(22): 6560-4, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20933419

ABSTRACT

High-throughput screening of 66,000 compounds using competitive binding of peptides comprising the BH3 domain to anti-apoptotic Bfl-1 led to the identification of 14 validated 'hits' as inhibitors of Bfl-1. N-Aryl maleimide 1 was among the validated 'hits'. A chemical library encompassing over 280 analogs of 1 was prepared following a two-step synthesis. Structure-activity studies for inhibition of Bfl-1 by analogs of N-aryl maleimide 1 revealed a preference for electron-withdrawing substituents in the N-aryl ring and hydrophilic amines appended to the maleimide core. Inhibitors of Bfl-1 are potential development candidates for anti-cancer therapeutics.


Subject(s)
Maleimides/pharmacology , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Humans , Maleimides/chemistry , Minor Histocompatibility Antigens , Structure-Activity Relationship
11.
Biochem Biophys Res Commun ; 394(1): 194-9, 2010 Mar 26.
Article in English | MEDLINE | ID: mdl-20188705

ABSTRACT

Binding of leukocyte specific integrin CD11b/CD18 to its physiologic ligands is important for the development of normal immune response in vivo. Integrin CD11b/CD18 is also a key cellular effector of various inflammatory and autoimmune diseases. However, small molecules selectively inhibiting the function of integrin CD11b/CD18 are currently lacking. We used a newly described cell-based high-throughput screening assay to identify a number of highly potent antagonists of integrin CD11b/CD18 from chemical libraries containing >100,000 unique compounds. Computational analyses suggest that the identified compounds cluster into several different chemical classes. A number of the newly identified compounds blocked adhesion of wild-type mouse neutrophils to CD11b/CD18 ligand fibrinogen. Mapping the most active compounds against chemical fingerprints of known antagonists of related integrin CD11a/CD18 shows little structural similarity, suggesting that the newly identified compounds are novel and unique.


Subject(s)
CD11b Antigen/metabolism , CD18 Antigens/metabolism , High-Throughput Screening Assays , Small Molecule Libraries , Animals , Ligands , Mice , Mice, Inbred Strains , Neutrophils/drug effects , Neutrophils/immunology
12.
J Med Chem ; 52(21): 6919-25, 2009 Nov 12.
Article in English | MEDLINE | ID: mdl-19821572

ABSTRACT

We report the characterization and optimization of drug-like small molecule inhibitors of tissue-nonspecific alkaline phosphatase (TNAP), an enzyme critical for the regulation of extracellular matrix calcification during bone formation and growth. High-throughput screening (HTS) of a small molecule library led to the identification of arylsulfonamides as potent and selective inhibitors of TNAP. Critical structural requirements for activity were determined, and the compounds were subsequently profiled for in vitro activity and bioavailability parameters including metabolic stability and permeability. The plasma levels following subcutaneous administration of a member of the lead series in rat was determined, demonstrating the potential of these TNAP inhibitors as systemically active therapeutic agents to target various diseases involving soft tissue calcification. A representative member of the series was also characterized in mechanistic and kinetic studies.


Subject(s)
Alkaline Phosphatase/antagonists & inhibitors , Quinolines/chemical synthesis , Sulfonamides/chemical synthesis , Animals , Biological Availability , COS Cells , Calcinosis/prevention & control , Catalysis , Chlorocebus aethiops , In Vitro Techniques , Microsomes, Liver/metabolism , Permeability , Quinolines/pharmacokinetics , Quinolines/pharmacology , Rats , Solubility , Structure-Activity Relationship , Sulfonamides/pharmacokinetics , Sulfonamides/pharmacology
13.
J Biomol Screen ; 14(7): 824-37, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19556612

ABSTRACT

Tissue-nonspecific alkaline phosphatase (TNAP) is a ubiquitous enzyme expressed at high levels in bone, liver, and kidney. It appears involved in dephosphorylation of numerous phosphate monoesters, but only 2 of them, pyrophosphate and pyridoxal phosphate, have yet been unequivocally documented. Discovery and characterization of other substrates could be considerably facilitated if specific and potent modulators of TNAP activity with various modes of action were available. Here, the authors describe in detail a high-throughput screening campaign to identify inhibitors of TNAP, performed within the Molecular Library Screening Center Network (MLSCN). A novel homogeneous luminescent TNAP assay was developed and optimized with respect to the enzyme and substrate concentrations, enabling identification of a large number of compounds overlooked by a conventional colorimetric assay. Several new chemical series were identified from screening the Molecular Libraries Small Molecule Repository (MLSMR) collection and demonstrated to have diverse selectivity and mode of inhibition profiles. The nanomolar potency of some of these scaffolds surpasses currently known inhibitors. This article provides an example of a success where the Roadmap Initiative collaborative model, sponsored by the National Institutes of Health, brought together a deep knowledge of target biology from a principal investigator's laboratory, a well-designed compound collection from the MLSMR, and an industrial-level screening facility and staff at the MLSCN center to identify pharmacologically active compounds, with outstanding selectivity data from a panel of more than 200 publicly accessible assays, through a high-throughput screen.


Subject(s)
Alkaline Phosphatase/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/analysis , Enzyme Inhibitors/pharmacology , Animals , Biocatalysis/drug effects , COS Cells , Chlorocebus aethiops , Colorimetry , Enzyme Inhibitors/chemistry , High-Throughput Screening Assays , Luminescent Measurements , Organ Specificity/drug effects , Reproducibility of Results , Substrate Specificity/drug effects , Sulfanilamide , Sulfanilamides/chemistry
14.
Bioorg Med Chem Lett ; 19(1): 222-5, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19038545

ABSTRACT

Tissue-nonspecific alkaline phosphatase (TNAP) plays a central role in regulating extracellular matrix calcification during bone formation and growth. High-throughput screening (HTS) for small molecule TNAP inhibitors led to the identification of hits in the sub-micromolar potency range. We report the design, synthesis and in vitro evaluation of a series of pyrazole derivatives of a screening hit which are potent TNAP inhibitors exhibiting IC(50) values as low as 5nM. A representative of the series was characterized in kinetic studies and determined to have a mode of inhibition not previously observed for TNAP inhibitors.


Subject(s)
Alkaline Phosphatase/antagonists & inhibitors , Enzyme Inhibitors/chemical synthesis , Pyrazoles/chemical synthesis , Drug Design , Drug Evaluation, Preclinical , Humans , Inhibitory Concentration 50 , Kinetics , Pyrazoles/pharmacology
15.
Biochem Pharmacol ; 72(2): 244-55, 2006 Jul 14.
Article in English | MEDLINE | ID: mdl-16750175

ABSTRACT

The molecular interactions between non-peptide antagonists and the corticotropin-releasing factor type 1 (CRF1) receptor are poorly understood. A CRF1 receptor mutation has been identified that reduces binding affinity of the non-peptide antagonist NBI 27914 (M276I in transmembrane domain 5). We have investigated the mechanism of the mutation's effect using a combination of peptide and non-peptide ligands and receptor mutations. The M276I mutation reduced binding affinity of standard non-peptide antagonists 5-75-fold while having no effect on peptide ligand binding. We hypothesized that the side chain of isoleucine, beta-branched and so rotationally constrained when within an alpha-helix, introduces a barrier to non-peptide antagonist binding. In agreement with this hypothesis, mutation of M276 to the rotationally constrained valine produced similar reductions of affinity as M276I mutation, whereas mutation to leucine (with an unbranched beta-carbon) minimally affected non-peptide antagonist affinity. Mutation to alanine did not appreciably affect non-peptide antagonist affinity, implying the methionine side chain does not contribute directly to binding. Three observations suggested M276I/V mutations interfere with binding of the heterocyclic core of the compounds: (1) all compounds affected by M276I/V mutations possess a planar heterocyclic core. (2) None of the M276 mutations affected binding of an acylic compound. (3) The mutations differentially affected affinity of two compounds that differ only by core methylation. These findings imply that non-peptide antagonists, and specifically the heterocyclic core of such molecules, bind in the vicinity of M276 of the CRF1 receptor. M276 mutations did not affect peptide ligand binding and this residue is distant from determinants of peptide binding (predominantly in the extracellular regions), providing molecular evidence for non-overlapping (allosteric) binding sites for peptide and non-peptide ligands within the CRF1 receptor.


Subject(s)
Amino Acids/chemistry , Receptors, Corticotropin-Releasing Hormone/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Molecular Sequence Data , Mutation , Radioligand Assay , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/chemistry
16.
Endocrinology ; 146(6): 2766-73, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15774557

ABSTRACT

Cachexia is metabolic disorder characterized by anorexia, an increased metabolic rate, and loss of lean body mass. It is a relatively common disorder, and is a pathological feature of diseases such as cancer, HIV infection, and renal failure. Recent studies have demonstrated that cachexia brought about by a variety of illnesses can be attenuated or reversed by blocking activation of the melanocortin 4 subtype receptor (MC4-R) within the central nervous system. Although the potential use of central MC4-R antagonists for the treatment of cachexia was supported by these studies, utility was limited by the need to deliver these agents intracerebroventricularly. In the current study, we present a series of experiments demonstrating that peripheral administration of a small molecule MC4-R antagonist can effectively stimulate daytime (satiated) food intake as well as decrease basal metabolic rate in normal animals. Furthermore, this compound attenuated cachexia and preserved lean body mass in a murine cancer model. These data clearly demonstrate the potential of small molecule MC4-R antagonists in the treatment of cachexia and underscore the importance of melanocortin signaling in the development of this metabolic disorder.


Subject(s)
Cachexia/drug therapy , Carcinoma, Lewis Lung/complications , Lung Neoplasms/complications , Receptor, Melanocortin, Type 4/antagonists & inhibitors , alpha-MSH/analogs & derivatives , Animals , Cachexia/etiology , Cachexia/prevention & control , Cell Line , Energy Metabolism/drug effects , Feeding Behavior/drug effects , Humans , Iodine Radioisotopes , Kidney/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Transplantation , Radioligand Assay , Receptor, Melanocortin, Type 3/genetics , Receptor, Melanocortin, Type 4/genetics , alpha-MSH/metabolism , alpha-MSH/pharmacology
17.
Bioorg Med Chem Lett ; 14(5): 1151-4, 2004 Mar 08.
Article in English | MEDLINE | ID: mdl-14980654

ABSTRACT

Diarylimidazolecarboxamides and diaryltriazolecarboxamides related to SR141716 were synthesized and tested for binding to the human CB(1) receptor. Suitably substituted imidazoles are comparably potent to the clinical candidate, whereas the analogous triazoles are less so due to the absence of an additional substituent on the azole ring.


Subject(s)
Imidazoles/metabolism , Piperidines/metabolism , Pyrazoles/metabolism , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Triazoles/metabolism , Imidazoles/chemistry , Piperidines/chemistry , Protein Binding , Pyrazoles/chemistry , Rimonabant , Triazoles/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...