Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
J Org Chem ; 86(3): 2499-2521, 2021 02 05.
Article in English | MEDLINE | ID: mdl-33417458

ABSTRACT

Thailanstatin A and spliceostatin D, two naturally occurring molecules endowed with potent antitumor activities by virtue of their ability to bind and inhibit the function of the spliceosome, and their natural siblings and designed analogues, constitute an appealing family of compounds for further evaluation and optimization as potential drug candidates for cancer therapies. In this article, the design, synthesis, and biological investigation of a number of novel thailanstatin A analogues, including some accommodating 1,1-difluorocyclopropyl and tetrahydrooxazine structural motifs within their structures, are described. Important findings from these studies paving the way for further investigations include the identification of several highly potent compounds for advancement as payloads for antibody-drug conjugates (ADCs) as potential targeted cancer therapies and/or small molecule drugs, either alone or in combination with other anticancer agents.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Antineoplastic Agents/pharmacology , Pyrans/pharmacology
2.
J Med Chem ; 59(10): 4926-47, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27077528

ABSTRACT

Transient receptor potential vanilloid 3 (TRPV3) is a Ca(2+)- and Na(+)-permeable channel with a unique expression pattern. TRPV3 is found in both neuronal and non-neuronal tissues, including dorsal root ganglia, spinal cord, and keratinocytes. Recent studies suggest that TRPV3 may play a role in inflammation, pain sensation, and skin disorders. TRPV3 studies have been challenging, in part due to a lack of research tools such as selective antagonists. Herein, we provide the first detailed report on the development of potent and selective TRPV3 antagonists featuring a pyridinyl methanol moiety. Systematic optimization of pharmacological, physicochemical, and ADME properties of original lead 5a resulted in identification of a novel and selective TRPV3 antagonist 74a, which demonstrated a favorable preclinical profile in two different models of neuropathic pain as well as in a reserpine model of central pain.


Subject(s)
Cyclobutanes/chemical synthesis , Cyclobutanes/pharmacology , Pyridines/chemical synthesis , Pyridines/pharmacology , TRPV Cation Channels/antagonists & inhibitors , Calcium/metabolism , Cyclobutanes/chemistry , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Molecular Conformation , Pyridines/chemistry , Structure-Activity Relationship , TRPV Cation Channels/metabolism
3.
Temperature (Austin) ; 2(2): 297-301, 2015.
Article in English | MEDLINE | ID: mdl-27227030

ABSTRACT

Transient receptor potential vanilloid 1 (TRPV1) is a multifunctional ion channel playing important roles in a numerous biological processes including the regulation of body temperature. Within distinct and tight chemical space of chromanyl ureas TRPV1 ligands were identified that exhibit distinctive pharmacology and a spectrum of thermoregulatory effects ranging from hypothermia to hyperthermia. The ability to manipulate these effects by subtle structural modifications of chromanyl ureas may serve as a productive approach in TRPV1 drug discovery programs addressing either side effect or desired target profiles of the compounds. Because chromanyl ureas in the TRPV1 context are generally antagonists, we verified observed partial agonist effects of a subset of compounds within that chemotype by comparing the in vitro profile of Compound 3 with known partial agonist 5'-I-RTX.

4.
J Med Chem ; 57(17): 7412-24, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25100568

ABSTRACT

The synthesis and characterization of a series of selective, orally bioavailable 1-(chroman-4-yl)urea TRPV1 antagonists is described. Whereas first-generation antagonists that inhibit all modes of TRPV1 activation can elicit hyperthermia, the compounds disclosed herein do not elevate core body temperature in preclinical models and only partially block acid activation of TRPV1. Advancing the SAR of this series led to the eventual identification of (R)-1-(7-chloro-2,2-bis(fluoromethyl)chroman-4-yl)-3-(3-methylisoquinolin-5-yl)urea (A-1165442, 52), an analogue that possesses excellent pharmacological selectivity, has a favorable pharmacokinetic profile, and demonstrates good efficacy against osteoarthritis pain in rodents.


Subject(s)
Analgesics/chemistry , Body Temperature/drug effects , TRPV Cation Channels/antagonists & inhibitors , Urea/chemistry , Analgesics/pharmacokinetics , Analgesics/pharmacology , Animals , Area Under Curve , Body Temperature/physiology , Dogs , Dose-Response Relationship, Drug , Drug Discovery , HEK293 Cells , Humans , Isoquinolines/chemistry , Isoquinolines/pharmacokinetics , Isoquinolines/pharmacology , Metabolic Clearance Rate , Models, Chemical , Molecular Structure , Rats , Structure-Activity Relationship , TRPV Cation Channels/chemistry , TRPV Cation Channels/metabolism , Urea/analogs & derivatives , Urea/pharmacokinetics , Urea/pharmacology
5.
J Pharmacol Exp Ther ; 342(2): 416-28, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22570364

ABSTRACT

The transient receptor potential vanilloid-1 (TRPV1) channel is involved in the development and maintenance of pain and participates in the regulation of temperature. The channel is activated by diverse agents, including capsaicin, noxious heat (≥ 43°C), acidic pH (< 6), and endogenous lipids including N-arachidonoyl dopamine (NADA). Antagonists that block all modes of TRPV1 activation elicit hyperthermia. To identify efficacious TRPV1 antagonists that do not affect temperature antagonists representing multiple TRPV1 pharmacophores were evaluated at recombinant rat and human TRPV1 channels with Ca(2+) flux assays, and two classes of antagonists were identified based on their differential ability to inhibit acid activation. Although both classes of antagonists completely blocked capsaicin- and NADA-induced activation of TRPV1, select compounds only partially inhibited activation of the channel by protons. Electrophysiology and calcitonin gene-related peptide release studies confirmed the differential pharmacology of these antagonists at native TRPV1 channels in the rat. Comparison of the in vitro pharmacological properties of these TRPV1 antagonists with their in vivo effects on core body temperature confirms and expands earlier observations that acid-sparing TRPV1 antagonists do not significantly increase core body temperature. Although both classes of compounds elicit equivalent analgesia in a rat model of knee joint pain, the acid-sparing antagonist tested is not effective in a mouse model of bone cancer pain.


Subject(s)
Body Temperature/drug effects , TRPV Cation Channels/antagonists & inhibitors , Analgesics/pharmacology , Animals , Calcitonin Gene-Related Peptide/metabolism , Calcium/metabolism , Capsaicin/pharmacology , Cell Line, Transformed , Fever/drug therapy , Fever/physiopathology , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C3H , Neurons/drug effects , Neurons/metabolism , Pain/drug therapy , Pain/metabolism , Pain/physiopathology , Protons , Rats , Rats, Sprague-Dawley , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , TRPV Cation Channels/metabolism
6.
J Pharmacol Exp Ther ; 341(2): 360-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22319196

ABSTRACT

The transient receptor potential ankyrin-1 (TRPA1) channel has emerged as an attractive target for development of analgesic and anti-inflammatory drugs. However, drug discovery efforts targeting TRPA1 have been hampered by differences between human and rodent species. Many compounds have been identified to have antagonist activity at human TRPA1 (hTRPA1), but when tested at rat TRPA1 (rTRPA1) and mouse TRPA1 (mTRPA1), they show reduced potency as antagonists, no effect, or agonist activity. These compounds are excluded from further drug development because they cannot be tested in preclinical studies using conventional rat/mouse models. To broaden our understanding of species-specific differences, we cloned and functionally characterized rhesus monkey TRPA1 (rhTRPA1) and compared its pharmacological profile to hTRPA1, rTRPA1, and mTRPA1 channels. The functional activities of a diverse group of TRPA1 ligands (both reactive and nonreactive) were determined in a fluorescent Ca²âº influx assay, using transiently transfected human embryonic kidney 293-F cells. 4-Methyl-N-[2,2,2-trichloro-1-(4-nitro-phenylsulfanyl)-ethyl]-benzamide, menthol, and caffeine displayed species-specific differential pharmacology at TRPA1. The pharmacological profile of the rhTRPA1 channel was found to be similar to the hTRPA1 channel. In contrast, the rTRPA1 and mTRPA1 channels closely resembled each other but were pharmacologically distinct from either hTRPA1 or rhTRPA1 channels. Our findings reveal that TRPA1 function differs between primate and rodent species and suggest that rhesus monkey could serve as a surrogate species for humans in preclinical studies.


Subject(s)
Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism , Animals , Calcium/metabolism , Cell Line , DNA, Complementary/genetics , Drug Delivery Systems , Drug Discovery , HEK293 Cells , Haplorhini , Humans , Ligands , Mice , Rats , Species Specificity , Transfection , Transient Receptor Potential Channels/agonists , Transient Receptor Potential Channels/antagonists & inhibitors
7.
J Med Chem ; 55(4): 1751-7, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-22263872

ABSTRACT

A high-throughput screen against human DGAT-1 led to the identification of a core structure that was subsequently optimized to afford the potent, selective, and orally bioavailable compound 14. Oral administration at doses ≥0.03 mg/kg significantly reduced postprandial triglycerides in mice following an oral lipid challenge. Further assessment in both acute and chronic safety pharmacology and toxicology studies demonstrated a clean profile up to high plasma levels, thus culminating in the nomination of 14 as clinical candidate ABT-046.


Subject(s)
Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Pyrazoles/chemical synthesis , Pyrimidines/chemical synthesis , Administration, Oral , Animals , Biological Availability , Caco-2 Cells , Databases, Factual , Diacylglycerol O-Acyltransferase/chemistry , Dogs , Female , Ferrets , Gastrointestinal Transit/drug effects , HeLa Cells , Hemodynamics/drug effects , Humans , Hyperlipidemias/blood , Hyperlipidemias/drug therapy , Male , Mice , Mice, Inbred C57BL , Microsomes, Liver/metabolism , Postprandial Period , Pyrazoles/pharmacokinetics , Pyrazoles/pharmacology , Pyrimidines/pharmacokinetics , Pyrimidines/pharmacology , Rats , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Structure-Activity Relationship , Triglycerides/blood , Vomiting/chemically induced
8.
Synapse ; 66(3): 187-95, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21953601

ABSTRACT

TRPV1 (transient receptor potential vanilloid family type 1) is a nonselective cation channel that is activated and/or sensitized by noxious heat, protons, and other endogenous molecules released following tissue injury. In addition, a role for TRPV1 in mechanotransmission is emerging. We have recently reported that a selective TRPV1 receptor antagonist, A-889425, reduces mechanical allodynia and spinal neuron responses to mechanical stimulation of complete Freund's adjuvant (CFA)-inflamed rat hind paws. The population of peripheral nerve fibers through which TRPV1 antagonists mediate their effect on mechanotransmission have not yet been described. The objective of this study was to characterize TRPV1-mediated modulation of mechanically evoked activity in sensory axons innervating rat hind paws. We used an in vitro skin-nerve preparation to record neural activity from single axons isolated from rat tibial nerve. Single fibers were classified by conduction velocity, mechanical threshold, and stimulus-response relationships. We used A-889425 to investigate uninjured and inflamed skin afferent neuron populations to evoked mechanical stimulation. Application of A-889425 had no effect on the mechanical responsiveness of Aδ and C-fiber units innervating uninjured skin. In contrast, A-889425 inhibited responses of slowly conducting Aδ fiber units to noxious mechanical stimulation in a population of axons innervating CFA-inflamed hind paws. These data support a role for TRPV1 in mechanotransmission following peripheral inflammation, and highlight the importance of a distinct subclass of primary afferent neurons in mediating this effect.


Subject(s)
Mechanotransduction, Cellular/physiology , Neurogenic Inflammation/physiopathology , Neurons, Afferent/physiology , Pyridines/pharmacology , TRPV Cation Channels/metabolism , Animals , Evoked Potentials/drug effects , Evoked Potentials/physiology , Male , Mechanotransduction, Cellular/drug effects , Neural Conduction/drug effects , Neural Conduction/physiology , Nociception/drug effects , Nociception/physiology , Rats , Rats, Sprague-Dawley , Skin/injuries , Skin/innervation , TRPV Cation Channels/antagonists & inhibitors , Tibial Nerve/physiology
9.
Curr Top Med Chem ; 11(17): 2210-5, 2011.
Article in English | MEDLINE | ID: mdl-21671874

ABSTRACT

The vanilloid subfamily of transient receptor potential (TRPV) ion channels serves critical functions in sensory signaling in specialized cells and intact organisms ranging from yeast to primates. As thermosensors, chemosensors, and/or mechanosensors, these channels monitor the local environment and integrate and respond to multiple stimuli distinctively. More than a decade of research on the founding member of the subclass, TRPV1, has led to advancement of multiple antagonists into the clinic for the treatment of chronic pain. In recent years the comprehensive knowledge accessed through these studies has been applied to enhance understanding of other TRPV isoforms and, in particular, to determine whether they, too, represent promising targets for drug discovery. This review focuses on emerging data that define a role for TRPV3 in transducing signals in pain pathways and identify antagonists that demonstrate efficacy in relevant preclinical behavioral models.


Subject(s)
Analgesics/pharmacology , Analgesics/therapeutic use , Pain/drug therapy , TRPV Cation Channels/antagonists & inhibitors , Analgesics/chemistry , Animals , Drug Discovery , Humans , Signal Transduction/drug effects , TRPV Cation Channels/metabolism
10.
Pain ; 152(5): 1165-1172, 2011 May.
Article in English | MEDLINE | ID: mdl-21402443

ABSTRACT

Despite the increasing interest in TRPA1 channel as a pain target, its role in cold sensation and body temperature regulation is not clear; the efficacy and particularly side effects resulting from channel blockade remain poorly understood. Here we use a potent, selective, and bioavailable antagonist to address these issues. A-967079 potently blocks human (IC(50): 51 nmol/L, electrophysiology, 67 nmol/L, Ca(2+) assay) and rat TRPA1 (IC(50): 101 nmol/L, electrophysiology, 289 nmol/L, Ca(2+) assay). It is >1000-fold selective over other TRP channels, and is >150-fold selective over 75 other ion channels, enzymes, and G-protein-coupled receptors. Oral dosing of A-967079 produces robust drug exposure in rodents, and exhibits analgesic efficacy in allyl isothiocyanate-induced nocifensive response and osteoarthritic pain in rats (ED(50): 23.2 mg/kg, p.o.). A-967079 attenuates cold allodynia produced by nerve injury but does not alter noxious cold sensation in naive animals, suggesting distinct roles of TRPA1 in physiological and pathological states. Unlike TRPV1 antagonists, A-967079 does not alter body temperature. It also does not produce locomotor or cardiovascular side effects. Collectively, these data provide novel insights into TRPA1 function and suggest that the selective TRPA1 blockade may present a viable strategy for alleviating pain without untoward side effects.


Subject(s)
Body Temperature Regulation/drug effects , Calcium Channels/metabolism , Cold Temperature/adverse effects , Hyperalgesia/drug therapy , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Pain/physiopathology , Sensation/physiology , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/metabolism , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Body Temperature/drug effects , Body Temperature/physiology , Body Temperature Regulation/genetics , Body Temperature Regulation/physiology , Calcitonin Gene-Related Peptide/metabolism , Calcium/metabolism , Calcium Channels/genetics , Cells, Cultured , Disease Models, Animal , Drug Interactions , Ganglia, Spinal/pathology , Heart Rate/drug effects , Heart Rate/physiology , Humans , Hyperalgesia/physiopathology , Inhibitory Concentration 50 , Isothiocyanates/pharmacology , Magnetic Resonance Imaging/methods , Male , Mice , Nerve Tissue Proteins/genetics , Neurons/drug effects , Oximes/pharmacology , Oximes/therapeutic use , Pain/drug therapy , Pain/genetics , Pain/metabolism , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Reaction Time/drug effects , Sensation/drug effects , Sensory Thresholds/drug effects , TRPA1 Cation Channel , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/genetics , Tritium
11.
Methods Enzymol ; 485: 293-309, 2010.
Article in English | MEDLINE | ID: mdl-21050924

ABSTRACT

Despite increasing use of cell-based assays in biomedical research and drug discovery, one challenge is the adequate supply of high-quality cells expressing the target of interest. To this end, stable cell lines expressing the target are often established, maintained, and expanded in large-scale cell culture. These steps require significant investment of time and resources. Moreover, variability occurs regularly in cell yield, viability, expression, and target activities. In particular, stable expression of many targets, such as ion channels, causes toxicity, cell line degeneration, and loss of functional activity. To circumvent these problems, we utilize large-scale transient transfection (LSTT) to generate a large quantity of cells, which are cryopreserved and readily available for use in cell-based functional assays. Here we describe the application of LSTT cells to ion channel and G protein-coupled receptor (GPCR) assays in a drug discovery setting. This approach can also be applied to many other assay formats and target classes.


Subject(s)
Drug Evaluation, Preclinical/methods , Ion Channels/metabolism , Receptors, G-Protein-Coupled/metabolism , Transfection/methods , Animals , Calcium/analysis , Calcium/metabolism , Cell Line , Cryopreservation/methods , Drug Evaluation, Preclinical/economics , Electrophysiology/methods , Fluorescence Resonance Energy Transfer/methods , Humans , Ion Channels/genetics , Receptors, G-Protein-Coupled/genetics , Transfection/economics
12.
Pain ; 150(2): 319-326, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20621685

ABSTRACT

The TRPV1 antagonist A-995662 demonstrates analgesic efficacy in monoiodoacetate-induced osteoarthritic (OA) pain in rat, and repeated dosing results in increased in vivo potency and a prolonged duration of action. To identify possible mechanism(s) underlying these observations, release of neuropeptides and the neurotransmitter glutamate from isolated spinal cord was measured. In OA rats, basal release of glutamate, bradykinin and calcitonin gene-related peptide (CGRP) was significantly elevated compared to naïve levels, whereas substance P (SP) levels were not changed. In vitro studies showed that capsaicin-evoked TRPV1-dependent CGRP release was 54.7+/-7.7% higher in OA, relative to levels measured for naïve rats, suggesting that TRPV1 activity was higher under OA conditions. The efficacy of A-995662 in OA corresponded with its ability to inhibit glutamate and CGRP release from the spinal cord. A single, fully efficacious dose of A-995662, 100 micromol/kg, reduced spinal glutamate and CGRP release, while a single sub-efficacious dose of A-995662 (25 micromol/kg) was ineffective. Multiple dosing with A-995662 increased the potency and duration of efficacy in OA rats. Changes in efficacy did not correlate with plasma concentrations of A-995662, but were accompanied with reductions in spinal glutamate release. These findings suggest that repeated dosing of TRPV1 antagonists enhances therapeutic potency and duration of action against OA pain, at least in part, by the sustained reduction in release of glutamate and CGRP from the spinal cord.


Subject(s)
Calcitonin Gene-Related Peptide/metabolism , Glutamic Acid/metabolism , Osteoarthritis, Knee/metabolism , Pain/metabolism , Spinal Cord/drug effects , TRPV Cation Channels/antagonists & inhibitors , Tetrahydronaphthalenes/pharmacology , Analysis of Variance , Animals , Bradykinin/metabolism , Osteoarthritis, Knee/chemically induced , Pain/chemically induced , Pain Measurement , Rats , Rats, Sprague-Dawley , Spinal Cord/metabolism , Substance P/metabolism
13.
Bioorg Med Chem ; 18(13): 4821-9, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20570528

ABSTRACT

The synthesis and structure-activity relationships of a series of 5-monosubstituted and 4,5-disubstituted 2-arylaminooxazoles as novel antagonists of the transient receptor potential vanilloid 1 (TRPV1) receptor are described. The 7-hydroxy group of the tetrahydronaphthyl moiety on the 2-amino substituent of the oxazole ring was important for obtaining excellent in vitro potency at the human TRPV1 receptor, while a variety of alkyl and phenyl substituents at the 4- and 5-positions of the oxazole ring were well tolerated and yielded potent TRPV1 antagonists. Despite excellent in vitro potency, the 5-monosubstituted compounds suffered from poor pharmacokinetics. It was found that 4,5-disubstitution on the oxazole ring was critical to the improvement of the overall pharmacokinetic profile of these analogues, which led to the discovery of compound (R)-27, a novel TRPV1 antagonist with good oral activity in preclinical animal models of pain.


Subject(s)
Naphthols/chemical synthesis , Oxazoles/chemistry , TRPV Cation Channels/antagonists & inhibitors , Cell Line , Crystallography, X-Ray , Humans , Molecular Conformation , Naphthols/chemistry , Naphthols/pharmacokinetics , Oxazoles/chemical synthesis , Oxazoles/pharmacokinetics , TRPV Cation Channels/metabolism
14.
Eur J Pharmacol ; 637(1-3): 155-61, 2010 Jul 10.
Article in English | MEDLINE | ID: mdl-20385122

ABSTRACT

Postprandial serum triglyceride concentrations have recently been identified as a major, independent risk factor for future cardiovascular events. As a result, postprandial hyperlipidemia has emerged as a potential therapeutic target. The purpose of this study was two-fold. Firstly, to describe and characterize a standardized model of postprandial hyperlipidemia in multiple rodent species; and secondly, apply these rodent models to the evaluation of a novel class of pharmacologic agent; acyl CoA:diacylglycerol acyltransferase (DGAT) 1 inhibitors. Serum triglycerides were measured before and for 4h after oral administration of a standardized volume of corn oil, to fasted C57BL/6, ob/ob, apoE(-/-) and CD-1 mice; Sprague-Dawley and JCR/LA-cp rats; and normolipidemic and hyperlipidemic hamsters. Intragastric administration of corn oil increased serum triglycerides in all animals evaluated, however the magnitude and time-course of the postprandial triglyceride excursion varied. The potent and selective DGAT-1 inhibitor A-922500 (0.03, 0.3 and 3 mg/kg, p.o.), dose-dependently attenuated the maximal postprandial rise in serum triglyceride concentrations in all species tested. At the highest dose of DGAT-1 inhibitor, the postprandial triglyceride response was abolished. This study provides a comprehensive characterization of the time-course of postprandial hyperlipidemia in rodents. In addition, the ability of DGAT-1 inhibitors to attenuate postprandial hyperlipidemia in multiple rodent models, including those that feature insulin resistance, is documented. Exaggerated postprandial hyperlipidemia is inherent to insulin-resistant states in humans and contributes to the substantially elevated cardiovascular risk observed in these patients. Therefore, by attenuating postprandial hyperlipidemia, DGAT-1 inhibition may represent a novel therapeutic approach to reduce cardiovascular risk.


Subject(s)
Acyl Coenzyme A/metabolism , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Hyperlipidemias/drug therapy , Postprandial Period , Animals , Cardiovascular Diseases/prevention & control , Corn Oil/pharmacology , Cricetinae , Diacylglycerol O-Acyltransferase/metabolism , Dose-Response Relationship, Drug , Hyperlipidemias/blood , Hyperlipidemias/enzymology , Hyperlipidemias/metabolism , Male , Mice , Rats , Risk Factors , Rodentia/classification , Triglycerides/blood , Triglycerides/metabolism
15.
J Pharmacol Exp Ther ; 330(2): 526-31, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19478132

ABSTRACT

Acyl CoA/diacylglycerol acyltransferase (DGAT) 1 is one of two known DGAT enzymes that catalyze the final and only committed step in triglyceride biosynthesis. The purpose of this study was to test the hypothesis that chronic inhibition of DGAT-1 with a small-molecule inhibitor will reduce serum triglyceride concentrations in both genetic and diet-induced models of hypertriglyceridemia. Zucker fatty rats and diet-induced dyslipidemic hamsters were dosed orally with A-922500 (0.03, 0.3, and 3-mg/kg), a potent and selective DGAT-1 inhibitor, for 14 days. Serum triglycerides were significantly reduced by the 3 mg/kg dose of the DGAT-1 inhibitor in both the Zucker fatty rat (39%) and hyperlipidemic hamster (53%). These serum triglyceride changes were accompanied by significant reductions in free fatty acid levels by 32% in the Zucker fatty rat and 55% in the hyperlipidemic hamster. In addition, high-density lipoprotein-cholesterol was significantly increased (25%) in the Zucker fatty rat by A-922500 administered at 3 mg/kg. This study provides the first report that inhibition of DGAT-1, the final and only committed step of triglyceride synthesis, with a selective small-molecule inhibitor, significantly reduces serum triglyceride levels in both genetic and diet-induced animal models of hypertriglyceridemia. The results of this study support further investigation of DGAT-1 inhibition as a novel therapeutic approach to the treatment of hypertriglyceridemia in humans, and they suggest that inhibition of triglyceride synthesis may have more diverse beneficial effects on serum lipid profiles beyond triglyceride lowering.


Subject(s)
Biphenyl Compounds/pharmacology , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Hyperlipidemias/drug therapy , Hyperlipidemias/enzymology , Phenylurea Compounds/pharmacology , Triglycerides/blood , Animals , Biphenyl Compounds/therapeutic use , Body Weight/drug effects , Body Weight/physiology , Cricetinae , Diacylglycerol O-Acyltransferase/blood , Diacylglycerol O-Acyltransferase/physiology , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Hyperlipidemias/blood , Male , Mesocricetus , Phenylurea Compounds/therapeutic use , Rats , Rats, Zucker , Triglycerides/antagonists & inhibitors , Triglycerides/biosynthesis
16.
Pain ; 142(1-2): 27-35, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19135797

ABSTRACT

Transient receptor potential vanilloid type 1 (TRPV1) is a ligand-gated ion channel that functions as an integrator of multiple pain stimuli including heat, acid, capsaicin and a variety of putative endogenous lipid ligands. TRPV1 antagonists have been shown to decrease inflammatory pain in animal models and to produce limited hyperthermia at analgesic doses. Here, we report that ABT-102, which is a potent and selective TRPV1 antagonist, is effective in blocking nociception in rodent models of inflammatory, post-operative, osteoarthritic, and bone cancer pain. ABT-102 decreased both spontaneous pain behaviors and those evoked by thermal and mechanical stimuli in these models. Moreover, we have found that repeated administration of ABT-102 for 5-12 days increased its analgesic activity in models of post-operative, osteoarthritic, and bone cancer pain without an associated accumulation of ABT-102 concentration in plasma or brain. Similar effects were also observed with a structurally distinct TRPV1 antagonist, A-993610. Although a single dose of ABT-102 produced a self-limiting increase in core body temperature that remained in the normal range, the hyperthermic effects of ABT-102 effectively tolerated following twice-daily dosing for 2 days. Therefore, the present data demonstrate that, following repeated administration, the analgesic activity of TRPV1 receptor antagonists is enhanced, while the associated hyperthermic effects are attenuated. The analgesic efficacy of ABT-102 supports its advancement into clinical studies.


Subject(s)
Analgesics/administration & dosage , Fever/drug therapy , Indazoles/administration & dosage , Pain Threshold/drug effects , Pain/drug therapy , TRPV Cation Channels/metabolism , Urea/analogs & derivatives , Animals , Body Temperature/drug effects , Bone Neoplasms/complications , Calcium/metabolism , Disease Models, Animal , Drug Interactions , Fever/chemically induced , Inflammation/complications , Male , Mice , Mice, Inbred C3H , Motor Activity/drug effects , Osteoarthritis/complications , Pain/etiology , Pain Measurement , Rats , Rats, Sprague-Dawley , TRPV Cation Channels/antagonists & inhibitors , Urea/administration & dosage
17.
Mol Pain ; 5: 3, 2009 Jan 21.
Article in English | MEDLINE | ID: mdl-19159452

ABSTRACT

Abundantly expressed in pain-sensing neurons, TRPV1, TRPA1 and TRPM8 are major cellular sensors of thermal, chemical and mechanical stimuli. The function of these ion channels has been attributed to their selective permeation of small cations (e.g., Ca2+, Na+ and K+), and the ion selectivity has been assumed to be an invariant fingerprint to a given channel. However, for TRPV1, the notion of invariant ion selectivity has been revised recently. When activated, TRPV1 undergoes time and agonist-dependent pore dilation, allowing permeation of large organic cations such as Yo-Pro and NMDG+. The pore dilation is of physiological importance, and has been exploited to specifically silence TRPV1-positive sensory neurons. It is unknown whether TRPA1 and TRPM8 undergo pore dilation. Here we show that TRPA1 activation by reactive or non-reactive agonists induces Yo-Pro uptake, which can be blocked by TRPA1 antagonists. In outside-out patch recordings using NMDG+ as the sole external cation and Na+ as the internal cation, TRPA1 activation results in dynamic changes in permeability to NMDG+. In contrast, TRPM8 activation does not produce either Yo-Pro uptake or significant change in ion selectivity. Hence, pore dilation occurs in TRPA1, but not in TRPM8 channels.


Subject(s)
Calcium Channels/physiology , Ion Channel Gating/physiology , Nerve Tissue Proteins/physiology , Porins/metabolism , TRPM Cation Channels/physiology , Transient Receptor Potential Channels/physiology , Aldehydes/pharmacology , Allyl Compounds/pharmacology , Anesthetics, Local/pharmacology , Animals , Benzamides/pharmacology , Benzoxazoles/pharmacokinetics , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Channels/metabolism , Calcium Signaling/physiology , Carbamates/pharmacology , Cells, Cultured , HeLa Cells , Humans , Ion Channel Gating/drug effects , Lidocaine/analogs & derivatives , Lidocaine/pharmacology , Membrane Potentials/drug effects , Membrane Potentials/physiology , Movement/physiology , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Quinolinium Compounds/pharmacokinetics , Rats , TRPA1 Cation Channel , TRPM Cation Channels/agonists , TRPM Cation Channels/metabolism , Thiocyanates/pharmacology , Transient Receptor Potential Channels/agonists , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/metabolism
18.
J Neurosci ; 28(19): 5063-71, 2008 May 07.
Article in English | MEDLINE | ID: mdl-18463259

ABSTRACT

TRPA1 is an excitatory, nonselective cation channel implicated in somatosensory function, pain, and neurogenic inflammation. Through covalent modification of cysteine and lysine residues, TRPA1 can be activated by electrophilic compounds, including active ingredients of pungent natural products (e.g., allyl isothiocyanate), environmental irritants (e.g., acrolein), and endogenous ligands (4-hydroxynonenal). However, how covalent modification leads to channel opening is not understood. Here, we report that electrophilic, thioaminal-containing compounds [e.g., CMP1 (4-methyl-N-[2,2,2-trichloro-1-(4-nitro-phenylsulfanyl)-ethyl]-benzamide)] covalently modify cysteine residues but produce striking species-specific effects [i.e., activation of rat TRPA1 (rTRPA1) and blockade of human TRPA1 (hTRPA1) activation by reactive and nonreactive agonists]. Through characterizing rTRPA1 and hTRPA1 chimeric channels and point mutations, we identified several residues in the upper portion of the S6 transmembrane domains as critical determinants of the opposite channel gating: Ala-946 and Met-949 of rTRPA1 determine channel activation, whereas equivalent residues of hTRPA1 (Ser-943 and Ile-946) determine channel block. Furthermore, side-chain replacements at these critical residues profoundly affect channel function. Therefore, our findings reveal a molecular basis of species-specific channel gating and provide novel insights into how TRPA1 respond to stimuli.


Subject(s)
Benzamides/pharmacology , Calcium Channels/metabolism , Ion Channel Gating/drug effects , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/metabolism , Transient Receptor Potential Channels/antagonists & inhibitors , Transient Receptor Potential Channels/metabolism , Animals , Ankyrins , Calcium Channels/chemistry , Calcium Channels/genetics , Cell Line , Humans , Ion Channel Gating/physiology , Mutation , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Protein Structure, Tertiary , Rats , Species Specificity , TRPA1 Cation Channel , TRPC Cation Channels , Transient Receptor Potential Channels/chemistry , Transient Receptor Potential Channels/genetics
19.
J Med Chem ; 51(3): 380-3, 2008 Feb 14.
Article in English | MEDLINE | ID: mdl-18183944

ABSTRACT

A highly potent and selective DGAT-1 inhibitor was identified and used in rodent models of obesity and postprandial chylomicron excursion to validate DGAT-1 inhibition as a novel approach for the treatment of metabolic diseases. Specifically, compound 4a conferred weight loss and a reduction in liver triglycerides when dosed chronically in DIO mice and depleted serum triglycerides following a lipid challenge in a dose-dependent manner, thus, reproducing major phenotypical characteristics of DGAT-1(-/-) mice.


Subject(s)
Anti-Obesity Agents/chemical synthesis , Cycloheptanes/chemical synthesis , Diacylglycerol O-Acyltransferase/antagonists & inhibitors , Hypolipidemic Agents/chemical synthesis , Keto Acids/chemical synthesis , Urea/analogs & derivatives , Urea/chemical synthesis , Animals , Anti-Obesity Agents/pharmacokinetics , Anti-Obesity Agents/pharmacology , Biphenyl Compounds/chemical synthesis , Biphenyl Compounds/pharmacokinetics , Biphenyl Compounds/pharmacology , Cycloheptanes/pharmacokinetics , Cycloheptanes/pharmacology , Diacylglycerol O-Acyltransferase/genetics , Eating/drug effects , Humans , Hypolipidemic Agents/pharmacokinetics , Hypolipidemic Agents/pharmacology , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Keto Acids/pharmacokinetics , Keto Acids/pharmacology , Liver/metabolism , Mice , Mice, Mutant Strains , Stereoisomerism , Structure-Activity Relationship , Triglycerides/metabolism , Urea/pharmacokinetics , Urea/pharmacology , Weight Loss
20.
Front Biosci ; 12: 3781-94, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17485339

ABSTRACT

Stearoyl-CoA desaturases (SCDs) catalyze the biosynthesis of monounsaturated fatty acids from saturated fatty acids. Four scd genes have been identified in mice and three in human (including one pseudogene). Among the four mouse SCD isoforms, SCD1 is predominantly expressed in liver and adipose tissue. Mice null for the scd1 gene have reduced adiposity, increased energy expenditure and altered lipid profiles. To further evaluate the specific role of hepatic SCD1 and the potential to achieve similar desirable phenotypic changes in adult obese mice, adenovirus-mediated short hairpin interfering RNA (shRNA) was used to acutely knock down hepatic scd1 expression in ob/ob mice. Robust reductions in hepatic SCD1 mRNA and SCD1 enzymatic activity were achieved, sustained up to 2 weeks. Reduced hepatic content of neutral lipids and robust lowering of lipid desaturation indexes, but increased content of liver phosphotidylcholine were observed with SCD1 knockdown. Increased total plasma cholesterol levels were also observed. No significant changes in body weight were observed. Expression levels of several lipogenic and lipid oxidation genes were not significantly altered by short term SCD1 reduction, but UCP2 expression was increased. Our results demonstrate that significant changes to both hepatic and systemic lipid profiles can be achieved through specific knockdown of liver-expressed SCD1 in the ob/ob mouse model. However, hepatic SCD1 knockdown does not result in significant changes in body weight in the short term.


Subject(s)
Fatty Acids/chemistry , Lipids/chemistry , Liver/enzymology , Obesity/enzymology , RNA Interference , Stearoyl-CoA Desaturase/metabolism , Animals , Mice , Obesity/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...