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










Database
Language
Publication year range
1.
Medchemcomm ; 5(6): 826-830, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-25068033

ABSTRACT

Despite a rising demand for anti-obesity therapeutics, few effective pharmacological options are clinically available that target the synthesis and accumulation of body fat. Moderate inhibition of mammalian glycerol-3-phosphate acyltransferase (GPAT) with 2-(alkanesulfonamido)benzoic acids has recently been described in vitro, accompanied by promising weight loss in vivo. In silico docking studies with 2-(octanesulfonamido)benzoic acid modeled into the active site of squash GPAT revealed an unoccupied volume lined with hydrophobic residues proximal to C-4 and C-5 of the benzoic acid ring. In an effort to produce more potent GPAT inhibitors, a series of 4- and 5-substituted analogs were designed, synthesized, and evaluated for inhibitory activity. In general, compounds containing hydrophobic substituents at the 4- and 5-positions, such as biphenyl and alkylphenyl hydrocarbons, exhibited an improved inhibitory activity against GPAT in vitro. The most active compound, 4-([1,1'-biphenyl]-4-carbonyl)-2-(octanesulfonamido)benzoic acid, demonstrated an IC50 of 8.5 µM and represents the best GPAT inhibitor discovered to date. Conversely, further substitution with hydroxyl or fluoro groups, led to a 3-fold decrease in activity. These results are consistent with the presence of a hydrophobic pocket and may support the binding model as a potential tool for developing more potent inhibitors.

2.
Am J Physiol Regul Integr Comp Physiol ; 301(1): R116-30, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21490364

ABSTRACT

Storage of excess calories as triglycerides is central to obesity and its associated disorders. Glycerol-3-phosphate acyltransferases (GPATs) catalyze the initial step in acylglyceride syntheses, including triglyceride synthesis. We utilized a novel small-molecule GPAT inhibitor, FSG67, to investigate metabolic consequences of systemic pharmacological GPAT inhibition in lean and diet-induced obese (DIO) mice. FSG67 administered intraperitoneally decreased body weight and energy intake, without producing conditioned taste aversion. Daily FSG67 (5 mg/kg, 15.3 µmol/kg) produced gradual 12% weight loss in DIO mice beyond that due to transient 9- to 10-day hypophagia (6% weight loss in pair-fed controls). Continued FSG67 maintained the weight loss despite return to baseline energy intake. Weight was lost specifically from fat mass. Indirect calorimetry showed partial protection by FSG67 against decreased rates of oxygen consumption seen with hypophagia. Despite low respiratory exchange ratio due to a high-fat diet, FSG67-treated mice showed further decreased respiratory exchange ratio, beyond pair-fed controls, indicating enhanced fat oxidation. Chronic FSG67 increased glucose tolerance and insulin sensitivity in DIO mice. Chronic FSG67 decreased gene expression for lipogenic enzymes in white adipose tissue and liver and decreased lipid accumulation in white adipose, brown adipose, and liver tissues without signs of damage. RT-PCR showed decreased gene expression for orexigenic hypothalamic neuropeptides AgRP or NPY after acute and chronic systemic FSG67. FSG67 given intracerebroventricularly (100 and 320 nmol icv) produced 24-h weight loss and feeding suppression, indicating contributions from direct central nervous system sites of action. Together, these data point to GPAT as a new potential therapeutic target for the management of obesity and its comorbidities.


Subject(s)
Adiposity/physiology , Eating/physiology , Enzyme Inhibitors/pharmacology , Glycerol-3-Phosphate O-Acyltransferase/antagonists & inhibitors , Insulin Resistance/physiology , Obesity/physiopathology , Adiposity/drug effects , Agouti-Related Protein/metabolism , Animals , Dietary Fats/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Eating/drug effects , Fatty Liver/metabolism , Fatty Liver/physiopathology , Glycerol-3-Phosphate O-Acyltransferase/physiology , Mice , Mice, Inbred Strains , Mitochondria, Liver/drug effects , Mitochondria, Liver/enzymology , Neuropeptide Y/metabolism , Obesity/etiology , Obesity/metabolism , Oxygen Consumption/drug effects , Oxygen Consumption/physiology , Thinness/metabolism , Thinness/physiopathology , Triglycerides/metabolism
3.
Bioorg Med Chem ; 18(17): 6470-9, 2010 Sep 01.
Article in English | MEDLINE | ID: mdl-20692840

ABSTRACT

Glycerol 3-phosphate acyltransferase (GPAT) isozymes are central control points for fat synthesis in mammals. Development of inhibitors of these membrane-bound enzymes could lead to an effective treatment for obesity, but is thwarted by an absence of direct structural information. Based on a highly successful study involving conformationally constrained glycerol 3-phosphate analogs functioning as potent glycerol 3-phosphate dehydrogenase inhibitors, several series of cyclic bisubstrate and transition state analogs were designed, synthesized, and tested as GPAT inhibitors. The weaker in vitro inhibitory activity of these compounds compared to a previously described benzoic acid series was then examined in docking experiments with the soluble squash chloroplast GPAT crystal structure. These in silico experiments indicate that cyclopentyl and cyclohexyl scaffolds prepared in this study may be occluded from the enzyme active site by two protein loops that sterically guard the phosphate binding region. In view of these findings, future GPAT inhibitor design will be driven toward compounds based on planar frameworks able to slide between these loops and enter the active site, resulting in improved inhibitory activity.


Subject(s)
Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Glycerol-3-Phosphate O-Acyltransferase/antagonists & inhibitors , Drug Design , Enzyme Inhibitors/chemical synthesis , Glycerol-3-Phosphate O-Acyltransferase/chemistry , Glycerol-3-Phosphate O-Acyltransferase/metabolism , Humans , Molecular Conformation , Molecular Structure , Obesity/drug therapy , Obesity/enzymology , Structure-Activity Relationship
4.
J Med Chem ; 52(10): 3317-27, 2009 May 28.
Article in English | MEDLINE | ID: mdl-19388675

ABSTRACT

The incidence of obesity and other diseases associated with an increased triacylglycerol mass is growing rapidly, particularly in the United States. Glycerol 3-phosphate acyltransferase (GPAT) catalyzes the rate-limiting step of glycerolipid biosynthesis, the acylation of glycerol 3-phosphate with saturated long-chain acyl-CoAs. In an effort to produce small molecule inhibitors of this enzyme, a series of benzoic and phosphonic acids was designed and synthesized. In vitro testing of this series has led to the identification of several compounds, in particular 2-(nonylsulfonamido)benzoic acid (15g), possessing moderate GPAT inhibitory activity in an intact mitochondrial assay.


Subject(s)
Glycerol-3-Phosphate O-Acyltransferase/antagonists & inhibitors , Obesity/drug therapy , Organophosphonates/chemical synthesis , Sulfonamides/chemical synthesis , ortho-Aminobenzoates/chemical synthesis , Acylation , Animals , Drug Design , Drug Evaluation, Preclinical , Glycerides/biosynthesis , Glycerophosphates/metabolism , Mice , Mitochondria/enzymology , Mitochondria/metabolism , Organophosphonates/pharmacology , Structure-Activity Relationship , Sulfonamides/pharmacology , ortho-Aminobenzoates/pharmacology
5.
Am J Physiol Regul Integr Comp Physiol ; 294(2): R352-61, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18056987

ABSTRACT

Inhibition of brain carnitine palmitoyl-transferase-1 (CPT-1) is reported to decrease food intake and body weight in rats. Yet, the fatty acid synthase (FAS) inhibitor and CPT-1 stimulator C75 produces hypophagia and weight loss when given to rodents intracerebroventricularly (icv). Thus roles and relative contributions of altered brain CPT-1 activity and fatty acid oxidation in these phenomena remain unclarified. We administered compounds that target FAS or CPT-1 to mice by single icv bolus and examined acute and prolonged effects on feeding and body weight. C75 decreased food intake rapidly and potently at all doses (1-56 nmol) and dose dependently inhibited intake on day 1. Dose-dependent weight loss on day 1 persisted through 4 days of postinjection monitoring. The FAS inhibitor cerulenin produced dose-dependent (560 nmol) hypophagia for 1 day, weight loss for 2 days, and weight regain to vehicle control by day 3. The CPT-1 inhibitor etomoxir (32, 320 nmol) did not alter overall day 1 feeding. However, etomoxir attenuated the hypophagia produced by C75, indicating that CPT-1 stimulation is important for C75's effect. A novel compound, C89b, was characterized in vitro as a selective stimulator of CPT-1 that does not affect fatty acid synthesis. C89b (100, 320 nmol) decreased feeding in mice for 3 days and produced persistent weight loss for 6 days without producing conditioned taste aversion. Similarly, intraperitoneal administration decreased feeding and body weight without producing conditioned taste aversion. These results suggest a role for brain CPT-1 in the regulation of energy balance and implicate CPT-1 stimulation as a pharmacological approach to weight loss.


Subject(s)
Body Weight/physiology , Carnitine O-Palmitoyltransferase/metabolism , Eating/physiology , Hypothalamus/enzymology , 4-Butyrolactone/analogs & derivatives , 4-Butyrolactone/pharmacology , Animals , Body Weight/drug effects , Cell Line, Tumor , Dose-Response Relationship, Drug , Eating/drug effects , Energy Metabolism/drug effects , Energy Metabolism/physiology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Epoxy Compounds/pharmacology , Fatty Acid Synthesis Inhibitors/metabolism , Fatty Acids/metabolism , Female , Hypothalamus/cytology , Hypothalamus/drug effects , Injections, Intraventricular , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neurons/cytology , Neurons/drug effects , Neurons/enzymology , Pregnancy , Rats
6.
Clin Cancer Res ; 13(23): 7139-45, 2007 Dec 01.
Article in English | MEDLINE | ID: mdl-18056164

ABSTRACT

PURPOSE: Fatty acid synthase (FAS) is overexpressed in many human cancers and is considered to be a promising target for therapy. However, in vitro use of previous generations of FAS inhibitors has been limited by severe, but reversible, anorexia in treated animals, which is thought to be related to a parallel stimulation of fatty acid oxidation by these agents. This study investigated pharmacologic inhibition of FAS using C93, a rationally designed molecule that inhibits FAS activity without affecting fatty acid oxidation in preclinical models of lung cancer. EXPERIMENTAL DESIGN: Activity of C93 on FAS and fatty acid oxidation was evaluated in cultured non-small cell lung cancer (NSCLC) cells. Antineoplastic activity of the compound, given orally or by i.p. injection, was evaluated in s.c. and orthotopic NSCLC xenografts. RESULTS: Our experiments confirm that C93 effectively inhibits FAS without stimulating fatty acid oxidation in lung cancer cells. More importantly, C93 significantly inhibits the growth of both s.c. and orthotopic xenograft tumors from human NSCLC cell lines without causing anorexia and weight loss in the treated animals. CONCLUSIONS: We conclude that inhibition of FAS can be achieved without parallel stimulation of fatty acid oxidation and that inhibition of tumor growth in vivo can be achieved without anorexia and weight loss. Thus, this therapeutic strategy holds promise for clinical treatment of cancers, including non-small cell lung cancer, the leading cause of cancer mortality in the United States and Europe.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/enzymology , Fatty Acid Synthases/antagonists & inhibitors , Fatty Acid Synthesis Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Administration, Oral , Animals , Anorexia/chemically induced , Antineoplastic Agents/toxicity , Cell Line, Tumor , Fatty Acid Synthases/biosynthesis , Fatty Acid Synthases/metabolism , Fatty Acid Synthesis Inhibitors/toxicity , Fatty Acids/metabolism , Humans , Mice , Mice, Nude , Oxidation-Reduction/drug effects , Weight Loss/drug effects , Xenograft Model Antitumor Assays
7.
Cancer Res ; 67(7): 2964-71, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17409402

ABSTRACT

Fatty acid synthase (FAS), the enzyme responsible for the de novo synthesis of fatty acids, is highly expressed in ovarian cancers and most common human carcinomas. Inhibition of FAS and activation of AMP-activated protein kinase (AMPK) have been shown to be cytotoxic to human cancer cells in vitro and in vivo. In this report, we explore the cytotoxic mechanism of action of FAS inhibition and show that C93, a synthetic FAS inhibitor, increases the AMP/ATP ratio, activating AMPK in SKOV3 human ovarian cancer cells, which leads to cytotoxicity. As a physiologic consequence of AMPK activation, acetyl-CoA carboxylase (ACC), the rate-limiting enzyme of fatty acid synthesis, was phosphorylated and inhibited whereas glucose oxidation was increased. Despite these attempts to conserve energy, the AMP/ATP ratio increased with worsening cellular redox status. Pretreatment of SKOV3 cells with compound C, an AMPK inhibitor, substantially rescued the cells from C93 cytotoxicity, indicating its dependence on AMPK activation. 5-(Tetradecyloxy)-2-furoic acid, an ACC inhibitor, did not activate AMPK despite inhibiting fatty acid synthesis pathway activity and was not significantly cytotoxic to SKOV3 cells. This indicates that substrate accumulation from FAS inhibition triggering AMPK activation, not end-product depletion of fatty acids, is likely responsible for AMPK activation. C93 also exhibited significant antitumor activity and apoptosis against SKOV3 xenografts in athymic mice without significant weight loss or cytotoxicity to proliferating cellular compartments such as bone marrow, gastrointestinal tract, or skin. Thus, pharmacologic FAS inhibition selectively activates AMPK in ovarian cancer cells, inducing cytotoxicity while sparing most normal human tissues from the pleiotropic effects of AMPK activation.


Subject(s)
Enzyme Inhibitors/pharmacology , Fatty Acid Synthases/antagonists & inhibitors , Multienzyme Complexes/metabolism , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/enzymology , Protein Serine-Threonine Kinases/metabolism , AMP-Activated Protein Kinases , Amino Acid Sequence , Animals , Cell Line, Tumor , Enzyme Activation , Fatty Acids/metabolism , Female , Furans/pharmacology , Glucose/metabolism , Humans , Mice , Mice, Inbred BALB C , Molecular Sequence Data , NAD/metabolism , Ovarian Neoplasms/metabolism , Oxidation-Reduction , Xenograft Model Antitumor Assays
8.
J Med Chem ; 48(4): 946-61, 2005 Feb 24.
Article in English | MEDLINE | ID: mdl-15715465

ABSTRACT

Fatty acid synthase (FAS) catalyzes the synthesis of palmitate from the sequential condensation of an acetyl primer with two carbon units added from malonyl-CoA. Inhibition of the beta-ketoacyl synthase domain of mammalian FAS leads to selective cytotoxicity to various cancer cell lines in vitro and in vivo. Also, inhibitors of FAS can cause reduced food intake and body weight in mice. Naturally occurring thiolactomycin (TLM) was used as a template to develop a new class of type I FAS inhibitors. Using a flexible synthesis, families of TLM structural analogues were obtained that possess selective FAS activity and display anticancer and weight loss effects. Compounds 13a and 13d inhibit pure FAS (ZR-75-1 breast cancer, IC(50) = 50 microg/mL), and display effective weight loss in BalbC mice (>5%). Another subclass of TLM derivatives (23b-d, 31a) exhibits FAS activity (IC(50) = 5%), and is cytotoxic to cancer cells (IC(50) < 38 microg/mL). Finally, a third subclass (16b, 29, 30) is also active against FAS (IC(50) =

Subject(s)
Anti-Obesity Agents/chemical synthesis , Antineoplastic Agents/chemical synthesis , Fatty Acid Synthases/antagonists & inhibitors , Thiophenes/chemical synthesis , Animals , Anti-Obesity Agents/chemistry , Anti-Obesity Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Body Weight/drug effects , Cell Line, Tumor , Drug Screening Assays, Antitumor , Fatty Acid Synthases/chemistry , Humans , Mice , Mice, Inbred BALB C , Models, Molecular , Stereoisomerism , Structure-Activity Relationship , Thiophenes/chemistry , Thiophenes/pharmacology
9.
Cancer Res ; 63(21): 7330-7, 2003 Nov 01.
Article in English | MEDLINE | ID: mdl-14612531

ABSTRACT

C75, an inhibitor of fatty acid synthase (FAS), induces apoptosis in cultured human cancer cells. Its proposed mechanism of action linked high levels of malonyl-CoA after FAS inhibition to potential downstream effects including inhibition of carnitine palmitoyltransferase-1 (CPT-1) with resultant inhibition of fatty acid oxidation. Recent data has shown that C75 directly stimulates CPT-1 increasing fatty acid oxidation in MCF-7 human breast cancer cells despite inhibitory concentrations of malonyl-CoA. In light of these findings, we have studied fatty acid metabolism in MCF7 human breast cancer cells to elucidate the mechanism of action of C75. We now report that: (a) in the setting of increased fatty acid oxidation, C75 inhibits fatty acid synthesis; (b) C273, a reduced form of C75, is unable to inhibit fatty acid synthesis and is nontoxic to MCF7 cells; (c) C75 and 5-(tetradecyloxy)-2-furoic acid (TOFA), an inhibitor of acetyl-CoA carboxylase, both cause a significant reduction of fatty acid incorporation into phosphatidylcholine, the major membrane phospholipid, within 2 h; (d) pulse chase studies with [(14)C]acetate labeling of membrane lipids show that both C75 and TOFA accelerate the decay of (14)C-labeled lipid from membranes within 2 h; (e) C75 also promotes a 2-3-fold increase in oxidation of membrane lipids within 2 h; and (f) because interference with phospholipid synthesis during S phase is known to trigger apoptosis in cycling cells, we performed double-labeled terminal deoxynucleotidyltransferase-mediated nick end labeling and BrdUrd analysis with both TOFA and C75. C75 triggered apoptosis during S phase, whereas TOFA did not. Moreover, application of TOFA 2 h before C75 blocked the C75 induced apoptosis, whereas etomoxir did not. Taken together these data indicate that FAS inhibition and its downstream inhibition of phospholipid production is a necessary part of the mechanism of action of C75. CPT-1 stimulation does not likely play a role in the cytotoxic response. The continued ability of TOFA to rescue cancer cells from C75 cytotoxicity implies a proapoptotic role for malonyl-CoA independent of CPT-1 that selectively targets cancer cells as they progress into S phase.


Subject(s)
4-Butyrolactone/analogs & derivatives , 4-Butyrolactone/pharmacology , Apoptosis/drug effects , Fatty Acid Synthases/antagonists & inhibitors , Furans/pharmacology , Apoptosis/physiology , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , Breast Neoplasms/pathology , Carnitine O-Palmitoyltransferase/metabolism , Cell Division/drug effects , Cell Line, Tumor , Drug Interactions , Epoxy Compounds/pharmacology , Humans , Membrane Lipids/biosynthesis , Membrane Lipids/metabolism , Phospholipids/biosynthesis , S Phase/drug effects , S Phase/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...