当前位置: 首页 > 期刊 > 《内分泌学杂志》 > 2006年第2期 > 正文
编号:11416419
Biological Characterization of a Heterodimer-Selective Retinoid X Receptor Modulator: Potential Benefits for the Treatment of Type
http://www.100md.com 《内分泌学杂志》
     Ligand Pharmaceuticals (M.D.L., R.J.A., M.F.B., D.L.C., J.D., N.I.H., R.A.H., D.J., K.K., S.L., D.E.M., C.M.M., K.B.M., P.-Y.M., K.M.O., B.P., D.R., J.S.T., M.S.U., M.W.), San Diego, California 92121

    Lilly Research Laboratories (C.L.B., M.A.C., G.J.E., M.M.F., T.A.G., H.H., C.M.-R., N.Y., A.R.-M.), Indianapolis, Indiana 46285

    Abstract

    Specific retinoid X receptor (RXR) agonists, such as LG100268 (LG268), and the thiazolidinedione (TZD) PPAR agonists, such as rosiglitazone, produce insulin sensitization in rodent models of insulin resistance and type 2 diabetes. In sharp contrast to the TZDs that produce significant increases in body weight gain, RXR agonists reduce body weight gain and food consumption. Unfortunately, RXR agonists also suppress the thyroid hormone axis and generally produce hypertriglyceridemia. Heterodimer-selective RXR modulators have been identified that, in rodents, retain the metabolic benefits of RXR agonists with reduced side effects. These modulators bind specifically to RXR with high affinity and are RXR homodimer partial agonists. Although RXR agonists activate many heterodimer partners, these modulators selectively activate RXR:PPAR and RXR:PPAR, but not RXR:RAR, RXR:LXR, RXR:LXR, or RXR:FXR. We report the in vivo characterization of one RXR modulator, LG101506 (LG1506). In Zucker fatty (fa/fa) rats, LG1506 is a potent insulin sensitizer that also enhances the insulin-sensitizing activities of rosiglitazone. Administration of LG1506 reduces both body weight gain and food consumption and blocks the TZD-induced weight gain when coadministered with rosiglitazone. LG1506 does not significantly suppress the thyroid hormone axis in rats, nor does it elevate triglycerides in Sprague Dawley rats. However, LG1506 produces a unique pattern of triglycerides elevation in Zucker rats. LG1506 elevates high-density lipoprotein cholesterol in humanized apolipoprotein A-1-transgenic mice. Therefore, selective RXR modulators are a promising approach for developing improved therapies for type 2 diabetes, although additional studies are needed to understand the strain-specific effects on triglycerides.

    Introduction

    THE RETINOID X receptor (RXR) plays a unique and central role in the activity of many members of the nuclear hormone receptor superfamily. It functions as an obligate heterodimer partner for retinoic acid receptors (RARs), thyroid receptor, vitamin D receptor, peroxisome proliferator-activated receptors (PPARs), liver X receptors (LXRs), farnesoid X receptor (FXR), pregnane X receptor, constitutively active receptor, nerve growth factor 1B-like receptor, and nuclear receptor related receptor (reviewed in Ref.1). This unique position within the superfamily allows the RXR to impact many regulatory and metabolic systems. Such a broad spectrum of actions is particularly well suited to modulatory and integrative functions. We discuss the biological activity of novel synthetic RXR ligands (rexinoids) that exhibit heterodimer selectivity. Because these rexinoids activate only a specific heterodimer subset in vitro, we refer to them as heterodimer-selective RXR modulators.

    Synthetic ligands for both RXR and PPAR produce insulin sensitization in animal models of type 2 diabetes (2, 3, 4). Although the well-characterized PPAR agonist rosiglitazone is specific for the RXR:PPAR heterodimer (5), the prototypic RXR-selective agonist LG100268 (LG268) (6) activates many RXR heterodimers and RXR homodimers (2, 7). Thus, rexinoids, when compared with a PPAR agonist, can signal through multiple RXR-dependent pathways (involving different partner receptors) with the potential to elicit not only broad therapeutic benefits, but also unwanted side effects. Tissue-selective modulators of other nuclear receptors have been identified and exploited clinically. The classic successful examples of this approach are the selective estrogen receptor modulators, such as raloxifene and tamoxifen.

    The antidiabetic profile of LG268 in rodent models of insulin resistance is superior to that of thiazolidinedione (TZD) PPAR agonists that increase both fat mass and body weight. Rexinoid agonists reduce plasma glucose and insulin, body weight gain, and food consumption while preserving lean body mass (2, 8, 9). Unfortunately, they also suppress the thyroid hormone axis (10) and can produce rapid and dramatic increases in triglycerides (11). Thus, synthetic ligands for RXR that lack these side-effects have the potential to significantly improve upon the currently used thiazolidinedione insulin sensitizers for the treatment of type 2 diabetes. We chose to explore the identification of heterodimer-selective rexinoids to dissect the desirable effects from the undesirable side-effects.

    In this communication, we report the biological characterization of the heterodimer-selective RXR modulator LG101506 [LG1506; (2E,4E,6Z)-7-(2-(2,2-difluoroethoxy)-3,5-di-tert-butylbenzene)-3-methylocta-2,4,6-trienoic acid] (12). This compound binds to the RXR with high affinity and induces a receptor conformation that results in selective activation of RXR:PPAR, RXR:PPAR, and RXR:PPAR, but not RXR:RAR, RXR:LXR, or RXR:FXR heterodimers. In rodent models of insulin resistance, LG1506 administration produces antidiabetic activities similar to those seen after rosiglitazone administration. When administered in combination with rosiglitazone, LG1506 enhances the insulin-sensitizing activity of the TZD and blocks TZD-induced body weight gain. In addition, administration of LG1506 to humanized apolipoprotein A1-transgenic (hApoA1tg) mice leads to an elevation in high-density lipoprotein cholesterol (HDL-C) and enhances the activity of fenofibrate. LG1506 retains all the antidiabetic benefits of LG268 without suppressing the thyroid hormone axis. Interestingly, a unique pattern of triglycerides elevation is produced after the administration of LG1506 to Zucker rats.

    Materials and Methods

    Binding assays

    Heterodimer binding assays were performed using scintillation proximity assay technology with appropriate receptors and corresponding radiolabeled ligands (13). Briefly, receptors (RXR, -, or - and RAR, -, or -) were produced using a baculovirus expression system. Biotinylated oligonucleotides containing RXR or RAR response elements were used to couple the corresponding receptor dimers to yttrium silicate streptavidin-coated scintillation proximity assay beads. Receptor binding assays for RARs and RXRs were performed in a similar manner, as described by Boehm et al. (14) using [3H]-9-cis-retinoic acid as the radioligand for RXRs, and [3H]all-trans-retinoic acid (NEN Life Science Products-DuPont, Boston, MA) for the RARs. Ki values were determined by application of the Cheng-Prusoff equation (15). Test compounds were evaluated using an 11-point dose-response curve with concentrations ranging from 0.169 nM to 10 μM.

    Cotransfection (CTF) assays

    PPAR, PPAR, PPAR, LXR, LXR, FXR, RAR, or RXR were constitutively expressed using plasmids containing the CMV promoter. The luciferase reporter constructs contained three to five copies of the cellular retinol-binding protein II response element for RXR (16), a thyroid receptor response element for RAR (17, 18), an LXR response element for LXR (19), an ecdysone receptor response element for FXR (20), an acyl coenzyme A oxidase-PPAR response element for PPAR and PPAR (21), or a cytochrome P450 4A1 PPAR response element for PPAR (22). Synergy assays for PPAR, PPAR, and RAR were performed as the standard CTF assays with the addition of EC20 concentrations of the appropriate specific ligand {rosiglitazone, GW501516, and (E)-4-[2-(5,6,7,8-tetrahydro-5,5,8,8-tetramethyl-2-naphthylenyl)-1-propenyl] benzoic acid (TTNPB), respectively}. All assays were performed in CV-1 cells, and compounds were tested in full log dilution, from 0.1 nM to 10 μM in duplicate. Efficacy was determined relative to reference molecules. The median effective concentration (EC50) values were determined by computer fit to a concentration-response curve. An EC50 value was not calculated if the maximum observed efficacy for the compound was less than 20%.

    In vivo studies

    All in vivo procedures were approved by the Ligand or Eli Lilly Institutional animal care and use committees, and principles of laboratory animal care (National Institutes of Health publication 85-23, revised 1985) were followed. Animals were housed in temperature-controlled rooms (70–74 F) with lights on from 0600–1800 h, with free access to water and food, except where noted below.

    Zucker fatty (fa/fa) rat studies

    Female Zucker obese (fa/fa) or lean rats were obtained from Harlan (Indianapolis, IN) at 6 wk of age. After a 2-wk acclimation period, rats for efficacy studies were prebled and assigned to experimental groups to minimize the variance between groups based on the measured plasma glucose levels and body weight (six animals per group). Compounds were administered by oral gavage once daily between 0730 and 0830 h for 14 d: LG268 (30 mg/kg), rosiglitazone (10 mg/kg), LG1506 (3, 10, or 30 mg/kg), or LG1506 (3 or 30 mg/kg) plus rosiglitazone (1 mg/kg). The dosing vehicle was 0.085% povidone (ISP Technologies, Inc., New Milford, CT), 1.5% lactose (Quest International, New York, NY), 0.026% Tween 80 (Sigma-Aldrich Corp., St. Louis, MO), and 0.2% (vol/vol) Antifoam (Dow Corning, Midland, MI) with control animals receiving dosing vehicle only. Insulin, TSH, and triglycerides levels were determined from blood samples collected from the tail vein of unfasted conscious animals 3 h after treatment on d 2, 7, and 14. Body weight gain was determined by subtracting the starting body weight of each animal on d –1 from its weight on d 14. The rats were fasted overnight after the last dose and were subjected to an oral glucose tolerance test (OGTT) the following morning. Glucose and insulin levels were measured at 0 min (immediately before the glucose challenge) and 15, 30, 60, and 120 min after challenge (2 g glucose/kg body weight). The insulin resistance index was calculated by multiplying the insulin area under the curve (AUC) by the glucose AUC. For the study described in Fig. 5, lean or obese Zucker rats were sorted based upon body weight and pretreatment triglycerides (six animals per group). Animals were treated for 7 d as described above with vehicle or LG1506 (1, 3, or 30 mg/kg).

    Homozygous hApoA-1tg mouse studies

    The hApoA1tg mice (23), 6.5 wk old, were purchased from The Jackson Laboratory (Bar Harbor, ME). After a 2-wk acclimation period, the mice were assigned (based on body weight) to treatment groups (five animals per group). Compounds were administered daily by oral gavage between 0700 and 0800 h for 7 d: fenofibrate (100 mg/kg), LG1506 (3, 10, 30, and 100 mg/kg), and LG1506 plus fenofibrate (3, 10, 30, and 100 mg/kg LG1506 plus 100 mg/kg fenofibrate). The dosing vehicle was 0.085% povidone (ISP Technologies, Inc.), 1.5% lactose (Quest International), 0.026% Tween 80 (Sigma-Aldrich Corp.), and 0.2% (vol/vol) Antifoam (Dow Corning) with control animals receiving dosing vehicle only. Three hours after the final dose, mice were killed under CO2, and blood was collected by cardiac puncture and placed into serum separator tubes.

    Lipoproteins were separated by fast protein liquid chromatography, and cholesterol was quantitated with an in-line detection system based on that described by Kieft et al. (24). Briefly, 35-μl serum samples (from 50-μl pooled samples) were applied to a Superose 6 HR 10/30 size exclusion column (Pharmacia Biotech, Piscataway, NJ) and eluted with PBS (pH 7.4; diluted 1:10), containing 5 mM EDTA, at 0.5 ml/min. Cholesterol reagent from Roche Diagnostics (Indianapolis, IN) at 0.16 ml/min was mixed with the column effluent through a T-connection; the mixture was then passed through a 15-m x 0.5-mm knitted tubing reactor (Aura Industries, New York, NY) immersed in a 37 C water bath. The colored product produced in the presence of cholesterol was monitored in the flow stream at 505 nm, and the analog voltage from the monitor was converted to a digital signal for collection and analysis. The change in voltage corresponding to the change in cholesterol concentration was plotted vs. time, and the AUC corresponding to the elution of HDL-C was calculated using Turbochrome software (version 4.12F12, PerkinElmer, Norwalk, CT).

    Sprague Dawley rat studies

    Sprague Dawley rats were purchased from Harlan (San Diego, CA) at 6 wk of age. After a 2-wk acclimation period, the rats were assigned (based on weight) to individual groups (seven animals per group). An acute study was performed in which TSH and triglycerides levels were measured 2 h after a single dose of vehicle, rosiglitazone (30 mg/kg), LG268 (3, 10, 30 mg), or LG1506 (30 mg/kg). An additional study focused on triglycerides was performed in which rats were administered vehicle, LG268 (30 mg/kg), or LG1506 (1, 3, 10, or 30 mg/kg) for 7 d, with triglycerides levels measured 2 h after treatment on d 1, 3, 5, and 7. An acute LG1506 dose-response study focused on TSH was conducted using eight animals per group. TSH was measured 2 h after a single dose of vehicle, LG268 (30 mg/kg), or LG1506 (1, 3, 10, or 30 mg/kg). The dosing vehicle was 1% (wt/vol) carboxymethylcellulose and 0.25% Tween-80.

    Statistical analysis

    All results are expressed as the mean ± SEM. Data were analyzed by either one- or two-way ANOVA, followed by Dunnett’s or Tukey’s test. In the two-way analysis, the time factor was considered a repeated measure. Data that did not meet the assumptions for parametric statistics (normal distribution and equality of variance) were transformed using Box-Cox transformations (25) before analysis. Any specific transformations used are described in the figure legends. All statistical analysis was performed using JMP 5.1 (SAS Institute, Inc., Cary, NC), with the level set at 0.05.

    Results

    LG1506 is a heterodimer-selective RXR modulator

    Binding and CTF assays were conducted to define the in vitro properties of selective RXR modulators. The RXR agonist LG268 (2, 6, 8) was included in these assays as a comparator compound. Both the RXR modulator LG1506 and LG268 are high-affinity ligands for RXR, -, or -, with minimal affinity for RAR, -, or - (Table 1), thus defining both compounds as selective RXR ligands (rexinoids). We have not identified compounds that are selective for RXR, -, or -. As this is the case, we have used the RXR isotype for all further molecular profiling. In standard CTF assays, LG268 functioned as a PPAR, PPAR, LXR, and FXR agonist, whereas LG1506 was active only in the PPAR assay (Table 2). In the LXR assay, LG1506 was a weak antagonist (data not shown). To more clearly define the transcriptional potential of LG1506, CTF synergy assays were developed. In the synergy assays, the EC20 concentration of a known ligand for the RXR heterodimeric partner was added to the standard CTF assay. Using this approach, LG268 was active in the PPAR, PPAR, and RAR synergy assays. In contrast, the modulator LG1506 did not activate the RXR:RAR heterodimer in the synergy assay and thus demonstrated heterodimer selectivity (Table 2). These data differentiated LG1506 from rexinoid agonists (exemplified here by LG268) and prompted us to investigate whether LG1506 retained the superior antidiabetic properties of an RXR agonist, but had reduced negative side effects.

    Studies in obese Zucker fatty (fa/fa) rats were conducted to investigate the insulin-sensitizing properties of LG1506. Based upon the in vitro synergy data, a fa/fa rat study was conducted including both LG1506 monotherapy and LG1506 plus TZD combination therapy arms. Rats in the monotherapy arm received three dose levels of LG1506 (3, 10, or 30 mg/kg), a maximally efficacious dose of LG268 (30 mg/kg), or the PPAR agonist rosiglitazone (10 mg/kg). The combination arm included LG1506 plus a submaximal dose of rosiglitazone (3 or 30 mg/kg LG1506 plus 1 mg/kg rosiglitazone). The study also included fa/fa and lean control groups administered the dosing vehicle only. After 14 d of compound administration, the rats were fasted overnight and subjected to an OGTT. As shown in Fig. 1A, results from the monotherapy arm of the study demonstrated that LG1506, LG268, and rosiglitazone all reduced the hyperinsulinemia associated with this rodent model of insulin resistance. In contrast, insulin levels remained elevated in the control fa/fa rats, whereas the lean control rats maintained normal insulin levels throughout the study period. LG1506 treatment resulted in a dose-dependent reduction in both glucose and insulin excursions after the oral glucose challenge, whereas both LG268 and rosiglitazone reduced the excursions to levels observed in the lean control rats (Fig. 1, B and C).

    When the insulin resistance index (insulin AUC x glucose AUC) was calculated, LG1506 produced a statistically significant and dose-dependent decrease, approaching that achieved with maximally efficacious doses of either rosiglitazone or LG268 (Fig. 1D). In the combination arm of the study, both the 3 and 30 mg/kg doses of LG1506 enhanced the activity of a submaximal dose of rosiglitazone (Fig. 1E). In fact, the 1 mg/kg dose of rosiglitazone plus the 30 mg/kg dose of LG1506 were as efficacious as the maximally efficacious dose of rosiglitazone (10 mg/kg), both of which normalized the insulin resistance index to that of the lean control animals.

    Administration of a high dose of rosiglitazone (10 mg/kg) for 14 d to Zucker fa/fa rats increased body weight gain significantly (Table 3). In contrast, a fully efficacious dose of LG268 significantly reduced weight gain, as we previously reported (8). Although combined administration of rosiglitazone and LG268 did not further enhance insulin-lowering efficacy, body weight gain was restrained to the same extent observed in animals dosed with LG268 alone (Table 3).

    In the experiment shown in Fig. 1, the 1 mg/kg dose of rosiglitazone was a submaximally efficacious dose, although it produced an increase in weight gain (Fig. 2). In comparison, neither the 3 nor the 30 mg/kg dose of LG1506, alone or in combination with rosiglitazone, increased weight gain compared with the obese control animals. In fact, the 30 mg/kg dose of LG1506 in combination with rosiglitazone produced significantly less weight gain than that in the fa/fa controls. The combination study revealed that administration of 3 mg/kg LG1506 plus a submaximal dose of rosiglitazone led to significant insulin sensitization, with no drug-induced weight gain, and a higher dose of LG1506 in combination with rosiglitazone normalized the insulin resistance index while reducing weight gain. All animals in this study remained healthy and had weight gain at least comparable with that of the control lean animals.

    Based upon the PPAR CTF activity of LG1506, a study was conducted in hApoA1tg mice to investigate the lipid-altering properties of the compound. Administration of LG1506 to hApoA1tg mice increased HDL-C to the level seen after treatment with 100 mg/kg fenofibrate. Combined treatment with LG1506 and fenofibrate produced greater increases in HDL-C at all doses compared with the monotherapy arm of the study (Table 4). These results demonstrate that the RXR modulator LG1506 can have significant impact on the RXR:PPAR heterodimer in vivo, and in vivo activation of both sides of the heterodimer produces increased activity.

    RXR agonists such as LG268 produce side effects in vivo, including alterations in hormones of the thyroid hormone axis (10, 26). Previous work demonstrated that in rats, TSH levels fall before T3 or T4 levels decrease (10). Therefore, we structured our experiments around the use of TSH as an indicator of the rexinoid-induced suppression of the thyroid hormone axis. In Sprague Dawley rats, a dose-dependent decrease in TSH levels was observed 2 h after a single administration of LG268 (3, 10, or 30 mg/kg), whereas neither rosiglitazone (30 mg/kg) nor LG1506 (30 mg/kg) significantly altered TSH levels (Fig. 3A). To evaluate this effect, we conducted an LG1506 dose-response study using 1, 3, 10, or 30 mg/kg LG1506 and 30 mg/kg LG268 as the positive control (Fig. 3B). Although the absolute values are different in these two studies, LG268 reduced TSH by the same percentage in both experiments (49% and 50%, respectively). Animals treated with all doses of LG1506 in excess of 1 mg/kg showed a trend for reduced TSH levels, although none of the decreases was statistically significant.

    Another undesirable metabolic consequence seen after the administration of RXR agonists is an elevation in triglycerides levels (11, 27). We conducted both acute (single administration) and 7-d chronic studies in Sprague Dawley rats to investigate triglycerides levels after the administration of RXR ligands. Acute administration of LG268 (3, 10, or 30 mg/kg) produced a dose-dependent increase in triglycerides 2 h after administration, whereas neither LG1506 nor rosiglitazone administration (both at 30 mg/kg) altered triglycerides levels (Fig. 3C). To explore these results in more detail, Sprague Dawley rats were treated for 7 d with LG1506 (1, 3, 10, or 30 mg/kg) or a maximally efficacious dose of LG268 (30 mg/kg). Triglycerides levels were determined from plasma collected 2 h after treatment on d 1, 3, 5, and 7. LG1506 did not alter triglycerides levels at any dose on any day of this study; however, LG268 administration led to a rapid and sustained increase in triglycerides (Fig. 3D).

    During the fa/fa rat efficacy study described in Fig. 1 we determined both TSH and triglycerides levels. As we had seen in Sprague Dawley rats, administration of LG268 (30 mg/kg) to fa/fa rats for 14 d decreased TSH levels for the duration of the study (Fig. 4A). Although the mean TSH levels in animals treated with LG1506 (3, 10, or 30 mg/kg) are lower than those in the vehicle-treated animals, none of the reductions was statistically significant (Fig. 4A). The selective RXR modulator LG1506 does not significantly alter TSH levels in two different rat strains, suggesting that such a selective modulator might not cause this side effect in man. As expected based upon the previous 7-d Sprague Dawley rat study, LG268 administration led to a rapid and sustained increase in triglycerides (Fig. 4B). Interestingly, although fa/fa rats receiving LG1506 had control levels of triglycerides after three doses, triglycerides were higher after seven doses. Furthermore, this increase in triglycerides had an inverted dose response; the greatest increase in triglycerides was at the lowest dose of the modulator. Similar results were found in other studies and when other selective RXR modulators were studied (data not shown).

    To examine the unexpected triglycerides elevation in the fa/fa rats, a study was conducted in age-matched lean and obese Zucker animals. Rats received daily oral doses of LG1506 (1, 3, or 30 mg/kg) for 7 d. Both lean and obese animals receiving 30 mg/kg LG1506 had triglycerides levels indistinguishable from those in the control animals at all times (Fig. 5). On d 3, triglycerides were elevated (but not statistically different from the control level) in lean rats after the administration of both the 1 and 3 mg/kg doses of LG1506. On d 7, lean animals receiving 3 mg/kg LG1506 had triglycerides levels statistically higher than those in control animals. In the obese fa/fa rats, both 1 and 3 mg/kg LG1506 doses produced significant increases in triglycerides on d 3, although only the 3 mg/kg dose was significant on d 7. These data indicate that at low doses, LG1506 retains the unwanted side effect of triglycerides elevation in the Zucker strain of rat (regardless of obesity phenotype).

    Data from these studies clearly indicate that selective RXR modulators are devoid of the hypertriglyceridemia associated with the administration of LG268 to Sprague Dawley rats. In addition, the kinetic pattern of the elevation in triglycerides seen after administration of the RXR modulator LG1506 to fa/fa rats is distinct from that seen after administration of the RXR agonist LG268, suggesting that different and/or multiple mechanisms underlie the effects.

    Discussion

    RXR occupies a unique domain within the nuclear receptor superfamily, because it is required as an obligate heterodimer partner for many other receptors. This multiplicity of actions poses both the "promise" and the "curse" associated with the potential therapeutic exploitation of RXR ligands. The challenge is to identify rexinoids that, after binding to the receptor, induce conformations of the receptor that produce the desired outcome without undesired activities. This approach has been successful for the selective estrogen receptor modulators, raloxifene and tamoxifen. Heterodimer-selective RXR modulators represent a promising approach for the future treatment of type 2 diabetes and its associated dyslipidemias. We describe in this report the biological characterization of a representative heterodimer-selective RXR modulator (LG1506) and compare its activity with both that of the well-characterized RXR agonist LG268 and that of the thiazolidinedione PPAR agonist rosiglitazone.

    We reasoned that an appropriate heterodimer-selective RXR modulator would need to retain high-affinity binding to RXR to maintain receptor specificity. The molecule would have to retain the desirable antidiabetic/metabolic efficacy and reduce/eliminate the undesirable side effects, such as suppression of the thyroid hormone axis and elevation of triglycerides. We believed that an RXR partial agonist could sensitize the in vivo homeostatic network to the activity of endogenous ligands for partner receptors such as the PPARs (28). This hypothesis led us to use in vitro synergy assays to explore in a sensitive manner the activities of our compounds on various heterodimers. We used a PPAR synergy assay to screen for desirable antidiabetic activities and an RAR synergy assay as a marker to screen against undesirable side effects. The synergy assays are more sensitive at detecting agonist activity and/or reflect mechanistically relevant receptor conformations that are distinct from those identified in the standard CTF assays. As expected based upon this increased sensitivity, numerous compounds are active in the synergy assays, but negative in the corresponding standard CTF assays. The most promising selective RXR modulators would be those that activate only RXR:PPAR and RXR:PPAR, thus mimicking the effects of the thiazolidinediones and fibrates, respectively. The selective RXR modulator LG1506 described in this report does not activate RXR:RAR, RXR:LXR, or RXR:FXR heterodimers. The ability of modulators to be selectively devoid of agonist activity on RXR:RAR and RXR:LXR is important, because both RAR and LXR agonists have been shown to increase triglycerides (29, 30, 31, 32, 33). It is important to note, however, that the kinetics of the increase in triglycerides induced after the administration of an RXR agonist are not the same as those seen after the administration of an RAR agonist, suggesting that the RXR:RAR heterodimer cannot fully account for all the observed increases.

    All our CTF assays were conducted using RXR. It is possible that RXR or RXR would produce different results. However, we think that this is unlikely, because we have been unable to identify rexinoids that are selective for any of the RXR subtypes, and the binding of both LG268 and LG1506 is essentially the same to all three RXRs. Because the RXR isotypes are not identical in either sequence or tissue distribution, it is still possible that they could interact in heterodimer-specific ways, although we are not aware of any demonstration of such specificity.

    The in vivo activity of LG1506 is compared with that of the RXR agonist LG268 that activates all four heterodimers. LG1506 demonstrates an efficacious antidiabetic profile when administered alone and has the ability to enhance the activity of the PPAR agonist rosiglitazone when administered in combination. Although LG1506 was positive in the PPAR CTF synergy assay, it was inactive in the standard CTF assay. It has been shown that full PPAR agonist activity, as measured in CTF and cofactor recruitment assays, is not needed to obtain significant insulin-sensitizing benefits in vivo (28, 34). Therefore, it is not surprising that administration of LG1506 resulted in significant insulin sensitization in the absence of in vitro PPAR agonist activity in the standard CTF assay. Alternatively, one could argue that the insulin-sensitizing benefit is due to the PPAR agonist activity of LG1506, because a number of reports have shown that PPAR agonists can function as insulin-sensitizing agents (35, 36, 37). With these alternatives offered, the most likely explanation is that both PPAR and PPAR agonist activities of LG1506 contribute to the insulin-sensitizing benefits of the selective RXR modulator.

    The third member of the PPAR family, PPAR, could also play a role in the observed activity of LG1506 and/or LG268. We did not explore this possibility in detail. Although the initial demonstration that PPAR activation influenced cholesterol metabolism was made in db/db mice (38), the more dramatic metabolic effects that have been demonstrated recently have been in nonhuman primates (39) or have used molecular manipulations to dramatically increase receptor activation (40). Nevertheless, it is possible that some fraction of the observed metabolic activity of the rexinoids could be mediated through PPAR. In particular, it is possible that some of the advantageous activity with respect to body weight gain could result from activation of PPAR.

    One of the potential advantages of the rexinoid approach is that multiple RXR partners can be activated simultaneously. This benefit is clearly demonstrated in the combination study with rosiglitazone and LG1506 in Zucker fa/fa rats. In the respective monotherapy studies, rosiglitazone normalized the insulin resistance index when administered at 10 mg/kg, and LG1506 caused a dose-dependent decrease at the doses administered (3, 10, and 30 mg/kg). However, combined administration of a submaximal dose of rosiglitazone (1 mg/kg) plus LG1506 (30 mg/kg) normalized the insulin resistance index. Even more interesting and exciting is the finding that LG1506 blocked the drug-induced weight gain associated with rosiglitazone treatment. A significant increase in body weight gain was seen after the administration of a submaximal dose of rosiglitazone (1 mg/kg), whereas this dose of rosiglitazone plus LG1506 (30 mg/kg) resulted in a significant decrease in body weight gain. Importantly, all animals in this study remained healthy and achieved at least the weight gain of the lean control animals. The lack of drug-induced weight gain after mono- and combination therapy with LG1506 can result from multiple pathways. Ogilvie et al. (8) demonstrated, using intracerebroventricular injections, that RXR agonists act directly in the central nervous system to regulate food intake and satiety. When administered intracerebroventricularly, LG1506 also has these effects (data not shown). In addition, numerous reports have shown that PPAR activity is associated with increased fatty acid -oxidation, and a lack of weight gain has been observed when a PPAR agonist is administered to insulin-resistant rodents and nonhuman primates (35, 36, 37).

    Our data demonstrate that the dose of a PPAR agonist required to achieve maximal efficacy can be reduced when used in combination with a selective RXR modulator. Thus, combination therapy may reduce the incidence of additional side effects associated with PPAR agonist therapy, such as edema and the potential for congestive heart failure.

    The selective RXR modulators identified and characterized to date demonstrate PPAR agonist activity in the standard CTF assay. These compounds demonstrate a lipid-altering effect in vivo, as shown in the hApoA1 transgenic mouse. Although lipid metabolism in rodents is not identical with that in humans (e.g. rodents do not express cholesterol ester transfer protein), rodent models have been and continue to be used extensively for both basic research and drug discovery. For example, the fibrate class of hypolipidemic drugs raise HDL-C and lower triglycerides levels in the hApoA1-transgenic mouse and in humans. When used alone, LG1506 produced a dose-dependent increase in HDL-C that was equivalent to the elevation produced by 100 mg/kg fenofibrate. Coadministration of LG1506 with 100 mg/kg fenofibrate produced greater increases in HDL-C at all doses of LG1506 tested.

    Administration of the heterodimer-selective RXR modulator LG1506 did not significantly suppress the thyroid hormone axis, as seen after the administration of RXR agonists (10, 26). Because the LG268-mediated reduction in thyroid hormones has been shown to result from a decrease in TSH secretion from the anterior pituitary, which then leads to a reduction in T3 and T4 levels (10), we carefully monitored TSH levels in the Zucker fa/fa rat efficacy study and in the acute studies in Sprague Dawley rats. No significant changes in TSH levels were observed in any of the in vivo studies with LG1506. The mechanism by which rexinoids alter TSH levels is not understood. Although neither LG268 nor LG1506 binds to the RAR, LG268 clearly has agonist activity in the RXR:RAR CTF synergy assay, whereas LG1506 has absolutely no agonist activity in that assay. These data clearly demonstrate the importance of the sensitive RXR:RAR CTF synergy assay to differentiate between rexinoid agonists (such as LG268) that suppress the thyroid axis and heterodimer-selective modulators (such as LG1506) that do not.

    Administration of RXR agonists, including LG268, to naive animals is known to cause a rapid elevation in triglycerides levels (11, 27). Therefore, triglycerides levels were carefully monitored after the administration of selective RXR modulators to Sprague Dawley rats under conditions where the RXR agonist LG268 raised triglycerides levels. Both acute and chronic studies were conducted, and no alteration in levels of triglycerides were seen after the administration of LG1506 to Sprague Dawley rats. These data definitively demonstrate in normal rats that the activities of LG1506, with respect to triglycerides, are distinctly differentiated from those of the RXR agonists.

    RXR agonists, such as LG268, produce the same pattern of triglycerides elevation in both Sprague Dawley and Zucker fa/fa rats. It was therefore totally unexpected that LG1506 would produce a different pattern in Zucker compared with Sprague Dawley rats. The fact that triglycerides levels in Zucker rats are increased more at lower doses of LG1506 suggests that the compound activates multiple pathways that impact triglycerides. Furthermore, the compound appears to activate a pathway(s) that increases levels of triglycerides at low doses and to activate another pathway(s) that opposes the increase at higher doses.

    These observations are not specific to LG1506, because analogous delayed increases in triglycerides have been observed after the administration of other heterodimer-selective RXR modulators to Zucker rats (data not shown). Additional studies will be required to understand the mechanism of this unexpected difference between strains. The insulin resistance phenotype of the fa/fa rat may contribute to the unusual lipid findings. We presume that the increase in triglycerides observed in Zucker rats after treatment with LG1506 is not mediated through activation of LXR. Although agonist activity at either partner of the RXR:LXR heterodimer is associated with hypertriglyceridemia in rodents (11, 33), LG1506 is a weak LXR antagonist in CTF assays.

    In conclusion, selective RXR modulators offer an interesting and potentially beneficial approach to the identification and development of novel therapies for type 2 diabetes. Although we do not fully understand the precise molecular mechanisms involved, heterodimer-selective RXR modulators demonstrate a number of advantages when used alone and in combination with PPAR and/or PPAR agonists. At the same time, one must be cautious, because all our work relied upon rodent models and, as such, may not precisely reflect the activity of the compounds in man. Although we have identified RXR modulators that do not significantly suppress the thyroid hormone axis, the unexpected pattern of triglycerides elevation that has been unmasked remains an issue that requires additional study to obtain a solution.

    Acknowledgments

    We are grateful to Dr. Peter Davies for his insightful comments and suggestions as this work progressed, and to Drs. David Seyler, Donald Karanewsky, and Sharon Dana for their involvement in and contributions to the modulator project. These studies relied upon excellent technical support from the vivarium staff at both Ligand Pharmaceuticals and Lilly Research Laboratories.

    Footnotes

    Current address of M.F.B.: Conforma Therapeutics, San Diego, California 92121.

    Current address of M.M.F.: Amgen, Thousand Oaks, California 91320.

    Current address of R.A.H.: Exelixis, San Diego, California 92121.

    Current address of D.J.: Neurocrine Biosciences, San Diego, California 92130.

    Current address of C.M.M.: Johnson & Johnson Pharmaceutical Research and Development, San Diego, California 92121.

    Current address of P.-Y.M.: Genomics Institute of the Novartis Research Foundation, San Diego, California 92121.

    Current address of K.M.O. and B.P.: Pfizer Global Research and Development, La Jolla, California 92121.

    First Published Online November 3, 2005

    Abbreviations: Apo, Apolipoprotein; AUC, area under the curve; CTF, cotransfection; FXR, farnesoid X receptor; h, humanized; HDL-C, high-density lipoprotein cholesterol; LG268, LG100268; LXR, liver X receptor; OGTT, oral glucose tolerance test; PPAR, peroxisome proliferator-activated receptor; RAR, retinoic acid receptor; RXR, retinoid X receptor; tg, transgenic; TZD, thiazolidinedione.

    Accepted for publication October 21, 2005.

    References

    Laudet V, Gronemeyer H 2001 The nuclear receptor facts book. London: Academic Press

    Mukherjee R, Davies PJ, Crombie DL, Bischoff ED, Cesario RM, Jow L, Hamann LG, Boehm MF, Mondon CE, Nadzan AM, Paterniti Jr JR, Heyman RA 1997 Sensitization of diabetic and obese mice to insulin by retinoid X receptor agonists. Nature 386:407–410

    Lenhard JM, Lancaster ME, Paulik MA, Weiel JE, Binz JG, Sundseth SS, Gaskill BA, Lightfoot RM, Brown HR 1999 The RXR agonist LG100268 causes hepatomegaly, improves glycaemic control and decreases cardiovascular risk and cachexia in diabetic mice suffering from pancreatic -cell dysfunction. Diabetologia 42:545–554

    Singh Ahuja H, Liu S, Crombie DL, Boehm M, Leibowitz MD, Heyman RA, Depre C, Nagy L, Tontonoz P, Davies PJA 2001 Differential effects of rexinoids and thiazolidinediones on metabolic gene expression in diabetic rodents. Mol Pharmacol 59:765–773

    Lehmann JM, Moore LB, Smith-Oliver TA, Wilkison WO, Willson TM, Kliewer SA 1995 An antidiabetic thiazolidinedione is a high affinity ligand for peroxisome proliferator-activated receptor (PPAR). J Biol Chem 270:12953–12956

    Boehm MF, Zhang L, Zhi L, McClurg MR, Berger E, Wagoner M, Mais DE, Suto CM, Davies PJA, Heyman RA, Nadzan AM 1995 Design and synthesis of potent retinoid X receptor selective ligands that induce apoptosis in leukemia cells. J Med Chem 38:3146–3155

    Lala DS, Mukherjee R, Schulman IG, Koch SS, Dardashti LJ, Nadzan AM, Croston GE, Evans RM, Heyman RA 1996 Activation of specific RXR heterodimers by an antagonist of RXR homodimers. Nature 383:450–453

    Ogilvie KM, Saladin R, Nagy TR, Urcan MS, Heyman RA, Leibowitz MD 2004 Activation of the retinoid X receptor suppresses appetite in the rat. Endocrinology 145:565–573

    Liu YL, Sennitt MV, Hislop DC, Crombie DL, Heyman RA, Cawthorne MA 2000 Retinoid X receptor agonists have anti-obesity effects and improve insulin sensitivity in Zucker fa/fa rats. Int J Obes Relat Metab Disord 24:997–1004

    Liu S, Ogilvie KM, Klausing K, Lawson MA, Jolley D, Li D, Bilakovics J, Pascual B, Hein N, Urcan M, Leibowitz MD 2002 Mechanism of selective retinoid X receptor agonist-induced hypothyroidism in the rat. Endocrinology 143:2880–2885

    Davies PJ, Berry SA, Shipley GL, Eckel RH, Hennuyer N, Crombie DL, Ogilvie KM, Peinado-Onsurbe J, Fievet C, Leibowitz MD, Heyman RA, Auwerx J 2001 Metabolic effects of rexinoids: tissue-specific regulation of lipoprotein lipase activity. Mol Pharmacol 59:170–176

    Michellys P-Y, Ardecky RJ, Chen JH, Crombie DL, Etgen GJ, Faul MM, Faulkner AL, Grese TA, Heyman RA, Karanewsky DS, Klausing K, Leibowitz MD, Liu S, Mais DA, Mapes CM, Marschke KB, Reifel-Miller A, Ogilvie KM, Rungta D, Thompson AW, Tyhonas JS, Boehm MF 2003 Novel (2E,4E,6Z)-7-(2-alkoxy-3,5-dialkylbenzene)-3-methylocta-2,4,6-trienoic acid retinoid X receptor modulators are active in models of type 2 diabetes. J Med Chem 46:2683–2696

    Mais DE, Hamann L, Klausing K, Paterniti J, Mukherjee R 1997 Characterization of a synthetic tritium-labeled peroxisome proliferator-activated receptor (hPPAR) ligand. Medicinal Chem Res 7:325–334

    Boehm MF, Zhang L, Badea BA, White SK, Mais DE, Berger E, Suto CM, Goldman ME, Heyman RA 1994 Synthesis and structure-activity relationships of novel retinoid X receptor-selective retinoids. J Med Chem 37:2930–2941

    Cheng Y, Prusoff WH 1973 Relationship between the inhibition constant (K1) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem Pharmacol 22:3099–3108

    Mangelsdorf DJ, Umesono K, Kliewer SA, Borgmeyer U, Ong ES, Evans RM 1991 A direct repeat in the cellular retinol-binding protein type II gene confers differential regulation by RXR and RAR. Cell 66:555–561

    Bissonnette RP, Brunner T, Lazarchik SB, Yoo NJ, Boehm MF, Green DR, Heyman RA 1995 9-cis retinoic acid inhibition of activation-induced apoptosis is mediated via regulation of fas ligand and requires retinoic acid receptor and retinoid X receptor activation. Mol Cell Biol 15:5576–5585

    Umesono K, Giguere V, Glass CK, Rosenfeld MG, Evans RM 1988 Retinoic acid and thyroid hormone induce gene expression through a common responsive element. Nature 336:262–265

    Willy PJ, Umesono K, Ong ES, Evans RM, Heyman RA, Mangelsdorf DJ 1995 LXR, a nuclear receptor that defines a distinct retinoid response pathway. Genes Dev 9:1033–1045

    Forman BM, Goode E, Chen J, Oro AE, Bradley DJ, Perlmann T, Noonan DJ, Burka LT, McMorris T, Lamph WW, Evans RM, Weinberger C 1995 Identification of a nuclear receptor that is activated by farnesol metabolites. Cell 81:687–693

    Kliewer SA, Umesono K, Mangelsdorf DJ, Evans RM 1992 Retinoid X receptor interacts with nuclear receptors in retinoic acid, thyroid hormone and vitamin D3 signalling. Nature 355:446–449

    Mukherjee R, Jow L, Noonan D, McDonnell DP 1994 Human and rat peroxisome proliferator activated receptors (PPARs) demonstrate similar tissue distribution but different responsiveness to PPAR activators. J Steroid Biochem Mol Biol 51:157–166

    Rubin EM, Ishida BY, Clift SM, Krauss RM 1991 Expression of human apolipoprotein A-I in transgenic mice results in reduced plasma levels of murine apolipoprotein A-I and the appearance of two new high density lipoprotein size subclasses. Proc Natl Acad Sci USA 88:434–438

    Kieft KA, Bocan TM, Krause BR 1991 Rapid on-line determination of cholesterol distribution among plasma lipoproteins after high-performance gel filtration chromatography. J Lipid Res 32:859–866

    Box GEP, Cox DR 1964 An analysis of transformations. J R Stat Soc B 26:211–252

    Sherman SI, Gopal J, Haugen BR, Chiu AC, Whaley K, Nowlakha P, Duvic M 1999 Central hypothyroidism associated with retinoid X receptor-selective ligands. N Engl J Med 340:1075–1079

    Miller VA, Benedetti FM, Rigas JR, Verret AL, Pfister DG, Straus D, Kris MG, Crisp M, Heyman R, Loewen GR, Truglia JA, Warrell Jr RP 1997 Initial clinical trial of a selective retinoid X receptor ligand, LGD1069. J Clin Oncol 15:790–795

    Forman BM 2002 The antidiabetic agent LG100754 sensitizes cells to low concentrations of peroxisome proliferator-activated receptor ligands. J Biol Chem 277:12503–12506

    Lyons F, Laker MF, Marsden JR, Manuel R, Shuster S 1982 Effect of oral 13-cis-retinoic acid on serum lipids. Br J Dermatol 107:591–595

    McMaster J, Rogers MP, Sherratt HS, Shuster S 1989 Effects of isotretinoin on lipid metabolism in the rat. Arch Dermatol Res 281:116–118

    Standeven AM, Beard RL, Johnson AT, Boehm MF, Escobar M, Heyman RA, Chandraratna RA 1996 Retinoid-induced hypertriglyceridemia in rats is mediated by retinoic acid receptors. Fund Appl Toxicol 33:264–271

    Grefhorst A, Elzinga BM, Voshol PJ, Plosch T, Kok T, Bloks VW, van der Sluijs FH, Havekes LM, Romijn JA, Verkade HJ, Kuipers F 2002 Stimulation of lipogenesis by pharmacological activation of the liver X receptor leads to production of large, triglyceride-rich very low density lipoprotein particles. J Biol Chem 277:34182–34190

    Schultz JR, Tu H, Luk A, Repa JJ, Medina JC, Li L, Schwendner S, Wang S, Thoolen M, Mangelsdorf DJ, Lustig KD, Shan B 2000 Role of LXRs in control of lipogenesis. Genes Dev 14:2831–2838

    Ljung B, Bamberg K, Dahllof B, Kjellstedt A, Oakes ND, Ostling J, Svensson L, Camejo G 2002 AZ 242, a novel PPAR/ agonist with beneficial effects on insulin resistance and carbohydrate and lipid metabolism in ob/ob mice and obese Zucker rats. J Lipid Res 43:1855–1863

    Guerre-Millo M, Gervois P, Raspe E, Madsen L, Poulain P, Derudas B, Herbert JM, Winegar DA, Willson TM, Fruchart JC, Berge RK, Staels B 2000 Peroxisome proliferator-activated receptor activators improve insulin sensitivity and reduce adiposity. J Biol Chem 275:16638–16642

    Bodkin NL, Pill J, Meyer K, Hansen BC 2003 The effects of K-111, a new insulin-sensitizer, on metabolic syndrome in obese prediabetic rhesus monkeys. Horm Metab Res 35:617–624

    Aasum E, Belke DD, Severson DL, Riemersma RA, Cooper M, Andreassen M, Larsen TS 2002 Cardiac function and metabolism in type 2 diabetic mice after treatment with BM 17.0744, a novel PPAR- activator. Am J Physiol 283:H949–H9457

    Leibowitz MD, Fievet C, Hennuyer N, Peinado-Onsurbe J, Duez H, Bergera J, Cullinan CA, Sparrow CP, Baffic J, Berger GD, Santini C, Marquis RW, Tolman RL, Smith RG, Moller DE, Auwerx J 2000 Activation of PPAR alters lipid metabolism in db/db mice. FEBS Lett 473:333–336

    Oliver Jr WR, Shenk JL, Snaith MR, Russell CS, Plunket KD, Bodkin NL, Lewis MC, Winegar DA, Sznaidman ML, Lambert MH, Xu HE, Sternbach DD, Kliewer SA, Hansen BC, Willson TM 2001 A selective peroxisome proliferator-activated receptor agonist promotes reverse cholesterol transport. Proc Natl Acad Sci USA 98:5306–5311

    Wang YX, Zhang CL, Yu RT, Cho HK, Nelson MC, Bayuga-Ocampo CR, Ham J, Kang H, Evans RM 2004 Regulation of muscle fiber type and running endurance by PPAR. PLOS Biol 2:e294(Mark D. Leibowitz, Robert J. Ardecky, Ma)