15 research outputs found

    DGAT1 inhibitors with high lipophilicity induce sebaceous gland atrophy.

    No full text
    <p>Shown are hematoxylin and eosin stains of dorsal skin biopsies from DIO mice treated with either vehicle (A), Cpd1 (B), Cpd2 (C), or Cpd3 (D) for 14 days at doses indicated. Scoring refers to the histological adverse effect score as described in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0088908#pone-0088908-t001" target="_blank">Table 1</a>. Bar  = 50 µm. The corresponding sebaceous gland sizes (area) are plotted in (E) (and shown in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0088908#pone.0088908.s002" target="_blank">Table S1</a>).</p

    RNA biomarkers for sebaceous gland atrophy in skin. Listed are the 41 unique genes from the 42 probesets identified in the Training Set as shown in Figure 4 (Cxcl16 had 2 probesets).

    No full text
    <p>Fold change and ANOVA p values for compound treatments compared to their respective vehicle treatments, for both the Training and Test Sets, are included. The 26 probesets that are also significantly regulated in the Test Set are shown in bold. **  = ANOVA p<0.01; *  = ANOVA p<0.05; $ = ANOVA p<0.1.</p

    RNA biomarkers for sebaceous gland atrophy in skin.

    No full text
    <p>Shown are the 42 probesets, identified in the Training Set (Studies 1 and 2), that were regulated by skin-positive compound treatments (those that produced sebaceous gland atrophy) but not by the skin-negative compound treatments (the one that did <i>not</i> produce sebaceous gland atrophy). After excluding the absent probes (low intensity), these 42 probesets met the following cutoffs: 1.2 fold change and ANOVA p<0.01 between all 3 skin-positive compound treatments (red arrows) and their respective vehicle treatments, and ANOVA p>0.1 between the skin negative compound treatment (black arrow) and its respective vehicle treatment. The probesets for RIKEN genes were excluded. Plotted are the LogRatio values (+/− 4 fold fold scale) with magenta representing up-regulated probesets and cyan representing down-regulated probesets. Treatments from the independent Test Set (Study 4) are included for comparison but were not used to identify the 42 probesets.</p

    Immune-regulated genes are up-regulated, while lipid metabolism genes are down-regulated, with sebaceous gland atrophy.

    No full text
    <p>Box plots of probesets regulated by the skin-positive DGAT1 inhibitors (those that produce sebaceous gland atrophy) but not by the skin-negative compounds (those that do <i>not</i> produce sebaceous gland atrophy). Plotted are the LogIntensity values across the replicates in each group, and across the three studies. Ccl1 (A; chemokine (C-C motif) ligand 1) is involved in the recruitment of T cells in skin inflammation; and Scd3 (B; stearoyl-coenzyme A desaturase 3) is a sebaceous gland specific gene.</p

    Validation of the 42 probeset-composite score in an independent Test Set.

    No full text
    <p>The 42 probesets from the Training Set, shown in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0088908#pone-0088908-g005" target="_blank">Figure 5</a>, were used to generate a composite score across the treatments from the independent Test Set (Study 4). The p value between Cpd6 treatment (skin-positive) and Cpd18 treatment (skin-negative), in Study 4, is less than 0.0001. Expression data from this set of RNA biomarkers is predictive for sebaceous gland atrophy in mice following DGAT inhibitor treatment.</p

    Compound characteristics. Cpd1 is the same as DGAT1i in Lin, H et al (2013) [4], while Cpd2 is the same as Compound L in Liu, J et al (2013) [3].

    No full text
    <p>Plasma  =  plasma concentration; Skin  =  skin concentration; Skin/Plasma Ratio  =  skin to plasma concentration ratio; Fu Plasma  =  unbound fraction in plasma; mlogD  =  measured logD; clogD  =  calculated logD using ACD (measure of lipophilicity); mDGAT1 IC50  =  in vitro potency; Skin/IC50 =  skin concentration vs in vitro potency; Scoring  =  histological adverse effect score; clogD/mIC50 =  calculated logD vs in vitro potency; BLQ  =  below detection. Bold  =  compounds causing atrophy.</p

    Stimulation of Glucose-Dependent Insulin Secretion by a Potent, Selective sst<sub>3</sub> Antagonist

    No full text
    This letter provides the first pharmacological proof of principle that the sst<sub>3</sub> receptor mediates glucose-stimulated insulin secretion (GSIS) from pancreatic β-cells. To enable these studies, we identified the selective sst<sub>3</sub> antagonist (1<i>R</i>,3<i>R</i>)-3-(5-phenyl-1<i>H</i>-imidazol-2-yl)-1-(tetrahydro-2<i>H</i>-pyran-4-yl)-2,3,4,9-tetrahydro-1<i>H</i>-β-carboline (<b>5a</b>), with improved ion channel selectivity and mouse pharmacokinetic properties as compared to previously described tetrahydro-β-carboline imidazole sst3 antagonists. We demonstrated that compound <b>5a</b> enhances GSIS in pancreatic β-cells and blocks glucose excursion induced by dextrose challenge in ipGTT and OGTT models in mice. Finally, we provided strong evidence that these effects are mechanism-based in an ipGTT study, showing reduction of glucose excursion in wild-type but not sst<sub>3</sub> knockout mice. Thus, we have shown that antagonism of sst<sub>3</sub> represents a new mechanism with potential in treating type 2 diabetes mellitus

    Diamine Derivatives as Novel Small-Molecule, Potent, and Subtype-Selective Somatostatin SST3 Receptor Agonists

    No full text
    A novel class of small-molecule, highly potent, and subtype-selective somatostatin SST3 agonists was discovered through modification of a SST3 antagonist. As an example, (1<i>R</i>,2<i>S</i>)-<b>9</b> demonstrated not only potent in vitro SST3 agonist activity but also in vivo SST3 agonist activity in a mouse oral glucose tolerance test (OGTT). These agonists may be useful reagents for studying the physiological roles of the SST3 receptor and may potentially be useful as therapeutic agents
    corecore