9 research outputs found

    <i>dhs-16</i> modulates feedback regulation of hypodermal <i>daf-9</i> expression.

    No full text
    <p>(A) In response to mild stress (e.g., growth at 25°C), hypodermal <i>daf-9</i> is upregulated in N2 wild-type animals (WT) carrying an integrated <i>dhIs64(daf-9::gfp)</i> array (shown at left). This is rescued upon growth with 33 µM Δ<sup>7</sup>-DA (shown at right). Arrowheads indicate the XXX R/L neuroendocrine cells in which <i>daf-9</i> expression is relatively unchanged, and arrows indicate hypodermal expression. The expression levels are displayed quantitatively to the right of each image, as the percentage of animals observed with strong (green), weak (yellow), or no (red) hypodermal GFP expression (<i>N</i>≥3, M ± SD; ***<i>p</i><0.0001). (B) Under stressful growth conditions at 27°C N2 wild-type animals still undergo reproductive development but have high hypodermal <i>daf-9::gfp</i> expression (left), whereas <i>dhs-16</i> mutants mostly develop as dauer larvae and shut off hypodermal <i>daf-9::gfp</i> expression (right) (**<i>p</i><0.01). (C) Under normal growth conditions at 20°C, hypodermal <i>daf-9</i> upregulation is low in wild-type (left), whereas upregulation is seen in the <i>dhs-16(tm1890)</i> mutant background (right), suggesting <i>daf-9</i> upregulation in response to DA deficiency (***<i>p</i><0.0001). (D) Upregulation of hypodermal <i>daf-9::gfp</i> seen in <i>dhs-16</i> mutants is not rescued by provision of 33 µM of the upstream DA precursor lathosterol (left) but is rescued by the downstream product lathosterone (right) (**<i>p</i><0.01). (E) Upregulation of hypodermal <i>daf-9::gfp</i> seen in reproductively growing <i>daf-7(e1372)</i>/TGF-β mutants (left) is rescued by DA (right) (**<i>p</i><0.01). (F) Upregulation of hypodermal <i>daf-9::gfp</i> seen in reproductively growing <i>daf-2</i>/InsR mutants (left) is also rescued by DA (right) (***<i>p</i><0.0001).</p

    <i>dhs-16</i> epistasis and synergy experiments.

    No full text
    <p>nd, not determined.</p>a<p>Dauers formed under reproductive growth conditions.</p>b<p>Number of experiments is given in parentheses.</p>c<p>Hermaphrodite distal tip cells that fail to turn in L3, <i>n</i>>50 cells, grown on NGM without added cholesterol.</p>d<p>Measured at 20°C.</p>e<p>Percentage Mig on normal NGM plates with cholesterol.</p

    <i>dhs-16</i> expression pattern and regulation.

    No full text
    <p>(A) A functional <i>dhs-16::gfp</i> is expressed in the hypodermis (arrowheads), head neurons (arrows), and posterior pharyngeal bulb (arrowheads). (B) Reduction of IIS in the <i>daf-2(e1368)</i> background results in 2-fold upregulation of <i>dhs-16::gfp</i> in the hypodermis of L3 stage animals at 20°C (*<i>p</i><0.05), although no significant change was seen in the absence of <i>daf-16</i>/FOXO, <i>daf-12</i>/NHR, or <i>daf-7</i>/TGFβ. Similar upregulation was seen in the <i>daf-36(k114)</i>/Rieske oxygenase mutant background, consistent with a role of <i>dhs-16</i> downstream of <i>daf-36</i> in DA production (<i>N</i>≥3, M ± SEM, *<i>p</i><0.05).</p

    Biosynthesis and regulation of nematode bile acids.

    No full text
    <p>(A) A revised model of the dafachronic acid biosynthetic pathway from dietary cholesterol, with newly identified activities shown in red. Although <i>dhs-16</i> is required for lathosterone production, mutant animals still produce low levels of Δ<sup>7</sup>-DA. An alternative pathway for Δ<sup>7</sup>-DA synthesis is therefore likely. In addition, <i>hsd-1</i> is not required for 4-cholesten-3-one production as previously proposed, but may be involved in producing alternative dafachronic acids. These ligands may have complex regulation and influence the synthesis of one another. Comparison to mammalian bile acid synthesis (right) reveals conserved aspects of bile acid biochemistry. Nematode and mammalian bile acid synthesis involves modification at the 7-position (shown in pink), which speculatively may partition cholesterol towards bile acid synthesis, and in both pathways oxidation of the 3-alchohol and oxidation of the sidechain at the 27-position occurs (shown in orange). (B) Model of hormonal feedback on hypodermal <i>daf-9::gfp</i> expression. (i) Stressful environmental conditions result in downregulation of IIS and TGF-β signaling, suppression of DA synthesis, and hypodermal <i>daf-9</i> expression by the DAF-12/DIN-1 repressor complex, resulting in dauer formation. (ii) Moderately stressful environments result in modest downregulation of dauer signaling pathways and DA synthesis, with compensatory upregulation of hypodermal <i>daf-9</i>, allowing for reproductive development. (iii) In favorable environments with low levels of stress, active IIS and TGF-β signaling results in ample DA production, with low expression of hypodermal <i>daf-9</i>. Note it is unknown whether DAF-12 regulates <i>daf-9</i> directly or indirectly.</p

    Newly identified loci display phenotypes resembling DA deficiency.

    No full text
    <p>(A) Enhancement of <i>daf-36(k114)</i> gonadal Mig defects is seen upon knockdown of <i>daf-9</i>, <i>dhs-16</i>, and <i>emb-8</i>. Arrowheads indicate the arms of the gonad, visible along the body due to failure of proper distal tip cell migration. (B) Mig and Daf-c phenotypes of <i>dhs-16(tm1890)</i> deletion mutants, under conditions of cholesterol deprivation (upper image) and 27°C (lower image). (C) Analysis of dauer formation at 27°C on NGM. <i>dhs-16(tm1890)</i> mutants show Daf-c phenotypes similar to <i>daf-36(k114)</i> (<i>N</i> = 3, M±SD; **<i>p</i><0.01). (D) Analysis of the gonadal Mig defects of <i>dhs-16</i> null animals on NGM without added cholesterol (<i>N</i> = 3, M ± SD; **<i>p</i><0.01, *<i>p</i><0.05). (E) RNAi knockdown of <i>dhs-16</i> and <i>emb-8</i> in wild-type worms at 27°C induces Daf-c phenotypes, similar to knockdown of <i>daf-36</i> (<i>N</i>≥4, M ± SD; **<i>p</i><0.005). (F) Genetic epistasis analysis of <i>dhs-16(tm1890)</i> (Daf-c) together with Daf-d mutations in transcription factors of insulin/IGF, TGF-β, and DA signaling show that DHS-16 works downstream of DAF-16/FOXO and DAF-5/SKI, but upstream of DAF-12/NHR (<i>N</i> = 3, M ± SD; **<i>p</i><0.01). (G) Similar genetic epistasis analysis of <i>emb-8</i> RNAi-induced dauer phenotypes at 27°C suggests that EMB-8 acts downstream of DAF-16 and upstream of DAF-12 (<i>N</i> = 3, M ± SD; **<i>p</i><0.01). (H) The DAF-12 target gene <i>mir-241</i>, a <i>let-7</i> related microRNA, shows reduced expression in <i>dhs-16</i> mutants under low cholesterol conditions at 25°C (<i>N</i> = 3, M ± SD; **<i>p</i><0.01, *<i>p</i><0.05).</p

    <i>dhs-16</i> mutant animals are deficient in lathosterone.

    No full text
    <p>(A) <i>daf-9(k182)</i> rescue is seen only with the DAs. Experiments were carried out at 27°C with 33 µM concentration of supplemented compounds (<i>N</i>≥3, M ± SD; ***<i>p</i><0.0001). (B) <i>dhs-16(tm1890)</i> rescue is seen with lathosterone and dafachronic acids, but not by cholesterol, 7-dehydrocholesterol, and lathosterol. Similarly, 4-cholesten-3-one rescues, but not cholesterol or 4-cholesten-3β-ol. This indicates that <i>dhs-16</i> may function in the conversion of lathosterol to lathosterone in the production of Δ<sup>7</sup>-DA and in the formation of 4-cholestene-3-one in the production of Δ<sup>4</sup>-DA (N≥3, M ± SD; ***<i>p</i><0.0001). (C) <i>emb-8</i> RNAi rescue is seen only with the DAs, and not with lathosterone or 4-cholesten-3-one. (<i>N</i>≥3, M ± SD; **<i>p</i><0.01). (D) LC/MS/MS analysis of lipid extracts from L3 stage animals reveals that lathosterone levels are reduced in <i>dhs-16(tm1890)</i> mutant animals compared to N2 wild-type (WT) animals, shown quantitatively in (E). Lathosterone levels are significantly reduced in <i>dhs-16</i> animals relative to N2 wild-type (3.5-fold decrease; <i>N</i> = 7, M ± SD; **<i>p</i><0.001). (F) GC/MS/MS analysis of sterol levels in <i>dhs-16</i> mutants reveals deficiencies in lathosterone, Δ<sup>7</sup>-dafachronic acid, and 4-methyl sterols compared to N2 wild-type (<i>N</i>≥10, M ± SEM; *<i>p</i><0.05, **below detection limit).</p

    <i>dhs-16</i> is partially required for longevity in the absence of the germline.

    No full text
    <p>(A) Lifespan of <i>dhs-16(tm1890)</i> animals after ablation of germline precursor cells by laser microsurgery. One representative experiment is shown. N2 wild-type (WT) animals live twice as long when the germline is ablated. Longevity is significantly attenuated in <i>dhs-16</i> ablated animals (<i>N</i> = 2, <i>p</i><0.0001). (B) DAF-16::GFP strongly localizes to intestinal nuclei of day 1 adults after ablation of the germline by laser microsurgery in control animals, whereas the degree of localization is reduced in germline-ablated <i>dhs-16</i> mutants. Magnified views of boxed regions are shown to the right. (C) The degree of DAF-16::GFP nuclear localization after germline ablation is shown quantitatively as the percentage of animals with strong (green) or weak (yellow) localization (<i>N</i> = 3, M ± SD; *<i>p</i><0.05). (D) The ratio of intestinal DAF-16::GFP intensity in the nucleus versus cytoplasm reveals increased levels of nuclear expression in germline deficient <i>glp-1(e2141ts)</i> mutants at the restrictive temperature of 25°C compared to control animals. Localization is significantly decreased in <i>dhs-16;glp-1</i> double mutant animals, and is restored upon provision of lathosterone or Δ<sup>7</sup>-dafachronic acid, but not lathosterol (<i>N</i> = 3, M ± SD; ***<i>p</i><0.0001, **<i>p</i><0.001).</p

    DHS-16 acts as a 3-hydroxysteroid dehydrogenase in DA biosynthesis.

    No full text
    <p>(A) LC/MS/MS analysis of lipid extracts from DHS-16 and control microsomes incubated with the proposed substrate lathosterol, shown quantitatively in (B). Significantly more lathosterone is detected in incubations of DHS-16 microsomes with lathosterol than in incubations with empty vehicle ethanol controls. DHS-16 microsomes also do not produce lathosterone when incubated with cholesterol, demonstrating that specific products are made depending upon the substrate provided (<i>N</i>≥3, M ± SD; **<i>p</i><0.005). (C) LC/MS/MS analysis of 4-cholesten-3-one levels in lipid extracts from DHS-16 and control microsomes incubated with cholesterol, shown quantitatively in (D). Concentrations of 4-cholesten-3-one in incubations of DHS-16 microsomes with cholesterol are significantly greater than that seen in control microsomes or incubations with lathosterol (<i>N</i> = 3, M ± SD; *<i>p</i><0.05). (E) Rescue of the Daf-c phenotype of <i>dhs-16(tm1890)</i> mutants at 27°C is seen when fed lipid extracts of DHS-16 microsomes incubated with the proposed substrate lathosterol. Rescue is not seen with extracts from DHS-16 microsomes incubated with ethanol vehicle alone or with cholesterol, and extracts from empty vector pCMV control microsomes do not rescue in any condition (estimated concentrations of 300 nM in plates; <i>N</i> = 3, M ± SEM; *<i>p</i><0.05). (F) GC/MS/MS analysis of sterol levels in N2 wild-type, <i>dhs-16(tm1890)</i>, <i>hsd-1(mg433)</i>, and <i>dhs-16;hsd-1</i> double mutants reveals that <i>hsd-1</i> is not required for 4-cholesten-3-one production, as previously proposed, suggesting HSD-1 may act in an alternative parallel pathway. In addition, although the <i>dhs-16;hsd-1</i> double mutants did not contain measurable lathosterone levels, Δ<sup>7</sup>-DA levels were not reduced, suggesting that lathosterone is not required for its production and that alternate pathways must exist that are independent of <i>dhs-16</i> (<i>N</i>≥6, M ± SEM; **below detection limit, *<i>p</i><0.05).</p
    corecore