The LXXLL motif of murine forkhead transcription factor FoxO1 mediates Sirt1-dependent transcriptional activity (2024)

The LXXLL motif is conserved among FoxO family members. We identified an LXXLL motif at amino acids 459–463 in murine FoxO1, which is highly conserved among FoxO family members in human, murine, and Xenopus laevis and weakly conserved in Drosophila melanogaster and Caenorhabditis elegans (Table 1). This finding suggests that the LXXLL motif may have an important role in the function of FoxO proteins.

Table 1

Sequence alignment of LXXLL motifs in members of FoxO family

Mutagenesis of the LXXLL motif of FoxO1 abolishes Igfbp-1 and G6Pase promoter activity. At first, in order to investigate roles of the LXXLL motif of FoxO1, we constructed an LXXAA mutant in which leucine 462 and 463 were substituted by alanine (Figure 1A) and performed Igfbp-1 and G6Pase promoter assays using this mutant FoxO1 in SV40-transformed hepatocytes (25). Both Igfbp-1 and G6Pase have already been identified as target genes of FoxO1 (16, 26). In the absence of serum, WT FoxO1 increased Igfbp-1 and G6Pase promoter activity by 2.5- and 3.2-fold compared with control, respectively (Figure 1, B and C). However, the LXXAA mutant increased Igfbp-1 and G6Pase promoter activity by only 1.3- and 1.9-fold, respectively (Figure 1, and C). These data indicate that disruption of the LXXAA motif of FoxO1 decreases its transcriptional activity on its target genes in the absence of serum.

Figure 1

Scheme and subcellular localization of FoxO1 mutants in SV40-transformed hepatocytes and expression of an Igfbp-1 /luciferase or G6Pase/luciferase reporter gene in SV40-transformed hepatocytes transfected with WT or LXXAA FoxO1. (A) Scheme of WT, LXXAA FoxO1, 3A FoxO1, and 3A/LXXAA FoxO1. (B) Expression of cMyc-WT (lane 2) and LXXAA FoxO1 (lane 3) in SV40-transformed hepatocytes was detected. (C) After overnight serum deprivation and induction with dexamethasone, 8-Br-cAMP, and IBMX in the presence or absence of insulin (100 nM) as described in Methods, cells were harvested and luciferase activity was measured. *Statistically significant difference between control and WT-transfected cells, P < 0.001 by 1-way ANOVA; **statistically significant difference between WT-and LXXAA-transfected cells, P < 0.005 by 1-way ANOVA. Data represent mean ± SEM from 3 independent transfection experiments. Following transient transfection, hepatocytes were seeded onto 4-well slide culture chambers and incubated in various conditions, including serum deprivation, for 24 hours (D), stimulation with insulin (100 nM, 20 minutes) (E), incubation with serum (F), and stimulation with H2O2 (250 μM, 4 hours) (G). Myc epitope–tagged FoxO1 was visualized with anti-cMyc monoclonal antibody and FITC-conjugated anti-mouse IgG.

Transcriptional activity of FoxO1 is largely dependent on its subcellular localization (1). Although serum deprivation increases nuclear localization of FoxO1, the amount of FoxO1 that is localized in the nucleus is about 60% and cannot reach 100% in this cell line (data not shown). In order to investigate effects of disruption of the LXXAA motif on transcriptional regulation without considering subcellular localization of FoxO1, we constructed 3A/LXXAA mutant in which all Akt phosphorylation sites and leucines 462 and 463 were also substituted by alanine (Figure 1A). Immunofluorescence studies demonstrated that subcellular localization of the 3A/LXXAA mutant was the same as in the 3A mutant, in which only 3 Akt phosphorylation sites were substituted by alanine (Figure 1A) under several conditions, i.e., it is constitutively nuclear (Figure 1, D–G). Using these mutant FoxO1, we investigated effects of disruption of the LXXLL motif of FoxO1 on transcriptional regulation of FoxO1 target genes without considering subcellular localization. We performed Igfbp-1 and G6Pase promoter assays to investigate the effects of mutagenesis of the LXXLL motif on target gene expression. We used adenovirus vectors encoding LacZ, WT, 3A, and 3A/LXXAA in order to fix FoxO1 expression level (Figure 2A). WT FoxO1 and 3A FoxO1 increased Igfbp-1 promoter activity by 2.3- and 5.4-fold compared with LacZ, respectively (Figure 2B). However, the 3A/LXXAA mutant increased Igfbp-1 promoter activity by only 2.8-fold (Figure 2B). Therefore, disruption of the LXXLL motif of 3A FoxO1 decreased Igfbp-1 promoter activity by 47%. Furthermore, WT FoxO1 and 3A FoxO1 increased G6Pase promoter activity by 5.0- and 11.6-fold, respectively (Figure 2C). However, the 3A/LXXAA mutant increased G6Pase promoter activity by only 5.7-fold (Figure 2C). Therefore, disruption of the LXXLL motif of 3A FoxO1 decreased G6Pase promoter activity by 50%. These data suggest that disruption of the LXXLL motif of FoxO1 abolishes Igfbp-1 and G6Pase promoter activity and that the LXXLL motif of FoxO1 plays an important role in mediating the transcriptional activity of FoxO1.

Figure 2

Expression of an Igfbp-1 /luciferase or G6Pase /luciferase reporter gene in SV40-transformed hepatocytes transduced with adenovirus encoding LacZ, WT FoxO1, 3A FoxO1, or 3A/LXXAA FoxO1. (A) Expression of HA-WT FoxO1, Flag 3A FoxO1, or 3A/LXXAA FoxO1 in SV40-transformed hepatocytes was detected. Lane 1, hepatocytes transduced with adenovirus encoding LacZ; lane 2, with WT; lane 3, with 3A FoxO1; lane 4, with 3A/LXXAA FoxO1 at 10 MOI. After hepatocytes were transfected with Igfbp-1/(p925GL3) (B) or G6Pase/(PicaGene/ human G6Pase promoter-luciferase) (C), cells were infected with indicated adenovirus. Synthetic Renilla luciferase reporter vector (phRL-SV40) was used as an internal control of transfection efficiency. After overnight serum deprivation and induction with dexamethasone, 8-Br-cAMP, and IBMX as described in Methods, cells were harvested and luciferase activity was measured. *Statistically significant difference between 3A FoxO1– and WT FoxO1– or 3A/LXXAA FoxO1–transduced cells, P < 0.001 by 1-way ANOVA. Data represent mean ± SEM from 3 independent experiments.

Mutagenesis of the LXXLL motif of FoxO1 inhibits endogenous Igfbp-1 and G6Pase gene expression. In order to investigate effects of disrupting the LXXLL motif of FoxO1 on endogenous gene expression, we transduced SV40-transformed hepatocytes with adenovirus vector encoding WT FoxO1, 3A FoxO1, or 3A/LXXAA FoxO1 (Figure 3A). In this cell line, Igfbp-1 and G6Pase are induced in a dexamethasone/8-Br-cAMP/IBMX–dependent manner (8-Br-cAMP, 8-bromoadenosine cAMP; IBMX, 3-isobutyl-1-methylxanthine). After serum deprivation, dexamethasone/8-Br-cAMP/IBMX increased endogenous Igfbp-1 gene expression by 43-fold compared with basal condition (data not shown). Furthermore, transduction with 3A FoxO1 increased dexamethasone/8-Br-cAMP/IBMX–induced Igfbp-1 gene expression by 23-fold compared with LacZ-transduced conditions (Figure 3B). However, transduction with 3A/LXXAA FoxO1 increased Igfbp-1 gene expression by only 5.2-fold compared with LacZ-transduced conditions (Figure 3B). Furthermore, we investigated the effects of overexpression of mutant FoxO1 protein on endogenous G6Pase gene expression in this cell line. Transduction with 3A FoxO1 increased dexamethasone/8-Br-cAMP/IBMX–induced endogenous G6Pase by 2.6-fold compared with LacZ-transduced condition. However, transduction with 3A/LXXAA FoxO1 increased by only 1.8-fold (Figure 3C). These data indicate that FoxO1 regulates endogenous Igfbp-1 and G6Pase gene expression in SV40-transformed hepatocytes in an LXXLL motif–dependent manner.

Figure 3

Effects of 3A FoxO1 and 3A/LXXAA FoxO1 mutants on endogenous Igfbp-1 or G6Pase gene expression in SV40-transformed hepatocytes. (A) Expression of HA-WT, Flag 3A FoxO1, or 3A/LXXAA FoxO1 in SV40-transformed hepatocytes was detected. Lane 1, hepatocytes transduced with adenovirus encoding LacZ; lane 2, with WT; lane 3, with 3A mutant; lane 4, with 3A/LXXAA mutant at 10 MOI. Endogenous Igfbp-1 (B) or G6Pase (C) gene expression in SV40-transformed hepatocytes transduced with adenovirus encoding LacZ, WT FoxO1, 3A FoxO1, or 3A/LXXAA FoxO1. Hepatocytes transduced with each adenovirus were incubated in serum-free medium overnight before addition of dexamethasone/8-Br-cAMP/IBMX and incubation for 8 hours. Isolation of total RNA and real-time PCR were performed as described in Methods. Data (mean ± SEM) are from 3 independent experiments and are normalized by the amount of β-actin mRNA and expressed relative to the corresponding LacZ value. *Statistically significant difference between 3A- and WT- or 3A/LXXAA-transduced cells at each MOI (P < 0.001 by 1-way ANOVA). (D) Effects of 3A FoxO1, 3A/LXXAA FoxO1, and Δ256 FoxO1 mutants on endogenous Igfbp-1 gene expression in SV40-transformed hepatocytes. Hepatocytes transduced with each adenovirus at 10 MOI were incubated in serum-free medium overnight before addition of dexamethasone/8-Br-cAMP/IBMX for 8 hours. Isolation of total RNA and real-time PCR were performed as described in Methods. Data are from 3 independent experiments and are expressed as mean ± SEM of fold of relative gene expression for β-actin in LacZ-transduced hepatocytes in the absence of dexamethasone/8-Br-cAMP/IBMX.

Because the C terminal region of FoxO1 is a transactivation domain, truncation of the terminus (Δ256) abolishes its transcriptional activity. In order to determine how much transcriptional activity the LXXLL motif contributes to full transcriptional activity of the terminus of FoxO1, we compared endogenous Igfbp-1 gene expression levels in SV40-transformed hepatocytes infected with adenovirus encoding 3A FoxO1, 3A/LXXAA FoxO1, or Δ256 FoxO1. In the absence of dexamethasone/8-Br-cAMP/ IBMX, 3A FoxO1, 3A/LXXAA FoxO1, or Δ256 FoxO1 induced endogenous Igfbp-1 gene expression by 9.9-, 4.3-, or 1.4-fold, respectively (Figure 3D). In the presence of dexamethasone/8-Br-cAMP/ IBMX, transduction with 3A FoxO1, 3A/LXXAA FoxO1, or Δ256 FoxO1 induced gene expression by 98-, 30-, or 6.6-fold, respectively (Figure 3D). These data suggest that 70% of transcriptional activity of FoxO1 depends on the LXXLL motif.

Disruption of the LXXLL motif in constitutively active mutant FoxO1 confers dominant-negative effect onto endogenous Igfbp-1 gene expression. In order to investigate whether disruption of the LXXLL motif of FoxO1 confers dominant-negative effect onto endogenous FoxO1 target gene expression, we transduced SV40-transformed hepatocytes with adenoviruses encoding hemagglutinin–T24A/S253D/S316A (HA-ADA), which has already been reported to be constitutively active FoxO1 (16), and Flag-3A/LXXAA FoxO1 (Figure 4A), and examined endogenous Igfbp-1 gene expression by real-time PCR. In this cell line, HA-ADA FoxO1 induced endogenous Igfbp-1 gene expression by 25-fold compared with nontransduced cells in the presence of dexamethasone/8-Br-cAMP/ IBMX (Figure 4B). However, overexpression of 3A/LXXAA FoxO1 inhibited endogenous Igfbp-1 gene expression induced by overexpression of HA-ADA by 65% and 70% in a dose-dependent manner (Figure 4B). These data suggest that disruption of the LXXLL motif of FoxO1 confers dominant-negative effect onto endogenous Igfbp-1 gene expression in SV40-transformed hepatocytes.

Figure 4

Effects of 3A/LXXAA FoxO1 on endogenous Igfbp-1 gene expression in SV40-transformed hepatocytes. (A) Expression of Flag 3A/LXXAA FoxO1 and HA-ADA FoxO1 at indicated MOI in SV40-transformed hepatocytes. (B) The effects of 3A/LXXAA FoxO1 on HA-ADA–induced Igfbp-1 gene expression in the presence of dexamethasone/8-Br-cAMP/IBMX in SV40-transformed hepatocytes. Data from 3 independent experiments are expressed as mean ± SEM of the folds of endogenous Igfbp-1 gene expression in the absence of HA-ADA FoxO1. *Statistically significant difference between 3A/LXXAA FoxO1 at MOI 0 and at MOI 10 or 20. P < 0.001 by 1-way ANOVA.

The LXXLL motif of FoxO1 is indispensable for binding to Sirt1 in vivo. From previous studies, the LXXLL motif of FoxO1 appears to be quite important for its transcriptional activity. Therefore, we sought to identify the mechanism by which the LXXLL motif of FoxO1 is involved in its transcriptional regulation. The nicotinamide adenine dinucleotide–dependent (NAD-dependent) deacetylase Sirt1 is abundantly expressed in SV40-transformed hepatocytes (Figure 5A). After transduction of SV40-transformed hepatocytes with adenovirus vector encoding LacZ, 3A FoxO1, or 3A/LXXAA FoxO1, we immunoprecipitated 3A FoxO1 or 3A/LXXAA FoxO1 using anti-Flag antibody and blotted with anti–silent information regulator 2 (anti-Sir2; mammalian Sirt1) antibody. We showed that 3A FoxO1 bound to endogenous Sirt1. However, 3A/LXXAA FoxO1 failed to do so (Figure 5A). Furthermore, immunoprecipitation with anti-Sir2 antibody revealed that endogenous Sirt1 interacted with transduced Flag 3A FoxO1 but not with Flag 3A/LXXAA FoxO1 (Figure 5A). These data suggest that the LXXLL motif of FoxO1 is important for binding to Sirt1.

Figure 5

Sirt1 interacts with 3A FoxO1 but not with 3A/LXXAA FoxO1, and disruption of the LXXLL motif enhances acetylation of FoxO1. (A) SV40-transformed hepatocytes were transduced with adenovirus encoding Flag 3A FoxO1 (lane 1) or Flag 3A/LXXAA FoxO1 (lane 2). After 24 hours serum starvation, cells were cultured in AMEM supplemented with 0.1% bovine serum albumin, 0.5 mM 8-Br-cAMP, 0.5 mM IBMX, and 1 μM dexamethasone for 8 hours. Lysates were immunoprecipitated using anti-Flag monoclonal antibody (M2) and Western blotted with anti-Sir2 polyclonal (first panel) or anti-Flag monoclonal antibody (M2) (fourth panel) or immunoprecipitated using anti-Sir2 polyclonal antibody and Western blotted with anti-Sir2 antibody (second panel) or anti-Flag monoclonal antibody (third panel). (B) SV40-transformed hepatocytes transduced with adenovirus encoding Flag 3A FoxO1 (lane 1) or Flag 3A/LXXAA FoxO1 (lane 2) were incubated with 0.5 mM 8-Br-cAMP, 0.5 mM IBMX, and 1 μM dexamethasone for 8 hours after 24 hours of serum starvation. Lysates were immunoprecipitated using anti-Flag monoclonal antibody (M2) and Western blotted with anti-acetylated lysine polyclonal (top panel) or anti-Flag monoclonal antibody (M2) (bottom panel). (C) Quantification of the data in B. Mean ± SEM of the folds of acetylation of 3A/LXXAA was calculated from 3 independent experiments using NIH Image 1.62 (http://rsb.info.nih.gov/nih-image/). Subsequent blotting with anti-Flag antibody using the same filter normalized acetylation of each FoxO1. (D) HEK293 cells transfected with pCMV5/cMyc/3A FoxO1 (lanes 1–4) or 3A/LXXAA FoxO1 (lanes 5–8) were incubated for 8 hours in the absence or presence of nicotinamide (50 mM) or trichostatin A (TSA) (2 μM). cMyc-tagged FoxO1 was immunoprecipitated, and the acetylation of FoxO1 was assessed by Western blot with the antibody to acetylated lysine (upper panel). Total levels of FoxO1 were assessed with the antibody to cMyc (lower panel).

In order to investigate the effects of disruption of the LXXLL motif of FoxO1 on its acetylation, we performed Western blotting with anti-acetylated lysine antibody using the same sample. Mutagenesis of the LXXLL motif in 3A FoxO1 showed increased acetylation compared with that of 3A FoxO1 (Figure 5, B and C). Furthermore, in order to investigate effects of disruption of the LXXLL motif on acetylation of FoxO1 in the presence of nicotinamide, an inhibitor of Sirt1, and/or trichostatin A (TSA), an inhibitor of class I and class II histone deacetylases (HDACs), we transfected HEK293 cells with pCMV5/cMyc/3A FoxO1 or 3A/LXXAA FoxO1, immunoprecipitated cell lysates with anti-cMyc antibody, and blotted with anti-acetylated lysine polyclonal antibody. Although acetylation of 3A/LXXAA FoxO1 was detected in the presence of nicotinamide and TSA (Figure 5D), 3A FoxO1 was not acetylated under the same conditions (Figure 5D). These data suggest that disruption of the LXXLL motif enhances acetylation of FoxO1.

Sirt1 inhibitor inhibits Igfbp-1 and G6Pase promoter activity in an LXXLL motif–dependent manner. Nicotinamide is a Sirt1-specific deacetylase inhibitor. The effects of Sirt1 on transcriptional activity of FoxO family members are controversial. These effects seem to be dependent on FoxO’s target genes (2730). In order to investigate whether the LXXLL motif of FoxO1 mediates Sirt1-dependent promoter activity of FoxO1 target genes, promoter assay of Igfbp-1 or G6Pase was performed. Nicotinamide decreased Igfbp-1 promoter activity by 48% compared with promoter activity induced by both dexamethasone/8-Br-cAMP/IBMX and 3A FoxO1 (Figure 6A). In contrast, nicotinamide did not change Igfbp-1 promoter activity induced by both dexamethasone/8-Br-cAMP/IBMX and 3A/LXXAA FoxO1 (Figure 6A). Furthermore, nicotinamide decreased G6Pase promoter activity by 70% compared with promoter activity induced by both dexamethasone/8-Br-cAMP/IBMX and 3A FoxO1 (Figure 6B). However, nicotinamide did not change G6Pase promoter activity induced by both dexamethasone/8-Br-cAMP/IBMX and 3A/LXXAA FoxO1 (Figure 6B). Interestingly, Igfbp-1 and G6Pase promoter activity induced by both dexamethasone/8-Br-cAMP/IBMX and 3A/LXXAA FoxO1 was the same as the nicotinamide-treated promoter activity level of 3A FoxO1 (Figure 6, A and B). In order to confirm the effect of the Sirt1 inhibitor on transcriptional regulation of FoxO1, we used another Sirt1 inhibitor, BML-210 (27), and performed the same promoter assays. It has been reported that BML-210 was a less potent inhibitor of Sirt1 than nicotinamide (27). BML-210 significantly decreased Igfbp-1 promoter activity (by 25%) compared with promoter activity induced by both dexamethasone/8-Br-cAMP/IBMX and 3A FoxO1 (Figure 7A). In contrast, BML-210 did not change Igfbp-1 promoter activity induced by both dexamethasone/8-Br-cAMP/IBMX and 3A/LXXAA FoxO1 (Figure 7A). Furthermore, BML-210 significantly decreased G6Pase promoter activity (by 17%) compared with promoter activity induced by both dexamethasone/8-Br-cAMP/IBMX and 3A FoxO1 (Figure 7B). However, BML-210 did not change G6Pase promoter activity induced by both dexamethasone/8-Br-cAMP/IBMX and 3A/LXXAA FoxO1 (Figure 7B). These data demonstrate that nicotinamide can inhibit 3A FoxO1–induced promoter activity of Igfbp-1 and G6Pase to the same level as that induced by 3A/LXXAA FoxO1 and suggest that the LXXLL motif mediates the Sirt1-dependent transcriptional activity of FoxO1.

Figure 6

Nicotinamide inhibits FoxO1-dependent induction of a reporter construct under the control of an Igfbp-1 or G6Pase promoter. After hepatocytes were transfected with Igfbp-1/(p925GL3) (A) or G6Pase/luciferase reporter gene (PicaGene/human G6Pase promoter-luciferase) (B), cells were infected with indicated adenovirus. phRL-SV40 was used as an internal control of transfection efficiency. After overnight serum deprivation (black bars) and induction with dexamethasone/8-Br-cAMP/ IBMX as described in Methods in the presence (light gray bars) or absence (dark gray bars) of nicotinamide (50 mM), cells were harvested and luciferase activity was measured. *Statistically significant difference between luciferase activity in the absence and in the presence of nicotinamide in cells transfected with each vector. P < 0.005 by 1-way ANOVA. Data represent mean ± SEM from 3 independent experiments.

Figure 7

BML-210 inhibits FoxO1-dependent induction of a reporter construct under the control of an Igfbp-1 or G6Pase promoter. After hepatocytes were transfected with Igfbp-1/(p925GL3) (A) or G6Pase/luciferase reporter gene (PicaGene/human G6Pase promoter-luciferase) (B), cells were infected with indicated adenovirus. phRL-SV40 was used as an internal control of transfection efficiency. After overnight serum deprivation (black bars) and induction with dexamethasone/8-Br-cAMP/IBMX as described in Methods in the presence (light gray bars) or absence (dark gray bars) of BML-210 (1.5 mM), cells were harvested and luciferase activity was measured. *Statistically significant difference between luciferase activity in the absence and luciferase activity in the presence of BML-210 at cells transfected with each vector, P < 0.01 by 1-way ANOVA. Data represent mean ± SEM from 3 independent experiments.

Resveratrol activates Igfbp-1 and G6Pase promoter activity in an LXXLL motif–dependent manner. Resveratrol was found to be a potent activator of Sirt1 (31). Recently, it has been reported that resveratrol increases glucose production by increasing FoxO1-dependent transcription of gluconeogenic genes in H4IIE rat hepatoma cells (32). In order to determine whether the LXXLL motif in FoxO1 is involved in Sirt1-dependent transcriptional regulation, we also assessed effects of resveratrol on Igfbp-1 or G6Pase promoter activity induced by both dexamethasone/8-Br-cAMP/IBMX and 3A FoxO1 or 3A/LXXAA FoxO1. Resveratrol increased Igfbp-1 or G6Pase promoter activity induced by both dexamethasone/8-Br-cAMP/IBMX and 3A FoxO1 by 30% and 37%, respectively (P < 0.005 or P < 0.001 by 1-way ANOVA, respectively) (Figure 8, A and B). In contrast, resveratrol had no effects on both Igfbp-1 and G6Pase promoter activity levels induced by both dexamethasone/8-Br-cAMP/IBMX and 3A/ LXXAA FoxO1 (Figure 8, A and B). These data suggest that resveratrol, a Sirt1 activator, activates promoter activity of FoxO1 target genes in an LXXLL motif–dependent manner.

Figure 8

Resveratrol activates induction of a reporter construct under the control of an Igfbp-1 or G6Pase promoter. After hepatocytes were transfected with Igfbp-1/(p925GL3) (A) or G6Pase/luciferase reporter gene (PicaGene/human G6Pase promoter-luciferase) (B), cells were infected with indicated adenovirus. phRL-SV40 was used as an internal control of transfection efficiency. After overnight serum deprivation (black bars) and induction with dexamethasone/8-Br-cAMP/IBMX as described in Methods in the presence (light gray bars) or absence (dark gray bars) of resveratrol (10 μM), cells were harvested and luciferase activity was measured. *Statistically significant difference between luciferase activity in the absence and luciferase activity in the presence of resveratrol, *P < 0.005 and **P < 0.001 by 1-way ANOVA. Data represent mean ± SEM from 3 independent experiments.

Effects of acute hepatic overexpression of 3A/LXXAA FoxO1 on glucose metabolism in Leprdb/db mice. From the previous data, it appears that 3A/LXXAA FoxO1 acts as a dominant-negative type FoxO1. In order to confirm the dominant-negative effect of 3A/LXXAA FoxO1 in vivo, we administered adenovirus (4 × 108 pfu/g of body weight) encoding LacZ, 3A FoxO1, or 3A/LXXAA FoxO1 into leptin receptor–deficient Leprdb/db mice (Figure 9, A and B). These mice show hyperphagia, obesity, and insulin resistance in their basal state, and it has already been reported that about 50% of total intracellular FoxO1 proteins were localized in nucleus in livers from Leprdb/db mice (33). Therefore, it is appropriate to use Leprdb/db mice for investigation of a dominant-negative effect of 3A/LXXAA FoxO1. Immunohistochemistry using anti-FoxO1 serum demonstrated that both exogenous FoxO1 mutants were localized in nucleus of liver (Figure 9B). Five days after injection, we performed intraperitoneal glucose tolerance tests. Fasting blood glucose levels of mice injected with 3A FoxO1 increased significantly compared with those of mice injected with adenovirus encoding LacZ (469.6 ± 30.7 versus 311 ± 27.5; P < 0.001 by 1-way ANOVA). In contrast, fasting blood glucose of mice injected with 3A/LXXAA FoxO1 decreased significantly compared with mice injected with LacZ or 3A FoxO1 (176 ± 30.7 versus 311 ± 27.5 or 469.9 ± 30.7; P < 0.001 by 1-way ANOVA) (Figure 9C). Furthermore, intraperitoneal glucose tolerance testing showed that mice injected with 3A/LXXAA FoxO1 were significantly more glucose tolerant than mice injected with LacZ or 3A FoxO1 (Figure 9D). These data suggest that overexpression of 3A/LXXAA FoxO1 in liver can alleviate insulin resistance of Leprdb/db mice.

Figure 9

Effects of overexpression of 3A/LXXAA FoxO1 on fasted blood glucose and glucose tolerance in Leprdb/db mice. (A) Expression of 3A FoxO1 or 3A/LXXAA FoxO1 mutant in liver. Liver was removed from mice at day 5 after injection with adenovirus encoding LacZ (lane 1), 3A FoxO1 (lane 2), or 3A/LXXAA FoxO1 (lane 3) intravenously, and 15 μg of each liver lysate was subjected to immunoblot analysis with an anti-FOXO1 polyclonal antibody (N18). (B) Immunohistochemistry of liver from Leprdb/db mice injected with adenovirus encoding LacZ (left panel), 3A FoxO1 (middle panel), or 3A/LXXAA FoxO1 (right panel). Each panel shows representative sections. Magnification, ×40. Scale bar: 10 μm. (C) Effect of overexpression of 3A FoxO1 or 3A/LXXAA FoxO1 in liver from Leprdb/db mice on fasted blood glucose. Blood glucose levels were measured as described in Methods after overnight fasting at day 5 after injection with each adenovirus encoding LacZ (white bar), 3A FoxO1 (gray bar) or 3A/LXXAA FoxO1 (black bar). Data represent mean ± SEM of 6 mice for each group. *Statistically significant difference between mice injected with each adenovirus encoding LacZ, 3A FoxO1, or 3A/LXXAA FoxO1. P < 0.001 by 1-way ANOVA. (D) Intraperitoneal glucose tolerance test was performed in mice injected with the adenoviruses indicated. The groups of mice indicated were injected with a 1.2 g/kg of body weight of glucose intraperitoneally as follows: LacZ (open circles, n = 6), 3A FoxO1 (filled circles, n = 6), and 3A/LXXAA FoxO1 (filled squares, n = 6). *Statistically significant difference between 3A and 3A/LXXAA, P < 0.01 by 1-way ANOVA; **statistically significant difference between 3A/LXXAA and LacZ or 3A, P < 0.01 by 1-way ANOVA.

Overexpression of 3A/LXXAA FoxO1 in liver from Leprdb/db mice inhibits glucose production through inhibition of G6Pase gene expression. Real-time PCR shows that G6Pase and Igfbp-1 gene expression levels in liver from fasted mice injected with 3A/ LXXAA FoxO1 are significantly decreased compared with those in mice injected with 3A FoxO1. In contrast, Pepck gene expression level was not different statistically among LacZ, 3A FoxO1, and 3A/LXXAA FoxO1–treated mice (Figure 10A). In order to confirm whether both 3A FoxO1 and 3A/LXXAA FoxO1 can bind to promoter regions of FoxO1 target genes, which include Igfbp-1, G6Pase, and Pepck, chromatin immunoprecipitation (ChIP) assay was performed using liver from Leprdb/db mice injected with adenovirus. Both 3A FoxO1 and 3A/LXXAA FoxO1 bound to promoter regions of Igfbp-1 and G6Pase (Figure 10B). Interestingly, although FoxO1 didn’t affect Pepck gene expression in liver in the present study, both 3A FoxO1 and 3A/LXXAA FoxO1 can bind to Pepck promoter region (Figure 10B). Furthermore, hepatic glycogen content of mice injected with 3A/LXXAA FoxO1 increased significantly compared with mice injected with the LacZ or 3A FoxO1 (0.0162 ± 0.0014 versus 0.0071 ± 0.0008 or 0.0065 ± 0.0009 mg glycogen/mg liver, respectively, P < 0.01 by 1-way ANOVA) (Figure 10C). These data suggest that 3A/LXXAA FoxO1 can bind to the promoter region of FoxO1 target genes in liver and that acute over-expression of 3A/LXXAA FoxO1 inhibits G6Pase gene expression and suppresses glucose production in liver from Leprdb/db mice.

Figure 10

Effects of overexpression of 3A/LXXAA FoxO1 in liver on gene expression and hepatic glycogen content. (A) Real-time PCR. Following overnight fasting, mRNA from the liver of Leprdb/db mice injected with adenovirus encoding LacZ (white bars), 3A FoxO1 (gray bars), or 3A/LXXAA FoxO1 (black bars) was isolated at day 5 after injection. Real-time PCR was performed with primers encoding the genes indicated. Data represent mean ± SEM of 3 independent experiments (n = 4 for each group). *Statistically significant difference between 3A FoxO1 and 3A/LXXAA FoxO1, P < 0.02 by 1-way ANOVA; **statistically significant difference between LacZ and 3A FoxO1, P < 0.05 by 1-way ANOVA. (B) ChIP assay of Igfbp-1 (lanes 1–3), G6Pase (lanes 4–6), and Pepck (lanes 7–9) promoter. ChIP assay was performed using liver sections from Leprdb/db mice injected with adenovirus encoding LacZ, 3A FoxO1, or 3A/LXXAA FoxO1 with the indicated antibody. The cross-linked DNA was amplified by PCR using a set of primers spanning the forkhead binding sites in the Igfbp-1, G6Pase, or Pepck promoter. (C) Hepatic glycogen content. Leprdb/db mice at day 4 after injection with adenovirus encoding LacZ (n = 3, white bar), 3A FoxO1 (n = 3, gray bar), or 3A/LXXAA FoxO1 (n = 3, black bar) were subjected to an overnight fast and killed the next morning for determination of glycogen levels in liver extracts. *Statistically significant difference between 3A and 3A/LXXAA, P < 0.02 by 1-way ANOVA; **statistically significant difference between LacZ and 3A/LXXAA, P < 0.05 by 1-way ANOVA.

Disruption of the LXXLL motif enhances acetylation of FoxO1 in liver. As described above, we demonstrated that disruption of the LXXLL motif inhibits binding of FoxO1 to Sirt1 and enhances acetylation of FoxO1 in SV40-transformed hepatocytes or HEK293 cells. In order to confirm effects of disruption of the LXXLL motif in FoxO1 on in vivo acetylation in liver, we performed immunoprecipitation of liver lysates injected with adenovirus encoding 3A FoxO1 or 3A/LXXAA FoxO1 using anti-Flag antibody and Western blotting using anti-acetylated lysine antibody. Although both 3A FoxO1 and 3A/LXXAA FoxO1 can be coimmunoprecipitated with endogenous Cbp and Pgc-1α, 3A/LXXAA FoxO1 binds to endogenous Sirt1 weakly compared with 3A FoxO1 (Figure 11). Consistent with data described above, acetylation of 3A/LXXAA FoxO1 is enhanced compared with that of 3A FoxO1 (Figure 11). These data suggest that disruption of the LXXLL motif in FoxO1 doesn’t affect binding to Cbp but inhibits interaction with Sirt1 and finally enhances acetylation of FoxO1 in vivo.

Figure 11

Acetylation of transduced FoxO1 in liver from Leprdb/db mice. We sacrificed Leprdb/db mice at day 5 after injection of adenovirus encoding 3A (lane 1) or 3A/LXXAA FoxO1 (lane 2) and lysed liver. Liver lysates were immunoprecipitated with anti-Flag monoclonal antibody and Western blotted with the indicated antibodies.

The LXXLL motif of murine forkhead transcription factor FoxO1 mediates Sirt1-dependent transcriptional activity (2024)

FAQs

What is the function of the FOXO1 gene? ›

FoxO1 is a cell-specific core transcription factor for endometrial remodeling and homeostasis during menstrual cycle and early pregnancy. FOXO1 inhibits prostate cancer cell proliferation via suppressing E2F1 activated NPRL2 expression. FOXO1 forkhead domain mutants in B-cell lymphoma lack transcriptional activity.

What is the mechanism of action of FOXO1? ›

To date, it has been identified that the acetylation of FOXO1 regulates its activity through three different mechanisms: (1) acetylation of lysine residues in the forkhead domain of FOXO1 reduces its binding ability to DNA; (2) FOXO1 acetylation promotes the phosphorylation of S256 of human FOXO1 mediated by AKT, which ...

What are forkhead box O transcription factors? ›

Forkhead (Foxo) Transcription Factors

The forkhead transcription factors (FOXO) have roles in cell cycle arrest and apoptosis, and, as demonstrated by the embryonic lethality of a Foxo1 knockout, these proteins are important in embryogenesis.

What do forkhead transcription factors do? ›

Forkhead transcription factor (FoxM1) is an important constituent of a myriad of biological activities, including regulation of G1/S and G2/M transitions of cell cycle, maintenance of mitotic spindle integrity, angiogenesis, metastasis, apoptosis, DNA damage repair, and tissue regeneration and deregulation of either of ...

What does FoxO1 do in the liver? ›

FOXO1 plays a major role in regulating the insulin response and the liver is one of its critical sites of action. The liver adapts to feeding through several insulin mediated events including increasing glucose uptake into hepatocytes, suppressing gluconeogenesis and glycogenolysis, and upregulating glycogen synthesis.

What does FoxO1 do in the muscles? ›

Previous studies have revealed that FoxO1 provokes myotube fusion of primary mouse myoblasts [21, 22]. But more recent researches support FoxO1 as an inhibitor of muscle differentiation [9, 13, 19]. Moreover, FoxO1 is believed to play a role in skeletal muscle fiber type specification [23–26].

What effect does insulin have on FOXO1? ›

FoxO's control of gluconeogenesis and glycogenolysis is mainly regulated by insulin signaling. For example, in the fed state, insulin levels increase and stimulate the PI3K/AKT pathway, which will export FoxO out of the nucleus and reduce FoxO's transcriptional activity (Figure 2).

What activates FOXO1? ›

Phosphatase and tensin hom*olog (PTEN) promotes FOXO activation and nuclear localization by inhibiting the activation of PI3K. In contrast to inhibition by insulin/IGF signaling, reactive oxygen species (ROS) promote nuclear localization and activation of FOXO by activating the c-Jun N-terminal kinase (JNK).

What is the response element of FOXO1? ›

FOXOs recognize two response elements: Daf-16 family member binding element (5′-GTAAA(T/C)AA) and insulin-responsive element (5′-(C/A)(A/C)AAA(C/T)AA). The core DNA sequence 5′-(A/C)AA(C/T)A-3′ is recognized by all FOX-family members.

What is Fox O transcription factors? ›

FOXO proteins are a subgroup of the Forkhead family of transcription factors. This family is characterized by a conserved DNA-binding domain (the 'Forkhead box', or FOX) and comprises more than 100 members in humans, classified from FOXA to FOXR on the basis of sequence similarity.

What do forkhead box proteins do? ›

Forkhead box (FOX) proteins are multifaceted transcription factors that are responsible for fine-tuning the spatial and temporal expression of a broad range of genes both during development and in adult tissues.

Is foxp1 a transcription factor? ›

It is primarily found within the nucleus, where it acts as a transcription regulator. Studies have shown FUBP1 can bind to the FUSE domain of c-Myc and other genes, increasing their expression.

What is the function of the foxa gene? ›

FOXA protein is required for the maintenance of enhancer activity, chromatin accessibility, nucleosome localization, and HNF4α binding. Therefore, the FOXA factor continues to function and maintain the liver gene regulatory network [10].

What is the meaning of forkhead? ›

: an arrowhead with two or occasionally three prongs pointing forward.

What is the role of FOXP3? ›

On the basis of its role in controlling gene activity, the FOXP3 protein is called a transcription factor. This protein is essential for the production and normal function of certain immune cells called regulatory T cells, which play an important role in preventing autoimmunity.

FOXO Transcription Factors: Their Clinical ...National Institutes of Health (NIH) (.gov)https://www.ncbi.nlm.nih.gov ›

The forkhead transcription factor family is characterized by a winged-helix DNA binding motif and the forkhead domain [1]. The mammalian forkhead transcription ...
The binding of 14-3-3s to FOXO transcription factors both prevents nuclear import and potentiates nuclear export, thereby blocking FOXO-mediated gene expression...
The forkhead box O (FoxO) transcription factors are known to be involved in many physiological and pathological processes including apoptosis, cell cycle arrest...

What is the function of the Fox 1 gene? ›

This gene provides instructions for making a protein called forkhead box G1. This protein plays an important role in brain development before birth, particularly in a region of the embryonic brain known as the telencephalon.

What is the function of the Fis1 gene? ›

GeneRIFs: Gene References Into Functions

Hepatic Fis1 regulates mitochondrial integrated stress response and improves metabolic homeostasis. Fis1 ablation in the male germline disrupts mitochondrial morphology and mitophagy, and arrests spermatid maturation.

What is the function of the FOXO pathway? ›

FOXO transcription factors and proteins are involved in cell death and survival and multiple signal transduction pathways which are essential for cell proliferation, angiogenesis and tumor suppressor gene functions.

What is the function of the C1q gene? ›

C1q directly promotes neuronal survival, thereby demonstrating new interactions between immune proteins and neuronal cells that may facilitate neuroprotection.

Top Articles
The Funniest LeBron James Memes and Tweets
LeBron James "You Are My Sunshine" Edits
Chren, inaugural chair of the Department of Dermatology, to step down
Brett Cooper Wikifeet
The Fappening Blgo
BEL MOONEY: Should I leave this boorish, bullying layabout?
Best Jewelry Laser Engraving Machine to Elevate Your Design
R/Sellingsunset
Www.craigslist.com Springfield Mo
Sir Mo Farah says 'sport saved me' after finishing final race of illustrious career at Great North Run
Tammi Light Obituary
U-Haul Customer Service & Support
102 Weatherby Dr Greenville Sc 29615
Bomei Massage
1 Bedroom Apartment For Rent Private Landlord
Childersburg Busted Mugshots
Making a Docker Container Use a VPN – Natural Born Coder
Jacy Nittolo Ex Husband
Elektrische Arbeit W (Kilowattstunden kWh Strompreis Berechnen Berechnung)
Party City Enfield Photos
Usccb 1 John 4
All Obituaries | Dante Jelks Funeral Home LLC. | Birmingham AL funeral home and cremation Gadsden AL funeral home and cremation
102Km To Mph
Top Songs On Octane 2022
A vintage funfair / fairground
How To Level Up Intellect Tarkov
Define Percosivism
Huadu Cn Fedex
Creator League Standings
352-730-1982
Pipa Mountain Hot Pot渝味晓宇重庆老火锅 Menu
Arsenal’s Auston Trusty: Inspired by Ronaldinho, World Cup dreams and Birmingham loan
Galen Rupp Net Worth
Natick Mall Directory Map
The Untold Truth Of 'Counting Cars' Star - Danny Koker
24 Hour Pharmacy Berkeley
Missing 2023 Showtimes Near Mjr Partridge Creek Digital Cinema 14
Craigslist Hawley Pa
Used Cars for Sale in Phoenix, AZ (with Photos)
8 Common Things That are 7 Centimeters Long | Measuringly
Experity Installer
Pokimane Boob Flash
Exploring The Craigslist Washington DC Marketplace - A Complete Overview
What is Landshark Beer?
2015 | Ducati 1299 Panigale S Test
Craigslist Free Stuff Columbus Ga
Lifetime Benefits Login
Minecraft Skin Tynker
Espn Masters Leaderboard
Fishing Report - Southwest Zone
La tarifa "Go Hilton" para los amigos y familiares de los miembros del equipo - Lo que debe saber
Highplainsobserverperryton
Latest Posts
Article information

Author: Carlyn Walter

Last Updated:

Views: 6485

Rating: 5 / 5 (70 voted)

Reviews: 85% of readers found this page helpful

Author information

Name: Carlyn Walter

Birthday: 1996-01-03

Address: Suite 452 40815 Denyse Extensions, Sengermouth, OR 42374

Phone: +8501809515404

Job: Manufacturing Technician

Hobby: Table tennis, Archery, Vacation, Metal detecting, Yo-yoing, Crocheting, Creative writing

Introduction: My name is Carlyn Walter, I am a lively, glamorous, healthy, clean, powerful, calm, combative person who loves writing and wants to share my knowledge and understanding with you.