It is well established that chronic glucocorticoid exposure causes hyperglycemia. While glucocorticoid receptor (GR) stimulates hepatic gluconeogenic gene transcription, additional mechanisms are activated by chronic glucocorticoid exposure to enhance gluconeogenesis. We found that chronic glucocorticoid treatment activated sphingosine-1-phosphate (S1P)–mediated signaling. Hepatic knockdown of hepatic S1P receptor 1 (S1PR1) had no effect on chronic glucocorticoid-induced glucose intolerance but elevated fasting plasma insulin levels. In contrast, hepatic S1PR3 knockdown exacerbated chronic glucocorticoid-induced glucose intolerance without affecting fasting plasma insulin levels. Finally, hepatic S1PR2 knockdown attenuated chronic glucocorticoid-induced glucose intolerance and reduced fasting plasma insulin levels. Here, we focused on dissecting the role of S1PR2 signaling in chronic glucocorticoid response on glucose homeostasis. We found that chronic glucocorticoid-induced hepatic gluconeogenesis, gluconeogenic gene expression, and GR recruitment to the glucocorticoid response elements (GREs) of gluconeogenic genes were all reduced in hepatic S1PR2 knockdown male mice. Hepatic S1PR2 knockdown also enhanced glucocorticoid suppression of RAR-related orphan receptor γ (RORγ) expression. Hepatic RORγ overexpression in hepatic S1PR2 knockdown mice restored glucocorticoid-induced glucose intolerance, gluconeogenic gene expression, and GR recruitment to their GREs. Conversely, RORγ antagonist and the reduction of hepatic RORγ expression attenuated such glucocorticoid effects. Thus, chronic glucocorticoid exposure induces an S1PR2-RORγ axis to cooperate with GR to enhance hepatic gluconeogenesis. Overall, this work provides novel mechanisms of and pharmaceutical targets against steroid-induced hyperglycemia.

Article Highlights
  • Chronic glucocorticoid exposure increased hepatic sphingosine-1-phosphate (S1P) levels in male mice, and reduced hepatic sphingosine-1-phosphate receptor 1, 2, and 3 (S1PR1–3) expression modulated glucocorticoid-regulated glucose homeostasis.

  • Hepatic S1PR2 knockdown blunted chronic glucocorticoid-induced glucose intolerance and gluconeogenesis and enhanced glucocorticoid-reduced RAR-related orphan receptor γ (RORγ) expression.

  • RORγ antagonist and hepatic RORγ knockdown reduced, whereas overexpressing RORγ in hepatic S1PR2 knockdown mice enhanced, glucocorticoid response on gluconeogenesis and potentiated glucocorticoid receptor occupancy at the glucocorticoid response elements of hepatic gluconeogenic genes to enhance their transcription.

  • Overall, upon chronic glucocorticoid exposure, hepatic S1PR2 signaling attenuates glucocorticoid-reduced RORγ expression to enhance glucocorticoid-induced gluconeogenesis in male mice.

It is well established that chronic and excess glucocorticoid exposure, through multiple mechanisms, including prolonged stress, long-term glucocorticoid use for treating inflammatory and autoimmune disease, depression, chronic stress, or Cushing syndrome, cause hyperglycemia and insulin resistance (13). Glucocorticoids act through their intracellular glucocorticoid receptor (GR), promoting transcription of genes encoding enzymes in hepatic gluconeogenesis, such as phosphoenolpyruvate carboxykinase (Pck1) and the catalytic subunit of glucose 6 phosphatase (G6pc1) (1,4,5). Studies have shown that glucocorticoids induce additional mechanisms to enhance hepatic glucose production. We previously showed that prolonged glucocorticoid treatment increases ceramide production, a process that requires angiopoietin-like 4 (Angptl4), a glucocorticoid-induced gene encoding a secreted protein that inhibits lipoprotein lipase activity and promotes adipose tissue lipolysis (68). Ceramide-initiated signaling in the liver activates protein phosphatase 2A (PP2A) and protein kinase Cζ (PKCζ) to suppress Akt, a key component in insulin signaling, causing insulin resistance and elevating hepatic gluconeogenesis (9).

In addition to ceramides, hepatic sphingosine-1-phosphate (S1P) levels are also augmented by glucocorticoid treatment (9). Ceramides can be converted to sphingosine, which can be further metabolized to S1P. S1P is a signaling molecule whose role in glucocorticoid-mediated metabolic regulation has not been reported. S1P can exert its actions intracellularly and extracellularly (10,11). Here, we explored the potential role of extracellular S1P in chronic glucocorticoid exposure–induced glucose homeostasis disorder. Extracellular S1P binds to its cognate membrane G-protein–coupled receptors S1PR1–5 to exert its actions (12). S1PR1–3 are highly expressed in the liver (13). We found that hepatic knockdown of S1PR1, 2, and 3 in male mice all modulated glucocorticoid-regulated glucose homeostasis. Specifically, reducing hepatic S1PR2 expression blunted glucocorticoid-induced glucose intolerance and hepatic gluconeogenesis. RNA sequencing (RNA-seq) found that glucocorticoid suppression of RAR-related orphan receptor γ (Rorc) (called RORγ henceforward) gene expression was enhanced in glucocorticoid-treated hepatic S1PR2 knockdown mouse liver. Because RORγ has been previously shown to positively regulate hepatic gluconeogenic gene transcription (14), we further analyzed its role in chronic glucocorticoid exposure–enhanced gluconeogenesis.

Animals

Andras Nagy (Samuel Lunenfeld Research Institute, Mount Sinai Hospital) and Jeff Gordon (Washington University) provided Angptl4+/+ and Angptl4−/− mixed C57BL/6J:129 SV background male mice, which were cohoused as previously described (15). All experiments were approved by the University of California, Berkeley (AUP-2014-07-6617). This study was performed entirely in male mice.

Adeno-Associated Virus

Adeno-associated virus serotype 8 (AAV8) expressing shRNA scramble, shRNA targeting S1PR1, S1PR2, S1PR3, RORγ, and green fluorescent protein (GFP), and mouse RORγ cDNA were purchased from Vector Biolabs (Malvern, PA) and were injected in the tail vein (3 × 1011 genome copies/mouse). The shRNA sequences are provided in Supplementary File 1.

Dexamethasone Water Supplementation

Dexamethasone (Dex) water 0.84 mg/kg body weight (PHR 1768; Sigma) was previously described (15). AAV mice were switched to Dex-supplemented water 2 days after injection for 4 weeks.

Glucose, Insulin, and Pyruvate Tolerance Tests

Mice were fasted for 16 h for intraperitoneal glucose tolerance tests (IPGTTs) (1 mg/kg d-glucose, 50-99-7; Sigma) and pyruvate tolerance tests (PTTs) (2 mg/kg sodium pyruvate, P2256; Sigma) and fasted 2 h for insulin tolerance tests (1 unit/kg of insulin, 5 mL I0516; Sigma). A Contour Next glucometer was used to measure blood glucose.

Blood and Tissue Collection

Fasted blood was collected in EDTA tubes (16.44.100; Sarstedt). CO2 euthanasia was performed at 3–5 h into the light cycle (10:00 a.m.–12:00 p.m.). Tissues were snap frozen in liquid nitrogen and stored at −80°C.

ELISA

Plasma insulin, liver phosphorylated Akt at serine 473 (pAkt) and Akt levels, and liver and plasma S1P levels were measured using ELISA. The detailed protocols are provided in Supplementary File 1.

RNA-Seq

RNA-seq was performed by BGI Americas (Cambridge, MA). The protocol of the analysis of RNA-seq is provided in Supplementary File 1.

Western Blot

Tissues were prepared for SDS-PAGE as previously described (15). Antibody information is provided in Supplementary File 1. Images were analyzed using ImageJ software.

Gene Expression

Gene expression was performed as previously described (15), and Rpl19 was used for internal normalization. Primers are listed in Table 1.

Table 1

Primers

Primer nameForwardReverse
Quantitative PCR   
 rpl19 tcc ttg gtc tta gac ctg cg atg gag cac atc aca ag c 
 pck1 ctg cat aac ggt ctg gac ttc cag caa ctg ccc gta ctc c 
 g6pc1 gac cat aac ata gta tac acc tgc tgc gac cat aac ata gta tac acc tgc tgc 
ChIP   
 rpl19 tcc ttg gtc tta gac ctg cg atg gag cac atc aca ag c 
 Pck1-GBR gca ggc tct tgc ctt aat tg ggt gtt ttg aca acc agc ag 
 G6pc1-GBR tgc aag agt cat ggt tga aac ag cac act gct tga cag tgc ta 
Primer nameForwardReverse
Quantitative PCR   
 rpl19 tcc ttg gtc tta gac ctg cg atg gag cac atc aca ag c 
 pck1 ctg cat aac ggt ctg gac ttc cag caa ctg ccc gta ctc c 
 g6pc1 gac cat aac ata gta tac acc tgc tgc gac cat aac ata gta tac acc tgc tgc 
ChIP   
 rpl19 tcc ttg gtc tta gac ctg cg atg gag cac atc aca ag c 
 Pck1-GBR gca ggc tct tgc ctt aat tg ggt gtt ttg aca acc agc ag 
 G6pc1-GBR tgc aag agt cat ggt tga aac ag cac act gct tga cag tgc ta 

Liver Chromatin Immunoprecipitation

Liver chromatin immunoprecipitation (ChIP) was performed as previously described (15) with the following modifications: 10 strokes with dounce homogenizer, sonication 60% amplitude (5 min total), and 10-s bursts (40-s rests). Four micrograms of the following antibodies were used: purified rabbit IgG (whole molecule) control (A01008; GeneScript), GR (IA-1 [a polyclonal rabbit antibody against human GR amino acids 84–112 QPDLSKAVSLSMGLYMGETETKVMGNDLG]; a gift from Dr. Miles Pufall, University of Iowa), and RORγ (14-6988-82; Invitrogen). Forty microliters of 25% protein A/G sepharose beads (Santa Cruz Biotechnology) were used (15).

Cell Culture and Transfection

H4IIE cells were cultured in DMEM with 10% FBS and serum starved overnight before treatment. For 4 h, 100 nmol/L Dex (D1756; Sigma), 100 nmol/L Ly3214996 (501932152; MedChemExpress), and/or 2.5 μmol/L XY018 were added. During the last 2 h, 0.5 μmol/L CYM-5520 was added (5418/10; R&D Systems). For transfection, H4IIE cells were plated at 0.5 × 106 cells/well and cultured in DMEM containing 5% FBS. The detailed protocol is provided in Supplementary File 1.

Quantification and Statistical Analysis

GraphPad Prism 9.0 software was used for the data analysis. Data are presented as mean with SD and SEM using Student t test, one-way ANOVA, and two-way ANOVA with Fisher least significant difference post hoc and Benjamini and Hochberg false discovery rate correction tests as indicated.

Data and Resource Availability

All data generated or analyzed during this study are included in the published article and its online supplementary files. RNA-seq data are deposited in the National Center for Biotechnology Information Gene Expression Omnibus database (accession no. GSE150626).

Reducing Hepatic S1PR1–3 Signaling Modulated Glucocorticoid-Regulated Glucose Homeostasis

Wild-type (WT) male mice infected with AAV8 expressing scramble shRNA (AAV8-shScr) were treated with or without Dex (a synthetic glucocorticoid) for 4 weeks. Dex treatment resulted in glucose intolerance (Fig.1A). As previously reported, hepatic, but not plasma, S1P levels were elevated by Dex treatment (9) (Fig.1B). To learn whether extracellular S1P is involved in glucocorticoid-regulated glucose homeostasis, WT mice were infected with AAV8 expressing shRNA targeting S1PR1 (AAV8-shS1PR1), AAV8-shS1PR2, or AAV8-shS1PR3. These mice, along with AAV8-shScr mice, were treated with Dex for 4 weeks. The expression of S1PR1, 2, and 3 was reduced in the liver but not in gastrocnemius muscle, inguinal white adipose tissue, or brown adipose tissue (Fig.1C and Supplementary Fig. 1A). Hepatic S1PR1 knockdown had no effect, whereas hepatic S1PR3 knockdown further exacerbated Dex-induced glucose intolerance (Fig.1D). In contrast, hepatic S1PR2 knockdown blunted the Dex response (Fig.1D). Similarly, hepatic S1PR1 knockdown had no effect on Dex-induced fasting hyperglycemia, while hepatic S1PR3 knockdown worsened and hepatic S1PR2 knockdown improved Dex-induced fasting hyperglycemia (Fig.1E). Compared with AAV8-shScr mice, fasting plasma insulin levels were lower in AAV8-shS1PR2 mice and higher in AAV8-shS1PR1 mice, but fasting insulin levels were not different between AAV8-shS1PR3 and AAV8-shScr mice (Fig.1F). Overall, these results demonstrate that reducing hepatic S1PR1, 2, and 3 expression all affected glucose homeostasis in the presence of Dex. Importantly, reducing their hepatic expression did not affect plasma ALT levels (Supplementary Fig. 1B). Thus, hepatic S1PR1, 2, and 3 knockdowns did not cause liver injury. Of note, we found that without Dex treatment, AAV8-shS1PR2 mice were also more glucose tolerant than AAV8-shScr mice (Fig.1G), though there were no differences in fasting plasma insulin levels (Fig.1H). Next, we specifically investigated the mechanism underlying the effects of altered hepatic S1PR2 signaling in the context of chronic glucocorticoid exposure–induced glucose disorder.

Figure 1

Hepatic S1PR2 knockdown but not S1PR1 and S1PR3 improved chronic glucocorticoid exposure–induced glucose intolerance. A: IPGTT and relative area under the curve (AUC) of WT male AAV8-shScr mice treated without Dex and with Dex (0.84 mg/kg body weight) for 4 weeks after a 16-h fast (n = 7–12). **P < 0.005 using Student t test with Welch correction. Error bars represent SD and SEM for the tolerance test. B: Hepatic and plasma S1P levels in WT male mice undergoing Dex treatment (4 weeks) (n = 6–10). C: Western blots of S1PR1, S1PR2, and S1PR3 in AAV8-shScr, AAV8-shS1PR1, AAV8-shS1PR2, and AAV8-shS1PR3 male mouse liver. Images are from the same blot, and groups were cropped for ease of visualization (n = 3–9). D: IPGTT and relative AUC of AAV8-shScr, AAV8-shS1PR1, AAV8-shS1PR2, and AAV8-shS1PR3 mice treated with Dex (0.84 mg/kg body weight) for 4 weeks after 16-h fast (n = 14–23). E: Fasting blood glucose levels of AAV8-shScr, AAV8-shS1PR1, AAV8-shS1PR2, and AAV8-shS1PR3 mice treated with Dex after 16-h fast. F: Plasma insulin of AAV8-shScr, AAV8-shS1PR1, AAV8-shS1PR2, and AAV8-shS1PR3 mice treated with Dex after 16-h fast (n = 7–13). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 using ANOVA with Welch correction. Error bars represent SEM for the tolerance test and SD for the rest. G: IPGTT and relative AUC of AAV8-shScr and AAV8-shS1PR2 without Dex (n = 7). H: Plasma insulin of AAV8-shScr and AAV8-shS1PR2 mice without Dex after 16-h fast (n = 7). **P < 0.005 using Student t test with Welch correction. Error bars represent SEM for the tolerance test and SD for the rest.

Figure 1

Hepatic S1PR2 knockdown but not S1PR1 and S1PR3 improved chronic glucocorticoid exposure–induced glucose intolerance. A: IPGTT and relative area under the curve (AUC) of WT male AAV8-shScr mice treated without Dex and with Dex (0.84 mg/kg body weight) for 4 weeks after a 16-h fast (n = 7–12). **P < 0.005 using Student t test with Welch correction. Error bars represent SD and SEM for the tolerance test. B: Hepatic and plasma S1P levels in WT male mice undergoing Dex treatment (4 weeks) (n = 6–10). C: Western blots of S1PR1, S1PR2, and S1PR3 in AAV8-shScr, AAV8-shS1PR1, AAV8-shS1PR2, and AAV8-shS1PR3 male mouse liver. Images are from the same blot, and groups were cropped for ease of visualization (n = 3–9). D: IPGTT and relative AUC of AAV8-shScr, AAV8-shS1PR1, AAV8-shS1PR2, and AAV8-shS1PR3 mice treated with Dex (0.84 mg/kg body weight) for 4 weeks after 16-h fast (n = 14–23). E: Fasting blood glucose levels of AAV8-shScr, AAV8-shS1PR1, AAV8-shS1PR2, and AAV8-shS1PR3 mice treated with Dex after 16-h fast. F: Plasma insulin of AAV8-shScr, AAV8-shS1PR1, AAV8-shS1PR2, and AAV8-shS1PR3 mice treated with Dex after 16-h fast (n = 7–13). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 using ANOVA with Welch correction. Error bars represent SEM for the tolerance test and SD for the rest. G: IPGTT and relative AUC of AAV8-shScr and AAV8-shS1PR2 without Dex (n = 7). H: Plasma insulin of AAV8-shScr and AAV8-shS1PR2 mice without Dex after 16-h fast (n = 7). **P < 0.005 using Student t test with Welch correction. Error bars represent SEM for the tolerance test and SD for the rest.

Close modal

Because hepatic S1P induction by Dex depends on the presence of ANGPTL4 in male mice (9), we tested whether the effects of S1PR2 knockdown on glucose tolerance also requires ANGPTL4. As previously reported, Dex-induced glucose intolerance was compromised in Angptl4−/− mice (9) (Supplementary Fig. 2). In contrast to the observations in WT mice, hepatic S1PR2 knockdown did not affect glucose tolerance in Dex-treated Angptl4−/− mice (Supplementary Fig. 2), and a trend toward improved glucose tolerance was observed in Angptl4−/− mice not treated with Dex (Supplementary Fig. 2). These results demonstrate that while ANGPTL4 is required for the effects of S1PR2 signaling in the context of chronic Dex treatment–induced glucose intolerance, it perhaps is not required for the effects of S1PR2 signaling with respect to the regulation of basal glucose tolerance.

Hepatic S1PR2 Knockdown Did Not Affect Insulin Tolerance but Reduced Gluconeogenesis in Dex-Treated Mice

We performed an insulin tolerance test on AAV8-shScr and AAV8-shS1PR2 mice treated with or without Dex. Without Dex, these mice had similar insulin tolerance (Fig.2A). Dex treatment induced insulin intolerance, but, interestingly, hepatic S1PR2 knockdown did not affect Dex-induced insulin intolerance (Fig.2A). This observation is consistent with analyses of the ratio of pAkt and total Akt (pAkt/Akt), an indicator of insulin signaling activity (16) in liver, gastrocnemius muscle, and epididymal white adipose tissue in AAV8-shScr and AAV8-shS1PR2 mice treated with Dex and injected with insulin or PBS. pAkt/Akt levels were similar in all three tissues examined between AAV8-shScr and AAV8-shS1PR2 mice (Fig.2B). Thus, reducing hepatic S1PR2 signaling did not affect Dex-modulated insulin signaling in these tissues.

Figure 2

Hepatic S1PR2 knockdown reduced Dex-induced pyruvate tolerance but not insulin intolerance. A: Insulin tolerance test of AAV8-shScr and AAV8-shS1PR2 male mice treated with or without Dex for 4 weeks after 2-h fast (n = 6–12). B: pAkt/Akt ratio determined through ELISA of the liver, epidydimal white adipose tissue (eWAT), and gastrocnemius muscle (GA) in AAV8-shScr and AAV8-shS1PR2 mice treated with Dex and injected with or without insulin (n = 4–5). C: PTT in AAV8-shScr and AAV8-shS1PR2 mice treated with or without Dex after 16-h fast (n = 6–10). D: Gene expression in AAV8-shScr and AAV8-shS1PR2 mice treated with or without Dex (n = 4–5). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 using two-way ANOVA with Benjamini and Hochberg false discovery rate correction. Error bars represent the SD. E: GR ChIP on Pck1 and G6pc1 (n = 3–5). *P < 0.05, **P < 0.01, using a two-way ANOVA with Fisher least significant difference post hoc test. Error bars represent SEM for the tolerance tests and SD for the rest. AUC, area under the curve.

Figure 2

Hepatic S1PR2 knockdown reduced Dex-induced pyruvate tolerance but not insulin intolerance. A: Insulin tolerance test of AAV8-shScr and AAV8-shS1PR2 male mice treated with or without Dex for 4 weeks after 2-h fast (n = 6–12). B: pAkt/Akt ratio determined through ELISA of the liver, epidydimal white adipose tissue (eWAT), and gastrocnemius muscle (GA) in AAV8-shScr and AAV8-shS1PR2 mice treated with Dex and injected with or without insulin (n = 4–5). C: PTT in AAV8-shScr and AAV8-shS1PR2 mice treated with or without Dex after 16-h fast (n = 6–10). D: Gene expression in AAV8-shScr and AAV8-shS1PR2 mice treated with or without Dex (n = 4–5). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 using two-way ANOVA with Benjamini and Hochberg false discovery rate correction. Error bars represent the SD. E: GR ChIP on Pck1 and G6pc1 (n = 3–5). *P < 0.05, **P < 0.01, using a two-way ANOVA with Fisher least significant difference post hoc test. Error bars represent SEM for the tolerance tests and SD for the rest. AUC, area under the curve.

Close modal

We next performed a PTT to monitor the hepatic gluconeogenic capacity of AAV8-shScr and AAV8-shS1PR2 mice treated with or without Dex. Dex treatment increased fasting glucose levels and gluconeogenesis in AAV8-shScr mice (Fig.2C). Hepatic S1PR2 knockdown reduced gluconeogenic capacity in both control and Dex-treated mice (Fig.2C). Gene expression analysis revealed that hepatic S1PR2 knockdown reduced the expression of gluconeogenic genes Pck1 and G6pc1 (Fig.2D). Notably, without Dex, Pck1 and G6pc1 expressions were similar between these mice (Fig.2D). These indicate that S1PR2 signaling is involved in Dex-stimulated and basal gluconeogenesis. However, the mechanisms underlying S1PR2’s role in these two processes are different.

One potential mechanism to explain the lower induction of gluconeogenic genes by Dex in hepatic S1PR2 knockdown mouse liver would be through reduced Dex-induced recruitment of GR to the GREs of gluconeogenic genes. We performed GR ChIP to examine this model. Without Dex, GR occupancy on the GREs was minimal in AAV8-shScr and AAV8-shS1PR2 mouse liver (Fig.2E). Dex treatment significantly increased GR recruitment to the GREs of Pck1 (4) and G6pc1 (17) genes in AAV8-shScr mouse liver (Fig.2E). However, GR recruitment was significantly lower in Dex-treated AAV-shS1PR2 mice than Dex-treated AAV8-shScr mice (Fig.2E).

Hepatic S1PR2 Knockdown Reduced the Expression of RORγ

To probe potential mechanisms for the effects of S1PR2 knockdown, we performed RNA-seq to identify differentially expressed genes in Dex-treated AAV8-shScr versus Dex-treated AAV8-shS1PR2 mouse liver. Using a twofold difference and adjusted P value of 0.05 as the cutoff, we found that 238 genes had lower expression, and 182 genes were higher in Dex-treated AAV8-shS1PR2 versus Dex-treated AAV8-shScr mouse liver (Fig.3A). A list of these genes is presented in Supplementary File 2. Gluconeogenic genes were among the list of downregulated genes in AAV8-shS1PR2 liver, including G6pc1, fructose bisphosphatase 1 (Fbp1), and tyrosine aminotransferase (Tat). Pck1 was not scored based on the cutoff standard, but conventional real-time PCR analysis clearly showed a lower Pck1 expression in the liver of Dex-treated AAV8-shS1PR2 mice (Fig.2D). Moreover, RNA-seq also found that S1PR2 knockdown reduced the expression of Rorc, which encodes a nuclear factor that can potentiate Pck1 and G6pc1 transcription (14). Gene ontology analysis found that genes encoding the epoxygenase p450 pathway, lipid, steroid, acetyl-CoA, and glycolytic metabolic processes were highly represented among S1PR2 knockdown–reduced genes (Fig.3B). In contrast, genes involved in immune response, antiviral defense, estrogen responses, lipid transport, and lipoprotein metabolism were highly represented among S1PR2 knockdown–induced genes (Fig.3B). Thus, S1PR2 signaling participates in many facets of glucocorticoid actions. The role of RORγ in glucocorticoid-regulated hepatic gluconeogenesis was of particular interest since this had not been previously reported. Immunoblotting showed that without Dex, S1PR2 knockdown did not affect RORγ expression (Fig.3C). While Dex treatment reduced RORγ expression in the liver AAV8-shScr mice, Dex suppression of RORγ expression was even more pronounced in the liver of AAV8-shS1PR2 mice (Fig.3C). Similarly, nuclear levels of RORγ were suppressed in Dex-treated AAV8-shScr and further suppressed in AAV8-shS1PR2 mice (Supplementary Fig. 3A). RORα has also been shown to activate the Pck1 gene (18,19). However, protein levels of RORα were not affected by hepatic S1PR2 knockdown (Supplementary Fig. 3B).

Figure 3

Hepatic knockdown of S1PR2 reduces RORγ expression. A: MA plot of RNA-seq analysis of genes in the liver of Dex-treated AAV8-shScr and Dex-treated AAV8-shS1PR2 male mice with twofold difference and adjusted P value of 0.05 as the cutoff. B: Gene ontology analysis of hepatic genes induced and reduced by hepatic S1PR2 knockdown. C: Western blot for RORγ in AAV8-shScr and AAV8-shS1PR2 mice treated with and without Dex. Relative expression was normalized to GAPDH using ImageJ software (n = 3). *P < 0.05, ****P < 0.0001 using two-way ANOVA with Fisher least significant difference post hoc test. Error bars represent the SD. no-DEGs, number of differentially expressed genes.

Figure 3

Hepatic knockdown of S1PR2 reduces RORγ expression. A: MA plot of RNA-seq analysis of genes in the liver of Dex-treated AAV8-shScr and Dex-treated AAV8-shS1PR2 male mice with twofold difference and adjusted P value of 0.05 as the cutoff. B: Gene ontology analysis of hepatic genes induced and reduced by hepatic S1PR2 knockdown. C: Western blot for RORγ in AAV8-shScr and AAV8-shS1PR2 mice treated with and without Dex. Relative expression was normalized to GAPDH using ImageJ software (n = 3). *P < 0.05, ****P < 0.0001 using two-way ANOVA with Fisher least significant difference post hoc test. Error bars represent the SD. no-DEGs, number of differentially expressed genes.

Close modal

RORγ Was Involved in Dex-Induced Gluconeogenic Gene Transcription

To explore whether RORγ is involved in Dex-regulated gluconeogenic gene expression, H4IIE rat hepatoma cells were treated with 1) DMSO (vehicle), 2) Dex, 3) CYM-5520 (an agonist of S1PR2) (20), 4) Dex plus CYM-5520, 5) XY018 (an RORγ antagonist) (21), 6) Dex plus XY108, 7) CYM-5520 plus XY018, and 8) Dex plus CYM-5520 plus XY108 for 4 h, then Pck1 and G6pc1 expression were analyzed. Pck1 and G6pc1 expression were induced by Dex but not by CYM-5520 (Fig.4A). However, CYM-5520 enhanced Dex-induced Pck1 and G6pc1 expression (Fig.4A). XY018 did not affect basal Pck1 and G6pc1 expression but suppressed both Dex and Dex plus CYM-5520–induced Pck1 and G6pc1 expression (Fig.4A). Thus, RORγ activity was required for maximal Dex and S1PR2 signaling response on gluconeogenic gene expression.

Figure 4

RORγ antagonist reduces Dex-activated hepatic gluconeogenesis. A: RORγ antagonist (XY018) reduces Dex-induced gluconeogenic gene expression. These experiments were repeated three times, and each time had triplicates: 100 nmol/L Dex and/or 0.5 μmol/L S1PR2 agonist CYM-5520 and/or 2.5 μmol/L RORγ antagonist (XY018). B: XY018 5 mg/kg was intraperitoneally injected daily in male mice for the last 2 weeks of the 4-week Dex treatment and an IPGTT was done after a 16-h fast (n = 3–4). C: XY018 5 mg/kg was intraperitoneally injected twice per every 3 days of the last 2 weeks of the 4-week Dex treatment, and a PTT was done after a 16-h fast (n = 5–8). D: Hepatic gluconeogenic gene expression WT mice treated without Dex (−) or with 0.84 mg/kg body weight Dex (+) and with PBS or 5 mg/kg RORγ antagonist (XY018) (n = 3–4). E: The location of mouse Pck1 and G6pc1 GREs and ROREs and ChIP of GR and RORγ on the GRE-RORE region of mouse Pck1 and G6pc1 genes of liver of WT mice treated without Dex (−) or with Dex (+) and with PBS or 5 mg/kg RORγ antagonist (XY018) (n = 3–4). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 by two-way ANOVA with Fisher least significant difference post hoc test. Error bars represent SEM for the tolerance tests and SD for the rest. AUC, area under the curve.

Figure 4

RORγ antagonist reduces Dex-activated hepatic gluconeogenesis. A: RORγ antagonist (XY018) reduces Dex-induced gluconeogenic gene expression. These experiments were repeated three times, and each time had triplicates: 100 nmol/L Dex and/or 0.5 μmol/L S1PR2 agonist CYM-5520 and/or 2.5 μmol/L RORγ antagonist (XY018). B: XY018 5 mg/kg was intraperitoneally injected daily in male mice for the last 2 weeks of the 4-week Dex treatment and an IPGTT was done after a 16-h fast (n = 3–4). C: XY018 5 mg/kg was intraperitoneally injected twice per every 3 days of the last 2 weeks of the 4-week Dex treatment, and a PTT was done after a 16-h fast (n = 5–8). D: Hepatic gluconeogenic gene expression WT mice treated without Dex (−) or with 0.84 mg/kg body weight Dex (+) and with PBS or 5 mg/kg RORγ antagonist (XY018) (n = 3–4). E: The location of mouse Pck1 and G6pc1 GREs and ROREs and ChIP of GR and RORγ on the GRE-RORE region of mouse Pck1 and G6pc1 genes of liver of WT mice treated without Dex (−) or with Dex (+) and with PBS or 5 mg/kg RORγ antagonist (XY018) (n = 3–4). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 by two-way ANOVA with Fisher least significant difference post hoc test. Error bars represent SEM for the tolerance tests and SD for the rest. AUC, area under the curve.

Close modal

RORγ Antagonist Improved Dex-Induced Glucose Intolerance and Hepatic Gluconeogenesis

We next tested RORγ’s role in Dex-induced glucose intolerance in vivo. WT male mice were treated with or without Dex for 4 weeks. For the final 2 weeks, one-half of each group was treated with PBS or XY018. Dex-treated mice with PBS injections were more glucose intolerant than mice that only received PBS injections (Fig.4B). XY018 treatment alleviated Dex-induced glucose intolerance but did not affect basal glucose tolerance (Fig.4B). PTTs showed that Dex treatment augmented glucose production in WT mice, whereas XY018 alone had no effect (Fig.4C). Dex-treated mice that also received XY018 had a lower gluconeogenic capacity than mice treated with Dex alone (Fig.4C). Not surprisingly, XY018 reduced Pck1 and G6pc1 expression in Dex-treated WT mice (Fig.4D). XY018 did not affect Pck1 and G6pc1 gene expression in mice not treated with Dex (Fig.4D). These results were consistent with the results observed in H4IIE cells.

We performed liver ChIP in these mice to monitor the GR and RORγ occupancy at the RORγ response elements (ROREs) (18) and GREs (4) of Pck1 and G6pc1 (Fig.4E). Without treatment, GR was modestly recruited to the GREs (Fig.4E). This occupancy was likely due to endogenous corticosterone. RORγ, however, was not found at the ROREs (Fig.4E). Dex treatment increased both GR and RORγ recruitment to the GRE-RORE regions of these two genes (Fig.4E). XY018 did not affect basal GR and RORγ occupancy but attenuated Dex-induced GR and RORγ recruitment (Fig.4E).

Hepatic RORγ Knockdown Improved Dex-Induced Glucose Intolerance and Reduced Gluconeogenesis

WT mice were infected with AAV8-shScr or shRNA targeting Rorc (AAV8-shRORγ) and treated with Dex. Immunoblotting confirmed a significantly reduced RORγ expression in the liver, but not brown adipose tissue or gastrocnemius muscle, in AAV8-shRORγ mice (Fig.5A). Dex-treated AAV8-shRORγ mice showed a trend toward better glucose tolerance than AAV8-shScr mice (Fig.5B). Moreover, PTT showed that hepatic gluconeogenesis was significantly lower in Dex-treated AAV8-shRORγ mice (Fig.5C). Fasting plasma insulin levels were also lower in AAV8-shRORγ mice (Fig.5D). Interestingly, while Pck1 expression was reduced in livers of AAV8-shRORγ mice, there were no differences in G6pc1 expression (Fig.5E). Nonetheless, the results from hepatic RORγ knockdown mice generally agreed with the observation from the RORγ antagonist studies in Fig. 4. Notably, hepatic RORγ knockdown did not affect plasma ALT levels (Supplementary Fig. 4B).

Figure 5

Hepatic RORγ knockdown reduced Dex-induced gluconeogenesis in male mice. A: Western blots of RORγ in AAV8-shScr and AAV8-shRORγ liver, brown adipose tissue (BAT), and gastrocnemius muscle (GA). Relative expression normalized to GAPDH using ImageJ software (n = 3–7). B: IPGTT and relative area under the curve (AUC) of AAV8-shScr and AAV8-shRORγ mice treated with Dex after a 16-h fast (n = 9). C: PTT and relative AUC of AAV8-shScr and AAV8-shRORγ mice treated with Dex after a 16-h fast (n = 12–15). D: Plasma insulin levels of AAV8-shScr and AAV8-shRORγ mice treated with Dex after a 16-h fast (n = 11–13). E: Hepatic gluconeogenic gene expression AAV8-shScr and AAV8-shRORγ mice treated with Dex (n = 6–8). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 by Student t test with Welch correction. The error bars represent the SD. F: Rat Pck1 promoter luciferase activity in H4IIE cells transfected with porcine cytomegalovirus (pCMV) (control), and 100 ng, 200 ng, or 300 ng hRORγ treated with or without 500 nmol/L Dex (n = 4). G: Rat Pck1 promoter luciferase activity in H4IIE cells transfected with 300 ng hRORγ, treated with or without 500 nmol/L Dex, and treated with DMSO, 50 nmol/L, or 100 nmol/L RORγ agonist (LYC-55716) (n = 4). *P < 0.05, **P < 0.01, ***P < 0.001 by two-way ANOVA with Fisher least significant difference post hoc test. Error bars represent SEM for the tolerance tests and SD for the rest.

Figure 5

Hepatic RORγ knockdown reduced Dex-induced gluconeogenesis in male mice. A: Western blots of RORγ in AAV8-shScr and AAV8-shRORγ liver, brown adipose tissue (BAT), and gastrocnemius muscle (GA). Relative expression normalized to GAPDH using ImageJ software (n = 3–7). B: IPGTT and relative area under the curve (AUC) of AAV8-shScr and AAV8-shRORγ mice treated with Dex after a 16-h fast (n = 9). C: PTT and relative AUC of AAV8-shScr and AAV8-shRORγ mice treated with Dex after a 16-h fast (n = 12–15). D: Plasma insulin levels of AAV8-shScr and AAV8-shRORγ mice treated with Dex after a 16-h fast (n = 11–13). E: Hepatic gluconeogenic gene expression AAV8-shScr and AAV8-shRORγ mice treated with Dex (n = 6–8). *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001 by Student t test with Welch correction. The error bars represent the SD. F: Rat Pck1 promoter luciferase activity in H4IIE cells transfected with porcine cytomegalovirus (pCMV) (control), and 100 ng, 200 ng, or 300 ng hRORγ treated with or without 500 nmol/L Dex (n = 4). G: Rat Pck1 promoter luciferase activity in H4IIE cells transfected with 300 ng hRORγ, treated with or without 500 nmol/L Dex, and treated with DMSO, 50 nmol/L, or 100 nmol/L RORγ agonist (LYC-55716) (n = 4). *P < 0.05, **P < 0.01, ***P < 0.001 by two-way ANOVA with Fisher least significant difference post hoc test. Error bars represent SEM for the tolerance tests and SD for the rest.

Close modal

To further test the functional interaction between RORγ and GR, a rat Pck1 promoter luciferase reporter was transfected into H4IIE cells along with expression plasmids of human GR and human RORγ (hRORγ). Without hRORγ overexpression, Dex treatment induced the reporter activity (Fig.5F). hRORγ overexpression did not affect the reporter activity. However, transfecting 300 ng of hRORγ expression plasmid significantly potentiated the Dex response (Fig.5F). We tested whether an RORγ agonist can further potentiate this response. In separate experiments, H4IIE cells were transfected with a rat Pck1 promoter reporter, expression plasmids of human GR, and 300 ng hRORγ. Twenty-four hours after transfection, cells were treated with DMSO, Dex, LYC-55716 (an RORγ agonist) (22), and Dex plus LYC-55716. Interestingly, 100 nmol/L LYC-55716 treatment alone showed a trend toward increased basal reporter activity. Both 50 nmol/L and 100 nmol/L LYC-55716 significantly enhanced the Dex response (Fig.5G). Overall, these results suggest that RORγ enhances GR activation of rat Pck1 promoter.

Hepatic RORγ Overexpression Restored Glucose Intolerance and Gluconeogenesis Induced by Dex Treatment in AAV8-shS1PR2 Mice

Next, we tested whether RORγ overexpression in hepatic S1PR2 knockdown mice restored Dex-induced glucose intolerance and gluconeogenesis. Male WT mice infected with AAV8-shS1PR2 were treated with or without Dex. One week later, they were infected with AAV8 expressing GFP (AAV8-shS1PR2-GFP) or mouse RORγ (AAV8-shS1PR2-RORγ). Dex treatment was continued for 3 more weeks, and IPGTT and PTT were performed. Western blotting validated liver specificity of RORγ overexpression (Fig.6A). Dex treatment caused glucose intolerance and increased gluconeogenesis in AAV8-shS1PR2-GFP mice (Fig.6B and C). Overexpression of RORγ further enhanced these phenotypes (Fig.6B and C). Hepatic Pck1 and G6pc1 expression was also higher in Dex-treated AAV8-shS1PR2-RORγ mice (Fig.6D). Without Dex, RORγ overexpression did not increase glucose intolerance or gluconeogenesis of AAV8-shS1PR2 mice (Fig.6B and C). Thus, RORγ is not involved in the regulation of basal glucose tolerance and gluconeogenesis regulated by S1PR2 signaling.

Figure 6

RORγ overexpression in AAV8-shS1PR2 male mice worsens 4 weeks Dex treatment–induced glucose and pyruvate tolerance. A: RORγ protein expression by Western blot in liver, gastrocnemius muscle (GA), and epididymal white adipose tissue (eWAT) (n = 4). **P < 0.01 by Student t test. B: IPGTT after a 16-h fast. C: PTT after a 16-h fast. D: Hepatic gluconeogenic gene expression. E: ChIP of GR and RORγ on the GRE-RORE region of mouse Pck1 and G6pc1 genes of the liver of AAV8-shS1PR2 mice overexpressing IgG, GR, or RORγ and treated without Dex (−) or with Dex (+) (n = 3–6). *P < 0.05, **P < 0.01, ***P < 0.001, using two-way ANOVA with Fisher least significant difference post hoc test. Error bars represent SEM for the tolerance tests and SD for the rest.

Figure 6

RORγ overexpression in AAV8-shS1PR2 male mice worsens 4 weeks Dex treatment–induced glucose and pyruvate tolerance. A: RORγ protein expression by Western blot in liver, gastrocnemius muscle (GA), and epididymal white adipose tissue (eWAT) (n = 4). **P < 0.01 by Student t test. B: IPGTT after a 16-h fast. C: PTT after a 16-h fast. D: Hepatic gluconeogenic gene expression. E: ChIP of GR and RORγ on the GRE-RORE region of mouse Pck1 and G6pc1 genes of the liver of AAV8-shS1PR2 mice overexpressing IgG, GR, or RORγ and treated without Dex (−) or with Dex (+) (n = 3–6). *P < 0.05, **P < 0.01, ***P < 0.001, using two-way ANOVA with Fisher least significant difference post hoc test. Error bars represent SEM for the tolerance tests and SD for the rest.

Close modal

The results of ChIP assays showed that the Dex effect on GR recruitment to the Pck1 and the G6pc1 GREs in AAV8-shS1PR2-GFP mice was minimal (Fig.6E), similar to the observation in AAV8-shS1PR2 mice (Fig.2E). Overexpression of RORγ enhanced GR recruitment (Fig.6E). RORγ was not present in the Pck1 and G6pc1 ROREs in AAV8-shS1PR2-GFP mice (Fig.6E). Surprisingly, without Dex, RORγ was also not present in the Pck1 and G6pc1 ROREs in AAV8-shS1PR2-RORγ mouse liver (Fig.6E). RORγ was only recruited to the ROREs in AAV8-shS1PR2-RORγ mouse liver upon Dex treatment (Fig.6E).

Overall, these results demonstrate that upon chronic Dex treatment, RORγ is recruited to the ROREs of Pck1 and G6pc1 genes and is required for maximal GR recruitment to the GREs. This augments the expression of hepatic gluconeogenic genes, which enhances gluconeogenesis and glucose intolerance (Fig. 7).

Figure 7

Model of chronic glucocorticoid exposure–induced S1PR2-RORγ axis on gluconeogenic gene expression regulation. Short-term glucocorticoid exposure, such as 6 h or an overnight fast, induces hepatic glucose production through GR binding to GREs of gluconeogenic genes while suppressing RORγ expression. Chronic glucocorticoid exposure increases hepatic production of S1P, which can be exported extracellularly where S1P binds to S1PR2 on its own cell surface or neighboring cells. S1PR2-initiated signaling antagonizes glucocorticoid-suppressed RORγ expression. The increased RORγ then enhances gluconeogenic gene transcription, as RORγ can potentiate GR recruitment to GREs of gluconeogenic genes. Hepatic S1PR2 signaling also reduces basal gluconeogenesis by unknown mechanisms.

Figure 7

Model of chronic glucocorticoid exposure–induced S1PR2-RORγ axis on gluconeogenic gene expression regulation. Short-term glucocorticoid exposure, such as 6 h or an overnight fast, induces hepatic glucose production through GR binding to GREs of gluconeogenic genes while suppressing RORγ expression. Chronic glucocorticoid exposure increases hepatic production of S1P, which can be exported extracellularly where S1P binds to S1PR2 on its own cell surface or neighboring cells. S1PR2-initiated signaling antagonizes glucocorticoid-suppressed RORγ expression. The increased RORγ then enhances gluconeogenic gene transcription, as RORγ can potentiate GR recruitment to GREs of gluconeogenic genes. Hepatic S1PR2 signaling also reduces basal gluconeogenesis by unknown mechanisms.

Close modal

In this study, we showed that chronic Dex treatment increased liver S1P levels in male mice and that reducing the expression of hepatic S1PR1, 2, and 3 all modulated the effect of Dex treatment on glucose metabolism, though with distinct effects. Hepatic S1PR3 knockdown exacerbated Dex-induced glucose intolerance and fasting hyperglycemia but had no effect on fasting hyperinsulinemia. In contrast, hepatic S1PR1 knockdown did not affect glucose tolerance and fasting glucose levels but increased fasting insulin levels. We speculate that S1PR3 signaling negatively regulates chronic glucocorticoid-induced hepatic gluconeogenesis, whereas S1PR1 signaling attenuates chronic glucocorticoid-induced insulin resistance. These models will be examined in our future studies. We also found that hepatic S1PR2 knockdown attenuated Dex-induced glucose intolerance. However, hepatic S1PR2 knockdown had no effect on insulin tolerance, and Dex effect on insulin stimulated Akt activity in the metabolic tissues tested. PTTs showed that S1PR2 knockdown reduced Dex-induced gluconeogenesis. Therefore, prolonged Dex treatment induced hepatic S1PR2 signaling to enhance gluconeogenesis without affecting insulin signaling. This report focuses on elucidating the role of hepatic S1PR2 in chronic glucocorticoid-induced glucose disorders.

RNA-seq identified Rorc as a gene whose expression was reduced in the liver by hepatic S1PR2 knockdown in Dex-treated mice. RORγ was previously shown to potentiate transcription of gluconeogenic genes (14), but its role in glucocorticoid’s effects has not been explored. We found that RORγ overexpression in hepatic S1PR2 knockdown mouse liver restored Dex-induced glucose intolerance and increased gluconeogenesis and gluconeogenic gene expression. In contrast, RORγ antagonist XY018 treatment attenuated Dex-induced glucose intolerance and reduced gluconeogenic gene expression and hepatic gluconeogenesis. Similarly, Dex-induced hepatic gluconeogenesis was lower in hepatic RORγ knockdown mice, and they were trending toward having better glucose tolerance under Dex treatment. These results demonstrated RORγ’s participation in chronic Dex-induced hepatic gluconeogenesis. RORγ overexpression in mouse liver did not affect Pck1 and G6pc1 basal expression or basal glucose and pyruvate tolerance. Similarly, XY018 did not affect basal glucose tolerance or hepatic gluconeogenesis. The studies of Pck1 and G6pc1 mRNA in H4IIE cells were consistent with these in vivo results. A previous report showed that liver-specific Rorc knockout mice had lower gluconeogenic activity and gluconeogenic gene expression during the daytime (14). Our results did not exactly contradict these results. Perhaps the dose and/or duration of XY018 we administered was sufficient to improve Dex-induced glucose intolerance but not enough to affect basal glucose tolerance in control mice. In fact, in rat Pck1 promoter reporter assay, we found that RORγ agonist LYC-55176 was trending toward increasing the reporter gene activity. Interestingly, reporter gene studies showed that even without adding LYC-55716, overexpressing RORγ was able to potentiate Dex response, and LYC-55176 further enhanced this effect. This suggests that endogenous ligands of RORγ are enough to potentiate the Dex response to a certain degree and that more agonists further enhanced the effect.

In any case, because hepatic S1PR2 knockdown improved basal glucose tolerance and reduced basal gluconeogenesis, we propose that S1PR2 signaling exerts at least two mechanisms that enhance chronic glucocorticoid exposure–induced hepatic gluconeogenesis. The first mechanism is through suppressing glucocorticoid-reduced RORγ expression. The other is to regulate basal gluconeogenesis, for which the mechanisms remain unclear. Although S1PR2 signaling did not regulate basal gluconeogenic gene expression, other mechanisms could modulate the rate of gluconeogenesis (23). Notably, the regulation of basal glucose tolerance by S1PR2 signaling was likely independent of the presence of ANGPTL4. In contrast, ANGPTL4 was required for S1PR2 signaling to enhance glucocorticoid-induced glucose intolerance. This is not surprising because ANGPTL4 was required for Dex-induced hepatic S1P production (9). A previous study showed that basal glucose tolerance was not different between WT and S1pr2 null mice fed a chow diet (24). Here, we specifically reduced S1PR2 in the liver of male mice. Perhaps S1PR2 signaling in different tissues has distinct roles on whole-body glucose homeostasis. In S1pr2-null mice, S1PR2 signaling in certain tissues somehow offsets its role in liver gluconeogenesis.

The recruitment of GR to Pck1 and G6pc1 GREs was decreased in hepatic S1PR2 knockdown mouse liver. When RORγ was overexpressed in the liver of these mice, it restored GR occupancy. In contrast, XY018 attenuated Dex-induced GR occupancy. Thus, mechanistically, RORγ assists GR recruitment to the GREs of gluconeogenic genes. Previous studies of glucocorticoid activation of rat Pck1 gene found that HNF4 and FoxA1/2 (also known as HNF3α/β) bind upstream of GREs (25,26) and enhance GR binding to the GREs (27). The RORE in the mouse Pck1 gene overlaps with another accessory element required for maximal glucocorticoid response located downstream of GREs that has been previously shown to bind to chicken ovalbumin upstream promoter transcription factor (COUP-TF) (28). The fact that XY018 reduced GR and RORγ recruitment to the GRE-ROREs of gluconeogenic genes suggests that RORγ’s transcriptional activation activity is required for its enhancement of Dex response. This is reminiscent of the requirement of HNF4 and FoxA’s transactivation domains to assist GR with activating rat Pck1 gene transcription (29). RORγ likely participates in Dex-stimulated gluconeogenic gene transcription through recruiting transcriptional coactivators into a GR-containing transcriptional complex.

Overall, the novel GR-SIPR2-RORγ axis identified here provides valuable insights to improve glucocorticoid therapy. However, two important questions remain to be explored. First, glucocorticoids are potent anti-inflammatory and immunomodulatory agents, but their applications are limited by their adverse effects, including hyperglycemia. This study suggests that including the antagonists (ideally liver specific) of S1PR2 and/or RORγ in glucocorticoid therapy to dampen hyperglycemia could provide improved flexibility of glucocorticoid use. Notably, there are active studies ongoing for designing antagonists of both S1PR2 (10,30) and RORγ (31,32) for other diseases. Obviously, this approach requires that S1PR2 and/or RORγ antagonists do not affect the anti-inflammatory response of glucocorticoids. This notion should be confirmed. Second, how hepatic S1PR2 signaling antagonizes glucocorticoid-suppressed RORγ expression is unclear. This mechanism should be explored in future studies.

Acknowledgments. The authors thank Dr. Donald Scott (Icahn School of Medicine at Mount Sinai) for his comments. Model design was performed using BioRender.

Funding. This work was supported by National Institute of Diabetes and Digestive and Kidney Diseases (NIDDK) grants DK113019, DK124866, and DK131194. J.-C.W. is a member of the UCSF Liver Center (NIDDK grant DK026743). Work in the laboratory of R.M.O. is supported by NIDDK grant DK132259.

Duality of Interest. No potential conflicts of interest relevant to this article were reported.

Author Contributions. R.A.L. and M.C. analyzed the data. R.A.L., M.C., A.T., Y.R.L., D.L., N.Y., S.T., and M.Z. executed the experiments. R.A.L., M.C., R.M.O., and J.-C.W. wrote the manuscript. R.A.L., M.C., and J.-C.W designed and supervised the experiments. R.A.L and J.-C.W. planned and conceptualized the study. J.-C.W. is the guarantor of this work and, as such, had full access to all data in the study and takes responsibility for the integrity of the data and accuracy of data analysis.

Prior Presentation. Parts of this study were presented in abstract form at the Endocrine Society Conference, Chicago, IL, 15–18 June 2023.

This article contains supplementary material online at https://doi.org/10.2337/figshare.23877852.

R.A.L. and M.C. contributed equally.

1.
Kuo
T
,
McQueen
A
,
Chen
TC
,
Wang
JC.
Regulation of glucose homeostasis by glucocorticoids
.
Adv Exp Med Biol
2015
;
872
:
99
126
2.
Kwon
S
,
Hermayer
KL
,
Hermayer
K.
Glucocorticoid-induced hyperglycemia
.
Am J Med Sci
2013
;
345
:
274
277
3.
Li
JX
,
Cummins
CL.
Fresh insights into glucocorticoid-induced diabetes mellitus and new therapeutic directions
.
Nat Rev Endocrinol
2022
;
18
:
540
557
4.
Granner
DK
,
Wang
JC
,
Yamamoto
KR.
Regulatory actions of glucocorticoid hormones: from organisms to mechanisms
.
Adv Exp Med Biol
2015
;
872
:
3
31
5.
Pilkis
SJ
,
Granner
DK.
Molecular physiology of the regulation of hepatic gluconeogenesis and glycolysis
.
Annu Rev Physiol
1992
;
54
:
885
909
6.
McQueen
AE
,
Kanamaluru
D
,
Yan
K
, et al
.
The C-terminal fibrinogen-like domain of angiopoietin-like 4 stimulates adipose tissue lipolysis and promotes energy expenditure
.
J Biol Chem
2017
;
292
:
16122
16134
7.
Gray
NE
,
Lam
LN
,
Yang
K
,
Zhou
AY
,
Koliwad
S
,
Wang
JC.
Angiopoietin-like 4 (Angptl4) protein is a physiological mediator of intracellular lipolysis in murine adipocytes
.
J Biol Chem
2012
;
287
:
8444
8456
8.
Dijk
W
,
Kersten
S.
Regulation of lipoprotein lipase by Angptl4
.
Trends Endocrinol Metab
2014
;
25
:
146
155
9.
Chen
TC
,
Benjamin
DI
,
Kuo
T
, et al
.
The glucocorticoid-Angptl4-ceramide axis induces insulin resistance through PP2A and PKCζ
.
Sci Signal
2017
;
10
:eaai7905
10.
Cartier
A
,
Hla
T.
Sphingosine 1-phosphate: lipid signaling in pathology and therapy
.
Science
2019
;
366
:eaar5551
11.
Strub
GM
,
Paillard
M
,
Liang
J
, et al
.
Sphingosine-1-phosphate produced by sphingosine kinase 2 in mitochondria interacts with prohibitin 2 to regulate complex IV assembly and respiration
.
FASEB J
2011
;
25
:
600
612
12.
Patmanathan
SN
,
Wang
W
,
Yap
LF
,
Herr
DR
,
Paterson
IC.
Mechanisms of sphingosine 1-phosphate receptor signalling in cancer
.
Cell Signal
2017
;
34
:
66
75
13.
Fayyaz
S
,
Japtok
L
,
Kleuser
B.
Divergent role of sphingosine 1-phosphate on insulin resistance. Cell Physiol Biochem
2014
;
34
:
134
147
14.
Takeda
Y
,
Kang
HS
,
Freudenberg
J
,
DeGraff
LM
,
Jothi
R
,
Jetten
AM.
Retinoic acid-related orphan receptor γ (RORγ): a novel participant in the diurnal regulation of hepatic gluconeogenesis and insulin sensitivity
.
PLoS Genet
2014
;
10
:
e1004331
15.
Chen
TC
,
Lee
RA
,
Tsai
SL
, et al
.
An ANGPTL4-ceramide-protein kinase Cζ axis mediates chronic glucocorticoid exposure-induced hepatic steatosis and hypertriglyceridemia in mice
.
J Biol Chem
2019
;
294
:
9213
9224
16.
Alessi
DR
,
Andjelkovic
M
,
Caudwell
B
, et al
.
Mechanism of activation of protein kinase B by insulin and IGF-1
.
EMBO J
1996
;
15
:
6541
6551
17.
Vander Kooi
BT
,
Onuma
H
,
Oeser
JK
, et al
.
The glucose-6-phosphatase catalytic subunit gene promoter contains both positive and negative glucocorticoid response elements
.
Mol Endocrinol
2005
;
19
:
3001
3022
18.
Matsuoka
H
,
Shima
A
,
Kuramoto
D
,
Kikumoto
D
,
Matsui
T
,
Michihara
A.
Phosphoenolpyruvate carboxykinase, a key enzyme that controls blood glucose, is a target of retinoic acid receptor-related orphan receptor α
.
PLoS One
2015
;
10
:
e0137955
19.
Kadiri
S
,
Monnier
C
,
Ganbold
M
,
Ledent
T
,
Capeau
J
,
Antoine
B.
The nuclear retinoid-related orphan receptor-α regulates adipose tissue glyceroneogenesis in addition to hepatic gluconeogenesis
.
Am J Physiol Endocrinol Metab
2015
;
309
:
E105
E114
20.
Satsu
H
,
Schaeffer
MT
,
Guerrero
M
, et al
.
A sphingosine 1-phosphate receptor 2 selective allosteric agonist
.
Bioorg Med Chem
2013
;
21
:
5373
5382
21.
Ndagi
U
,
Mhlongo
NN
,
Soliman
ME.
Re-emergence of an orphan therapeutic target for the treatment of resistant prostate cancer - a thorough conformational and binding analysis for ROR-γ protein
.
J Biomol Struct Dyn
2018
;
36
:
335
350
22.
Aicher
TD
,
Van Huis
CA
,
Hurd
AR
, et al
.
Discovery of LYC-55716: a potent, selective, and orally bioavailable retinoic acid receptor-related orphan receptor-γ (RORγ) agonist for use in treating cancer
.
J Med Chem
2021
;
64
:
13410
13428
23.
Petersen
MC
,
Vatner
DF
,
Shulman
GI.
Regulation of hepatic glucose metabolism in health and disease
.
Nat Rev Endocrinol
2017
;
13
:
572
587
24.
Kitada
Y
,
Kajita
K
,
Taguchi
K
, et al
.
Blockade of sphingosine 1-phosphate receptor 2 signaling attenuates high-fat diet-induced adipocyte hypertrophy and systemic glucose intolerance in mice
.
Endocrinology
2016
;
157
:
1839
1851
25.
O’Brien
RM
,
Noisin
EL
,
Suwanichkul
A
, et al
.
Hepatic nuclear factor 3- and hormone-regulated expression of the phosphoenolpyruvate carboxykinase and insulin-like growth factor-binding protein 1 genes
.
Mol Cell Biol
1995
;
15
:
1747
1758
26.
Hall
RK
,
Sladek
FM
,
Granner
DK.
The orphan receptors COUP-TF and HNF-4 serve as accessory factors required for induction of phosphoenolpyruvate carboxykinase gene transcription by glucocorticoids
.
Proc Natl Acad Sci U S A
1995
;
92
:
412
416
27.
Stafford
JM
,
Wilkinson
JC
,
Beechem
JM
,
Granner
DK.
Accessory factors facilitate the binding of glucocorticoid receptor to the phosphoenolpyruvate carboxykinase gene promoter
.
J Biol Chem
2001
;
276
:
39885
39891
28.
Scott
DK
,
Mitchell
JA
,
Granner
DK.
The orphan receptor COUP-TF binds to a third glucocorticoid accessory factor element within the phosphoenolpyruvate carboxykinase gene promoter
.
J Biol Chem
1996
;
271
:
31909
31914
29.
Wang
JC
,
Strömstedt
PE
,
Sugiyama
T
,
Granner
DK.
The phosphoenolpyruvate carboxykinase gene glucocorticoid response unit: identification of the functional domains of accessory factors HNF3 beta (hepatic nuclear factor-3 beta) and HNF4 and the necessity of proper alignment of their cognate binding sites
.
Mol Endocrinol
1999
;
13
:
604
618
30.
Blankenbach
KV
,
Schwalm
S
,
Pfeilschifter
J
,
Meyer Zu Heringdorf
D.
Sphingosine-1-phosphate receptor-2 antagonists: therapeutic potential and potential risks
.
Front Pharmacol
2016
;
7
:
167
31.
Bronner
SM
,
Zbieg
JR
,
Crawford
JJ.
RORγ antagonists and inverse agonists: a patent review
.
Expert Opin Ther Pat
2017
;
27
:
101
112
32.
Pandya
VB
,
Kumar
S
, Sachchidanand, Sharma R, Desai RC.
Combating autoimmune diseases with retinoic acid receptor-related orphan receptor-gamma (RORgamma or RORc) inhibitors: hits and misses
.
J Med Chem
2018
;
61
:
10976
10995
Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered. More information is available at https://www.diabetesjournals.org/journals/pages/license.