79
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Retinoic Acid-Related Orphan Receptor γ (RORγ): A Novel Participant in the Diurnal Regulation of Hepatic Gluconeogenesis and Insulin Sensitivity

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          The hepatic circadian clock plays a key role in the daily regulation of glucose metabolism, but the precise molecular mechanisms that coordinate these two biological processes are not fully understood. In this study, we identify a novel connection between the regulation of RORγ by the clock machinery and the diurnal regulation of glucose metabolic networks. We demonstrate that particularly at daytime, mice deficient in RORγ exhibit improved insulin sensitivity and glucose tolerance due to reduced hepatic gluconeogenesis. This is associated with a reduced peak expression of several glucose metabolic genes critical in the control of gluconeogenesis and glycolysis. Genome-wide cistromic profiling, promoter and mutation analysis support the concept that RORγ regulates the transcription of several glucose metabolic genes directly by binding ROREs in their promoter regulatory region. Similar observations were made in liver-specific RORγ-deficient mice suggesting that the changes in glucose homeostasis were directly related to the loss of hepatic RORγ expression. Altogether, our study shows that RORγ regulates several glucose metabolic genes downstream of the hepatic clock and identifies a novel metabolic function for RORγ in the diurnal regulation of hepatic gluconeogenesis and insulin sensitivity. The inhibition of the activation of several metabolic gene promoters by an RORγ antagonist suggests that antagonists may provide a novel strategy in the management of metabolic diseases, including type 2 diabetes.

          Author Summary

          The circadian clock plays a critical role in the regulation of many physiological processes, including metabolism and energy homeostasis. The retinoic acid-related orphan receptor γ (RORγ) functions as a ligand-dependent transcription factor that regulates transcription by binding as a monomer to ROR-responsive elements. In liver, RORγ exhibits a robust circadian pattern of expression that is under direct control of the hepatic circadian clock. However, the connection between the circadian regulation of RORγ and its control of downstream metabolic processes is not well understood. In this study, by using ubiquitous and liver-specific RORγ-deficient mice as models, we demonstrate that hepatic RORγ modulates daily insulin sensitivity and glucose tolerance by regulating hepatic gluconeogenesis. Genome-wide cistromic profiling, gene expression, and promoter analysis revealed that RORγ is targeting and regulating a number of novel metabolic genes critical in the control of glycolysis and gluconeogenesis pathways. We provide evidence for a model in which RORγ regulates the circadian expression of glucose metabolic genes in the liver downstream of the hepatic circadian clock, thereby enhancing gluconeogenesis and decreasing insulin sensitivity and glucose tolerance. This study suggests that attenuating RORγ activity by antagonists might be beneficial for the management of glucose metabolic diseases including type 2 diabetes.

          Related collections

          Most cited references26

          • Record: found
          • Abstract: found
          • Article: not found

          T helper 17 lineage differentiation is programmed by orphan nuclear receptors ROR alpha and ROR gamma.

          T cell functional differentiation is mediated by lineage-specific transcription factors. T helper 17 (Th17) has been recently identified as a distinct Th lineage mediating tissue inflammation. Retinoic acid receptor-related orphan receptor gamma (ROR gamma) was shown to regulate Th17 differentiation; ROR gamma deficiency, however, did not completely abolish Th17 cytokine expression. Here, we report Th17 cells highly expressed another related nuclear receptor, ROR alpha, induced by transforming growth factor-beta and interleukin-6 (IL-6), which is dependent on signal transducer and activator of transcription 3. Overexpression of ROR alpha promoted Th17 differentiation, possibly through the conserved noncoding sequence 2 in Il17-Il17f locus. ROR alpha deficiency resulted in reduced IL-17 expression in vitro and in vivo. Furthermore, ROR alpha and ROR gamma coexpression synergistically led to greater Th17 differentiation. Double deficiencies in ROR alpha and ROR gamma globally impaired Th17 generation and completely protected mice against experimental autoimmune encephalomyelitis. Therefore, Th17 differentiation is directed by two lineage-specific nuclear receptors, ROR alpha and ROR gamma.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Regulation of Circadian Behavior and Metabolism by Rev-erbα and Rev-erbβ

            The circadian clock acts at the genomic level to coordinate internal behavioral and physiologic rhythms via the CLOCK-BMAL transcriptional heterodimer. Although the nuclear receptors REV-ERBα and β have been proposed to form an accessory feedback loop that contributes to clock function 1,2 , their precise roles and importance remain unresolved. To establish their regulatory potential we generated comparative cistromes of both REV-ERB isoforms, which revealed shared recognition at over 50% of their total sites and extensive overlap with the master circadian regulator BMAL1. While Rev-erbα has been shown to directly regulate Bmal1 expression 1,2 , the cistromic analysis reveals a direct connection between Bmal1 and Rev-erbα and β regulatory circuits than previously suspected. Genes within the intersection of the BMAL1, REV-ERBα and REV-ERBβ cistromes are highly enriched for both clock and metabolic functions. As predicted by the cistromic analysis, dual depletion of Rev-erbα/β function by creating double-knockout mice (DKOs) profoundly disrupted circadian expression of core circadian clock and lipid homeostatic gene networks. As a result, DKOs show strikingly altered circadian wheel-running behavior and deregulated lipid metabolism. These data now ally Rev-erbα/β with Per, Cry and other components of the principal feedback loop that drives circadian expression and suggest a more integral mechanism for the coordination of circadian rhythm and metabolism.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              BMAL1 and CLOCK, Two Essential Components of the Circadian Clock, Are Involved in Glucose Homeostasis

              Introduction The master clock, which, in mammals, resides in the hypothalamic suprachiasmatic nucleus (SCN), is thought to synchronize multiple peripheral oscillators to ensure temporal coordination of behavior and metabolism. Peripheral clocks amplify or dampen central rhythms or exhibit autonomous behavior to facilitate local adaptive responses (Hastings et al. 2003). The central clock may communicate to modulate or entrain rhythms in the periphery via hormones (McNamara et al. 2001) or hemodynamic cues. Asynchronous environmental cues, such as eating, also influence the autonomous behavior of peripheral clocks (Damiola et al. 2000; Stokkan et al. 2001). The variation in sleep and wakefulness (activity) is perhaps the most well-known circadian rhythm. Surgical ablation of the SCN in mice (Ibuka et al. 1980; Welsh et al. 1988) and rats (Ibuka et al. 1977; Mosko and Moore 1979) abolishes the nocturnal burst in locomotor activity. Similarly, disruption and/or mutation of Bmal1 (Bunger et al. 2000) or Clock (Vitaterna et al. 1994), transcription factors that compose the positive limb of an autoregulatory feedback loop in the core molecular clock (Young and Kay 2001; Reppert and Weaver 2002), also impairs circadian behavior. Bmal1 and Clock may influence behavioral rhythms by regulating the firing rate of SCN neurons (Herzog et al. 1998; Deboer et al. 2003). Genes relevant to the molecular clock are also expressed in peripheral tissues (Akhtar et al. 2002; Kita et al. 2002; Panda et al. 2002; Storch et al. 2002; Oishi et al. 2003) where approximately 5%–10% of the transcriptome is subject to circadian oscillation (Albrecht and Eichele 2003). Although the precise role of peripheral clocks and the mechanisms that link them to the SCN remain largely obscure, genetic mutation or deletion has implicated peripheral clocks in the regulation of some aspects of cellular function, including division (Matsuo et al. 2003), estrous cyclicity (Miller et al. 2004), and phospholipid metabolism (Marquez et al. 2004). Glucose and lipid homeostasis are also known to exhibit circadian variation (Seaman et al. 1965; Malherbe et al. 1969; Gagliardino and Hernandez 1971; Schlierf and Dorow 1973). Surgical ablation of the SCN impairs the control of glucose homeostasis (la Fleur et al. 2001). However, the proximity of satiety centres to the SCN has potentially confounded interpretation of these results. Indeed, there is no direct evidence implicating the molecular clock in the regulation of glycaemia or insulin sensitivity (Si). Our studies revealed a profound role for core clock genes—Bmal1 and Clock—in regulating recovery from insulin-induced hypoglycaemia. Furthermore, the impact of a high-fat diet (HF) was to amplify the diurnal variation in glucose tolerance and Si in a manner dependent on the Clock gene. These studies suggest that the temporal distribution of a caloric load may influence the response to insulin and that circadian variability in glucose homeostasis may be subject to modulation by asynchronous dietary cues. Results We examined the role of the molecular clock in glucose homeostasis by using mice in which core clock genes are impaired (Clockmut) or deficient (Bmal1−/−). Both plasma glucose and triglycerides were subject to circadian variation in wild-type (WT) mice, peaking at approximately circadian time point 4 (CT4) and CT28 (where CT0 is subjective day beginning at 7 AM, and CT12 is subjective night beginning at 7 PM) (Figure 1A and 1B), as reported previously (Seaman et al. 1965; Schlierf and Dorow 1973). We also observed that corticosterone (Figure 1C), which stimulates gluconeogenesis during hypoglycaemia (Cryer 1993), and adiponectin (Figure 1D), which has been associated with insulin resistance (Yamauchi et al. 2001; Maeda et al. 2002), oscillated significantly, but out of phase with the glucose and triglyceride rhythms. Diurnal variation in glucose and triglycerides, but not in corticosterone, was disrupted in the mutant mice (Table 1). Although there was no clear rhythm in the hypoglyacemic response to insulin, recovery of blood glucose exhibited a robust circadian variation (Figure 2A), with an excessive rebound from the effects of insulin evident at subjective dawn (CT19 and CT25) (Figure 2A). Insulin caused a profound hypoglyacemic response, independent of clock time, in both Bmal1 −/− and Clockmut mice (Figure 2B). This response was more pronounced in the former, consistent with the comparative severity of the molecular and behavioral phenotypes between the Bmal1−/− and Clockmut animals (King et al. 1997; Bunger et al. 2000). Despite exacerbation of the hypoglycaemic response to insulin in the mutants, the counterregulatory responses of both corticosterone and glucagon were retained (Figure 2C and 2D). Gluconeogenesis also contributes to restoration of blood glucose after insulin-induced hypoglycaemia. Consistent with this observation, conversion of exogenously administered pyruvate to glucose, which reflects gluconeogenesis (Miyake et al. 2002), was impaired in the Clockmut animals. This impairment was most marked in Bmal1−/− mice, while Bmal1+/− and Clockmut mice exhibited an intermediate phenotype when compared with WT littermate controls (Figure 3A). Furthermore, activity of the key rate-limiting enzyme of gluconeogenesis, phosphoenolpyruvate carboxykinase (PEPCK), exhibited diurnal variation in the liver and aorta that was blunted in Clockmut mice (Figure 3B). PEPCK activity in kidney was antiphasic to the rhythm in aorta and liver and was unimpaired in Clockmut mice (Figure 3B), suggesting tissue-specific regulation of enzyme activity. The frequent sampling intravenous glucose tolerance test (FSIGT) was performed to assess more precisely the impact of the molecular clock on sensitivity to insulin. This test provides an estimate of Si, consistent with that obtained by the euglycaemic clamp (Pacini et al. 2001). Additionally, data modeling provides estimates of glucose-mediated glucose disposal (Sg), insulin secretion, and Si. Si and insulin secretion, but not Sg, exhibited a diurnal variation in WT mice fed a regular chow diet (RC) (Table 2). Circadian variation of glucose and lipid homeostasis might condition the metabolic response to asynchronous environmental cues, such as diet, that impinge on Si. Dyslipidemia coincides with insulin resistance in the metabolic syndrome (Brotman and Girod 2002), and a diet high in fat impairs Si (Grundleger and Thenen 1982; Coulston et al. 1983). Both HF-fed WT and HF-fed Clockmut mice increased body weight significantly and to a similar degree in comparison to their age-matched, RC-fed controls (Table 3). Body fat composition averaged 17.6% of lean body mass in RC-fed WT mice, rising to 27.7% (p Clockmut). Thus, the most parsimonious interpretation is that the observed metabolic deficiencies in the Bmal1 −/− and Clockmut mice are due to their roles in the circadian clock, rather than to “off-clock” effects. We observed that the impact of HF on glucose homeostasis was apparently to emphasize the role of the molecular clock. Diet has previously been shown to interact with peripheral clocks. Changes in feeding shift the circadian pattern of gene expression in the liver, but not in the master clock in the SCN (Damiola et al. 2000), demonstrating the importance of food as a cue to circadian control. Individual constituents of food could also provide discrete stimuli. For example, glucose alone can induce rhythmic gene expression in isolated fibroblasts (Hirota et al. 2002). Thus, dietary composition, the size and timing of a feed might all be expected to interact differentially with an underlying circadian regulation of metabolic control. Alterations in dietary content, the availability of “fast food,” inactivity, and sociocultural factors have all been implicated in the emergence of the metabolic syndrome as a major challenge to the public health (Zimmet et al. 2001). However, while mechanistic integration of the diverse elements of the syndrome has proven elusive, our studies suggest that timing may influence the functional consequences of ingesting a caloric load. Materials and Methods Animals Mice were acclimatized for 2 wk in 12 h light–12 h dark cycles before being subjected to a 36-h period of constant darkness followed by experimentation in darkness. Experimental chronology is measured in CT, subjective day beginning at 7 AM (CT0), and subjective night beginning at 7 PM (CT12). Diet WT and Clockmut mice were placed on an HF (Teklad, TD02435) and compared to age-matched WT mice on a regular chow diet (RC). Mice were on RC for 8 wk except for those subjected to FSIGT where they received RC for 11 mo. Body mass composition was measured by dual energy X-ray absorptiometry at 10 mo. Intraperitoneal tolerance Tests were performed as described (Klaman et al. 2000) with a diminution in the glucose bolus (0.1 g/kg). Intravenous glucose tolerance test and minimal modeling The tolerance test was performed as described (Pacini et al. 2001) in unanesthetized mice, and the minimal model of Bergman et al. (1979) was applied to the data using MINMOD software (Boston et al. 2003). The derived values were Si, Sg, and acute insulin response to glucose, which measures insulin secretion. Si is the ratio of insulin delivery rate to the interstitium to insulin extraction rate from the interstitium. Long-term feeding of HF to WT mice resulted in imperceptibly small insulin sensitivity values. This could be the consequence of impaired delivery of insulin to the interstitium, exacerbated extraction rate, or a combination of both factors. Insulin secretion is derived from area under the insulin curve, above basal, from 0 to 10 min after glucose infusion; and disposition index, which equals the product of insulin sensitivity multiplied by insulin secretion and measures the degree to which insulin sensitivity can be compensated for by elevated insulin secretion (Pacini et al. 2001). Assay methods Insulin, leptin, corticosterone, and glucagon levels were measured by immunoassays from Crystalchem (Downers Grove, Illinois, United States), ICN Biochemicals (Costa Mesa, California, United States), and Linco Research (St. Charles, Missouri, United States). Plasma glucose was measured by the glucose oxidase method using a glucose analyzer machine for FSIGT and by glucometer for the intraperitoneal tolerance test. PEPCK activity was quantitated by a bioluminescent method (Wimmer 1988). Statistical analysis The significance of differences amongst the tolerance test curves was assessed by distribution-free two-way ANOVA with a Bonferroni correction. FSIGT data were tested by one-way ANOVA with the Kruskal-Wallis test. Paired Student's t-tests were used to perform comparisons of corticosterone levels before and after insulin injection in Bmal−/− mice and between WT and Clockmut mice. Plasma samples for glucagon analysis were pooled and were thus not compared by a formal statistical analysis. Results are presented as mean ± standard error of the mean (SEM), except for the FSIGT data (Table 2), presented as mean ± fractional standard deviation. Differences were considered significant when p < 0.05.
                Bookmark

                Author and article information

                Contributors
                Role: Editor
                Journal
                PLoS Genet
                PLoS Genet
                plos
                plosgen
                PLoS Genetics
                Public Library of Science (San Francisco, USA )
                1553-7390
                1553-7404
                May 2014
                15 May 2014
                : 10
                : 5
                : e1004331
                Affiliations
                [1 ]Cell Biology Section, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, United States of America
                [2 ]Systems Biology Group, Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, United States of America
                Charité - Universitätsmedizin Berlin, Germany
                Author notes

                The authors have declared that no competing interests exist.

                Conceived and designed the experiments: YT HSK RJ AMJ. Performed the experiments: YT HSK LMD. Analyzed the data: RJ JF. Contributed reagents/materials/analysis tools: YT HSK RJ JF. Wrote the paper: YT AMJ HSK RJ.

                Article
                PGENETICS-D-13-02938
                10.1371/journal.pgen.1004331
                4022472
                24831725
                414d3a32-22d8-434c-aa0f-fa6c08fa496d
                Copyright @ 2014

                This is an open-access article, free of all copyright, and may be freely reproduced, distributed, transmitted, modified, built upon, or otherwise used by anyone for any lawful purpose. The work is made available under the Creative Commons CC0 public domain dedication.

                History
                : 24 October 2013
                : 7 March 2014
                Page count
                Pages: 16
                Funding
                This research was supported by the Intramural Research Program of the National Institute of Environmental Health Sciences, the National Institutes of Health [Z01-ES-101586] and the Japanese Society for the Promotion of Science (JSPS). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.
                Categories
                Research Article
                Biology and life sciences
                Cell Biology
                Chromosome Biology
                Chromatin
                Molecular Cell Biology
                Computational Biology
                Gene Regulatory Networks
                Biochemistry
                Metabolism
                Carbohydrate Metabolism
                Proteins
                Regulatory Proteins
                Physiology
                Physiological Parameters
                Body Weight
                Obesity
                Genetics
                Gene expression
                DNA transcription
                Gene regulation
                Genomics
                Functional Genomics
                Molecular Genetics
                Nutrition
                Medicine and Health Sciences
                Endocrinology
                Diabetic Endocrinology
                Metabolic Disorders
                Diabetes Mellitus
                Type 2 Diabetes

                Genetics
                Genetics

                Comments

                Comment on this article