Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
EMBO Rep ; 24(2): e55363, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36520372

ABSTRACT

Macrophages are key cells after tissue damage since they mediate both acute inflammatory phase and regenerative inflammation by shifting from pro-inflammatory to restorative cells. Glucocorticoids (GCs) are the most potent anti-inflammatory hormone in clinical use, still their actions on macrophages are not fully understood. We show that the metabolic sensor AMP-activated protein kinase (AMPK) is required for GCs to induce restorative macrophages. GC Dexamethasone activates AMPK in macrophages and GC receptor (GR) phosphorylation is decreased in AMPK-deficient macrophages. Loss of AMPK in macrophages abrogates the GC-induced acquisition of their repair phenotype and impairs GC-induced resolution of inflammation in vivo during post-injury muscle regeneration and acute lung injury. Mechanistically, two categories of genes are impacted by GC treatment in macrophages. Firstly, canonical cytokine regulation by GCs is not affected by AMPK loss. Secondly, AMPK-dependent GC-induced genes required for the phenotypic transition of macrophages are co-regulated by the transcription factor FOXO3, an AMPK substrate. Thus, beyond cytokine regulation, GR requires AMPK-FOXO3 for immunomodulatory actions in macrophages, linking their metabolic status to transcriptional control in regenerative inflammation.


Subject(s)
AMP-Activated Protein Kinases , Glucocorticoids , Humans , Glucocorticoids/pharmacology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Macrophages/metabolism , Inflammation/metabolism , Cytokines/metabolism
2.
Diabetologia ; 64(8): 1850-1865, 2021 08.
Article in English | MEDLINE | ID: mdl-34014371

ABSTRACT

AIMS/HYPOTHESIS: Adipocytes are critical cornerstones of energy metabolism. While obesity-induced adipocyte dysfunction is associated with insulin resistance and systemic metabolic disturbances, adipogenesis, the formation of new adipocytes and healthy adipose tissue expansion are associated with metabolic benefits. Understanding the molecular mechanisms governing adipogenesis is of great clinical potential to efficiently restore metabolic health in obesity. Here we investigate the role of heart and neural crest derivatives-expressed 2 (HAND2) in adipogenesis. METHODS: Human white adipose tissue (WAT) was collected from two cross-sectional studies of 318 and 96 individuals. In vitro, for mechanistic experiments we used primary adipocytes from humans and mice as well as human multipotent adipose-derived stem (hMADS) cells. Gene silencing was performed using siRNA or genetic inactivation in primary adipocytes from loxP and or tamoxifen-inducible Cre-ERT2 mouse models with Cre-encoding mRNA or tamoxifen, respectively. Adipogenesis and adipocyte metabolism were measured by Oil Red O staining, quantitative PCR (qPCR), microarray, glucose uptake assay, western blot and lipolysis assay. A combinatorial RNA sequencing (RNAseq) and ChIP qPCR approach was used to identify target genes regulated by HAND2. In vivo, we created a conditional adipocyte Hand2 deletion mouse model using Cre under control of the Adipoq promoter (Hand2AdipoqCre) and performed a large panel of metabolic tests. RESULTS: We found that HAND2 is an obesity-linked white adipocyte transcription factor regulated by glucocorticoids that was necessary but insufficient for adipocyte differentiation in vitro. In a large cohort of humans, WAT HAND2 expression was correlated to BMI. The HAND2 gene was enriched in white adipocytes compared with brown, induced early in differentiation and responded to dexamethasone (DEX), a typical glucocorticoid receptor (GR, encoded by NR3C1) agonist. Silencing of NR3C1 in hMADS cells or deletion of GR in a transgenic conditional mouse model results in diminished HAND2 expression, establishing that adipocyte HAND2 is regulated by glucocorticoids via GR in vitro and in vivo. Furthermore, we identified gene clusters indirectly regulated by the GR-HAND2 pathway. Interestingly, silencing of HAND2 impaired adipocyte differentiation in hMADS and primary mouse adipocytes. However, a conditional adipocyte Hand2 deletion mouse model using Cre under control of the Adipoq promoter did not mirror these effects on adipose tissue differentiation, indicating that HAND2 was required at stages prior to Adipoq expression. CONCLUSIONS/INTERPRETATION: In summary, our study identifies HAND2 as a novel obesity-linked adipocyte transcription factor, highlighting new mechanisms of GR-dependent adipogenesis in humans and mice. DATA AVAILABILITY: Array data have been submitted to the GEO database at NCBI (GSE148699).


Subject(s)
Adipocytes/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation/physiology , Glucocorticoids/pharmacology , Obesity/genetics , Transcription Factors/genetics , Adipogenesis/physiology , Adipose Tissue, Brown/metabolism , Adult , Aged , Animals , Cross-Sectional Studies , Female , Gene Silencing , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Real-Time Polymerase Chain Reaction , Signal Transduction , Young Adult
3.
Ann Rheum Dis ; 77(11): 1610-1618, 2018 11.
Article in English | MEDLINE | ID: mdl-29997111

ABSTRACT

BACKGROUND: Glucocorticoid (GC) therapy is frequently used to treat rheumatoid arthritis due to potent anti-inflammatory actions of GCs. Direct actions of GCs on immune cells were suggested to suppress inflammation. OBJECTIVES: Define the role of the glucocorticoid receptor (GR) in stromal cells for suppression of inflammatory arthritis. METHODS: Bone marrow chimeric mice lacking the GR in the hematopoietic or stromal compartment, respectively, and mice with impaired GR dimerisation (GRdim) were analysed for their response to dexamethasone (DEX, 1 mg/kg) treatment in serum transfer-induced arthritis (STIA). Joint swelling, cell infiltration (histology), cytokines, cell composition (flow cytometry) and gene expression were analysed and RNASeq of wild type and GRdim primary murine fibroblast-like synoviocytes (FLS) was performed. RESULTS: GR deficiency in immune cells did not impair GC-mediated suppression of STIA. In contrast, mice with GR-deficient or GR dimerisation-impaired stromal cells were resistant to GC treatment, despite efficient suppression of cytokines. Intriguingly, in mice with impaired GR function in the stromal compartment, GCs failed to stimulate non-classical, non-activated macrophages (Ly6Cneg, MHCIIneg) and associated anti-inflammatory markers CD163, CD36, AnxA1, MerTK and Axl. Mice with GR deficiency in FLS were partially resistant to GC-induced suppression of STIA. Accordingly, RNASeq analysis of DEX-treated GRdim FLS revealed a distinct gene signature indicating enhanced activity and a failure to reduce macrophage inflammatory protein (Mip)-1α and Mip-1ß. CONCLUSION: We report a novel anti-inflammatory mechanism of GC action that involves GR dimerisation-dependent gene regulation in non-immune stromal cells, presumably FLS. FLS control non-classical, anti-inflammatory polarisation of macrophages that contributes to suppression of inflammation in arthritis.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Arthritis, Experimental/drug therapy , Arthritis, Rheumatoid/drug therapy , Dexamethasone/therapeutic use , Glucocorticoids/therapeutic use , Receptors, Glucocorticoid/physiology , Stromal Cells/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Arthritis, Rheumatoid/metabolism , Arthritis, Rheumatoid/pathology , Cytokines/biosynthesis , Dexamethasone/pharmacology , Dimerization , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Metabolism, Inborn Errors/metabolism , Metabolism, Inborn Errors/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Glucocorticoid/deficiency , Receptors, Glucocorticoid/metabolism , Stromal Cells/drug effects , Synoviocytes/drug effects , Synoviocytes/metabolism , Transplantation Chimera
4.
Clin Endocrinol (Oxf) ; 83(4): 441-8, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25627931

ABSTRACT

Glucocorticoid hormones are essential for life in vertebrates. They act through the glucocorticoid receptor (GR), which is expressed in virtually all cells of the human body. Yet the actions of glucocorticoids (GCs) are specific to particular cell types. Broadly GCs regulate carbohydrate metabolism, inflammation, stress and cell fate. Synthetic GCs are widely used in medicine and are by far the most frequent cause of Cushing's syndrome in routine practice. The advent of novel drugs targeting the GR offers new opportunities to treat patients with immune, or malignant disease, and may also offer new opportunities to manage patients with adrenal insufficiency also. This review covers the latest understanding of how GCs work, how their actions are affected by disease, and where the new drugs may take us.


Subject(s)
Receptors, Glucocorticoid/metabolism , Adrenal Insufficiency/genetics , Adrenal Insufficiency/metabolism , Animals , Glucocorticoids/metabolism , Humans , Receptors, Glucocorticoid/genetics
5.
Hypertension ; 80(11): 2333-2344, 2023 11.
Article in English | MEDLINE | ID: mdl-37646167

ABSTRACT

BACKGROUND: Cushing syndrome (CS) is a rare disease caused by excess cortisol levels with high cardiovascular morbidity and mortality. Hypertension in CS promotes hypercortisolism-associated cardiovascular events. Adipose tissue is a highly plastic tissue with most cell types strongly affected by the excess cortisol exposure. We hypothesized that the molecular and cellular changes of periadrenal adipose tissue in response to cortisol excess impact systemic blood pressure levels in patients with CS. METHODS: We investigated gene expression signatures in periadrenal adipose tissue from patients with adrenal CS collected during adrenal surgery. RESULTS: During active CS we observed a downregulation of gene programs associated with inflammation in periadrenal adipose tissue. In addition, we observed a clustering of the patients based on tissue gene expression profiles into 2 groups that differed in blood pressure levels (CS low blood pressure and CS high blood pressure). The 2 clusters showed significant differences in gene expression pattens of the renin-angiotensin-aldosterone-system. Renin was the strongest regulated gene compared with control patients and its expression correlated with increased blood pressure observed in our patients with CS. In the CS high blood pressure group, systemic renin plasma levels were suppressed indicative of an abnormal blood pressure associated with periadrenal adipose tissue renin-angiotensin-aldosterone-system activation. CONCLUSIONS: Here, we show for the first time a relevant association of the local renin-angiotensin-aldosterone-system and systemic blood pressure levels in patients with CS. Patients from the CS high blood pressure group still had increased blood pressure levels after 6 months in remission, highlighting the importance of local tissue effects on long-term systemic effects observed in CS.


Subject(s)
Cushing Syndrome , Hypertension , Humans , Renin , Cushing Syndrome/complications , Cushing Syndrome/genetics , Transcriptome , Aldosterone , Hydrocortisone , Renin-Angiotensin System/physiology , Hypertension/metabolism , Blood Pressure/genetics , Adipose Tissue , Angiotensins/metabolism
6.
Endocr Rev ; 44(6): 975-1011, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37253115

ABSTRACT

It is estimated that 2% to 3% of the population are currently prescribed systemic or topical glucocorticoid treatment. The potent anti-inflammatory action of glucocorticoids to deliver therapeutic benefit is not in doubt. However, the side effects associated with their use, including central weight gain, hypertension, insulin resistance, type 2 diabetes (T2D), and osteoporosis, often collectively termed iatrogenic Cushing's syndrome, are associated with a significant health and economic burden. The precise cellular mechanisms underpinning the differential action of glucocorticoids to drive the desirable and undesirable effects are still not completely understood. Faced with the unmet clinical need to limit glucocorticoid-induced adverse effects alongside ensuring the preservation of anti-inflammatory actions, several strategies have been pursued. The coprescription of existing licensed drugs to treat incident adverse effects can be effective, but data examining the prevention of adverse effects are limited. Novel selective glucocorticoid receptor agonists and selective glucocorticoid receptor modulators have been designed that aim to specifically and selectively activate anti-inflammatory responses based upon their interaction with the glucocorticoid receptor. Several of these compounds are currently in clinical trials to evaluate their efficacy. More recently, strategies exploiting tissue-specific glucocorticoid metabolism through the isoforms of 11ß-hydroxysteroid dehydrogenase has shown early potential, although data from clinical trials are limited. The aim of any treatment is to maximize benefit while minimizing risk, and within this review we define the adverse effect profile associated with glucocorticoid use and evaluate current and developing strategies that aim to limit side effects but preserve desirable therapeutic efficacy.


Subject(s)
Diabetes Mellitus, Type 2 , Insulin Resistance , Humans , Glucocorticoids/adverse effects , Glucocorticoids/metabolism , Receptors, Glucocorticoid , Diabetes Mellitus, Type 2/drug therapy , Anti-Inflammatory Agents/pharmacology , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism
7.
Nat Commun ; 14(1): 2271, 2023 04 20.
Article in English | MEDLINE | ID: mdl-37080971

ABSTRACT

Insulin resistance (IR) during obesity is linked to adipose tissue macrophage (ATM)-driven inflammation of adipose tissue. Whether anti-inflammatory glucocorticoids (GCs) at physiological levels modulate IR is unclear. Here, we report that deletion of the GC receptor (GR) in myeloid cells, including macrophages in mice, aggravates obesity-related IR by enhancing adipose tissue inflammation due to decreased anti-inflammatory ATM leading to exaggerated adipose tissue lipolysis and severe hepatic steatosis. In contrast, GR deletion in Kupffer cells alone does not alter IR. Co-culture experiments show that the absence of GR in macrophages directly causes reduced phospho-AKT and glucose uptake in adipocytes, suggesting an important function of GR in ATM. GR-deficient macrophages are refractory to alternative ATM-inducing IL-4 signaling, due to reduced STAT6 chromatin loading and diminished anti-inflammatory enhancer activation. We demonstrate that GR has an important function in macrophages during obesity by limiting adipose tissue inflammation and lipolysis to promote insulin sensitivity.


Subject(s)
Glucocorticoids , Insulin Resistance , Animals , Mice , Glucocorticoids/pharmacology , Insulin Resistance/genetics , Anti-Inflammatory Agents/pharmacology , Adipose Tissue , Macrophages , Obesity/genetics , Inflammation , Mice, Inbred C57BL
8.
FEBS Lett ; 596(20): 2617-2629, 2022 10.
Article in English | MEDLINE | ID: mdl-35997656

ABSTRACT

Regulation of cellular catabolic metabolism in immune cells has recently become a major concept for resolution of inflammation. Nuclear receptors (NRs), including peroxisome proliferator activator receptors, 1,25-dihydroxyvitamin D (3) receptor, liver X receptors, glucocorticoid receptors, oestrogen-related receptor α and nuclear receptor 4A1, have been identified as major modulators of inflammation, affecting innate immune cells, such as macrophages. Evidence emerges on how NRs regulate cellular metabolism in macrophages during inflammatory processes and contribute to the resolution of inflammation. This could have new implications for our understanding of how NRs shape immune responses and inform anti-inflammatory drug design. This review will highlight the recent developments about NRs and their role in cellular metabolism in macrophages.


Subject(s)
Orphan Nuclear Receptors , Receptors, Glucocorticoid , Humans , Orphan Nuclear Receptors/metabolism , Receptors, Glucocorticoid/metabolism , Peroxisome Proliferators/metabolism , Liver X Receptors/genetics , Liver X Receptors/metabolism , Macrophages/metabolism , Inflammation/metabolism
9.
Sci Signal ; 15(726): eabm4452, 2022 03 22.
Article in English | MEDLINE | ID: mdl-35316097

ABSTRACT

Mutations that activate members of the RAS family of GTPases are associated with various cancers and drive tumor growth. The glucocorticoid receptor (GR), a member of the nuclear receptor family, has been proposed to interact with and inhibit the activation of components of the PI3K-AKT and MAPK pathways downstream of RAS. In the absence of activating ligands, we found that GR was present in cytoplasmic KRAS-containing complexes and inhibited the activation of wild-type and oncogenic KRAS in mouse embryonic fibroblasts and human lung cancer A549 cells. The DNA binding domain of GR was involved in the interaction with KRAS, but GR-dependent inhibition of RAS activation did not depend on the nuclear translocation of GR. The addition of ligand released GR-dependent inhibition of RAS, AKT, the MAPK p38, and the MAPKK MEK. CRISPR-Cas9-mediated deletion of GR in A549 cells enhanced tumor growth in xenografts in mice. Patient samples of non-small cell lung carcinomas showed lower expression of NR3C1, the gene encoding GR, compared to adjacent normal tissues and lower NR3C1 expression correlated with a worse disease outcome. These results suggest that glucocorticoids prevent the ability of GR to limit tumor growth by inhibiting RAS activation, which has potential implications for the use of glucocorticoids in patients with cancer.


Subject(s)
Lung Neoplasms , Receptors, Glucocorticoid , Animals , Cell Proliferation , Fibroblasts/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Mice , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism
10.
Mol Metab ; 57: 101424, 2022 03.
Article in English | MEDLINE | ID: mdl-34954109

ABSTRACT

OBJECTIVES: Glucocorticoids (GCs) are one of the most widely prescribed anti-inflammatory drugs. By acting through their cognate receptor, the glucocorticoid receptor (GR), GCs downregulate the expression of pro-inflammatory genes and upregulate the expression of anti-inflammatory genes. Metabolic pathways have recently been identified as key parts of both the inflammatory activation and anti-inflammatory polarization of macrophages, immune cells responsible for acute inflammation and tissue repair. It is currently unknown whether GCs control macrophage metabolism, and if so, to what extent metabolic regulation by GCs confers anti-inflammatory activity. METHODS: Using transcriptomic and metabolomic profiling of macrophages, we identified GC-controlled pathways involved in metabolism, especially in mitochondrial function. RESULTS: Metabolic analyses revealed that GCs repress glycolysis in inflammatory myeloid cells and promote tricarboxylic acid (TCA) cycle flux, promoting succinate metabolism and preventing intracellular accumulation of succinate. Inhibition of ATP synthase attenuated GC-induced transcriptional changes, likely through stalling of TCA cycle anaplerosis. We further identified a glycolytic regulatory transcription factor, HIF1α, as regulated by GCs, and as a key regulator of GC responsiveness during inflammatory challenge. CONCLUSIONS: Our findings link metabolism to gene regulation by GCs in macrophages.


Subject(s)
Citric Acid Cycle , Glucocorticoids , Glucocorticoids/pharmacology , Humans , Inflammation/metabolism , Macrophages/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism
11.
Cell Metab ; 34(3): 473-486.e9, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35120589

ABSTRACT

Fasting metabolism and immunity are tightly linked; however, it is largely unknown how immune cells contribute to metabolic homeostasis during fasting in healthy subjects. Here, we combined cell-type-resolved genomics and computational approaches to map crosstalk between hepatocytes and liver macrophages during fasting. We identified the glucocorticoid receptor (GR) as a key driver of fasting-induced reprogramming of the macrophage secretome including fasting-suppressed cytokines and showed that lack of macrophage GR impaired induction of ketogenesis during fasting as well as endotoxemia. Mechanistically, macrophage GR suppressed the expression of tumor necrosis factor (TNF) and promoted nuclear translocation of hepatocyte GR to activate a fat oxidation/ketogenesis-related gene program, cooperatively induced by GR and peroxisome proliferator-activated receptor alpha (PPARα) in hepatocytes. Together, our results demonstrate how resident liver macrophages directly influence ketogenesis in hepatocytes, thereby also outlining a strategy by which the immune system can set the metabolic tone during inflammatory disease and infection.


Subject(s)
Fasting , Receptors, Glucocorticoid , Animals , Fasting/metabolism , Hepatocytes/metabolism , Humans , Ketone Bodies/metabolism , Liver/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , PPAR alpha/metabolism , Receptors, Glucocorticoid/metabolism
12.
Front Immunol ; 10: 1591, 2019.
Article in English | MEDLINE | ID: mdl-31354730

ABSTRACT

Inflammation is a complex process which is highly conserved among species. Inflammation occurs in response to injury, infection, and cancer, as an allostatic mechanism to return the tissue and to return the organism back to health and homeostasis. Excessive, or chronic inflammation is associated with numerous diseases, and thus strategies to combat run-away inflammation is required. Anti-inflammatory drugs were therefore developed to switch inflammation off. However, the inflammatory response may be beneficial for the organism, in particular in the case of sterile tissue injury. The inflammatory response can be divided into several parts. The first step is the mounting of the inflammatory reaction itself, characterized by the presence of pro-inflammatory cytokines, and the infiltration of immune cells into the injured area. The second step is the resolution phase, where immune cells move toward an anti-inflammatory phenotype and decrease the secretion of pro-inflammatory cytokines. The last stage of inflammation is the regeneration process, where the tissue is rebuilt. Innate immune cells are major actors in the inflammatory response, of which, macrophages play an important role. Macrophages are highly sensitive to a large number of environmental stimuli, and can adapt their phenotype and function on demand. This change in phenotype in response to the environment allow macrophages to be involved in all steps of inflammation, from the first mounting of the pro-inflammatory response to the post-damage tissue repair.


Subject(s)
Glucocorticoids/metabolism , Macrophages/metabolism , Wound Healing/physiology , Animals , Humans , Inflammation/metabolism , Phenotype , Signal Transduction/physiology
13.
Front Immunol ; 10: 1859, 2019.
Article in English | MEDLINE | ID: mdl-31440248

ABSTRACT

For many decades, glucocorticoids have been widely used as the gold standard treatment for inflammatory conditions. Unfortunately, their clinical use is limited by severe adverse effects such as insulin resistance, cardiometabolic diseases, muscle and skin atrophies, osteoporosis, and depression. Glucocorticoids exert their effects by binding to the Glucocorticoid Receptor (GR), a ligand-activated transcription factor which both positively, and negatively regulates gene expression. Extensive research during the past several years has uncovered novel mechanisms by which the GR activates and represses its target genes. Genome-wide studies and mouse models have provided valuable insight into the molecular mechanisms of inflammatory gene regulation by GR. This review focusses on newly identified target genes and GR co-regulators that are important for its anti-inflammatory effects in innate immune cells, as well as mutations within the GR itself that shed light on its transcriptional activity. This research progress will hopefully serve as the basis for the development of safer immune suppressants with reduced side effect profiles.


Subject(s)
Gene Expression Regulation/drug effects , Gene Expression Regulation/immunology , Glucocorticoids/immunology , Immunosuppressive Agents/immunology , Receptors, Glucocorticoid/immunology , Animals , Glucocorticoids/pharmacology , Humans , Immunosuppressive Agents/pharmacology
14.
J Clin Invest ; 128(10): 4454-4471, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30179226

ABSTRACT

The glucocorticoid receptor (GR) is a major drug target in inflammatory disease. However, chronic glucocorticoid (GC) treatment leads to disordered energy metabolism, including increased weight gain, adiposity, and hepatosteatosis - all programs modulated by the circadian clock. We demonstrated that while antiinflammatory GC actions were maintained irrespective of dosing time, the liver was significantly more GC sensitive during the day. Temporal segregation of GC action was underpinned by a physical interaction of GR with the circadian transcription factor REVERBa and co-binding with liver-specific hepatocyte nuclear transcription factors (HNFs) on chromatin. REVERBa promoted efficient GR recruitment to chromatin during the day, acting in part by maintaining histone acetylation, with REVERBa-dependent GC responses providing segregation of carbohydrate and lipid metabolism. Importantly, deletion of Reverba inverted circadian liver GC sensitivity and protected mice from hepatosteatosis induced by chronic GC administration. Our results reveal a mechanism by which the circadian clock acts through REVERBa in liver on elements bound by HNF4A/HNF6 to direct GR action on energy metabolism.


Subject(s)
Chromatin/metabolism , Circadian Clocks/drug effects , Fatty Liver/metabolism , Glucocorticoids/adverse effects , Liver/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Animals , Chromatin/genetics , Chromatin/pathology , Circadian Clocks/genetics , Energy Metabolism/drug effects , Energy Metabolism/genetics , Fatty Liver/chemically induced , Fatty Liver/genetics , Fatty Liver/pathology , Glucocorticoids/pharmacology , HEK293 Cells , Humans , Liver/pathology , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism
15.
J Endocrinol ; 223(2): 155-66, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25161081

ABSTRACT

Glucocorticoids (Gc) are potent anti-inflammatory agents with wide clinical application. We have previously shown that increased serum concentration significantly attenuates regulation of a simple Gc-responsive reporter. We now find that glucocorticoid receptor (GR) regulation of some endogenous transactivated but not transrepressed genes is impaired, suggesting template specificity. Serum did not directly affect GR expression, activity or trafficking, implicating GR crosstalk with other signalling pathways. Indeed, a JNK inhibitor completely abolished the serum effect. We identified the Gc modulating serum component as cholesterol. Cholesterol loading mimicked the serum effect, which was readily reversed by JNK inhibition. Chelation of serum cholesterol with methyl-ß-cyclodextrin or inhibition of cellular cholesterol synthesis with simvastatin potentiated the Gc response. To explore the effect in vivo we used ApoE(-/-) mice, a model of hypercholesterolaemia. Consistent with our in vitro studies, we find no impact of elevated cholesterol on the expression of GR, or on the hypothalamic-pituitary-adrenal axis, measured by dexamethasone suppression test. Instead we find selective Gc resistance on some hepatic target genes in ApoE(-/-) mice. Therefore, we have discovered an unexpected role for cholesterol as a selective modulator of Gc action in vivo. Taken together these findings reveal a new environmental constraint on Gc action with relevance to both inflammation and cancer.


Subject(s)
Cholesterol/blood , Drug Resistance , Glucocorticoids/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , Animals , Apolipoproteins E/genetics , Enzyme Activation/drug effects , Female , HeLa Cells , Humans , Metabolism, Inborn Errors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Glucocorticoid/deficiency , Receptors, Glucocorticoid/genetics
SELECTION OF CITATIONS
SEARCH DETAIL