Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.751
Filter
Add more filters

Publication year range
1.
Immunity ; 48(2): 286-298.e6, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29396162

ABSTRACT

Glucocorticoids are steroid hormones with strong anti-inflammatory and immunosuppressive effects that are produced in a diurnal fashion. Although glucocorticoids have the potential to induce interleukin-7 receptor (IL-7R) expression in T cells, whether they control T cell homeostasis and responses at physiological concentrations remains unclear. We found that glucocorticoid receptor signaling induces IL-7R expression in mouse T cells by binding to an enhancer of the IL-7Rα locus, with a peak at midnight and a trough at midday. This diurnal induction of IL-7R supported the survival of T cells and their redistribution between lymph nodes, spleen, and blood by controlling expression of the chemokine receptor CXCR4. In mice, T cell accumulation in the spleen at night enhanced immune responses against soluble antigens and systemic bacterial infection. Our results reveal the immunoenhancing role of glucocorticoids in adaptive immunity and provide insight into how immune function is regulated by the diurnal rhythm.


Subject(s)
Circadian Rhythm/physiology , Glucocorticoids/pharmacology , Receptors, CXCR4/physiology , Receptors, Interleukin-7/physiology , T-Lymphocytes/immunology , Animals , Cells, Cultured , Chemokine CXCL12/biosynthesis , Female , Immunologic Memory , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Receptors, Glucocorticoid/physiology
2.
PLoS Genet ; 17(8): e1009737, 2021 08.
Article in English | MEDLINE | ID: mdl-34375333

ABSTRACT

Ultradian glucocorticoid rhythms are highly conserved across mammalian species, however, their functional significance is not yet fully understood. Here we demonstrate that pulsatile corticosterone replacement in adrenalectomised rats induces a dynamic pattern of glucocorticoid receptor (GR) binding at ~3,000 genomic sites in liver at the pulse peak, subsequently not found during the pulse nadir. In contrast, constant corticosterone replacement induced prolonged binding at the majority of these sites. Additionally, each pattern further induced markedly different transcriptional responses. During pulsatile treatment, intragenic occupancy by active RNA polymerase II exhibited pulsatile dynamics with transient changes in enrichment, either decreased or increased depending on the gene, which mostly returned to baseline during the inter-pulse interval. In contrast, constant corticosterone exposure induced prolonged effects on RNA polymerase II occupancy at the majority of gene targets, thus acting as a sustained regulatory signal for both transactivation and repression of glucocorticoid target genes. The nett effect of these differences were consequently seen in the liver transcriptome as RNA-seq analysis indicated that despite the same overall amount of corticosterone infused, twice the number of transcripts were regulated by constant corticosterone infusion, when compared to pulsatile. Target genes that were found to be differentially regulated in a pattern-dependent manner were enriched in functional pathways including carbohydrate, cholesterol, glucose and fat metabolism as well as inflammation, suggesting a functional role for dysregulated glucocorticoid rhythms in the development of metabolic dysfunction.


Subject(s)
Corticosterone/pharmacology , Liver/pathology , Receptors, Glucocorticoid/metabolism , Animals , Gene Expression/genetics , Gene Expression Profiling/methods , Gene Expression Regulation/genetics , Glucocorticoids/metabolism , Liver/metabolism , Male , Periodicity , Protein Transport/genetics , RNA Polymerase II/genetics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptors, Glucocorticoid/physiology , Transcriptional Activation/genetics , Transcriptome/genetics
3.
Blood ; 137(4): 500-512, 2021 01 28.
Article in English | MEDLINE | ID: mdl-33507291

ABSTRACT

Glucocorticoid (GC) resistance remains a clinical challenge in pediatric acute lymphoblastic leukemia where response to GC is a reliable prognostic indicator. To identify GC resistance pathways, we conducted a genome-wide, survival-based, short hairpin RNA screen in murine T-cell acute lymphoblastic leukemia (T-ALL) cells. Genes identified in the screen interfere with cyclic adenosine monophosphate (cAMP) signaling and are underexpressed in GC-resistant or relapsed ALL patients. Silencing of the cAMP-activating Gnas gene interfered with GC-induced gene expression, resulting in dexamethasone resistance in vitro and in vivo. We demonstrate that cAMP signaling synergizes with dexamethasone to enhance cell death in GC-resistant human T-ALL cells. We find the E prostanoid receptor 4 expressed in T-ALL samples and demonstrate that prostaglandin E2 (PGE2) increases intracellular cAMP, potentiates GC-induced gene expression, and sensitizes human T-ALL samples to dexamethasone in vitro and in vivo. These findings identify PGE2 as a target for GC resensitization in relapsed pediatric T-ALL.


Subject(s)
Cyclic AMP/physiology , Dexamethasone/pharmacology , Dinoprostone/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Second Messenger Systems/drug effects , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Child , Chromogranins/antagonists & inhibitors , Colforsin/pharmacology , Cyclic AMP/pharmacology , Dexamethasone/administration & dosage , Dinoprostone/administration & dosage , Dinoprostone/antagonists & inhibitors , Dinoprostone/physiology , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/physiology , Female , GTP-Binding Protein alpha Subunits, Gs/antagonists & inhibitors , GTP-Binding Protein alpha Subunits, Gs/deficiency , Gene Expression Regulation, Leukemic/drug effects , Humans , Male , Mice , Models, Animal , Molecular Targeted Therapy , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Radiation Chimera , Receptors, Glucocorticoid/biosynthesis , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/physiology , Receptors, Prostaglandin E, EP4 Subtype/biosynthesis , Receptors, Prostaglandin E, EP4 Subtype/genetics , Xenograft Model Antitumor Assays
4.
Neurobiol Learn Mem ; 203: 107797, 2023 09.
Article in English | MEDLINE | ID: mdl-37385522

ABSTRACT

Glucocorticoid receptors (GRs) of the basolateral amygdala (BLA) play an important role in memory reconsolidation. The present study investigated the role of the BLA GRs in the late reconsolidation of fear memory using an inhibitory avoidance (IA) task in male Wistar rats. Stainless steel cannulae were implanted bilaterally into the BLA of the rats. After 7 days of recovery, the animals were trained in a one-trial IA task (1 mA, 3 s). In Experiment One, 48 h after the training session, the animals received 3 systemic doses of corticosterone (CORT; 1, 3, or 10 mg/kg, i.p.) followed by an intra-BLA microinjection of the vehicle (0.3 µl/side) at different time points (immediately, 12, or 24 h) after memory reactivation. Memory reactivation was performed by returning the animals to the light compartment while the sliding door was open. No shock was delivered during memory reactivation. CORT (10 mg/kg) injection 12 h after memory reactivation most effectively impaired the late memory reconsolidation (LMR). In the second part of Experiment One, immediately, 12, or 24 h after memory reactivation, GR antagonist RU38486 (RU; 1 ng/0.3 µl/side) was injected into BLA following a systemic injection of CORT (10 mg/kg) to examine whether it would block the CORT effect. RU inhibited the impairing effects of CORT on LMR. In Experiment Two, the animals received CORT (10 mg/kg) with time windows immediately, 3, 6, 12, and 24 h after memory reactivation. Again, CORT (10 mg/kg) injection 12 h after memory reactivation impaired LMR. Memory reactivation was performed in the third Experiment, 7, 14, 28, or 56 days after the training session. Injection of CORT (10 mg/kg) 12 h later had no significant effect on the LMR. The impairing effect of CORT was seen only in 2-day-old but not 7, 14, 28, and 56-day-old memories. GRs located in BLA seem to play an important role in the LMR of young memory, as with increasing the age of memories, they become less sensitive to manipulation.


Subject(s)
Basolateral Nuclear Complex , Rats , Male , Animals , Receptors, Glucocorticoid/physiology , Corticosterone/pharmacology , Rats, Wistar , Fear
5.
PLoS Genet ; 16(5): e1008757, 2020 05.
Article in English | MEDLINE | ID: mdl-32379754

ABSTRACT

In the last decades in vitro studies highlighted the potential for crosstalk between Hypoxia-Inducible Factor-(HIF) and glucocorticoid-(GC) signalling pathways. However, how this interplay precisely occurs in vivo is still debated. Here, we use zebrafish larvae (Danio rerio) to elucidate how and to what degree hypoxic signalling affects the endogenous glucocorticoid pathway and vice versa, in vivo. Firstly, our results demonstrate that in the presence of upregulated HIF signalling, both glucocorticoid receptor (Gr) responsiveness and endogenous cortisol levels are repressed in 5 days post fertilisation larvae. In addition, despite HIF activity being low at normoxia, our data show that it already impedes both glucocorticoid activity and levels. Secondly, we further analysed the in vivo contribution of glucocorticoids to HIF activity. Interestingly, our results show that both glucocorticoid receptor (GR) and mineralocorticoid receptor (MR) play a key role in enhancing it. Finally, we found indications that glucocorticoids promote HIF signalling via multiple routes. Cumulatively, our findings allowed us to suggest a model for how this crosstalk occurs in vivo.


Subject(s)
Glucocorticoids/pharmacology , Hypoxia-Inducible Factor 1/physiology , Receptor Cross-Talk/physiology , Zebrafish , Animals , Animals, Genetically Modified , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Embryo, Nonmammalian , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Glucocorticoids/metabolism , Hypoxia-Inducible Factor 1/metabolism , Larva/genetics , Larva/metabolism , Receptor Cross-Talk/drug effects , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Tumor Suppressor Proteins/genetics , Zebrafish/embryology , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/genetics
6.
Adv Exp Med Biol ; 1390: 109-122, 2022.
Article in English | MEDLINE | ID: mdl-36107315

ABSTRACT

Nuclear receptors play a central role in both energy metabolism and cardiomyocyte death and survival in the heart. Recent evidence suggests they may also influence cardiomyocyte endowment. Although several members of the nuclear receptor family play key roles in heart maturation (including thyroid hormone receptors) and cardiac metabolism, here, the focus will be on the corticosteroid receptors, the glucocorticoid receptor (GR) and mineralocorticoid receptor (MR). The heart is an important target for the actions of corticosteroids, yet the homeostatic role of GR and MR in the healthy heart has been elusive. However, MR antagonists are important in the treatment of heart failure, a condition associated with mitochondrial dysfunction and energy failure in cardiomyocytes leading to mitochondria-initiated cardiomyocyte death (Ingwall and Weiss, Circ Res 95:135-145, 2014; Ingwall , Cardiovasc Res 81:412-419, 2009; Zhou and Tian , J Clin Invest 128:3716-3726, 2018). In contrast, animal studies suggest GR activation in cardiomyocytes has a cardioprotective role, including in heart failure.


Subject(s)
Heart Failure , Receptors, Mineralocorticoid , Animals , Heart Failure/metabolism , Myocytes, Cardiac/metabolism , Receptors, Glucocorticoid/physiology , Receptors, Thyroid Hormone/metabolism
7.
J Neurosci ; 40(47): 9148-9162, 2020 11 18.
Article in English | MEDLINE | ID: mdl-33087471

ABSTRACT

A single stressful event can cause morphologic and functional changes in neurons and even malfunction of vascular systems, which can lead to acute stress disorder or post-traumatic stress disorder. However, there is a lack of evidence regarding how acute stress impacts neuronal activity, the concurrent vascular response, and the relationship between these two factors, which is defined as neurovascular coupling. Here, using in vivo two-photon imaging, we found that NMDA-evoked calcium transients of excitatory neurons were impaired and that vasodilation of penetrating arterioles was concomitantly disrupted in acutely stressed male mice. Furthermore, acute stress altered the relationship between excitatory neuronal calcium coherence and vascular responses. By measuring NMDA-evoked excitatory and inhibitory neuronal calcium activity in acute brain slices, we confirmed that neuronal coherence both between excitatory neurons and between excitatory and inhibitory neurons was reduced by acute stress but restored by blockade of glucocorticoid receptor signaling. Furthermore, the ratio of sEPSCs to sIPSCs was altered by acute stress, suggesting that the excitation-inhibition balance was disrupted by acute stress. In summary, in vivo, ex vivo, and whole-cell recording studies demonstrate that acute stress modifies excitatory-inhibitory neuronal coherence, disrupts the excitation-inhibition balance, and causes consequent neurovascular coupling changes, providing critical insights into the neural mechanism of stress-induced disorders.SIGNIFICANCE STATEMENT Acute stress can cause pathologic conditions, such as acute stress disorder and post-traumatic stress disorder, by affecting the functions of neurons and blood vessels. However, investigations into the impacts of acute stress on neurovascular coupling, the tight connection between local neural activity and subsequent blood flow changes, are lacking. Through investigations at the in vivo, ex vivo, and whole-cell recording levels, we found that acute stress alters the NMDA-evoked vascular response, impairs the function and coherence of excitatory and inhibitory neurons, and disrupts the excitatory and inhibitory balance. These novel findings provide insights into the relevance of the excitatory-inhibitory balance, neuronal coherence, and neurovascular coupling to stress-induced disorders.


Subject(s)
Neurons/pathology , Neurovascular Coupling/physiology , Stress, Psychological/pathology , Acute Disease , Animals , Calcium Signaling , Cerebrovascular Circulation/physiology , Corticosterone/physiology , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , N-Methylaspartate/pharmacology , Neural Inhibition , Patch-Clamp Techniques , Receptors, Glucocorticoid/physiology , Restraint, Physical
8.
Mol Cell ; 49(1): 158-71, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23159735

ABSTRACT

How the glucocorticoid receptor (GR) activates some genes while potently repressing others remains an open question. There are three current models for suppression: transrepression via GR tethering to AP-1/NF-κB sites, direct GR association with inhibitory elements (nGREs), and GR recruitment of the corepressor GRIP1. To gain insights into GR suppression, we used genomic analyses and genome-wide profiling of GR, p65, and c-Jun in LPS-stimulated macrophages. We show that GR mediates both activation and repression at tethered sites, GREs, and GRIP1-bound elements, indicating that motif classification is insufficient to predict regulatory polarity of GR binding. Interestingly, sites of GR repression utilize GRIP1's corepressor function and display reduced histone acetylation. Together, these findings suggest that while GR occupancy confers hormone responsiveness, the receptor itself may not participate in the regulatory effects. Furthermore, transcriptional outcome is not established by sequence but is influenced by epigenetic regulators, context, and other unrecognized regulatory determinants.


Subject(s)
Epigenesis, Genetic , Genome , Inflammation/genetics , Receptors, Glucocorticoid/physiology , Acetylation , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Sequence , Animals , Base Sequence , Binding Sites , Cells, Cultured , Chromosome Mapping , Cluster Analysis , Consensus Sequence , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Histones/metabolism , Inflammation/metabolism , Interferon Regulatory Factor-3/metabolism , Lipopolysaccharides/pharmacology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Nerve Tissue Proteins/metabolism , Protein Binding , Protein Processing, Post-Translational , Receptors, Glucocorticoid/agonists , Receptors, Glucocorticoid/metabolism , Response Elements , Transcription Factors/genetics , Transcriptome
9.
Int J Mol Sci ; 22(7)2021 Mar 31.
Article in English | MEDLINE | ID: mdl-33807481

ABSTRACT

Glucocorticoids (GCs) act via the GC receptor (GR), a receptor ubiquitously expressed in the body where it drives a broad spectrum of responses within distinct cell types and tissues, which vary in strength and specificity. The variability of GR-mediated cell responses is further extended by the existence of GR isoforms, such as GRα and GRß, generated through alternative splicing mechanisms. While GRα is the classic receptor responsible for GC actions, GRß has been implicated in the impairment of GRα-mediated activities. Interestingly, in contrast to the popular belief that GRß actions are restricted to its dominant-negative effects on GRα-mediated responses, GRß has been shown to have intrinsic activities and "directly" regulates a plethora of genes related to inflammatory process, cell communication, migration, and malignancy, each in a GRα-independent manner. Furthermore, GRß has been associated with increased cell migration, growth, and reduced sensitivity to GC-induced apoptosis. We will summarize the current knowledge of GRß-mediated responses, with a focus on the GRα-independent/intrinsic effects of GRß and the associated non-canonical signaling pathways. Where appropriate, potential links to airway inflammatory diseases will be highlighted.


Subject(s)
Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/physiology , Alternative Splicing/drug effects , Dexamethasone/pharmacology , Glucocorticoids/pharmacology , Humans , Protein Isoforms
10.
Int J Mol Sci ; 22(20)2021 Oct 16.
Article in English | MEDLINE | ID: mdl-34681832

ABSTRACT

Stress and depression increase the risk of Type 2 Diabetes (T2D) development. Evidence demonstrates that the Glucocorticoid (GC) negative feedback is impaired (GC resistance) in T2D patients resulting in Hypothalamic-Pituitary-Adrenal (HPA) axis hyperactivity and hypercortisolism. High GCs, in turn, activate multiple aspects of glucose homeostasis in peripheral tissues leading to hyperglycemia. Elucidation of the underlying molecular mechanisms revealed that Glucocorticoid Receptor (GR) mediates the GC-induced dysregulation of glucose production, uptake and insulin signaling in GC-sensitive peripheral tissues, such as liver, skeletal muscle, adipose tissue, and pancreas. In contrast to increased GR peripheral sensitivity, an impaired GR signaling in Peripheral Blood Mononuclear Cells (PBMCs) of T2D patients, associated with hyperglycemia, hyperlipidemia, and increased inflammation, has been shown. Given that GR changes in immune cells parallel those in brain, the above data implicate that a reduced brain GR function may be the biological link among stress, HPA hyperactivity, hypercortisolism and hyperglycemia. GR polymorphisms have also been associated with metabolic disturbances in T2D while dysregulation of micro-RNAs-known to target GR mRNA-has been described. Collectively, GR has a crucial role in T2D, acting in a cell-type and context-specific manner, leading to either GC sensitivity or GC resistance. Selective modulation of GR signaling in T2D therapy warrants further investigation.


Subject(s)
Diabetes Mellitus, Type 2/physiopathology , Glucocorticoids/physiology , MicroRNAs/physiology , Polymorphism, Genetic , Receptors, Glucocorticoid/physiology , Signal Transduction , Animals , Cushing Syndrome , Depression/metabolism , Glucose/metabolism , Homeostasis , Humans , Hypothalamo-Hypophyseal System/physiology , Insulin/metabolism , Stress, Physiological
11.
Toxicol Appl Pharmacol ; 395: 114979, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32234517

ABSTRACT

Epidemiology suggests that adverse environmental exposure during pregnancy may predispose children to hypercholesterolemia in adulthood. This study aimed to demonstrate hypercholesterolemia induced by prenatal dexamethasone exposure (PDE) in adult male offspring rats and explore the intrauterine programming mechanisms. Pregnant Wistar rats were injected subcutaneously with dexamethasone (0, 0.1, 0.2, and 0.4 mg/kg∙d) from gestational days (GD) 9 to 21, and the serum and liver of the male offsprings were collected at GD21, postnatal week (PW) 12 and 28. Furthermore, the effects of dexamethasone on the expression of low-density lipoprotein receptor (LDLR) and its epigenetic mechanism was confirmed in the bone marrow mesenchymal stem cells (BMSCs) hepatoid differentiated cells and continuous hepatocyte line. PDE could reduce the birth weight of male offsprings, increase the serum total cholesterol (TCH) level in adult rats, and decrease the liver low-density lipoprotein receptor (LDLR) expression. Serum TCH level and liver LDLR expression were decreased in PDE male fetuses in utero (GD21). Moreover, PDE increased the translocation of the glucocorticoid receptor (GR) in the fetal liver, the expression of DiGeorge syndrome critical region 8 gene (DGCR8), the pre- and post-natal expression of miR-148a. The results of PDE offspring in vivo and in vitro exhibited similar changes. These changes could be reversed by overexpressing LDLR, inhibiting miR-148a or GR. PDE caused hypercholesterolemia in male adult offspring rats, which was mediated via dexamethasone activated intrauterine hepatic GR, enhanced the expression of DGCR8 and miR-148a, thereby reducing the expression of LDLR, leading to impaired liver cholesterol reverse transport function, and finally causing hypercholesterolemia in adult rats.


Subject(s)
Dexamethasone/adverse effects , Hypercholesterolemia/etiology , Hypercholesterolemia/metabolism , MicroRNAs/physiology , Prenatal Exposure Delayed Effects , Receptors, LDL/physiology , Animals , Dexamethasone/administration & dosage , Epigenesis, Genetic/drug effects , Female , Gene Expression/drug effects , Gestational Age , Liver/chemistry , Liver/embryology , Liver/metabolism , Male , MicroRNAs/genetics , Pregnancy , Rats , Rats, Wistar , Receptors, Glucocorticoid/drug effects , Receptors, Glucocorticoid/physiology , Receptors, LDL/genetics
12.
FASEB J ; 33(6): 7387-7402, 2019 06.
Article in English | MEDLINE | ID: mdl-30860871

ABSTRACT

Glucocorticoids (GCs) potently induce T-cell apoptosis in a GC receptor (GR)-dependent manner and are used to control lymphocyte function in clinical practice. However, its downstream pathways remain controversial. Here, we showed that GC-induced transcript 1 (GLCCI1) is a novel downstream molecule of the GC-GR cascade that acts as an antiapoptotic mediator in thymic T cells. GLCCI1 was highly phosphorylated and colocalized with microtubules in GLCCI1-transfected human embryonic kidney QBI293A cells. GR-dependent up-regulation of GLCCI1 was associated with GC-induced proapoptotic events in a cultured thymocyte cell line. However, GLCCI1 knockdown in a thymocyte cell line led to apoptosis. Consistently, transgenic mice overexpressing human GLCCI1 displayed enlarged thymi that consisted of larger numbers of thymocytes. Further molecular characterization showed that GLCCI1 bound to both dynein light chain LC8-type 1 (LC8) and its functional kinase, p21-protein activated kinase 1 (PAK1), thereby inhibiting the kinase activity of PAK1 toward LC8 phosphorylation, a crucial event in apoptotic signaling. GLCCI1 induction facilitated LC8 dimer formation and reduced Bim expression. Thus, GLCCI1 is a candidate factor involved in apoptosis regulation of thymic T cells.-Kiuchi, Z., Nishibori, Y., Kutsuna, S., Kotani, M., Hada, I., Kimura, T., Fukutomi, T., Fukuhara, D., Ito-Nitta, N., Kudo, A., Takata, T., Ishigaki, Y., Tomosugi, N., Tanaka, H., Matsushima, S., Ogasawara, S., Hirayama, Y., Takematsu, H., Yan, K. GLCCI1 is a novel protector against glucocorticoid-induced apoptosis in T cells.


Subject(s)
Apoptosis/physiology , Glucocorticoids/physiology , Receptors, Glucocorticoid/physiology , T-Lymphocytes/cytology , Amino Acid Sequence , Animals , Apoptosis/drug effects , Bcl-2-Like Protein 11/biosynthesis , Bcl-2-Like Protein 11/genetics , Cell Line , Cytoplasmic Dyneins/metabolism , Dimerization , Down-Regulation , Gene Knockdown Techniques , Glucocorticoids/pharmacology , Humans , Hypertrophy , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microtubules/metabolism , Phosphorylation , Protein Interaction Mapping , Protein Processing, Post-Translational , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/pharmacology , Receptors, Glucocorticoid/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction/physiology , Thymus Gland/pathology , p21-Activated Kinases/metabolism
13.
FASEB J ; 33(1): 126-139, 2019 01.
Article in English | MEDLINE | ID: mdl-29965797

ABSTRACT

The circadian clock is a critical regulator of immune function. We recently highlighted a role for the circadian clock in a mouse model of pulmonary inflammation. The epithelial clock protein Bmal1 was required to regulate neutrophil recruitment in response to inflammatory challenge. Bmal1 regulated glucocorticoid receptor (GR) recruitment to the neutrophil chemokine, CXC chemokine ligand 5 (CXCL5), providing a candidate mechanism. We now show that clock control of pulmonary neutrophilia persists without rhythmic glucocorticoid availability. Epithelial GR-null mice had elevated expression of proinflammatory chemokines in the lung under homeostatic conditions. However, deletion of GR in the bronchial epithelium blocked rhythmic CXCL5 production, identifying GR as required to confer circadian control to CXCL5. Surprisingly, rhythmic pulmonary neutrophilia persisted, despite nonrhythmic CXCL5 responses, indicating additional circadian control mechanisms. Deletion of GR in myeloid cells alone did not prevent circadian variation in pulmonary neutrophilia and showed reduced neutrophilic inflammation in response to dexamethasone treatment. These new data show GR is required to confer circadian control to some inflammatory chemokines, but that this alone is insufficient to prevent circadian control of neutrophilic inflammation in response to inhaled LPS, with additional control mechanisms arising in the myeloid cell lineage.-Ince, L. M., Zhang, Z., Beesley, S., Vonslow, R. M., Saer, B. R., Matthews, L. C., Begley, N., Gibbs, J. E., Ray, D. W., Loudon, A. S. I. Circadian variation in pulmonary inflammatory responses is independent of rhythmic glucocorticoid signaling in airway epithelial cells.


Subject(s)
Circadian Rhythm/immunology , Epithelial Cells/immunology , Macrophages, Peritoneal/immunology , Neutrophils/immunology , Pneumonia/immunology , Receptors, Glucocorticoid/physiology , Respiratory System/immunology , Animals , Cells, Cultured , Chemokine CXCL5/metabolism , Circadian Rhythm/drug effects , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Glucocorticoids/pharmacology , Lipopolysaccharides/pharmacology , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Macrophages, Peritoneal/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/pathology , Pneumonia/drug therapy , Pneumonia/metabolism , Pneumonia/pathology , Respiratory System/drug effects , Respiratory System/metabolism , Respiratory System/pathology , Signal Transduction
14.
PLoS Biol ; 15(12): e2002690, 2017 12.
Article in English | MEDLINE | ID: mdl-29283992

ABSTRACT

Response to antidepressant treatment in major depressive disorder (MDD) cannot be predicted currently, leading to uncertainty in medication selection, increasing costs, and prolonged suffering for many patients. Despite tremendous efforts in identifying response-associated genes in large genome-wide association studies, the results have been fairly modest, underlining the need to establish conceptually novel strategies. For the identification of transcriptome signatures that can distinguish between treatment responders and nonresponders, we herein submit a novel animal experimental approach focusing on extreme phenotypes. We utilized the large variance in response to antidepressant treatment occurring in DBA/2J mice, enabling sample stratification into subpopulations of good and poor treatment responders to delineate response-associated signature transcript profiles in peripheral blood samples. As a proof of concept, we translated our murine data to the transcriptome data of a clinically relevant human cohort. A cluster of 259 differentially regulated genes was identified when peripheral transcriptome profiles of good and poor treatment responders were compared in the murine model. Differences in expression profiles from baseline to week 12 of the human orthologues selected on the basis of the murine transcript signature allowed prediction of response status with an accuracy of 76% in the patient population. Finally, we show that glucocorticoid receptor (GR)-regulated genes are significantly enriched in this cluster of antidepressant-response genes. Our findings point to the involvement of GR sensitivity as a potential key mechanism shaping response to antidepressant treatment and support the hypothesis that antidepressants could stimulate resilience-promoting molecular mechanisms. Our data highlight the suitability of an appropriate animal experimental approach for the discovery of treatment response-associated pathways across species.


Subject(s)
Antidepressive Agents/pharmacology , Depressive Disorder, Major/drug therapy , Paroxetine/pharmacology , Receptors, Glucocorticoid/physiology , Animals , Antidepressive Agents/therapeutic use , Biomarkers, Pharmacological , Brain/metabolism , Corticosterone/blood , Gene Expression Profiling , Gene Expression Regulation , Humans , Mice , Mice, Inbred DBA , Multigene Family , Paroxetine/metabolism , Paroxetine/therapeutic use , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism
15.
Alcohol Clin Exp Res ; 44(5): 1025-1036, 2020 05.
Article in English | MEDLINE | ID: mdl-32154593

ABSTRACT

BACKGROUND: Chronic alcohol exposure can alter glucocorticoid receptor (GR) function in some brain areas that promotes escalated and compulsive-like alcohol intake. GR antagonism can prevent dependence-induced escalation in drinking, but very little is known about the role of GR in regulating high-risk nondependent alcohol intake. Here, we investigate the role of GR in regulating binge-like drinking and aversive responses to alcohol in the High Drinking in the Dark (HDID-1) mice, which have been selectively bred for high blood ethanol (EtOH) concentrations (BECs) in the Drinking in the Dark (DID) test, and in their founder line, the HS/NPT. METHODS: In separate experiments, male and female HDID-1 mice were administered one of several compounds that inhibited GR or its negative regulator, FKBP51 (mifepristone [12.5, 25, 50, 100 mg/kg], CORT113176 [20, 40, 80 mg/kg], and SAFit2 [10, 20, 40 mg/kg]) during a 2-day DID task. EtOH consumption and BECs were measured. EtOH conditioned taste and place aversion (CTA and CPA, respectively) were measured in separate HDID-1 mice after mifepristone administration to assess GR's role in regulating the conditioned aversive effects of EtOH. Lastly, HS/NPT mice were administered CORT113176 during DID to assess whether dissimilar effects from those of HDID-1 would be observed, which could suggest that selective breeding had altered sensitivity to the effects of GR antagonism on binge-like drinking. RESULTS: GR antagonism (with both mifepristone and CORT113176) selectively reduced binge-like EtOH intake and BECs in the HDID-1 mice, while inhibition of FKBP51 did not alter intake or BECs. In contrast, GR antagonism had no effect on EtOH intake or BECs in the HS/NPT mice. Although HDID-1 mice exhibit attenuated EtOH CTA, mifepristone administration did not enhance the aversive effects of EtOH in either a CTA or CPA task. CONCLUSION: These data suggest that the selection process increased sensitivity to GR antagonism on EtOH intake in the HDID-1 mice, and support a role for the GR as a genetic risk factor for high-risk alcohol intake.


Subject(s)
Binge Drinking/physiopathology , Ethanol/administration & dosage , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/physiology , Alcohol Drinking/drug therapy , Alcohol Drinking/genetics , Animals , Aversive Agents , Binge Drinking/genetics , Binge Drinking/prevention & control , Female , Isoquinolines/pharmacology , Male , Mice , Mifepristone/pharmacology , Pyrazoles/pharmacology , Receptors, Glucocorticoid/genetics , Tacrolimus Binding Proteins/antagonists & inhibitors
16.
Biol Pharm Bull ; 43(8): 1279-1282, 2020.
Article in English | MEDLINE | ID: mdl-32741951

ABSTRACT

Clinical studies, especially those in animal models, have provided evidence that chronic stress may play a role in the etiology of psychiatric diseases, such as depression. Because chronic stress activates the hypothalamic-pituitary-adrenal (HPA) axis, resulting in the excessive secretion of glucocorticoids, the chronic stimulation of glucocorticoid receptors (GRs) may be involved in the pathogenesis of depression. To further investigate the relationship between GR activation and depression, we used the synthetic glucocorticoid dexamethasone (DEX) and the GR antagonist mifepristone to examine the effects of chronic GR stimulation on the circadian rhythms of locomotor activity and serotonergic neurotransmission in the basolateral amygdala (BLA) of rats. Chronic treatment with DEX reduced locomotor activity during the dark phase, without changing overall activity patterns. Measuring the basal release of serotonin in the BLA, using in vivo microdialysis, confirmed that chronic treatment with DEX induced serotonergic hypofunction in the BLA. The co-administration of DEX with mifepristone effectively suppressed the depressive-like symptoms caused by chronic treatment with DEX. Our results provided further evidence for a relationship between GR and depression and suggest that the pharmacological blockade of GR may increase the effectiveness of conventional pharmacotherapies used to treat depression.


Subject(s)
Basolateral Nuclear Complex/drug effects , Dexamethasone/pharmacology , Locomotion/drug effects , Receptors, Glucocorticoid/physiology , Serotonin/metabolism , Synaptic Transmission/drug effects , Animals , Basolateral Nuclear Complex/metabolism , Circadian Rhythm/physiology , Depression/drug therapy , Depression/etiology , Male , Rats , Rats, Sprague-Dawley
17.
Hippocampus ; 29(6): 491-499, 2019 06.
Article in English | MEDLINE | ID: mdl-30295349

ABSTRACT

Stress is known to have a critical impact on memory processes. In the present work, we focus on the effects of an acute stress event closely associated to an unrelated learning task. Here, we show that acute stress (elevated platform [EP] session) experienced 1 hr after a weak spatial object recognition (SOR) training, which only induces a short-term memory (STM), promoted the formation of SOR-long term memory (SOR-LTM) in rats. The effect induced by stress was dependent on the activation of glucocorticoid- and mineralocorticoid-receptors, brain-derived neurotrophic factor (BDNF) and protein synthesis in the dorsal hippocampus. In contrast, EP after a strong SOR impaired SOR-LTM probably by interfering with the use of necessary resources. Moreover, we show that the EP session before training induced anterograde interference, which it was not reversed by a subsequent exposure to an open field. Our findings provide novel insights into the impact of stress on LTM formation in rodents and they are discussed under the behavioral analogue of the synaptic tagging and capture hypothesis.


Subject(s)
Hippocampus/physiology , Memory, Long-Term/physiology , Recognition, Psychology/physiology , Stress, Physiological/physiology , Animals , Brain-Derived Neurotrophic Factor/physiology , Male , Memory, Short-Term/physiology , Rats , Rats, Wistar , Receptors, Glucocorticoid/physiology , Receptors, Mineralocorticoid/physiology , Spatial Learning/physiology , Spatial Memory/physiology
18.
Drug Metab Dispos ; 47(11): 1343-1351, 2019 11.
Article in English | MEDLINE | ID: mdl-31519697

ABSTRACT

Both nonsteroidal anti-inflammatory drugs (NSAIDs) and glucocorticoids have been widely used for the treatment of gout, a disease promoted by an excess body burden of uric acid (UA); however, their effects on the homeostasis of UA remain poorly understood. The present study showed that 1-week treatments with three NSAIDs (ibuprofen, diclofenac, and indomethacin) had little effect on UA homeostasis in mice, whereas 1-week low doses (1 and 5 mg/kg) of dexamethasone (DEX) significantly decreased serum UA by about 15%. Additionally, low doses of DEX also resulted in increases in hepatic UA concentration and urinary UA excretion, which were associated with an induction of xanthine oxidoreductase (XOR) in the liver and a downregulation of urate transporter 1 (URAT1) in the kidney, respectively. Neither 75 mg/kg DEX nor 100 mg/kg pregnenolone-16α-carbonitrile altered UA concentrations in serum and livers of mice, suggesting that the effect of DEX on UA homeostasis was not due to the pregnane X receptor pathway. Further in vitro studies demonstrated that glucocorticoid receptor (GR) was involved in DEX-mediated downregulation of URAT1. Knockdown of both p65 and c-Jun completely blocked the effect of DEX on URAT1, suggesting that GR regulates URAT1 via its interaction with both nuclear factor κB and activator protein 1 signaling pathways. To conclude, the present study identifies, for the first time, a critical role of glucocorticoids in regulating UA homeostasis and elucidates the mechanism for GR-mediated regulation of URAT1 in mice. SIGNIFICANCE STATEMENT: This study demonstrates, for the first time, a critical role of glucocorticoid receptor in regulating urate transporter 1 in mouse kidney.


Subject(s)
Dexamethasone/pharmacology , Kidney/metabolism , Organic Anion Transporters/genetics , Uric Acid/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Down-Regulation , Male , Mice , Mice, Inbred C57BL , NF-kappa B/physiology , Organic Anion Transporters/physiology , Pregnane X Receptor/physiology , Receptors, Glucocorticoid/physiology , Signal Transduction/physiology , Xanthine Dehydrogenase/physiology
19.
Respir Res ; 20(1): 37, 2019 Feb 18.
Article in English | MEDLINE | ID: mdl-30777086

ABSTRACT

BACKGROUND: Inhaled corticosteroids reduce inflammation in asthma but chronic use may cause adverse effects. AZD7594, an inhaled non-steroidal selective glucocorticoid receptor modulator, has the potential of an improved risk-benefit profile. We investigated the safety and efficacy of AZD7594 in asthma. METHODS: This phase 2a multi-center, randomized, double-blind, placebo-controlled crossover study enrolled adults with asthma aged 18 to 75 years. Patients were treated with budesonide 200 µg twice daily for 2-3 3 weeks (run in part one). If controlled, as demonstrated by an asthma control questionnaire-5 score of < 1.5, patients entered a three-week run-in (part two) where they received a short acting bronchodilator alone. Thereafter, patients with a fractional exhaled nitric oxide (FENO) ≥25 ppb and pre-dose FEV1 40 to 90% predicted were randomized to one of nine treatment sequences. Each patient received placebo and two of three dose levels of AZD7594 (58, 250, 800 µg) once daily via inhalation, in 14-day treatment periods, separated by three-week washout periods. The primary endpoint was the change from baseline in morning trough FEV1 versus placebo on day 15. Secondary endpoints included measures of airway inflammation and asthma control. RESULTS: Fifty-four patients were randomized and received at least 1 dose of treatment, 48 patients completed the study. Overall 52 patients received placebo, 34 received AZD7594 58 µg, 34 received AZD7594 250 µg, and 34 received AZD7594 800 µg. AZD7594 800 µg demonstrated a significant improvement in Day 15 morning trough FEV1versus placebo (LS means difference 0.148 L 95% CI 0.035-0.261, p = 0.011), with a dose-dependent response seen in the 250 µg (0.076 L -0·036-0·188, p = 0.183) and 58 µg (0·027 L -0·086-0·140, p = 0.683). All secondary endpoints showed statistically significant improvement at the 800 µg dose. All doses demonstrated a significant reduction in FENO at day 15 p < 0.01. No statistically significant difference in plasma cortisol level was observed between AZD7594 and placebo at any dose. AZD7594 was considered safe and well tolerated. CONCLUSIONS: Two-week treatment with AZD7594 demonstrated a favorable risk-benefit profile in patients with mild to moderate asthma. Further clinical studies are needed to fully characterize AZD7594. TRIAL REGISTRATION: ClinicalTrials.gov number NCT02479412 .


Subject(s)
Asthma/diagnosis , Asthma/drug therapy , Bronchodilator Agents/administration & dosage , Receptors, Glucocorticoid/physiology , Administration, Inhalation , Adult , Asthma/physiopathology , Cross-Over Studies , Double-Blind Method , Female , Forced Expiratory Volume/drug effects , Forced Expiratory Volume/physiology , Humans , Male , Middle Aged , Receptors, Glucocorticoid/agonists , Treatment Outcome
20.
Proc Natl Acad Sci U S A ; 113(12): 3377-82, 2016 Mar 22.
Article in English | MEDLINE | ID: mdl-26957608

ABSTRACT

Glucocorticoids (GCs) are widely prescribed anti-inflammatory agents, but their chronic use leads to undesirable side effects such as excessive expansion of adipose tissue. We have recently shown that the forkhead box protein A3 (Foxa3) is a calorie-hoarding factor that regulates the selective enlargement of epididymal fat depots and suppresses energy expenditure in a nutritional- and age-dependent manner. It has been demonstrated that Foxa3 levels are elevated in adipose depots in response to high-fat diet regimens and during the aging process; however no studies to date have elucidated the mechanisms that control Foxa3's expression in fat. Given the established effects of GCs in increasing visceral adiposity and in reducing thermogenesis, we assessed the existence of a possible link between GCs and Foxa3. Computational prediction analysis combined with molecular studies revealed that Foxa3 is regulated by the glucocorticoid receptor (GR) in preadipocytes, adipocytes, and adipose tissues and is required to facilitate the binding of the GR to its target gene promoters in fat depots. Analysis of the long-term effects of dexamethasone treatment in mice revealed that Foxa3 ablation protects mice specifically against fat accretion but not against other pathological side effects elicited by this synthetic GC in tissues such as liver, muscle, and spleen. In conclusion our studies provide the first demonstration, to our knowledge, that Foxa3 is a direct target of GC action in adipose tissues and point to a role of Foxa3 as a mediator of the side effects induced in fat tissues by chronic treatment with synthetic steroids.


Subject(s)
Adipose Tissue/physiology , Hepatocyte Nuclear Factor 3-gamma/physiology , Receptors, Glucocorticoid/physiology , Adipose Tissue/metabolism , Animals , Glucocorticoids/adverse effects , Hyperlipidemias/genetics , Mice , Promoter Regions, Genetic , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL