Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.720
Filter
1.
Respir Res ; 25(1): 227, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38812021

ABSTRACT

BACKGROUND: Steroid insensitivity in Chronic Obstructive Pulmonary Disease (COPD) presents a problem for controlling the chronic inflammation of the airways. The glucocorticoid receptor (GR) mediates the intracellular signaling of inhaled corticosteroids (ICS) by interacting with transcription factors and histone deacetylases (HDACs). The aim of this study was to assess if COPD patients' response to ICS in vivo, may be associated with the expression of GR, the complex of GR with transcription factors, and the expression of various HDACs in vitro. METHODS: Primary airway smooth muscle cells (ASMC) were established from endobronchial biopsies obtained from patients with asthma (n = 10), patients with COPD (n = 10) and subjects that underwent diagnostic bronchoscopy without pathological findings and served as controls (n = 6). ASMC were also established from 18 COPD patients, 10 responders and 8 non-responders to ICS, who participated in the HISTORIC study, an investigator-initiated and driven clinical trial that proved the hypothesis that COPD patients with high ASMC in their endobronchial biopsies respond better to ICS than patients with low ASMC. Expression of GR and its isoforms GRα and GRß and HDACs was investigated in primary ASMC in the absence or in the presence of dexamethasone (10- 8M) by western blotting. The complex formation of GR with transcription factors was assessed by co-immunoprecipitation. RESULTS: Expression of GR and its isoform GRα but not GRß was significantly reduced in ASMC from COPD patients as compared to controls. There were no significant differences in the expression of GR, GRα and GRß between responders and non-responders to ICS. However, treatment with dexamethasone upregulated the expression of total GR (p = 0.004) and GRα (p = 0.005) after 30 min in responders but not in non-responders. Τhe formation of the complex GR-c-Jun was increased 60 min after treatment with dexamethasone only in responders who exhibited significantly lower expression of HDAC3 (p = 0.005) and HDAC5 (p < 0.0001) as compared to non-responders. CONCLUSIONS: These data suggest that ASMC from COPD patients who do not respond to treatment with ICS, are characterized by reduced GR-c-Jun complex formation and increased expression of HDAC3 and HDAC5. TRIAL REGISTRATION: ISRCTN11017699 (Registration date: 15/11/2016).


Subject(s)
Histone Deacetylases , Myocytes, Smooth Muscle , Pulmonary Disease, Chronic Obstructive , Receptors, Glucocorticoid , Humans , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/drug therapy , Pulmonary Disease, Chronic Obstructive/pathology , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/biosynthesis , Histone Deacetylases/metabolism , Histone Deacetylases/biosynthesis , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Male , Middle Aged , Female , Aged , Cells, Cultured , Adrenal Cortex Hormones/therapeutic use , Glucocorticoids/pharmacology , Dexamethasone/pharmacology , Treatment Outcome , Administration, Inhalation , Bronchi/drug effects , Bronchi/metabolism , Bronchi/pathology , Bronchi/enzymology
2.
BMC Complement Med Ther ; 24(1): 186, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734604

ABSTRACT

BACKGROUND: Cepharanthin® alone or in combination with glucocorticoid (GC) has been used to treat chronic immune thrombocytopenia (ITP) since the 1990s. Cepharanthine (CEP) is one of the main active components of Cepharanthin®. The purpose of this study was to investigate the effects of CEP on GC pharmacodynamics on immune cells and analyse the possible action mechanism of their interactions. METHODS: Peripheral blood mononuclear cells (PBMCs), T lymphocytic leukemia MOLT-4 cells and daunorubicin resistant MOLT-4 cells (MOLT-4/DNR) were used to evaluate the pharmacodynamics and molecular mechanisms. Drug pharmacodynamics was evaluated by WST-8 assay. P-glycoprotein function was examined by rhodamine 123 assay. CD4+CD25+Foxp3+ regulatory T cells and Th1/Th2/Th17 cytokines were detected by flow cytometry. P-glycoprotein expression and GC receptor translocation were examined by Western blot. RESULTS: CEP synergistically increased methylprednisolone (MP) efficacy with the suppressive effect on the cell viability of PBMCs. 0.3 and 1 µM of CEP significantly inhibited P-glycoprotein efflux function of CD4+ cells, CD8+ cells, and lymphocytes (P<0.05). 0.03~3 µM of CEP also inhibited the P-glycoprotein efflux function in MOLT-4/DNR cells in a concentration-dependent manner (P<0.001). However, 0.03~3 µM of CEP did not influence P-glycoprotein expression. 0.03~0.3 µM of CEP significantly increased the GC receptor distribution from the cytoplasm to the nucleus in a concentration-dependent manner in MOLT-4/DNR cells. The combination did not influence the frequency of CD4+, CD4+CD25+ and CD4+CD25+Foxp3+ T cells or the secretion of Th1/Th2/Th17 cytokines from PBMCs. In contrast, CEP alone at 1 µM decreased the percentage of CD4+ T cell significantly (P<0.01). It also inhibited the secretion of IL-6, IL-10, IL-17, TNF-α, and IFN-γ. CONCLUSIONS: CEP synergistically promoted MP pharmacodynamics to decrease the cell viability of the mitogen-activated PBMCs, possibly via inhibiting P-glycoprotein function and potentiating GC receptor translocation. The present study provides new evidence of the therapeutic effect of Cepharanthin® alone or in combination with GC for the management of chronic ITP.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1 , Benzylisoquinolines , Drug Synergism , Leukocytes, Mononuclear , Methylprednisolone , Receptors, Glucocorticoid , Humans , Benzylisoquinolines/pharmacology , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Methylprednisolone/pharmacology , Receptors, Glucocorticoid/metabolism , Benzodioxoles
3.
Int J Mol Sci ; 25(10)2024 May 07.
Article in English | MEDLINE | ID: mdl-38791098

ABSTRACT

The similarity of the clinical picture of metabolic syndrome and hypercortisolemia supports the hypothesis that obesity may be associated with impaired expression of genes related to cortisol action and metabolism in adipose tissue. The expression of genes encoding the glucocorticoid receptor alpha (GR), cortisol metabolizing enzymes (HSD11B1, HSD11B2, H6PDH), and adipokines, as well as selected microRNAs, was measured by real-time PCR in adipose tissue from 75 patients with obesity, 19 patients following metabolic surgery, and 25 normal-weight subjects. Cortisol levels were analyzed by LC-MS/MS in 30 pairs of tissues. The mRNA levels of all genes studied were significantly (p < 0.05) decreased in the visceral adipose tissue (VAT) of patients with obesity and normalized by weight loss. In the subcutaneous adipose tissue (SAT), GR and HSD11B2 were affected by this phenomenon. Negative correlations were observed between the mRNA levels of the investigated genes and selected miRNAs (hsa-miR-142-3p, hsa-miR-561, and hsa-miR-579). However, the observed changes did not translate into differences in tissue cortisol concentrations, although levels of this hormone in the SAT of patients with obesity correlated negatively with mRNA levels for adiponectin. In conclusion, although the expression of genes related to cortisol action and metabolism in adipose tissue is altered in obesity and miRNAs may be involved in this process, these changes do not affect tissue cortisol concentrations.


Subject(s)
11-beta-Hydroxysteroid Dehydrogenase Type 1 , Hydrocortisone , MicroRNAs , Obesity , Receptors, Glucocorticoid , Humans , Hydrocortisone/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Obesity/metabolism , Obesity/genetics , Male , Female , Middle Aged , Adult , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 1/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 1/genetics , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Adipose Tissue/metabolism , Intra-Abdominal Fat/metabolism , Gene Expression Regulation , RNA, Messenger/metabolism , RNA, Messenger/genetics , Carbohydrate Dehydrogenases
4.
Int J Mol Sci ; 25(10)2024 May 16.
Article in English | MEDLINE | ID: mdl-38791468

ABSTRACT

Maternal type 2 diabetes mellitus (T2DM) has been shown to result in foetal programming of the hypothalamic-pituitary-adrenal (HPA) axis, leading to adverse foetal outcomes. T2DM is preceded by prediabetes and shares similar pathophysiological complications. However, no studies have investigated the effects of maternal prediabetes on foetal HPA axis function and postnatal offspring development. Hence, this study investigated the effects of pregestational prediabetes on maternal HPA axis function and postnatal offspring development. Pre-diabetic (PD) and non-pre-diabetic (NPD) female Sprague Dawley rats were mated with non-prediabetic males. After gestation, male pups born from the PD and NPD groups were collected. Markers of HPA axis function, adrenocorticotropin hormone (ACTH) and corticosterone, were measured in all dams and pups. Glucose tolerance, insulin and gene expressions of mineralocorticoid (MR) and glucocorticoid (GR) receptors were further measured in all pups at birth and their developmental milestones. The results demonstrated increased basal concentrations of ACTH and corticosterone in the dams from the PD group by comparison to NPD. Furthermore, the results show an increase basal ACTH and corticosterone concentrations, disturbed MR and GR gene expression, glucose intolerance and insulin resistance assessed via the Homeostasis Model Assessment (HOMA) indices in the pups born from the PD group compared to NPD group at all developmental milestones. These observations reveal that pregestational prediabetes is associated with maternal dysregulation of the HPA axis, impacting offspring HPA axis development along with impaired glucose handling.


Subject(s)
Adrenocorticotropic Hormone , Corticosterone , Hypothalamo-Hypophyseal System , Pituitary-Adrenal System , Prediabetic State , Rats, Sprague-Dawley , Animals , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Female , Pregnancy , Prediabetic State/metabolism , Rats , Adrenocorticotropic Hormone/blood , Adrenocorticotropic Hormone/metabolism , Corticosterone/blood , Corticosterone/metabolism , Male , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Receptors, Mineralocorticoid/genetics , Prenatal Exposure Delayed Effects/metabolism , Diabetes Mellitus, Type 2/metabolism , Insulin Resistance
5.
Dis Model Mech ; 17(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38770680

ABSTRACT

Absence of dystrophin results in muscular weakness, chronic inflammation and cardiomyopathy in Duchenne muscular dystrophy (DMD). Pharmacological corticosteroids are the DMD standard of care; however, they have harsh side effects and unclear molecular benefits. It is uncertain whether signaling by physiological corticosteroids and their receptors plays a modifying role in the natural etiology of DMD. Here, we knocked out the glucocorticoid receptor (GR, encoded by Nr3c1) specifically in myofibers and cardiomyocytes within wild-type and mdx52 mice to dissect its role in muscular dystrophy. Double-knockout mice showed significantly worse phenotypes than mdx52 littermate controls in measures of grip strength, hang time, inflammatory pathology and gene expression. In the heart, GR deletion acted additively with dystrophin loss to exacerbate cardiomyopathy, resulting in enlarged hearts, pathological gene expression and systolic dysfunction, consistent with imbalanced mineralocorticoid signaling. The results show that physiological GR functions provide a protective role during muscular dystrophy, directly contrasting its degenerative role in other disease states. These data provide new insights into corticosteroids in disease pathophysiology and establish a new model to investigate cell-autonomous roles of nuclear receptors and mechanisms of pharmacological corticosteroids.


Subject(s)
Dystrophin , Mice, Inbred mdx , Mice, Knockout , Receptors, Glucocorticoid , Animals , Receptors, Glucocorticoid/metabolism , Dystrophin/metabolism , Dystrophin/genetics , Dystrophin/deficiency , Myocardium/pathology , Myocardium/metabolism , Muscular Dystrophy, Duchenne/pathology , Muscular Dystrophy, Duchenne/metabolism , Muscle, Skeletal/pathology , Muscle, Skeletal/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/drug effects , Mice , Cardiomyopathies/pathology , Cardiomyopathies/metabolism , Mice, Inbred C57BL , Muscular Dystrophy, Animal/pathology , Muscular Dystrophy, Animal/metabolism , Phenotype , Systole/drug effects
6.
Cell Rep ; 43(5): 114200, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38717905

ABSTRACT

Innate lymphoid cells (ILCs), strategically positioned throughout the body, undergo population declines over time. A solution to counteract this problem is timely mobilization of multipotential progenitors from the bone marrow. It remains unknown what triggers the mobilization of bone marrow ILC progenitors (ILCPs). We report that ILCPs are regulated by the circadian clock to emigrate and generate mature ILCs in the periphery. We found that circadian-clock-defective ILCPs fail to normally emigrate and generate ILCs. We identified circadian-clock-controlled endocrine and cytokine cues that, respectively, regulate the retention and emigration of ILCPs at distinct times of each day. Activation of the stress-hormone-sensing glucocorticoid receptor upregulates CXCR4 on ILCPs for their retention in the bone marrow, while the interleukin-18 (IL-18) and RORα signals upregulate S1PR1 on ILCPs for their mobilization to the periphery. Our findings establish important roles of circadian signals for the homeostatic efflux of bone marrow ILCPs.


Subject(s)
Circadian Clocks , Animals , Mice , Cytokines/metabolism , Mice, Inbred C57BL , Bone Marrow/metabolism , Signal Transduction , Receptors, CXCR4/metabolism , Bone Marrow Cells/metabolism , Bone Marrow Cells/cytology , Lymphoid Progenitor Cells/metabolism , Lymphoid Progenitor Cells/cytology , Immunity, Innate , Cell Movement , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Receptors, Glucocorticoid/metabolism , Lymphocytes/metabolism , Lymphocytes/immunology
7.
Cancer Cell ; 42(5): 833-849.e12, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38701792

ABSTRACT

Glucocorticoids have been used for decades to treat lymphomas without an established mechanism of action. Using functional genomic, proteomic, and chemical screens, we discover that glucocorticoids inhibit oncogenic signaling by the B cell receptor (BCR), a recurrent feature of aggressive B cell malignancies, including diffuse large B cell lymphoma and Burkitt lymphoma. Glucocorticoids induce the glucocorticoid receptor (GR) to directly transactivate genes encoding negative regulators of BCR stability (LAPTM5; KLHL14) and the PI3 kinase pathway (INPP5D; DDIT4). GR directly represses transcription of CSK, a kinase that limits the activity of BCR-proximal Src-family kinases. CSK inhibition attenuates the constitutive BCR signaling of lymphomas by hyperactivating Src-family kinases, triggering their ubiquitination and degradation. With the knowledge that glucocorticoids disable oncogenic BCR signaling, they can now be deployed rationally to treat BCR-dependent aggressive lymphomas and used to construct mechanistically sound combination regimens with inhibitors of BTK, PI3 kinase, BCL2, and CSK.


Subject(s)
Glucocorticoids , Receptors, Antigen, B-Cell , Humans , Glucocorticoids/pharmacology , Receptors, Antigen, B-Cell/metabolism , Animals , Signal Transduction/drug effects , Receptors, Glucocorticoid/metabolism , Mice , Cell Line, Tumor , Lymphoma, Large B-Cell, Diffuse/drug therapy , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/metabolism , Burkitt Lymphoma/drug therapy , Burkitt Lymphoma/genetics , Burkitt Lymphoma/metabolism , Burkitt Lymphoma/pathology , Molecular Targeted Therapy/methods , Phosphatidylinositol 3-Kinases/metabolism , src-Family Kinases/metabolism , Gene Expression Regulation, Neoplastic/drug effects
8.
Sci Rep ; 14(1): 7759, 2024 04 02.
Article in English | MEDLINE | ID: mdl-38565594

ABSTRACT

The vertebrate stress response (SR) is mediated by the hypothalamic-pituitary-adrenal (HPA) axis and contributes to generating context appropriate physiological and behavioral changes. Although the HPA axis plays vital roles both in stressful and basal conditions, research has focused on the response under stress. To understand broader roles of the HPA axis in a changing environment, we characterized an adaptive behavior of larval zebrafish during ambient illumination changes. Genetic abrogation of glucocorticoid receptor (nr3c1) decreased basal locomotor activity in light and darkness. Some key HPI axis receptors (mc2r [ACTH receptor], nr3c1), but not nr3c2 (mineralocorticoid receptor), were required to adapt to light more efficiently but became dispensable when longer illumination was provided. Such light adaptation was more efficient in dimmer light. Our findings show that the HPI axis contributes to the SR, facilitating the phasic response and maintaining an adapted basal state, and that certain adaptations occur without HPI axis activity.


Subject(s)
Hypothalamo-Hypophyseal System , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/metabolism , Hypothalamo-Hypophyseal System/metabolism , Larva/genetics , Larva/metabolism , Pituitary-Adrenal System/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Adaptation, Psychological
9.
Gen Comp Endocrinol ; 352: 114516, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38593942

ABSTRACT

Cortisol is secreted from the adrenal cortex in response to stress, and its circulating levels are used as robust physiological indicators of stress intensity in various animals. Cortisol is also produced locally in adipose tissue by the conversion of steroid hormones such as cortisone, which is related to fat accumulation. Circulating cortisol levels, probably induced by cold stress, increase in cetaceans under cold conditions. However, whether cortisol production in subcutaneous adipose tissue is enhanced when fat accumulation is renewed during the cold season remains unclear. Therefore, in this study, we examine the effect of environmental temperature on the expression of cortisol synthesis-related enzymes and a glucocorticoid receptor in the subcutaneous fat (blubber) and explore the association between these expressions and fluctuations in circulating cortisol levels in common bottlenose dolphins (Tursiops truncatus). Skin biopsies were obtained seasonally from eight female dolphins, and seasonal differences in the expression of target genes in the blubber were analyzed. Blood samples were collected throughout the year, and cortisol levels were measured. We found that the expressions of cytochrome P450 family 21 subfamily A member 2 (CYP21A2) and nuclear receptor subfamily 3 group C member 1 (NR3C1), a glucocorticoid receptor, were increased in the cold season, and 11 beta-hydroxysteroid dehydrogenase type 1 (HSD11B1) showed a similar trend. Blood cortisol levels increased when the water temperature decreased. These results suggest that the conversion of 17-hydroxyprogesterone to cortisol via 11-deoxycortisol and/or of cortisone to cortisol is enhanced under cold conditions, and the physiological effects of cortisol in subcutaneous adipose tissue may contribute to on-site lipid accumulation and increase the circulating cortisol concentrations. The results obtained in this study highlight the role of cortisol in the regulation of the blubber that has developed to adapt to aquatic life.


Subject(s)
Bottle-Nosed Dolphin , Cortisone , Animals , Female , Hydrocortisone/metabolism , Seasons , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Cortisone/metabolism , Adipose Tissue/metabolism
10.
Front Endocrinol (Lausanne) ; 15: 1362573, 2024.
Article in English | MEDLINE | ID: mdl-38645426

ABSTRACT

Brain-derived neurotrophic factor (BDNF), a key neurotrophin within the brain, by selectively activating the TrkB receptor, exerts multimodal effects on neurodevelopment, synaptic plasticity, cellular integrity and neural network dynamics. In parallel, glucocorticoids (GCs), vital steroid hormones, which are secreted by adrenal glands and rapidly diffused across the mammalian body (including the brain), activate two different groups of intracellular receptors, the mineralocorticoid and the glucocorticoid receptors, modulating a wide range of genomic, epigenomic and postgenomic events, also expressed in the neural tissue and implicated in neurodevelopment, synaptic plasticity, cellular homeostasis, cognitive and emotional processing. Recent research evidences indicate that these two major regulatory systems interact at various levels: they share common intracellular downstream pathways, GCs differentially regulate BDNF expression, under certain conditions BDNF antagonises the GC-induced effects on long-term potentiation, neuritic outgrowth and cellular death, while GCs regulate the intraneuronal transportation and the lysosomal degradation of BDNF. Currently, the BDNF-GC crosstalk features have been mainly studied in neurons, although initial findings show that this crosstalk could be equally important for other brain cell types, such as astrocytes. Elucidating the precise neurobiological significance of BDNF-GC interactions in a tempospatial manner, is crucial for understanding the subtleties of brain function and dysfunction, with implications for neurodegenerative and neuroinflammatory diseases, mood disorders and cognitive enhancement strategies.


Subject(s)
Brain-Derived Neurotrophic Factor , Glucocorticoids , Humans , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/physiology , Glucocorticoids/metabolism , Animals , Brain/metabolism , Neuronal Plasticity/physiology , Receptors, Glucocorticoid/metabolism , Signal Transduction , Neurons/metabolism
11.
Front Biosci (Landmark Ed) ; 29(4): 163, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38682179

ABSTRACT

BACKGROUND: Glucocorticoids (GCs) are commonly used as the primary chemotherapy for lymphoid malignancies, including acute lymphoblastic leukemia (ALL). However, the development of GC resistance limits their prolonged use. METHODS: In this study, we investigated the potential of a newly synthesized indole derivative called LWX-473, in combination with the classic GC Dexamethasone (DEX), to enhance the responsiveness of Jurkat cells to GC treatment. RESULTS: Our findings demonstrate that LWX-473 alone or in combination with DEX significantly improves GC-induced cell apoptosis and arrests the cell cycle in the G1 phase. Notably, the combination of LWX-473 and DEX exhibits superior efficacy in killing Jurkat cells compared to LWX-473 alone. Importantly, this compound demonstrates reduced toxicity towards normal cells. CONCLUSIONS: Our study reveals that LWX-473 has the ability to restore the sensitivity of Jurkat cells to DEX by modulating the mitochondrial membrane potential, activating the expression of DEX-liganded glucocorticoid receptor (GR), and inhibiting key molecules in the JAK/STAT signaling pathway. These findings suggest that LWX-473 could be a potential therapeutic agent for overcoming GC resistance in lymphoid malignancies.


Subject(s)
Apoptosis , Dexamethasone , Drug Resistance, Neoplasm , Glucocorticoids , Indoles , Membrane Potential, Mitochondrial , Receptors, Glucocorticoid , Humans , Jurkat Cells , Apoptosis/drug effects , Dexamethasone/pharmacology , Drug Resistance, Neoplasm/drug effects , Glucocorticoids/pharmacology , Indoles/pharmacology , Receptors, Glucocorticoid/metabolism , Membrane Potential, Mitochondrial/drug effects , Signal Transduction/drug effects
12.
Lung ; 202(3): 235-243, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38641747

ABSTRACT

PURPOSE: The response to glucocorticoids is hampered in many COPD patients by a yet unknown mechanism. Earlier we reported that short-term heat exposure of primary human bronchial epithelial cells (BEC) and airway smooth muscle cells (ASMC) of asthma patients increased the expression and secretion of extracellular heat shock proteins (eHSPs) resulting in increased expression of glucocorticoid receptor (GR) in BEC and inhibition of ASMC remodeling. The aim of the present study was to assess if the same mechanism is also present in primary airway wall cells of COPD patients. METHODS: Primary BEC and ASMC were established from endobronchial biopsies obtained from COPD patients (n = 73), who participated in the HISTORIC study, an investigator-initiated and driven clinical trial. Secretion and protein expression of HSPs was assessed by ELISA and Western blotting. Expression of total GR, its isoforms GRα and GRß and toll-like receptor 4 (TLR4) was determined by Western-blotting. RESULTS: Short heat exposure (65 °C, 10 s) of BEC resulted in a significant increase of the secretion of eHSP70 and eHSP90, while the intracellular protein was not altered. Heat treatment or exposure to eHSP70 or eHSP90 had no effect on the expression of GR and GR-isoforms. However, eHSP70 and eHSP90 significantly reduced the expression of TLR4. CONCLUSIONS: The results of this study indicate that primary airway cells from COPD patients respond differently to heat exposure and extracellular HSP70 or HSP90 than cells from asthma patients regarding the expression of GR and this may explain the reduced response to glucocorticoids in patients with COPD. TRIAL REGISTRATION: ISRCTN11017699.


Subject(s)
Bronchi , HSP70 Heat-Shock Proteins , HSP90 Heat-Shock Proteins , Myocytes, Smooth Muscle , Pulmonary Disease, Chronic Obstructive , Receptors, Glucocorticoid , Toll-Like Receptor 4 , Humans , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , HSP70 Heat-Shock Proteins/metabolism , Receptors, Glucocorticoid/metabolism , HSP90 Heat-Shock Proteins/metabolism , Male , Middle Aged , Female , Aged , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/drug effects , Toll-Like Receptor 4/metabolism , Bronchi/metabolism , Bronchi/pathology , Cells, Cultured , Hot Temperature , Epithelial Cells/metabolism , Epithelial Cells/drug effects
13.
Dis Model Mech ; 17(4)2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38655653

ABSTRACT

Steroid myopathy is a clinically challenging condition exacerbated by prolonged corticosteroid use or adrenal tumors. In this study, we engineered a functional three-dimensional (3D) in vitro skeletal muscle model to investigate steroid myopathy. By subjecting our bioengineered muscle tissues to dexamethasone treatment, we reproduced the molecular and functional aspects of this disease. Dexamethasone caused a substantial reduction in muscle force, myotube diameter and induced fatigue. We observed nuclear translocation of the glucocorticoid receptor (GCR) and activation of the ubiquitin-proteasome system within our model, suggesting their coordinated role in muscle atrophy. We then examined the therapeutic potential of taurine in our 3D model for steroid myopathy. Our findings revealed an upregulation of phosphorylated AKT by taurine, effectively countering the hyperactivation of the ubiquitin-proteasomal pathway. Importantly, we demonstrate that discontinuing corticosteroid treatment was insufficient to restore muscle mass and function. Taurine treatment, when administered concurrently with corticosteroids, notably enhanced contractile strength and protein turnover by upregulating the AKT-mTOR axis. Our model not only identifies a promising therapeutic target, but also suggests combinatorial treatment that may benefit individuals undergoing corticosteroid treatment or those diagnosed with adrenal tumors.


Subject(s)
Dexamethasone , Models, Biological , Muscle Contraction , Muscular Diseases , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Taurine , Proto-Oncogene Proteins c-akt/metabolism , Humans , Taurine/pharmacology , TOR Serine-Threonine Kinases/metabolism , Muscle Contraction/drug effects , Dexamethasone/pharmacology , Muscular Diseases/pathology , Muscular Diseases/drug therapy , Signal Transduction/drug effects , Receptors, Glucocorticoid/metabolism , Muscle Strength/drug effects , Proteasome Endopeptidase Complex/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscle, Skeletal/metabolism , Organ Size/drug effects , Phosphorylation/drug effects , Adrenal Cortex Hormones/pharmacology , Ubiquitin/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/metabolism , Steroids/pharmacology
14.
J Steroid Biochem Mol Biol ; 241: 106521, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38631601

ABSTRACT

Increased cortisol levels in the preovulatory follicular fluid suggests a role of glucocorticoid in human ovulation. However, the mechanisms through which cortisol regulates the ovulatory process remain poorly understood. In this study, we examined the upregulation of f5 mRNA by glucocorticoid and its receptor (Gr) in the preovulatory follicles of zebrafish. Our findings demonstrate a significant increase in 11ß-hydroxysteroid dehydrogenase type 2 (hsd11b2), a cortisol response gene, in preovulatory follicles. Additionally, hydrocortisone exerts a dose- and time-dependent upregulation of f5 mRNA in these follicles. Importantly, this stimulatory effect is Gr-dependent, as it was completely abolished in gr-/- mutants. Furthermore, site-directed mutagenesis identified a glucocorticoid response element (GRE) in the promoter of zebrafish f5. Interestingly, successive incubation of hydrocortisone and the native ovulation-inducing steroid, progestin (17α,20ß-dihydroxy-4-pregnen-3-one, DHP), further enhanced f5 expression in preovulatory follicles. Overall, our results indicate that the dramatic increase of f5 expression in preovulatory follicles is partially attributable to the regulation of glucocorticoid and Gr.


Subject(s)
Glucocorticoids , Hydrocortisone , Ovarian Follicle , Receptors, Glucocorticoid , Up-Regulation , Zebrafish , Animals , Zebrafish/genetics , Zebrafish/metabolism , Ovarian Follicle/metabolism , Ovarian Follicle/drug effects , Female , Glucocorticoids/pharmacology , Up-Regulation/drug effects , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/genetics , Hydrocortisone/pharmacology , Hydrocortisone/metabolism , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism , Ovulation/drug effects , 11-beta-Hydroxysteroid Dehydrogenase Type 2/metabolism , 11-beta-Hydroxysteroid Dehydrogenase Type 2/genetics , Promoter Regions, Genetic
15.
Stress ; 27(1): 2312467, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38557197

ABSTRACT

Chronic stress exposure during development can have lasting behavioral consequences that differ in males and females. More specifically, increased depressive behaviors in females, but not males, are observed in both humans and rodent models of chronic stress. Despite these known stress-induced outcomes, the molecular consequences of chronic adolescent stress in the adult brain are less clear. The stress hormone corticosterone activates the glucocorticoid receptor, and activity of the receptor is regulated through interactions with co-chaperones-such as the immunophilin FK506 binding proteins 5 (FKBP5). Previously, it has been reported that the adult stress response is modified by a history of chronic stress; therefore, the current study assessed the impact of chronic adolescent stress on the interactions of the glucocorticoid receptor (GR) with its regulatory co-chaperone FKBP5 in response to acute stress in adulthood. Although protein presence for FKBP5 did not differ by group, assessment of GR-FKBP5 interactions demonstrated that adult females with a history of chronic adolescent stress had elevated GR-FKBP5 interactions in the hippocampus following an acute stress challenge which could potentially contribute to a reduced translocation pattern given previous literature describing the impact of FKBP5 on GR activity. Interestingly, the altered co-chaperone interactions of the GR in the stressed female hippocampus were not coupled to an observable difference in transcription of GR-regulated genes. Together, these studies show that chronic adolescent stress causes lasting changes to co-chaperone interactions with the glucocorticoid receptor following stress exposure in adulthood and highlight the potential role that FKBP5 plays in these modifications. Understanding the long-term implications of adolescent stress exposure will provide a mechanistic framework to guide the development of interventions for adult disorders related to early life stress exposures.


Subject(s)
Receptors, Glucocorticoid , Stress, Psychological , Tacrolimus Binding Proteins , Animals , Female , Male , Rats , Corticosterone/metabolism , Hippocampus/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Stress, Psychological/metabolism , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism
16.
Nature ; 629(8010): 184-192, 2024 May.
Article in English | MEDLINE | ID: mdl-38600378

ABSTRACT

Glucocorticoids represent the mainstay of therapy for a broad spectrum of immune-mediated inflammatory diseases. However, the molecular mechanisms underlying their anti-inflammatory mode of action have remained incompletely understood1. Here we show that the anti-inflammatory properties of glucocorticoids involve reprogramming of the mitochondrial metabolism of macrophages, resulting in increased and sustained production of the anti-inflammatory metabolite itaconate and consequent inhibition of the inflammatory response. The glucocorticoid receptor interacts with parts of the pyruvate dehydrogenase complex whereby glucocorticoids provoke an increase in activity and enable an accelerated and paradoxical flux of the tricarboxylic acid (TCA) cycle in otherwise pro-inflammatory macrophages. This glucocorticoid-mediated rewiring of mitochondrial metabolism potentiates TCA-cycle-dependent production of itaconate throughout the inflammatory response, thereby interfering with the production of pro-inflammatory cytokines. By contrast, artificial blocking of the TCA cycle or genetic deficiency in aconitate decarboxylase 1, the rate-limiting enzyme of itaconate synthesis, interferes with the anti-inflammatory effects of glucocorticoids and, accordingly, abrogates their beneficial effects during a diverse range of preclinical models of immune-mediated inflammatory diseases. Our findings provide important insights into the anti-inflammatory properties of glucocorticoids and have substantial implications for the design of new classes of anti-inflammatory drugs.


Subject(s)
Anti-Inflammatory Agents , Glucocorticoids , Inflammation , Macrophages , Mitochondria , Succinates , Animals , Female , Humans , Male , Mice , Anti-Inflammatory Agents/pharmacology , Carboxy-Lyases/metabolism , Carboxy-Lyases/antagonists & inhibitors , Citric Acid Cycle/drug effects , Citric Acid Cycle/genetics , Cytokines/immunology , Cytokines/metabolism , Glucocorticoids/pharmacology , Glucocorticoids/metabolism , Hydro-Lyases/deficiency , Hydro-Lyases/genetics , Inflammation/drug therapy , Inflammation/metabolism , Macrophages/cytology , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mitochondria/metabolism , Mitochondria/drug effects , Pyruvate Dehydrogenase Complex/metabolism , Receptors, Glucocorticoid/metabolism , Succinates/metabolism , Enzyme Activation/drug effects
17.
Eur J Med Chem ; 271: 116427, 2024 May 05.
Article in English | MEDLINE | ID: mdl-38657479

ABSTRACT

Glucocorticoids (GCs) have been used in the treatment of sepsis because of their potent anti-inflammatory effects. However, their clinical efficacy against sepsis remains controversial because of glucocorticoid receptor (GR) downregulation and side effects. Herein, we designed and synthesized 30 ocotillol derivatives and evaluated their anti-inflammatory activities. Ocotillol 24(R/S) differential isomers were stereoselective in their pharmacological action. Specifically, 24(S) derivatives had better anti-inflammatory activity than their corresponding 24(R) derivatives. Compound 20 most effectively inhibited NO release (85.97% reduction), and it exerted dose-dependent inhibitory effects on interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) levels. Mechanistic studies revealed that compound 20 reduces the degradation of GR mRNA and GR protein. Meanwhile, compound 20 inhibited the activation of nuclear factor-κB (NF-κB) signaling, thereby inhibiting the nuclear translocation of p65 and attenuating the inflammatory response. In vivo studies revealed that compound 20 attenuated hepatic, pulmonary, and renal pathology damage in mice with sepsis and suppressed the production of inflammatory mediators. These results indicated that compound 20 is a promising lead compound for designing and developing anti-sepsis drugs.


Subject(s)
NF-kappa B , Receptors, Glucocorticoid , Sepsis , Signal Transduction , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/antagonists & inhibitors , Sepsis/drug therapy , Animals , NF-kappa B/metabolism , NF-kappa B/antagonists & inhibitors , Mice , Signal Transduction/drug effects , Structure-Activity Relationship , Humans , Molecular Structure , RAW 264.7 Cells , Drug Discovery , Male , Dose-Response Relationship, Drug , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis
18.
Nat Metab ; 6(5): 825-836, 2024 May.
Article in English | MEDLINE | ID: mdl-38622413

ABSTRACT

Nuclear receptor corepressors (NCoRs) function in multiprotein complexes containing histone deacetylase 3 (HDAC3) to alter transcriptional output primarily through repressive chromatin remodelling at target loci1-5. In the liver, loss of HDAC3 causes a marked hepatosteatosis largely because of de-repression of genes involved in lipid metabolism6,7; however, the individual roles and contribution of other complex members to hepatic and systemic metabolic regulation are unclear. Here we show that adult loss of both NCoR1 and NCoR2 (double knockout (KO)) in hepatocytes phenocopied the hepatomegalic fatty liver phenotype of HDAC3 KO. In addition, double KO livers exhibited a dramatic reduction in glycogen storage and gluconeogenic gene expression that was not observed with hepatic KO of individual NCoRs or HDAC3, resulting in profound fasting hypoglycaemia. This surprising HDAC3-independent activation function of NCoR1 and NCoR2 is due to an unexpected loss of chromatin accessibility on deletion of NCoRs that prevented glucocorticoid receptor binding and stimulatory effect on gluconeogenic genes. These studies reveal an unanticipated, non-canonical activation function of NCoRs that is required for metabolic health.


Subject(s)
Gluconeogenesis , Histone Deacetylases , Liver , Mice, Knockout , Nuclear Receptor Co-Repressor 1 , Nuclear Receptor Co-Repressor 2 , Receptors, Glucocorticoid , Gluconeogenesis/genetics , Animals , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/genetics , Nuclear Receptor Co-Repressor 1/metabolism , Nuclear Receptor Co-Repressor 1/genetics , Mice , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Nuclear Receptor Co-Repressor 2/metabolism , Nuclear Receptor Co-Repressor 2/genetics , Liver/metabolism , Hepatocytes/metabolism , Nuclear Receptor Coactivator 2/metabolism , Nuclear Receptor Coactivator 2/genetics
20.
Sci Rep ; 14(1): 7844, 2024 04 03.
Article in English | MEDLINE | ID: mdl-38570726

ABSTRACT

Obesity, a worldwide epidemic, leads to various metabolic disorders threatening human health. In response to stress or fasting, glucocorticoid (GC) levels are elevated to promote food intake. This involves GC-induced expression of the orexigenic neuropeptides in agouti-related protein (AgRP) neurons of the hypothalamic arcuate nucleus (ARC) via the GC receptor (GR). Here, we report a selective GR modulator (SGRM) that suppresses GR-induced transcription of genes with non-classical glucocorticoid response elements (GREs) such as Agrp-GRE, but not with classical GREs, and via this way may serve as a novel anti-obesity agent. We have identified a novel SGRM, 2-O-trans-p-coumaroylalphitolic acid (Zj7), a triterpenoid extracted from the Ziziphus jujube plant, that selectively suppresses GR transcriptional activity in Agrp-GRE without affecting classical GREs. Zj7 reduces the expression of orexigenic genes in the ARC and exerts a significant anorexigenic effect with weight loss in both high fat diet-induced obese and genetically obese db/db mouse models. Transcriptome analysis showed that Zj7 represses the expression of a group of orexigenic genes including Agrp and Npy induced by the synthetic GR ligand dexamethasone (Dex) in the hypothalamus. Taken together, Zj7, as a selective GR modulator, showed beneficial metabolic activities, in part by suppressing GR activity in non-classical GREs in orexigenic genes. This study demonstrates that a potential anorexigenic molecule may allow GRE-specific inhibition of GR transcriptional activity, which is a promising approach for the treatment of metabolic disorders.


Subject(s)
Metabolic Diseases , Receptors, Glucocorticoid , Mice , Animals , Humans , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Glucocorticoids/pharmacology , Agouti-Related Protein/metabolism , Obesity/drug therapy , Obesity/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...