Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.429
Filter
1.
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
2.
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
3.
Eur J Endocrinol ; 190(4): 284-295, 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38584335

ABSTRACT

OBJECTIVE: Glucocorticoid resistance is a rare endocrine disease caused by variants of the NR3C1 gene encoding the glucocorticoid receptor (GR). We identified a novel heterozygous variant (GRR569Q) in a patient with uncommon reversible glucocorticoid resistance syndrome. METHODS: We performed ex vivo functional characterization of the variant in patient fibroblasts and in vitro through transient transfection in undifferentiated HEK 293T cells to assess transcriptional activity, affinity, and nuclear translocation. We studied the impact of the variant on the tertiary structure of the ligand-binding domain through 3D modeling. RESULTS: The patient presented initially with an adrenal adenoma with mild autonomous cortisol secretion and undetectable adrenocorticotropin hormone (ACTH) levels. Six months after surgery, biological investigations showed elevated cortisol and ACTH (urinary free cortisol 114 µg/24 h, ACTH 10.9 pmol/L) without clinical symptoms, evoking glucocorticoid resistance syndrome. Functional characterization of the GRR569Q showed decreased expression of target genes (in response to 100 nM cortisol: SGK1 control +97% vs patient +20%, P < .0001) and impaired nuclear translocation in patient fibroblasts compared to control. Similar observations were made in transiently transfected cells, but higher cortisol concentrations overcame glucocorticoid resistance. GRR569Q showed lower ligand affinity (Kd GRWT: 1.73 nM vs GRR569Q: 4.61 nM). Tertiary structure modeling suggested a loss of hydrogen bonds between H3 and the H1-H3 loop. CONCLUSION: This is the first description of a reversible glucocorticoid resistance syndrome with effective negative feedback on corticotroph cells regarding increased plasma cortisol concentrations due to the development of mild autonomous cortisol secretion.


Subject(s)
Glucocorticoids , Metabolism, Inborn Errors , Receptors, Glucocorticoid , Humans , Adrenocorticotropic Hormone/genetics , Glucocorticoids/pharmacology , Glucocorticoids/therapeutic use , Glucocorticoids/metabolism , Hydrocortisone , Ligands , Mutation , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/deficiency , Syndrome
4.
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
5.
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
6.
Cell Mol Life Sci ; 81(1): 160, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38564048

ABSTRACT

The androgen receptor (AR) is a primary target for treating prostate cancer (PCa), forming the bedrock of its clinical management. Despite their efficacy, resistance often hampers AR-targeted therapies, necessitating new strategies against therapy-resistant PCa. These resistances involve various mechanisms, including AR splice variant overexpression and altered activities of transcription factors like the glucocorticoid receptor (GR) and FOXA1. These factors rely on common coregulators, such as EP300/CREBBP, suggesting a rationale for coregulator-targeted therapies. Our study explores EP300/CREBBP acetyltransferase inhibition's impact on steroid receptor and FOXA1 signaling in PCa cells using genome-wide techniques. Results reveal that EP300/CREBBP inhibition significantly disrupts the AR-regulated transcriptome and receptor chromatin binding by reducing the AR-gene expression. Similarly, GR's regulated transcriptome and receptor binding were hindered, not linked to reduced GR expression but to diminished FOXA1 chromatin binding, restricting GR signaling. Overall, our findings highlight how EP300/CREBBP inhibition distinctively curtails oncogenic transcription factors' signaling, suggesting the potential of coregulatory-targeted therapies in PCa.


Subject(s)
Prostate , Prostatic Neoplasms , Male , Humans , Prostatic Neoplasms/genetics , Receptors, Glucocorticoid/genetics , Transcription Factors , Chromatin , Acetyltransferases , Hepatocyte Nuclear Factor 3-alpha/genetics , E1A-Associated p300 Protein/genetics , CREB-Binding Protein/genetics
7.
Nat Commun ; 15(1): 3572, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38670986

ABSTRACT

A regulated stress response is essential for healthy child growth and development trajectories. We conducted a cluster-randomized trial in rural Bangladesh (funded by the Bill & Melinda Gates Foundation, ClinicalTrials.gov NCT01590095) to assess the effects of an integrated nutritional, water, sanitation, and handwashing intervention on child health. We previously reported on the primary outcomes of the trial, linear growth and caregiver-reported diarrhea. Here, we assessed additional prespecified outcomes: physiological stress response, oxidative stress, and DNA methylation (N = 759, ages 1-2 years). Eight neighboring pregnant women were grouped into a study cluster. Eight geographically adjacent clusters were block-randomized into the control or the combined nutrition, water, sanitation, and handwashing (N + WSH) intervention group (receiving nutritional counseling and lipid-based nutrient supplements, chlorinated drinking water, upgraded sanitation, and handwashing with soap). Participants and data collectors were not masked, but analyses were masked. There were 358 children (68 clusters) in the control group and 401 children (63 clusters) in the intervention group. We measured four F2-isoprostanes isomers (iPF(2α)-III; 2,3-dinor-iPF(2α)-III; iPF(2α)-VI; 8,12-iso-iPF(2α)-VI), salivary alpha-amylase and cortisol, and methylation of the glucocorticoid receptor (NR3C1) exon 1F promoter including the NGFI-A binding site. Compared with control, the N + WSH group had lower concentrations of F2-isoprostanes isomers (differences ranging from -0.16 to -0.19 log ng/mg of creatinine, P < 0.01), elevated post-stressor cortisol (0.24 log µg/dl; P < 0.01), higher cortisol residualized gain scores (0.06 µg/dl; P = 0.023), and decreased methylation of the NGFI-A binding site (-0.04; P = 0.037). The N + WSH intervention enhanced adaptive responses of the physiological stress system in early childhood.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Hand Disinfection , Sanitation , Humans , Female , Bangladesh , Male , Infant , Child, Preschool , Pregnancy , Oxidative Stress , Stress, Physiological , Rural Population , Adult , Diarrhea/prevention & control , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/genetics
8.
Stress ; 27(1): 2321595, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38676353

ABSTRACT

Perinatal stress is associated with altered placental methylation, which plays a critical role in fetal development and infant outcomes. This proof-of-concept pilot study investigated the impact of lifetime trauma exposure and perinatal PTSD symptoms on epigenetic regulation of placenta glucocorticoid signaling genes (NR3C1 and FKBP5). Lifetime trauma exposure and PTSD symptoms during pregnancy were assessed in a racially/ethnically diverse sample of pregnant women (N = 198). Participants were categorized into three groups: (1) No Trauma (-T); (2) Trauma, No Symptoms (T - S); and (3) Trauma and Symptoms (T + S). Placental tissue was analyzed via bisulfite pyrosequencing for degree of methylation at the NR3C1 promoter and FKBP5 regulatory regions. Analyses of covariance were used to test group differences in percentages of NR3C1 and FKBP5 methylation overall and at each CpG site. We found a significant impact of PTSD symptoms on placental NR3C1 methylation. Compared to the -T group, the T + S group had greater NR3C1 methylation overall and at CpG6, CpG8, CpG9, and CpG13, but lower methylation at CpG5. The T + S group had significantly higher NR3C1 methylation overall and at CpG8 compared to the T - S group. There were no differences between the T - S group and - T group. Additionally, no group differences emerged for FKBP5 methylation. Pregnant trauma survivors with PTSD symptoms exhibited differential patterns of placental NR3C1 methylation compared to trauma survivors without PTSD symptoms and pregnant women unexposed to trauma. Results highlight the critical importance of interventions to address the mental health of pregnant trauma survivors.


Subject(s)
DNA Methylation , Receptors, Glucocorticoid , Stress Disorders, Post-Traumatic , Tacrolimus Binding Proteins , Adult , Female , Humans , Pregnancy , Young Adult , Epigenesis, Genetic , Pilot Projects , Placenta/metabolism , Pregnancy Complications/psychology , Receptors, Glucocorticoid/genetics , Stress Disorders, Post-Traumatic/genetics , Stress Disorders, Post-Traumatic/psychology , Tacrolimus Binding Proteins/genetics , Prenatal Exposure Delayed Effects/genetics
9.
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
10.
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
11.
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
12.
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
13.
Gen Comp Endocrinol ; 352: 114490, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38460737

ABSTRACT

Stressful experiences in early life can alter phenotypic expression later in life. For instance, in vertebrates, early life nutrient restriction can modify later life activity of the hypothalamic-pituitary-adrenal/interrenal axis (the HPI in amphibians), including the up- and downstream regulatory components of glucocorticoid signaling. Early life nutrient restriction can also influence later life behavior and metabolism (e.g., fat accumulation). Yet, less is known about whether nutrient stress-induced carryover effects on HPA/HPI axis regulation can vary across environmental contexts, such as the type of diet on which nutrient restriction occurs. Here, we experimentally address this question using the plains spadefoot toad (Spea bombifrons), whose larvae develop in ephemeral habitats that impose intense competition over access to two qualitatively distinct diet types: detritus and live shrimp prey. Consistent with diet type-specific carryover effects of early life nutrient restriction on later life HPI axis regulation, we found that temporary nutrient restriction at the larval stage reduced juvenile (i.e., post-metamorphic) brain gene expression of an upstream glucocorticoid regulator (corticotropin-releasing hormone) and two downstream regulators (glucocorticoid and mineralocorticoid receptors) only on the shrimp diet. These patterns are consistent with known diet type-specific effects of larval nutrient restriction on juvenile corticosterone and behavior. Additionally, larval nutrient restriction increased juvenile body fat levels. Our study indicates that HPA/HPI axis regulatory responses to nutrient restriction can vary remarkably across diet types. Such diet type-specific regulation of the HPA/HPI axis might provide a basis for developmental or evolutionary decoupling of stress-induced carryover effects.


Subject(s)
Corticotropin-Releasing Hormone , Glucocorticoids , Animals , Glucocorticoids/metabolism , Corticotropin-Releasing Hormone/metabolism , Hypothalamo-Hypophyseal System/metabolism , Corticosterone/metabolism , Anura/metabolism , Nutrients , Gene Expression , Pituitary-Adrenal System/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism
14.
J Mol Neurosci ; 74(1): 30, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38478195

ABSTRACT

Microglia are resident macrophages within the central nervous system, serving as the first responders to neuroinflammation. Glucocorticoids (GCs) may cause damage to brain tissue, but the specific mechanism remains unclear. This study was divided into two parts: a glucocorticoid receptor (GR) mitochondrial translocation intervention experiment and a mitochondrial oxidative stress inhibition experiment. BV-2 microglia were stimulated with dexamethasone (DEX) and treated with either tubastatin-A or mitoquinone (MitoQ) for 24 h. Our results showed that DEX increased the translocation of GRs to mitochondria, and this effect was accompanied by decreases in the expression of mitochondrially encoded cytochrome c oxidase 1 (MT-CO1) and mitochondrially encoded cytochrome c oxidase 3 (MT-CO3) and increases in the expression of NOD-like receptor thermal protein domain-associated protein 3 (NLRP3), caspase-1, and Gasdermin D (GSDMD). The level of mitochondrial respiratory chain complex IV (MRCC IV) and adenosine triphosphate (ATP) was decreased. An elevation in the level of mitochondrial oxidative stress and the opening of the mitochondrial permeability transition pore (mPTP) was also observed. Mechanistically, tubastatin-A significantly suppressed the mitochondrial translocation of GRs, improved the expression of mitochondrial genes, promoted the restoration of mitochondrial function, and inhibited pyroptosis. MitoQ significantly prevented mitochondrial oxidative stress, improved mitochondrial function, and reduced apoptosis and pyroptosis. Both tubastatin-A and MitoQ suppressed DEX-induced pyroptosis. This study substantiates that the increase in the mitochondrial translocation of GRs mediated by GCs exacerbates oxidative stress and pyroptosis in microglia, which indicates that the regulation of mitochondrial pathways by GCs is pathogenic to microglia.


Subject(s)
Glucocorticoids , Pyroptosis , Glucocorticoids/pharmacology , Glucocorticoids/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Microglia/metabolism , Electron Transport Complex IV/metabolism , Oxidative Stress , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
15.
J Endocrinol ; 261(2)2024 May 01.
Article in English | MEDLINE | ID: mdl-38451877

ABSTRACT

Glucocorticoids are steroid hormones, secreted by the adrenals to regulate a range of metabolic, immunologic, and homeostatic functions. Due to their potent anti-inflammatory effects, synthetic glucocorticoids are widely used to treat inflammatory disorders. However, their use especially at high doses and over the long-term is associated with several unwanted side effects that compromises their intended use (e.g. glucocorticoid-induced osteoporosis and/or diabetes, myopathy, and skin atrophy). Both endogenous and synthetic glucocorticoids exert their effects through the glucocorticoid receptor, a transcription factor present in nearly all nucleated cells. Glucocorticoid receptor knockout mouse models have proved to be valuable tools in understanding how glucocorticoids contribute to skeletal health and disease. These models, described in this review, have helped to establish that the effects of glucocorticoids on the skeleton are multifaceted, cell specific and concentration dependent. Intriguingly, while endogenous glucocorticoids are essential for bone formation, high-dose exogenous glucocorticoids may induce bone loss. Additionally, the actions of endogenous glucocorticoids vary greatly depending on the disease microenvironment. For example, endogenous glucocorticoids have predominately beneficial anti-inflammatory effects in rheumatoid arthritis, but detrimental actions in osteoarthritis by driving cartilage loss and abnormal bone formation. Studies in tissue-specific knockout models provide important insights that will aid the development of new glucocorticoid therapeutics that can specifically target certain cell types to minimise unwanted effects from current glucocorticoid therapy.


Subject(s)
Osteoporosis , Receptors, Glucocorticoid , Animals , Mice , Anti-Inflammatory Agents , Glucocorticoids/adverse effects , Mice, Knockout , Osteoporosis/chemically induced , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism
16.
J Investig Med ; 72(5): 449-456, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38494341

ABSTRACT

We aim to investigate the methylation of NR3C1 gene promotor and NR3C1 BclI polymorphism in schizophrenia (SCZ) patients with attempted suicide or non-suicidal self-injury (NSSI). A sample of 112 patients with SCZ was included in the study. Structured Clinical Interview for Diagnostic and Statistical Manual-Fourth Edition Axis I Disorders was used to confirm the diagnosis according to The Diagnostic and Statistical Manual of Mental Disorders, fourth edition, text revision criteria. The patients were evaluated by data forms that had sociodemographic, suicidal behavior, and NSSI information. Methylation-specific polymerase chain reaction (PCR) was used to identify the methylation of the NR3C1 gene. The analysis of the BclI polymorphism of the NR3C1 gene was evaluated by using the PCR restriction fragment length polymorphism. Our results revealed that although the NR3C1 gene methylation was not statistically significantly different, there was a significant difference in NR3C1 genotype distribution among the SCZ groups with and without attempted suicide. SCZ patients carrying the CC genotype had a lower risk of attempted suicide (Odds Ratio [OR]: 0.421; 95% Confidence Interval [CI]: 0.183-0.970; p = 0.040), while having the GG genotype in SCZ patients was associated with a higher risk of attempted suicide (OR: 3.785; 95% Cl: 1.107-12.945; p = 0.042). Additionally, due to NSSI in SCZ patients, there were no significant differences in NR3C1 gene methylation and NR3C1 genotype distribution among the groups. We propose that the NR3C1 BclI polymorphism may be associated with attempted suicide in Turkish patients diagnosed with SCZ.


Subject(s)
DNA Methylation , Receptors, Glucocorticoid , Schizophrenia , Self-Injurious Behavior , Suicide, Attempted , Humans , Male , Female , Receptors, Glucocorticoid/genetics , Adult , DNA Methylation/genetics , Schizophrenia/genetics , Self-Injurious Behavior/genetics , Genetic Predisposition to Disease , Polymorphism, Genetic , Middle Aged , Promoter Regions, Genetic
17.
Circ Res ; 134(10): 1306-1326, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38533639

ABSTRACT

BACKGROUND: Ventricular arrhythmias (VAs) demonstrate a prominent day-night rhythm, commonly presenting in the morning. Transcriptional rhythms in cardiac ion channels accompany this phenomenon, but their role in the morning vulnerability to VAs and the underlying mechanisms are not understood. We investigated the recruitment of transcription factors that underpins transcriptional rhythms in ion channels and assessed whether this mechanism was pertinent to the heart's intrinsic diurnal susceptibility to VA. METHODS AND RESULTS: Assay for transposase-accessible chromatin with sequencing performed in mouse ventricular myocyte nuclei at the beginning of the animals' inactive (ZT0) and active (ZT12) periods revealed differentially accessible chromatin sites annotating to rhythmically transcribed ion channels and distinct transcription factor binding motifs in these regions. Notably, motif enrichment for the glucocorticoid receptor (GR; transcriptional effector of corticosteroid signaling) in open chromatin profiles at ZT12 was observed, in line with the well-recognized ZT12 peak in circulating corticosteroids. Molecular, electrophysiological, and in silico biophysically-detailed modeling approaches demonstrated GR-mediated transcriptional control of ion channels (including Scn5a underlying the cardiac Na+ current, Kcnh2 underlying the rapid delayed rectifier K+ current, and Gja1 responsible for electrical coupling) and their contribution to the day-night rhythm in the vulnerability to VA. Strikingly, both pharmacological block of GR and cardiomyocyte-specific genetic knockout of GR blunted or abolished ion channel expression rhythms and abolished the ZT12 susceptibility to pacing-induced VA in isolated hearts. CONCLUSIONS: Our study registers a day-night rhythm in chromatin accessibility that accompanies diurnal cycles in ventricular myocytes. Our approaches directly implicate the cardiac GR in the myocyte excitability rhythm and mechanistically link the ZT12 surge in glucocorticoids to intrinsic VA propensity at this time.


Subject(s)
Circadian Rhythm , Myocytes, Cardiac , Receptors, Glucocorticoid , Animals , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/genetics , Mice , Myocytes, Cardiac/metabolism , Male , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/physiopathology , Arrhythmias, Cardiac/genetics , Mice, Inbred C57BL , NAV1.5 Voltage-Gated Sodium Channel/metabolism , NAV1.5 Voltage-Gated Sodium Channel/genetics , Connexin 43/metabolism , Connexin 43/genetics , Mice, Knockout , Action Potentials
18.
J Nutr Biochem ; 128: 109608, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38458474

ABSTRACT

Vitamin C (VitC) is maintained at high concentrations in the brain and is an essential micronutrient for brain function. VitC deficiency leads to neuropsychiatric scurvy, which is characterized by depression and cognitive impairment. However, the molecular mechanism by which mild VitC deficiency impairs brain function is currently unknown. In the present study, we conducted RNA sequencing analysis and found that a short-term VitC deficiency altered the brain transcriptome in ODS rats, which cannot synthesize VitC. Bioinformatic analysis indicated that VitC deficiency affected the expression of genes controlled by the glucocorticoid receptor in the brain. We confirmed an increased secretion of glucocorticoids from the adrenal gland during VitC deficiency. We found that non-neuronal cells, including microglia, which are resident immune cells in the brain, changed their transcriptional patterns in response to VitC deficiency. Immunohistochemical analysis revealed that the quiescent ramified microglia transform into the activated amoeboid microglia during three weeks of VitC deficiency. The morphological activation of microglia was accompanied by increased expression of proinflammatory cytokines such as interleukin-6 in the hippocampus. Furthermore, VitC deficiency decreased the number of newly born neurons in the dentate gyrus of the hippocampus, suggesting that VitC was required for adult neurogenesis that plays a crucial role in learning and memory. Our findings may provide insights into the molecular mechanisms underlying the maintenance of normal brain function by adequate levels of VitC.


Subject(s)
Ascorbic Acid Deficiency , Brain , Glucocorticoids , Microglia , Neurogenesis , Transcriptome , Animals , Microglia/metabolism , Rats , Brain/metabolism , Male , Glucocorticoids/metabolism , Ascorbic Acid Deficiency/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/genetics , Hippocampus/metabolism , Ascorbic Acid/metabolism , Ascorbic Acid/pharmacology
19.
Dev Psychobiol ; 66(3): e22482, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38482889

ABSTRACT

Early life deprivation and stress can contribute to life-long, problematic consequences, including epigenetic variations related to behavior and health. Domestic dogs share human environments and social-cognitive traits, making them a promising comparative model to examine developmental plasticity. We examined 47 owner-dog dyads, including dogs rescued from abusive or neglectful environments, and matched control dogs for changes in DNA methylation of glucocorticoid (NR3C1) and oxytocin (OXTR) receptor genes previously shown to be affected by early life stress in other species including humans. We used an attachment paradigm, which included a separation event to examine cortisol levels and owner-dog attachment styles. Overall, dogs with adverse histories had different NR3C1 methylation patterns as a function of age and less OXTR methylation than comparison dogs. Dogs with adverse histories did not differ in their cortisol change from baseline to poststressor from comparison dogs, but the change in cortisol was associated with NR3C1 methylation. In addition, dogs with a history of early life stress had more insecure attachment styles; for every unit increase of OXTR methylation, the odds increased for insecure attachment style. This study demonstrates that adverse life histories lead to methylation differences, resulting in the hypothalamic-pituitary-adrenal (HPA) axis's dysregulation and differences in behavioral phenotypes.


Subject(s)
Glucocorticoids , Receptors, Oxytocin , Humans , Dogs , Animals , Receptors, Oxytocin/genetics , Oxytocin/metabolism , Hydrocortisone , Receptors, Glucocorticoid/genetics , DNA Methylation
20.
Sci Rep ; 14(1): 5567, 2024 03 06.
Article in English | MEDLINE | ID: mdl-38448539

ABSTRACT

Progesterone receptor (PR)-interacting compounds in the environment are associated with serious health hazards. However, methods for their detection in environmental samples are cumbersome. We report a sensitive activity-based biosensor for rapid and reliable screening of progesterone receptor (PR)-interacting endocrine disrupting chemicals (EDCs). The biosensor is a cell line which expresses nuclear mCherry-NF1 and a green fluorescent protein (GFP)-tagged chimera of glucocorticoid receptor (GR) N terminus fused to the ligand binding domain (LBD) of PR (GFP-GR-PR). As this LBD is shared by the PRA and PRB, the biosensor reports on the activation of both PR isoforms. This GFP-GR-PR chimera is cytoplasmic in the absence of hormone and translocates rapidly to the nucleus in response to PR agonists or antagonists in concentration- and time-dependent manner. In live cells, presence of nuclear NF1 label eliminates cell fixation and nuclear staining resulting in efficient screening. The assay can be used in screens for novel PR ligands and PR-interacting contaminants in environmental samples. A limited screen of river water samples indicated a widespread, low-level contamination with PR-interacting contaminants in all tested samples.


Subject(s)
Endocrine Disruptors , Receptors, Progesterone/genetics , Biological Assay , Cell Line , Cytoplasm , Green Fluorescent Proteins/genetics , Receptors, Glucocorticoid/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...