Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.757
Filter
Add more filters

Publication year range
1.
Ecotoxicol Environ Saf ; 279: 116499, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38805828

ABSTRACT

There are various substances that can disrupt the homeostatic mechanisms of the body, defined as endocrine-disrupting chemicals (EDCs). The persistent nature of microplastics (MPs) is a cause for concern due to their ability to accumulate in food chains and widespread use, making their toxic effects particularly alarming. The potential of MPs for disrupting the endocrine system was observed in multiple tissues. Moreover, the adrenal gland is known to be extremely sensitive to EDCs, while with the effect of MPs on the adrenal gland has not previously been studied. This study aimed to highlight the potential polyethylene microplastics (PE-MPs) induced adreno-toxic effects rather than exploring the implicated mechanisms and concluding if melatonin (Mel) can afford protection against PE-MPs induced adreno-toxicity. To fulfill the goal, six groups of rats were used; control, Mel, PE-MPs (3.75 mg/kg), PE-MPs (15 mg/kg), PE-MPs (3.75 mg/kg) +Mel, and PE-MPs (15 mg/kg) +Mel. PE-MPs induced toxic changes in the adrenal cortex, which was evident by increased adrenal weight, histopathological examination, and ultrastructural changes detected by electron microscope. A reduction in serum cortisol and an increase in serum adrenocorticotropic hormone resulted from the adreno-toxic effects of PE-MPs. Mechanisms may include the reduction of steroidogenesis-related genes, as PE-MPs drastically reduce mRNA levels of StAR, Nr0b1, Cyp11A1, as well as Cyp11B1. Also, oxidative stress that results from PE-MPs is associated with higher rates of lipid peroxidation and decreased superoxide dismutase and glutathione. PE-MPs inflammatory effect was illustrated by elevated expression of IL-1ß and NF-kB, detected by immunohistochemical staining, in addition to increased expression of caspase-3 and mRNA of Bax, markers of proapoptotic activity. The impacts of PE-MPs were relatively dose-related, with the higher dose showing more significant toxicity than the lower one. Mel treatment was associated with a substantial amelioration of PE-MPs-induced toxic changes. Collectively, this study fills the knowledge gap about the MPs-induced adrenal cortex and elucidates various related toxic mechanisms. It also supports Mel's potential protective activity through antioxidant, anti-inflammatory, anti-apoptotic, and gene transcription regulatory effects.


Subject(s)
Melatonin , Microplastics , Polyethylene , Animals , Melatonin/pharmacology , Male , Rats , Polyethylene/toxicity , Microplastics/toxicity , Oxidative Stress/drug effects , Endocrine Disruptors/toxicity , Adrenal Cortex/drug effects , Adrenal Cortex/pathology , Antioxidants/metabolism , Antioxidants/pharmacology , Rats, Wistar
2.
Biochem Biophys Res Commun ; 534: 672-679, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33220920

ABSTRACT

Aldosterone is synthesized in the adrenal by the aldosterone synthase CYP11B2. Although the control of CYP11B2 expression is important to maintain the mineral homeostasis, its overexpression induced by the depolarization-induced calcium (Ca2+) signaling activation has been reported to increase the synthesis of aldosterone in primary aldosteronism (PA). The drug against PA focused on the suppression of CYP11B2 expression has not yet been developed, since the molecular mechanism of CYP11B2 transcriptional regulation activated via Ca2+ signaling remains unclear. To address the issue, we attempted to reveal the mechanism of the transcriptional regulation of CYP11B2 using chemical screening. We generated a cell line by inserting Nanoluc gene as a reporter into CYP11B2 locus in H295R adrenocortical cells using the CRSPR/Cas9 system, and established the high-throughput screening system using the cell line. We then identified 9 compounds that inhibited the CYP11B2 expression induced by potassium-mediated depolarization from the validated compound library (3399 compounds). Particularly, tacrolimus, an inhibitor of phosphatase calcineurin, strongly suppressed the CYP11B2 expression even at 10 nM. These results suggest that the system is effective in identifying drugs that suppress the depolarization-induced CYP11B2 expression. Our screening system may therefore be a useful tool for the development of novel medicines against PA.


Subject(s)
Cytochrome P-450 CYP11B2/antagonists & inhibitors , Cytochrome P-450 CYP11B2/genetics , Gene Editing/methods , High-Throughput Screening Assays/methods , Adrenal Cortex/drug effects , Adrenal Cortex/metabolism , Aldosterone/biosynthesis , Base Sequence , Calcium Signaling , Cell Line , Drug Evaluation, Preclinical/methods , Gene Expression Regulation, Enzymologic/drug effects , Genes, Reporter , Humans , Hyperaldosteronism/drug therapy , Hyperaldosteronism/genetics , Hyperaldosteronism/metabolism , RNA, Guide, Kinetoplastida/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Steroid 11-beta-Hydroxylase/genetics , Tacrolimus/pharmacology
3.
J Pharmacol Exp Ther ; 379(3): 324-330, 2021 12.
Article in English | MEDLINE | ID: mdl-34521699

ABSTRACT

Etomidate is a potent and rapidly acting anesthetic with high therapeutic index (TI) and superior hemodynamic stability. However, side effect of suppressing adrenocortical function limits its clinical use. To overcome this side effect, we designed a novel etomidate analog, EL-0052, aiming to retain beneficial properties of etomidate and avoid its disadvantage of suppressing adrenocortical steroid synthesis. Results exhibited that EL-0052 enhanced GABAA receptors currents with a concentration for EC50 of 0.98 ± 0.02 µM, which was about three times more potent than etomidate (3.07 ± 1.67 µM). Similar to hypnotic potency of etomidate, EL-0052 exhibited loss of righting reflex with ED50s of 1.02 (0.93-1.20) mg/kg in rats and 0.5 (0.45-0.56) mg/kg in dogs. The TI of EL-0052 in rats was 28, which was higher than 22 of etomidate. There was no significant difference in hypnotic onset time, recovery time, and walking time between EL-0052 and etomidate in rats. Both of them had minor effects on mean arterial pressure in dogs. EL-0052 had no significant effect on adrenocortical function in dogs even at a high dose (4.3 × ED50), whereas etomidate significantly inhibited corticosteroid secretion. The inhibition of cortisol synthesis assay showed that EL-0052 had a weak inhibition on cortisol biosynthesis in human H259 cells with an IC50 of 1050 ± 100 nM, which was 2.09 ± 0.27 nM for etomidate. EL-0052 retains the favorable properties of etomidate, including potent hypnotic effect, rapid onset and recovery, stable hemodynamics, and high therapeutic index without suppression of adrenocortical function. SIGNIFICANCE STATEMENT: The novel etomidate analog EL-0052 retains the favorable properties of etomidate without suppressing adrenocortical function and provides a new strategy to optimize the structure of etomidate.


Subject(s)
Adrenal Cortex/drug effects , Blood Pressure/drug effects , Etomidate/analogs & derivatives , Etomidate/pharmacology , Hemodynamics/drug effects , Hypnotics and Sedatives/pharmacology , Adrenal Cortex/metabolism , Animals , Blood Pressure/physiology , Corticosterone/blood , Dogs , Dose-Response Relationship, Drug , Female , HEK293 Cells , Hemodynamics/physiology , Humans , Male , Rats , Rats, Wistar , Reflex, Righting/drug effects , Reflex, Righting/physiology
4.
Endocr J ; 68(4): 441-450, 2021 Apr 28.
Article in English | MEDLINE | ID: mdl-33390420

ABSTRACT

Angiotensin II (Ang II) is a well-known peptide that maintains the balance of electrolytes in the higher vertebrates. Ang II stimulation in the adrenal gland induces the synthesis of mineralocorticoids, mainly aldosterone, through the up-regulation of aldosterone synthase (CYP11B2) gene expression. Additionally, it has been reported that Ang II activates multiple signaling pathways such as mitogen-activated protein kinase (MAPK) and Ca2+ signaling. Although Ang II has various effects on the cellular signaling in the adrenal cells, its biological significance, except for the aldosterone synthesis, is still unclear. In this study, we attempted to search the novel target gene(s) of Ang II in the human adrenal H295R cells using a proteomic approach combined with stable isotopic labeling using amino acid in cell culture (SILAC). Interestingly, we found that Ang II stimulation elevated the expression of phosphofructokinase type platelet (PFKP) in both protein and mRNA levels. Moreover, transactivation of PFKP by Ang II was dependent on extracellular-signal-regulated kinase (ERK) 1/2 activation. Finally, we observed that Ang II treatment facilitated glucose uptake in the H295R cells. Taken together, we here identified PFKP as a novel target gene of Ang II, indicating that Ang II not only stimulates steroidogenesis but also affects glucose metabolism.


Subject(s)
Adrenal Cortex/drug effects , Cytochrome P-450 CYP11B2/genetics , Gene Expression/drug effects , Adrenal Cortex/metabolism , Angiotensin II/pharmacology , Cell Line , Cytochrome P-450 CYP11B2/metabolism , Humans , Proteomics , Signal Transduction/drug effects , Up-Regulation/drug effects
5.
Endocr J ; 68(2): 243-250, 2021 Feb 28.
Article in English | MEDLINE | ID: mdl-33028758

ABSTRACT

Functional interactions between the levels of clock gene expression and adrenal steroidogenesis were studied in human adrenocortical H295R cells. Fluctuations of Bmal1, Clock, Per2 and Cry1 mRNA levels were found in H295R cells treated with forskolin (FSK) in a serum-free condition. The changes of clock gene expression levels were diverged, with Clock mRNA level being significantly higher than Cry1 and Per2 mRNA levels after 12-h stimulation with FSK. After FSK induction, mRNA levels of StAR and CYP11B2 were highest at 12 hours and CYP17 mRNA level reached a peak at 6 hours, but HSD3B1 mRNA level was transiently decreased at 3 hours. The expression levels of Clock mRNA showed a significant positive correlation with StAR among the interrelationships between mRNA levels of key steroidogenic factors and clock genes. Knockdown of Clock gene by siRNA led to a significant reduction of FSK-induced expression of StAR and CYP17 after 12-h treatment with FSK. BMP-6 and activin, which modulate adrenal steroidogenesis, had inhibitory effects on Clock mRNA expression, whereas treatment with follistatin, a binding protein of activin, increased Clock mRNA levels in the presence of FSK, suggesting an endogenous function of activin in regulation of Clock mRNA expression. Collectively, the results indicated that changes of Clock mRNA expression, being upregulated by FSK and suppressed by BMP-6 and activin, were tightly linked to StAR expression by human adrenocortical cells.


Subject(s)
Activins/metabolism , Adrenal Cortex/metabolism , Bone Morphogenetic Proteins/metabolism , CLOCK Proteins/metabolism , Activins/genetics , Adrenal Cortex/drug effects , Bone Morphogenetic Proteins/genetics , CLOCK Proteins/genetics , Cell Line, Tumor , Colforsin/pharmacology , Cytochrome P-450 CYP11B2/genetics , Cytochrome P-450 CYP11B2/metabolism , Gene Expression/drug effects , Gene Expression Regulation/drug effects , Humans , Phosphoproteins/genetics , Phosphoproteins/metabolism
6.
Article in English | MEDLINE | ID: mdl-34464741

ABSTRACT

The present study examined how food availability interacts with age to modulate lizard adrenal steroidogenic function at the cellular level. Adult male and juvenile male and female Eastern Fence Lizards (Sceloporus undulatus) underwent a period of food deprivation with or without a shorter re-feeding period. Lizards maintained on a full feeding regimen served as the controls. Across the feeding regimens, plasma corticosterone of adult lizards was unchanged whereas that of food-deprived juvenile lizards was increased nearly 7 times and this increase was normalized by a short re-feeding period. Freshly dispersed adrenocortical cells derived from these lizards were incubated with ACTH and the production of selected steroids was measured by highly specific radioimmunoassay. Net maximal steroid rates of juvenile cells were 161% greater than those of adult cells. Adult and juvenile progesterone rates were consistently suppressed by food deprivation (by nearly 48%) and were normalized by a re-feeding period, whereas divergent effects were seen with corticosterone and aldosterone rates. Food deprivation suppressed corticosterone rates of adult cells by 43% but not those of juvenile cells. In a reciprocal manner, food deprivation had no significant effect on aldosterone rates of adult cells, but it suppressed those of juvenile cells by 52%. A short re-feeding period normalized most rates in both adult and juvenile cells and further augmented the adult aldosterone rate by 54%. The effect of the feeding regimens on ACTH sensitivity varied with life stage and with steroid. The overall sensitivity of adult cells to ACTH was nearly three times that of juvenile cells. Collectively, the data presented here and data from previous work indicate that food restriction/deprivation in Sceloporus lizard species causes a functional remodeling of the adrenocortical tissue. Furthermore, life stage adds more complexity to this remodeling.


Subject(s)
Adrenal Cortex/drug effects , Adrenocorticotropic Hormone/pharmacology , Corticosterone/blood , Food Deprivation , Lizards/blood , Adrenal Cortex/metabolism , Age Factors , Animals , Female , Male
7.
J Cell Physiol ; 235(12): 9432-9444, 2020 12.
Article in English | MEDLINE | ID: mdl-32346900

ABSTRACT

The purpose of this study was to investigate the potential mechanism of interleukin-6 (IL-6) on the stimulation of excessive androgen secretion in human NCI-H295R adrenocortical cells. We performed transcriptome sequencing of cancer and paracancerous tissues obtained from functional adrenal cortical adenomas. The secretion of dehydroepiandrosterone sulfate (DHEAS) in NCI-H295R cells was detected by a chemiluminescence assay. The expression of messenger RNA (mRNA) was detected by real-time polymerase chain reaction and that of protein was detected by western blotting. The expression of secretogranin II (SCG2) and IL-6 were significantly increased in cancer tissues. Upregulation of mRNA and protein levels of AKR1C3, CYP11A, CYP17A1, 3ßHSD, and SULT2A1 was observed after stimulation with IL-6. IL-6 could also increase the expression of StAR mRNA and proteins. Our results suggest that IL-6 can promote androgen secretion by regulating the expression of genes related to androgen pathways.


Subject(s)
Adrenal Cortex/drug effects , Androgens/metabolism , Interleukin-6/pharmacology , Transcriptional Activation/drug effects , Adrenal Cortex/metabolism , Blotting, Western/methods , Cell Line, Tumor , Humans , Hydrocortisone/metabolism , Interleukin-6/metabolism , RNA, Messenger/genetics
8.
Biochem Biophys Res Commun ; 524(1): 184-189, 2020 03 26.
Article in English | MEDLINE | ID: mdl-31982132

ABSTRACT

Plasma aldosterone concentration increases in proportion to the severity of heart failure, even during treatment with renin-angiotensin system inhibitors. This study investigated alternative regulatory mechanisms of aldosterone production that are significant in heart failure. Dahl salt-sensitive rats on a high-salt diet, a rat model of heart failure with cardio-renal syndrome, had high plasma aldosterone levels and elevated ß3-adrenergic receptor expression in hypoxic zona glomerulosa cells. In H295R cells (a human adrenocortical cell line), hypoxia-induced ß3-adrenergic receptor expression. Hypoxia-mediated ß3-adrenergic receptor expression augmented aldosterone production by facilitating hydrolysis of lipid droplets though ERK-mediated phosphorylation of hormone-sensitive lipase, also known as cholesteryl ester hydrolase. Hypoxia also accelerated the synthesis of cholesterol esters by acyl-CoA:cholesterol acyltransferase, thereby increasing the cholesterol ester content in lipid droplets. Thus, hypoxia enhanced aldosterone production by zona glomerulosa cells via promotion of the accumulation and hydrolysis of cholesterol ester in lipid droplets. In conclusion, hypoxic zona glomerulosa cells with heart failure show enhanced aldosterone production via increased catecholamine responsiveness and activation of cholesterol trafficking, irrespective of the renin-angiotensin system.


Subject(s)
Adrenal Cortex/pathology , Aldosterone/biosynthesis , Heart Failure/metabolism , Heart Failure/pathology , Hypoxia/metabolism , Hypoxia/pathology , Adrenal Cortex/drug effects , Animals , Cardio-Renal Syndrome/complications , Catecholamines/pharmacology , Cell Hypoxia/drug effects , Cell Line , Cholesterol/metabolism , Disease Models, Animal , Humans , Hypoxia/complications , Male , Phosphorylation/drug effects , Rats, Inbred Dahl , Receptors, Adrenergic, beta-3/metabolism , Sterol Esterase/metabolism , Zona Glomerulosa/metabolism , Zona Glomerulosa/pathology
9.
FASEB J ; 33(9): 10218-10230, 2019 09.
Article in English | MEDLINE | ID: mdl-31208233

ABSTRACT

SUMOylation is a highly conserved and dynamic post-translational mechanism primarily affecting nuclear programs for adapting organisms to stressful challenges. Alteration of SUMOylation cycles leads to severe developmental and homeostatic defects and malignancy, but signals coordinating SUMOylation are still unidentified. The adrenal cortex is a zonated endocrine gland that controls body homeostasis and stress response. Here, we show that in human and in mouse adrenals, SUMOylation follows a decreasing centripetal gradient that mirrors cortical differentiation flow and delimits highly and weakly SUMOylated steroidogenic compartments, overlapping glomerulosa, and fasciculata zones. Activation of PKA signaling by acute hormonal treatment, mouse genetic engineering, or in Carney complex results in repression of small ubiquitin-like modifier (SUMO) conjugation in the inner cortex by coordinating expression of SUMO pathway inducers and repressors. Conversely, genetic activation of canonical wingless-related integration site signaling maintains high SUMOylation potential in the outer neoplastic cortex. Thus, SUMOylation is tightly regulated by signaling pathways that orchestrate adrenal zonation and diseases.-Dumontet, T., Sahut-Barnola, I., Dufour, D., Lefrançois-Martinez, A.-M., Berthon, A., Montanier, N., Ragazzon, B., Djari, C., Pointud, J.-C., Roucher-Boulez, F., Batisse-Lignier, M., Tauveron, I., Bertherat, J., Val, P., Martinez, A. Hormonal and spatial control of SUMOylation in the human and mouse adrenal cortex.


Subject(s)
Adrenal Cortex/metabolism , Adrenocorticotropic Hormone/pharmacology , Protein Processing, Post-Translational/physiology , Sumoylation/physiology , Adrenal Cortex/drug effects , Adrenal Cortex/ultrastructure , Adrenal Cortex Neoplasms/pathology , Adrenocorticotropic Hormone/administration & dosage , Animals , Carney Complex/metabolism , Cell Line, Tumor , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/physiology , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Delayed-Action Preparations , Dexamethasone/analogs & derivatives , Dexamethasone/pharmacology , Female , Humans , Mice , Mice, Knockout , Mice, Transgenic , Neoplasm Proteins/metabolism , Protein Processing, Post-Translational/drug effects , Signal Transduction/drug effects , Sumoylation/drug effects , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/physiology , Zona Fasciculata/drug effects , Zona Fasciculata/metabolism , Zona Glomerulosa/drug effects , Zona Glomerulosa/metabolism , beta Catenin/deficiency , beta Catenin/genetics
10.
J Endocrinol Invest ; 43(3): 357-367, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31587178

ABSTRACT

PURPOSE: Mitotane is the only chemotherapeutic agent available for the treatment of adrenocortical carcinoma (ACC), however, the anti-neoplastic efficacy is limited due to several side-effects in vivo. There is, therefore, a need of exploring for new anti-tumoral agents which can be used either alone or in combination with mitotane. The active vitamin D metabolite 1α,25-dihydroxyvitamin D3 (1α,25(OH)2D3) acts as an anti-proliferative agent in human cancer by inhibiting the Wnt/beta-catenin pathway through the vitamin D receptor (VDR). The aim of this study was to study the effects of mitotane and 1α,25(OH)2D3, individually or in combination, in an in vitro model with H295R ACC cells, and to elucidate the molecular events behind their effects involving the Wnt/beta-catenin signaling. METHODS AND RESULTS: Multiple concentrations of mitotane and 1α,25(OH)2D3, individually or in combination, were tested on H295R cells for 24-96 h, and the effects analysed by MTT. A reduction in cell growth was observed in a dose/time-dependent manner for both mitotane and 1α,25(OH)2D3. In addition, a combination of clinically sub-therapeutic concentrations of mitotane with 1α,25(OH)2D3, had an additive anti-proliferative effect (Combination Index = 1.02). In a wound healing assay, individual treatments of both mitotane and 1α,25(OH)2D3 reduced the migration ability of H295R cells, with the effect further enhanced on combining both the agents. Western blotting and qRT-PCR analysis showed a modulation of the Wnt/beta-catenin and VDR signaling pathways. CONCLUSION: Our results show an additive effect of mitotane and 1α,25(OH)2D3 on the inhibition of H295R ACC cell growth and viability, and suggest that molecular mechanisms of their effects involve a functional link between VDR and Wnt/beta-catenin pathways.


Subject(s)
Adrenal Cortex Neoplasms/metabolism , Adrenal Cortex/drug effects , Adrenocortical Carcinoma/metabolism , Calcitriol/pharmacology , Mitotane/pharmacology , Wnt Signaling Pathway/drug effects , Adrenal Cortex/metabolism , Cell Line, Tumor , Humans , beta Catenin/metabolism
11.
Microsc Microanal ; 26(3): 509-523, 2020 06.
Article in English | MEDLINE | ID: mdl-32366353

ABSTRACT

Tramadol is a centrally acting analgesic drug, used for the management of moderate to severe pain in a variety of diseases. The long-term use of tramadol can induce endocrinopathy. This study aimed to evaluate the effect of tramadol dependence on the adrenal cortex and the effect of its withdrawal. Thirty adult male rats were divided into three experimental groups: the control group, the tramadol-dependent group that received increasing therapeutic doses of tramadol orally for 1 month, and the recovery group that received tramadol in a dose and duration similar to the previous group followed by a withdrawal period for another month. Specimens from the adrenal cortex were processed for histological, immunohistochemical, enzyme assay, and quantitative real-time PCR (RT-qPCR) studies. Tramadol induced a significant increase in malondialdehyde level and a significant decrease in the levels of glutathione peroxidase and superoxide dismutase. A significant decrease in the levels of adrenocorticotrophic hormones, aldosterone, cortisol, corticosterone, and dehydroepiandrosterone sulfate was also detected. Severe histopathological changes in the adrenal cortex were demonstrated in the form of disturbed architecture, swollen cells, and shrunken cells with pyknotic nuclei. Inflammatory cellular infiltration and variable-sized homogenized areas were also detected. A significant increase in P53 and Bax immunoreaction was detected and confirmed by RT-qPCR. The ultrastructural examination showed irregular, shrunken adrenocorticocytes with dense nuclei. Dilated smooth endoplasmic reticulum, mitochondria with disrupted cristae, and numerous coalesced lipid droplets were also demonstrated. All these changes started to return to normal after the withdrawal of tramadol. Thus, it was confirmed that the long-term use of tramadol can induce severe adrenal changes with subsequent insufficiency.


Subject(s)
Adrenal Cortex/drug effects , Apoptosis/drug effects , Fibrosis/drug therapy , Oxidative Stress/drug effects , Tramadol/pharmacology , Adrenal Cortex/pathology , Animals , Fibrosis/pathology , Male , Microscopy, Electron , Rats , Testis/pathology
12.
Int J Toxicol ; 38(4): 279-290, 2019.
Article in English | MEDLINE | ID: mdl-31132918

ABSTRACT

The use of organophosphates phosphate flame retardants, particularly isopropylated triphenyl phosphate (IPTPP), has increased in recent years as replacements for polybrominated diphenyl ethers. This is despite limited understanding of the hazards of IPTPP. To examine the general and endocrine toxicity of IPTPP, adult Wistar rats were fed for 90 days on diets containing IPTPP estimated to deliver daily doses of 5 to 140 mg/kg/d. Exposure to IPTPP caused a dose-related increase in liver and adrenal gland weight in both sexes. Cells in the zona fasciculate (ZF) of the adrenal cortex were observed to be filled with droplets that stained with Nile red, suggesting they contained neutral lipid. Despite marked structural changes, there was no change in basal or stress-induced serum levels of their major secreted ZF product corticosterone (B), suggesting cell function was not altered. There were no effects on responses to glucose or insulin challenge, but serum levels of fructosamine were elevated by IPTPP exposure, suggesting a slight tendency of exposed animals to be hyperglycemic. Serum levels of total cholesterol and high-density lipoprotein cholesterol were significantly elevated in both sexes at the 2 highest doses. This study demonstrates that IPTPP exposure causes hypertrophy and neutral lipid accumulation in adrenal cortex ZF cells but does not result in impaired B production.


Subject(s)
Adrenal Cortex/drug effects , Flame Retardants/toxicity , Lipid Metabolism/drug effects , Liver/drug effects , Organophosphates/toxicity , Adrenal Cortex/metabolism , Adrenal Cortex/pathology , Animals , Corticosterone/blood , Cytochrome P-450 CYP1A1/metabolism , Cytochrome P-450 CYP2B1/metabolism , Female , Liver/enzymology , Liver/pathology , Male , Organophosphates/chemistry , Rats, Wistar
13.
Toxicol Appl Pharmacol ; 341: 64-76, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29343424

ABSTRACT

Our previous study proposed a glucocorticoid-insulin-like growth factor 1 (GC-IGF1) axis programming mechanism for prenatal caffeine exposure (PCE)-induced adrenal developmental dysfunction. Here, we focused on PCE-induced cell proliferation changes of the adrenal cortex in male offspring rats before and after birth and clarified the intrauterine programming mechanism. On gestational day (GD) 20, the PCE group had an elevated serum corticosterone level reduced fetal bodyweight, maximum adrenal sectional area, and elevated adrenal corticosterone and aldosterone contents. However, in postnatal week (PW) 6, the serum corticosterone level was decreased, and the bodyweight, with catch-up growth, adrenal cortex maximum cross-sectional area and aldosterone content were relatively increased, while the adrenal corticosterone content was lower. On GD20, the expression of adrenal IGF1, IGF1R and proliferating cell nuclear antigen (PCNA) were decreased, while the expression of these factors at PW6 were increased in the PCE group. Fetal adrenal gene chip analysis suggested that the mitogen-activated protein kinase/extracellular regulated protein kinase (MAPK/ERK) signal pathway was suppressed in the PCE group. Moreover, in the rat primary adrenal cells, corticosterone (rather than caffeine) was shown to significantly inhibit cell proliferation, IGF1 and PCNA expression, and ERK phosphorylation, which could be reversed by exogenous IGF1. Meanwhile, the effects of exogenous IGF1 were reversed by the ERK pathway inhibitor (PD184161). In conclusion, PCE could induce programming alterations in adrenal cortical cell proliferation before and after birth in male offspring rats. The underlying mechanism is associated with the inhibition of fetal adrenal IGF1-related MAPK/ERK signaling pathway caused by high glucocorticoid levels.


Subject(s)
Adrenal Cortex/metabolism , Caffeine/toxicity , Cell Proliferation/physiology , Insulin-Like Growth Factor I/metabolism , MAP Kinase Signaling System/physiology , Prenatal Exposure Delayed Effects/metabolism , Adrenal Cortex/cytology , Adrenal Cortex/drug effects , Animals , Body Weight/drug effects , Body Weight/physiology , Caffeine/administration & dosage , Cell Proliferation/drug effects , Cells, Cultured , Cellular Reprogramming/drug effects , Cellular Reprogramming/physiology , Dose-Response Relationship, Drug , Female , Insulin-Like Growth Factor I/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Male , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Random Allocation , Rats , Rats, Wistar
14.
Cell Mol Neurobiol ; 38(1): 25-35, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28895001

ABSTRACT

Hindbrain neurons in the nucleus of the solitary tract (NTS) are critical for regulation of hypothalamo-pituitary-adrenocortical (HPA) responses to stress. It is well known that noradrenergic (as well as adrenergic) neurons in the NTS send direct projections to hypophysiotropic corticotropin-releasing hormone (CRH) neurons and control activation of HPA axis responses to acute systemic (but not psychogenic) stressors. Norepinephrine (NE) signaling via alpha1 receptors is primarily excitatory, working either directly on CRH neurons or through presynaptic activation of glutamate release. However, there is also evidence for NE inhibition of CRH neurons (possibly via beta receptors), an effect that may occur at higher levels of stimulation, suggesting that NE effects on the HPA axis may be context-dependent. Lesions of ascending NE inputs to the paraventricular nucleus attenuate stress-induced ACTH but not corticosterone release after chronic stress, indicating reduction in central HPA drive and increased adrenal sensitivity. Non-catecholaminergic NTS glucagon-like peptide 1/glutamate neurons play a broader role in stress regulation, being important in HPA activation to both systemic and psychogenic stressors as well as HPA axis sensitization under conditions of chronic stress. Overall, the data highlight the importance of the NTS as a key regulatory node for coordination of acute and chronic stress.


Subject(s)
Adrenal Cortex/metabolism , Aortic Bodies/metabolism , Hypothalamo-Hypophyseal System/physiology , Pituitary-Adrenal System/physiology , Solitary Nucleus/metabolism , Stress, Psychological/metabolism , Adrenal Cortex/drug effects , Animals , Aortic Bodies/drug effects , Corticosterone/metabolism , Corticosterone/pharmacology , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide 1/pharmacology , Glutamic Acid/metabolism , Glutamic Acid/pharmacology , Humans , Hypothalamo-Hypophyseal System/drug effects , Pituitary-Adrenal System/drug effects , Solitary Nucleus/drug effects , Stress, Psychological/psychology
15.
Br J Anaesth ; 120(6): 1401-1411, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29793605

ABSTRACT

BACKGROUND: Cyclopropyl-methoxycarbonyl metomidate, or ABP-700, is a second generation analogue of etomidate, developed to retain etomidate's beneficial haemodynamic and respiratory profile but diminishing its suppression of the adrenocortical axis. The objective of this study was to characterise the safety and efficacy of 30-min continuous infusions of ABP-700, and to assess its effect on haemodynamics and the adrenocortical response in healthy human volunteers. METHODS: Five cohorts involving 40 subjects received increasing infusion doses of ABP-700, propofol 60 µg kg-1 min-1 or placebo. Safety was evaluated through adverse event (AE) monitoring, safety laboratory tests, and arterial blood gasses. Haemodynamic and respiratory stability were assessed by continuous monitoring. Adrenocortical function was analysed by adrenocorticotropic hormone (ACTH) stimulation tests. Clinical effect was measured using the modified observer's assessment of alertness/sedation (MOAA/S) and continuous bispectral index monitoring. RESULTS: No serious AEs were reported. Haemodynamic and respiratory effects included mild dose-dependent tachycardia, slightly elevated blood pressure, and no centrally mediated apnoea. Upon stimulation with ACTH, no adrenocortical depression was observed in any subject. Involuntary muscle movements (IMM) were reported, which were more extensive with higher dosing regimens. Higher dosages of ABP-700 were associated with deeper sedation and increased likelihood of sedation. Time to onset of clinical effect was variable throughout the cohorts and recovery was swift. CONCLUSIONS: Infusions of ABP-700 showed a dose-dependent hypnotic effect, and did not cause severe hypotension, severe respiratory depression, or adrenocortical suppression. The presentation and nature of IMM is a matter of concern. CLINICAL TRIAL REGISTRATION: NTR4735.


Subject(s)
Anesthetics, Intravenous/adverse effects , Etomidate/analogs & derivatives , Adolescent , Adrenal Cortex/drug effects , Adrenal Cortex/metabolism , Adult , Anesthetics, Intravenous/administration & dosage , Anesthetics, Intravenous/pharmacology , Blood Pressure/drug effects , Dose-Response Relationship, Drug , Etomidate/administration & dosage , Etomidate/adverse effects , Etomidate/pharmacology , Female , Healthy Volunteers , Heart Rate/drug effects , Humans , Infusions, Intravenous , Male , Middle Aged , Propofol/adverse effects , Propofol/pharmacology , Respiratory Mechanics/drug effects , Single-Blind Method , Young Adult
16.
Arch Toxicol ; 92(5): 1703-1715, 2018 05.
Article in English | MEDLINE | ID: mdl-29574565

ABSTRACT

Obesity is increasing worldwide, and since obesity is associated with dyslipidemia, the consumption of cholesterol-lowering pharmaceuticals has increased. The aim of this study was therefore to study potential endocrine disrupting effects of one of the world's most frequently prescribed drugs, the cholesterol-lowering drug, atorvastatin (ATO) in vitro using the H295R steroidogenesis assay and in vivo using male Sprague-Dawley rats. We analyzed all major steroids in the mammalian steroidogenesis using liquid chromatography-tandem mass spectrometry (LC-MS/MS). In vitro, ATO significantly decreased all steroids in the H295R steroidogenesis at concentrations close to human plasma Cmax values, with an IC50 value for testosterone of 0.093 ± 0.033 µM. Additionally, we determined steroid hormone levels in testis, adrenals, brain and plasma from rats after 14 days of exposure to three therapeutically relevant doses of ATO and observed pronounced decreasing steroid levels in particular in testis and adrenals but also in brain and plasma. In testis, all major steroidogenic enzymes were up-regulated, indicating autocrine and/or paracrine compensation for the decrease in steroid production by this tissue. In adrenals, StAR and CYP11A1 gene expression were decreased, whereas little effects were observed in the brain. Furthermore, we analyzed plasma LH and ACTH levels to investigate feedback via the PT and HPA axes. No effects were observed on LH levels, indicating little compensation via the PT axis. In contrast, ACTH levels increased during ATO exposure, indicating that the HPA axis to some extend compensated for the decrease in adrenal steroid production. Overall, ATO exerted pronounced effects on steroid production both in vitro and in vivo at therapeutically relevant doses. This clearly demonstrates the high potency of ATO to affect steroid homeostasis during therapeutic treatment. Further clinical and epidemiological studies should be conducted to investigate the relevance of these observations in patients treated with cholesterol-lowering pharmaceuticals.


Subject(s)
Atorvastatin/adverse effects , Endocrine Disruptors/adverse effects , Steroids/metabolism , Adrenal Cortex/cytology , Adrenal Cortex/drug effects , Adrenal Glands/drug effects , Adrenal Glands/metabolism , Adrenocorticotropic Hormone/blood , Animals , Anticholesteremic Agents/adverse effects , Brain/drug effects , Brain/metabolism , Cell Line , Cholesterol/metabolism , Enzymes/genetics , Gene Expression Regulation, Enzymologic/drug effects , Humans , Luteinizing Hormone/blood , Male , Rats, Sprague-Dawley , Testis/drug effects , Testis/metabolism
17.
J Neurochem ; 140(1): 37-52, 2017 01.
Article in English | MEDLINE | ID: mdl-27805736

ABSTRACT

Varenicline is a nicotinic acetylcholine receptor (nAChR) agonist used to treat nicotine addiction, but a live debate persists concerning its mechanism of action in reducing nicotine consumption. Although initially reported as α4ß2 selective, varenicline was subsequently shown to activate other nAChR subtypes implicated in nicotine addiction including α3ß4. However, it remains unclear whether activation of α3ß4 nAChRs by therapeutically relevant concentrations of varenicline is sufficient to affect the behavior of cells that express this subtype. We used patch-clamp electrophysiology to assess the effects of varenicline on native α3ß4* nAChRs (asterisk denotes the possible presence of other subunits) expressed in human adrenal chromaffin cells and compared its effects to those of nicotine. Varenicline and nicotine activated α3ß4* nAChRs with EC50 values of 1.8 (1.2-2.7) µM and 19.4 (11.1-33.9) µM, respectively. Stimulation of adrenal chromaffin cells with 10 ms pulses of 300 µM acetylcholine (ACh) in current-clamp mode evoked sodium channel-dependent action potentials (APs). Under these conditions, perfusion of 50 or 100 nM varenicline showed very little effect on AP firing compared to control conditions (ACh stimulation alone), but at higher concentrations (250 nM) varenicline increased the number of APs fired up to 436 ± 150%. These results demonstrate that therapeutic concentrations of varenicline are unlikely to alter AP firing in chromaffin cells. In contrast, nicotine showed no effect on AP firing at any of the concentrations tested (50, 100, 250, and 500 nM). However, perfusion of 50 nM nicotine simultaneously with 100 nM varenicline increased AP firing by 290 ± 104% indicating that exposure to varenicline and nicotine concurrently may alter cellular behavior such as excitability and neurotransmitter release.


Subject(s)
Action Potentials/drug effects , Adrenal Cortex/drug effects , Chromaffin Cells/drug effects , Nicotine/administration & dosage , Nicotinic Agonists/administration & dosage , Varenicline/administration & dosage , Action Potentials/physiology , Adrenal Cortex/cytology , Adrenal Cortex/physiology , Adult , Aged , Animals , Chromaffin Cells/physiology , Dose-Response Relationship, Drug , Drug Synergism , Female , Humans , Male , Middle Aged , Xenopus laevis
18.
Anesth Analg ; 125(2): 442-450, 2017 08.
Article in English | MEDLINE | ID: mdl-28514325

ABSTRACT

BACKGROUND: ET-26 hydrochloride (ET-26HCl) is a novel etomidate analogue designed to alleviate the adrenocortical suppression caused by etomidate while retaining the rapid sedative-hypnotic onset and stable hemodynamic features of etomidate. This study compared the anesthetic effect, hemodynamic stability, and recovery profiles of ET-26HCl, etomidate, and the sedative-hypnotic drug propofol in rats. METHODS: The metabolic half-life of ET-26HCl was determined in vitro using high performance liquid chromatography analysis of samples of rat plasma and liver homogenates taken from 3 animals. Hypnotic median effective doses (HD50) of ET-26HCl, etomidate, and propofol were determined by up-and-down methods. Anesthesia effect and mean arterial pressure were estimated using equivalent intravenous (IV) doses of propofol, etomidate, and ET-26HCl in the rats. Serum concentrations of corticosterone were analyzed by enzyme-linked immunosorbent assay. The ability of rats to recover from the sedative-hypnotic effects of the drugs was evaluated using open field and Morris water maze tests at equipotent doses of propofol, etomidate, ET-26HCl, and normal saline. RESULTS: The metabolic half-life of ET-26HCl was 81 ± 6 minutes in rat plasma and 126 ± 12 minutes in incubation liver homogenate (mean ± standard deviation), respectively. In vivo experiments showed that the potency of ET-26HCl to cause a loss of righting reflex in rats was 3 times lower than that of etomidate in the rats. IV propofol caused a greater decrease in mean arterial pressure relative to the baseline (-27.9 mm Hg) than did ET-26HCl (-10.7 mm Hg) and etomidate (-19.4 mm Hg) at equipotent doses. Serum corticosterone levels after drug administration were significantly higher in the ET-26HCl group than in the etomidate group at equivalent doses when measured 15 (P < .001), 30 (P < .001), and 60 (P = .002) minutes after stimulation with adrenocorticotropic hormone (ACTH1-24). Recovery of spatial orientation from anesthesia induced by an IV bolus injection was faster with ET-26HCl than with propofol, but recovery of spontaneous activity was slower. CONCLUSIONS: ET-26HCl has anesthetic potency and hemodynamic stability similar to etomidate, but it caused less adrenocortical hormone synthesis suppression than etomidate and faster spatial orientation recovery from anesthesia than propofol, which was similar to etomidate.


Subject(s)
Adrenal Cortex/drug effects , Behavior, Animal , Etomidate/analogs & derivatives , Etomidate/administration & dosage , Hemodynamics/drug effects , Hypnotics and Sedatives/administration & dosage , Adrenal Cortex Hormones/blood , Anesthetics/administration & dosage , Animals , Chromatography, High Pressure Liquid , Etomidate/pharmacology , Female , Male , Maze Learning , Propofol/administration & dosage , Rats , Rats, Sprague-Dawley , Reflex , Telemetry
19.
J Neurosci ; 35(34): 11897-910, 2015 Aug 26.
Article in English | MEDLINE | ID: mdl-26311772

ABSTRACT

The prelimbic region (PL) of the medial prefrontal cortex (mPFC) is implicated in the relapse of drug-seeking behavior. Optimal mPFC functioning relies on synaptic connections involving dendritic spines in pyramidal neurons, whereas prefrontal dysfunction resulting from elevated glucocorticoids, stress, aging, and mental illness are each linked to decreased apical dendritic branching and spine density in pyramidal neurons in these cortical fields. The fact that cocaine use induces activation of the stress-responsive hypothalamo-pituitary-adrenal axis raises the possibility that cocaine-related impairments in mPFC functioning may be manifested by similar changes in neuronal architecture in mPFC. Nevertheless, previous studies have generally identified increases, rather than decreases, in structural plasticity in mPFC after cocaine self-administration. Here, we use 3D imaging and analysis of dendritic spine morphometry to show that chronic cocaine self-administration leads to mild decreases of apical dendritic branching, prominent dendritic spine attrition in PL pyramidal neurons, and working memory deficits. Importantly, these impairments were largely accounted for in groups of rats that self-administered cocaine compared with yoked-cocaine- and saline-matched counterparts. Follow-up experiments failed to demonstrate any effects of either experimenter-administered cocaine or food self-administration on structural alterations in PL neurons. Finally, we verified that the cocaine self-administration group was distinguished by more protracted increases in adrenocortical activity compared with yoked-cocaine- and saline-matched controls. These studies suggest a mechanism whereby increased adrenocortical activity resulting from chronic cocaine self-administration may contribute to regressive prefrontal structural and functional plasticity. SIGNIFICANCE STATEMENT: Stress, aging, and mental illness are each linked to decreased prefrontal plasticity. Here, we show that chronic cocaine self-administration in rats leads to decrements in medial prefrontal structural and functional plasticity. Notably, these impairments were largely accounted for in rats that self-administered cocaine compared with yoked counterparts. Moreover, we verified previous reports showing that adrenocortical output is augmented by cocaine administration and is more protracted in rats that were permitted to receive the drug contingently instead of passively. These studies suggest that increased adrenocortical activity resulting from cocaine self-administration may contribute to regressive prefrontal structural and functional plasticity.


Subject(s)
Adrenal Cortex/drug effects , Adrenal Cortex/metabolism , Cocaine/administration & dosage , Neuronal Plasticity/drug effects , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Animals , Male , Neuronal Plasticity/physiology , Prefrontal Cortex/pathology , Rats , Rats, Sprague-Dawley , Self Administration
20.
FASEB J ; 29(7): 2930-42, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25818588

ABSTRACT

Hypothalamo-pituitary-adrenocortical dysfunction contributes to morbidity and mortality in a high proportion of patients with sepsis. Here, we provide new insights into the underlying adrenal pathology. Using a murine model of endotoxemia (LPS injection), we demonstrate that adrenal insufficiency is triggered early in the disease. LPS induced a local inflammatory response in the adrenal gland within 4 hours of administration, coupled with increased expression of mRNAs for annexin A1 (AnxA1) and the formyl peptide receptors [(Fprs) 1, 2, and 3], a loss of lipid droplets in cortical cells (index of availability of cholesterol, the substrate for steroidogenesis), and a failure to mount a steroidogenic response to ACTH. Deletion of AnxA1 or Fpr2/3 in mice prevented lipid droplet loss, but not leukocyte infiltration. LPS increased adrenal myeloid differentiation primary response gene 88 and TLR2 mRNA expression, but not lymphocyte antigen 96 or TLR4. By contrast, neutrophil depletion prevented leukocyte infiltration and increased AnxA1, Fpr1, and Fpr3 mRNAs but had no impact on lipid droplet loss. Our novel data demonstrate that AnxA1 and Fpr2 have a critical role in the manifestation of adrenal insufficiency in this model, through regulation of cholesterol ester storage, suggesting that pharmacologic interventions targeting the AnxA1/FPR/ALX pathway may provide a new approach for the maintenance of adrenal steroidogenesis in sepsis.


Subject(s)
Adrenal Cortex/drug effects , Adrenal Cortex/metabolism , Annexin A1/deficiency , Lipopolysaccharides/toxicity , Receptors, Formyl Peptide/deficiency , Adrenal Cortex/pathology , Adrenal Insufficiency/chemically induced , Adrenal Insufficiency/etiology , Adrenal Insufficiency/metabolism , Animals , Annexin A1/genetics , Annexin A1/metabolism , Cholesterol Esters , Corticosterone/biosynthesis , Cytokines/blood , Gene Expression/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neutrophils/drug effects , Neutrophils/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Formyl Peptide/genetics , Receptors, Formyl Peptide/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL