Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.468
Filter
Add more filters

Publication year range
1.
J Exp Zool A Ecol Integr Physiol ; 341(3): 242-255, 2024 04.
Article in English | MEDLINE | ID: mdl-38155514

ABSTRACT

The present study aimed to unravel the possible adverse effects of methomyl on the developing adrenal gland of rat fetuses and pups. Additionally, this study explored the potential improving effects of propolis against these possible hazards induced by methomyl exposure. To achieve that, pregnant rats were divided into four groups: control group, received 1 mL distilled water, propolis group, received 1 mL propolis at a dose of 300 mg/kg, methomyl group, received 1 mL methomyl at a dose of 2 mg/kg, and combined group, received 1 mL methomyl followed by 1 mL propolis, an hour later at the same previous doses. The results revealed that methomyl exposure, during pregnancy and lactation, induced many histological and ultrastructural changes, caused DNA damage and downregulated the expression of steroidogenic acute regulatory (StAR) and CYP11B2 genes in the adrenal glands of both rat fetuses and pups. Interestingly, propolis supplementation demonstrated a remarkable ability to mitigate these deleterious effects and restored the histology and ultrastructure architecture of the adrenal glands of both fetuses and pups, as well as decreased DNA damage and upregulated the expression of StAR and CYP11B2 genes in the adrenal gland of rat fetuses and pups. In conclusion, our study highlights the potential hazardous impact of methomyl exposure during pregnancy and lactation on the development of the adrenal gland in rat fetuses and pups, moreover, the study presents a new approach to alleviate these effects through propolis administration which could be used as a dietary supplement to mitigate the adverse effects of methomyl exposure.


Subject(s)
Methomyl , Propolis , Pregnancy , Female , Rats , Animals , Methomyl/metabolism , Methomyl/pharmacology , Propolis/pharmacology , Propolis/metabolism , Cytochrome P-450 CYP11B2/metabolism , Cytochrome P-450 CYP11B2/pharmacology , Adrenal Glands , Fetus , Dietary Supplements
2.
Genes (Basel) ; 14(11)2023 Nov 15.
Article in English | MEDLINE | ID: mdl-38003025

ABSTRACT

Knowledge of circadian rhythm clock gene expression outside the suprachiasmatic nucleus is increasing. The purpose of this study was to determine whether expression of circadian clock genes differed within or among the bovine stress axis tissues (e.g., amygdala, hypothalamus, pituitary, adrenal cortex, and adrenal medulla). Tissues were obtained at an abattoir from eight mature nonpregnant Brahman cows that had been maintained in the same pasture and nutritional conditions. Sample tissues were stored in RNase-free sterile cryovials at -80 °C until the total RNA was extracted, quantified, assessed, and sequenced (NovaSeq 6000 system; paired-end 150 bp cycles). The trimmed reads were then mapped to a Bos taurus (B. taurus) reference genome (Umd3.1). Further analysis used the edgeR package. Raw gene count tables were read into RStudio, and low-expression genes were filtered out using the criteria of three minimum reads per gene in at least five samples. Normalization factors were then calculated using the trimmed mean of M values method to produce normalized gene counts within each sample tissue. The normalized gene counts important for a circadian rhythm were analyzed within and between each tissue of the stress axis using the GLM and CORR procedures of the Statistical Analysis System (SAS). The relative expression profiles of circadian clock genes differed (p < 0.01) within each tissue, with neuronal PAS domain protein 2 (NPAS2) having greater expression in the amygdala (p < 0.01) and period circadian regulator (PER1) having greater expression in all other tissues (p < 0.01). The expression among tissues also differed (p < 0.01) for individual circadian clock genes, with circadian locomotor output cycles protein kaput (CLOCK) expression being greater within the adrenal tissues and nuclear receptor subfamily 1 group D member 1 (NR1D1) expression being greater within the other tissues (p < 0.01). Overall, the results indicate that within each tissue, the various circadian clock genes were differentially expressed, in addition to being differentially expressed among the stress tissues of mature Brahman cows. Future use of these findings may assist in improving livestock husbandry and welfare by understanding interactions of the environment, stress responsiveness, and peripheral circadian rhythms.


Subject(s)
Circadian Clocks , Female , Cattle/genetics , Animals , Circadian Clocks/genetics , Period Circadian Proteins , Circadian Rhythm/genetics , Hypothalamus , Adrenal Glands
3.
Article in English | MEDLINE | ID: mdl-36901116

ABSTRACT

Due to the many health-promoting properties of bee pollen and whey protein, both products are widely used as dietary supplements. According to these reports on their health-promoting properties, the aim of our study is to assess whether these products can influence the structure and function of the adrenal glands in rats. Thirty male Wistar rats were divided into six equal groups. Among them, there were three groups which included non-running rats and three groups which included running rats. Both of these running (n = 3) and non-running (n = 3) groups included non-supplemented (control groups), bee-pollen-supplemented groups, and whey-protein-supplemented groups. After 8 weeks, the rats were decapitated, their adrenal glands were collected, and paraffin slides were prepared. Then, staining according to the standard H&E and Masson's trichrome protocols was performed. Fecal and urine samples were collected prior to the end of the study to measure corticosterone levels. In the group of non-running rats, the consumption of bee pollen was noted to be significantly higher when compared to the group of running rats (p < 0.05). The thickness of the particular adrenal cortex layers was similar among all of the groups (p > 0.05). The statistically significant changes in the microscopic structure of the adrenal glands, especially regarding cell nuclei diameter and structure, as well as the architecture of sinusoids, were observed between the groups. Moreover, urine corticosterone concentrations were found to vary between all of the analyzed groups (p < 0.05). These results indicate that both bee pollen and whey protein have limited stress-reducing potential.


Subject(s)
Corticosterone , Dietary Supplements , Rats , Male , Bees , Animals , Rats, Wistar , Whey Proteins , Adrenal Glands
4.
J Affect Disord ; 329: 55-63, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36842648

ABSTRACT

BACKGROUND: Major depressive disorder (MDD) is a highly heterogeneous disease, which brings great difficulties to clinical diagnosis and therapy. Its mechanism is still unknown. Prior neuroimaging studies mainly focused on mean differences between patients and healthy controls (HC), largely ignoring individual differences between patients. METHODS: This study included 112 MDD patients and 93 HC subjects. Resting-state functional MRI data were obtained to examine the patterns of individual variability of brain functional connectivity (IVFC). The genetic risk of pathways including dopamine, 5-hydroxytryptamine (5-HT), norepinephrine (NE), hypothalamic-pituitary-adrenal (HPA) axis, and synaptic plasticity was assessed by multilocus genetic profile scores (MGPS), respectively. RESULTS: The IVFC pattern of the MDD group was similar but higher than that in HCs. The inter-network functional connectivity in the default mode network contributed to altered IVFC in MDD. 5-HT, NE, and HPA pathway genes affected IVFC in MDD patients. The age of onset, duration, severity, and treatment response, were correlated with IVFC. IVFC in the left ventromedial prefrontal cortex had a mediating effect between MGPS of the 5-HT pathway and baseline depression severity. LIMITATIONS: Environmental factors and differences in locations of functional areas across individuals were not taken into account. CONCLUSIONS: This study found MDD patients had significantly different inter-individual functional connectivity variations than healthy people, and genetic risk might affect clinical manifestations through brain function heterogeneity.


Subject(s)
Biological Variation, Individual , Brain , Depressive Disorder, Major , Genetic Predisposition to Disease , Multifactorial Inheritance , Neural Pathways , Depressive Disorder, Major/genetics , Depressive Disorder, Major/metabolism , Brain/metabolism , Serotonin/metabolism , Norepinephrine/metabolism , Humans , Male , Female , Adult , Adrenal Glands/metabolism , Pituitary Gland/metabolism , Hypothalamus/metabolism , Prefrontal Cortex/metabolism
5.
Drug Chem Toxicol ; 46(3): 482-490, 2023 May.
Article in English | MEDLINE | ID: mdl-35361025

ABSTRACT

N-acetyl cysteine (NAC) is a nutritional supplement and greatly applied as an antioxidant in vivo and in vitro. Therefore, this study aimed to assess the metabolic and antioxidant protective effect of NAC against selenium (Se) toxicity and gamma irradiation in rats by measuring biochemical and molecular parameters. This study was conducted on sixty rats divided into six equal different groups; control, NAC, Rad, Se, Rad + NAC, and Se + NAC groups. Oxidative/nitrosative makers (LPO, NO, and NOS), antioxidants status markers (GSH, GPx, and SOD), liver metabolic markers (LDH, SDH, and ATP), and plasma metabolic markers (Glucose, total cholesterol, and total proteins) were measured using commercial colorimetric kits while plasma corticosterone concentration was measured using commercial ELISA kit. Also, Levels of NR3C1 and Glut-2 genes expression using reverse transcription-quantitative polymerase chain reaction were done. Our results revealed that Se toxicity and gamma irradiation induced significant increases in oxidative/nitrosative stress markers and a significant decrease in antioxidant status markers in the liver and adrenal tissues. Moreover, metabolic disorders were recorded as manifested by elevation of plasma ALT, Albumin, glucose and cholesterol, and decrease in protein levels associated with a significant increase in corticosterone concentration. This was also accompanied by a significant decrease in SDH activity and ATP production in the hepatic tissue. Molecular analysis showed a marked increase in NR3C1 mRNA and decrease in Glut-2 mRNA in liver tissue. However, NAC supplementation attenuated the changes induced by these toxins. Finally, we could conclude that, oral supplementation of NAC can modulate the metabolic disturbances and has protective effects in rats exposed to Se toxicity and gamma irradiation.


Subject(s)
Acetylcysteine , Antioxidants , Gamma Rays , Liver , Selenium , Animals , Rats , Acetylcysteine/metabolism , Acetylcysteine/pharmacology , Adenosine Triphosphate/metabolism , Antioxidants/pharmacology , Antioxidants/metabolism , Cholesterol/metabolism , Cholesterol/pharmacology , Corticosterone/metabolism , Corticosterone/pharmacology , Liver/drug effects , Liver/metabolism , Liver/radiation effects , Oxidative Stress , Selenium/toxicity , Gamma Rays/adverse effects , Adrenal Glands/drug effects , Adrenal Glands/metabolism , Adrenal Glands/radiation effects
6.
J Laparoendosc Adv Surg Tech A ; 33(2): 129-136, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36318793

ABSTRACT

Introduction: Nearly half of the adult population in the United States has been diagnosed with hypertension. Adrenal hormonal hypersecretion is a leading cause of secondary hypertension. Adrenal vein sampling (AVS) may assist in differentiating between unilateral and bilateral adrenal hormonal hypersecretion to identify patients who are candidates for adrenalectomy. We reviewed the use of AVS at our institution along with associated outcomes after adrenalectomy. Materials and Methods: A retrospective chart review was conducted of patients with a diagnosis of primary hyperaldosteronism (PA) or adrenocorticotropic hormone-independent Cushing syndrome (AICS) and who underwent adrenalectomy between January 1, 2010, and December 1, 2021. Patient data of baseline characteristics, preoperative workup, including AVS, and postoperative outcomes were collected and analyzed. Results: Seventy-one patients were identified in the study period (48 PA and 23 AICS). Computed tomography scan identified unilateral adrenal nodules in 52 patients (29 left; and 23 right), bilateral nodules in 13 patients, and no nodules in 6 patients. AVS was performed in 45 patients with PA (93%) and 5 patients with AICS (21%). After surgery, the number of PA patients with hypokalemia or requiring potassium supplementation significantly decreased after adrenalectomy (before surgery: 33 [68.7%]; and after surgery: 5 [10.4%], P < .01). The number of medications required for hypertension in AICS patients also significantly decreased. No major adverse events were noted. Conclusions: Our long-term experience demonstrates the ongoing use of AVS during workup of patients with primary hyperaldosteronism and for select patients with adrenocorticotropic hormone-independent Cushing syndrome. However, a low level of discordance between imaging and AVS findings in PA patients suggests that there may be a subset of patients in whom preoperative AVS is not necessary.


Subject(s)
Adrenal Glands , Adrenocorticotropic Hormone , Cushing Syndrome , Hyperaldosteronism , Adult , Humans , Adrenal Glands/blood supply , Adrenal Glands/surgery , Adrenalectomy/methods , Adrenocorticotropic Hormone/blood , Cushing Syndrome/blood , Cushing Syndrome/diagnosis , Cushing Syndrome/etiology , Cushing Syndrome/surgery , Hyperaldosteronism/blood , Hyperaldosteronism/diagnosis , Hyperaldosteronism/etiology , Hyperaldosteronism/surgery , Hypertension , Retrospective Studies
7.
Clin Transl Med ; 12(11): e1091, 2022 11.
Article in English | MEDLINE | ID: mdl-36314066

ABSTRACT

BACKGROUND: The melanocortin receptor accessory proteins (MRAP1 and MRAP2) are well-known endocrine regulators for the trafficking and signalling of all five melanocortin receptors (MC1R-MC5R). The observation of MRAP2 on regulating several non-melanocortin G protein-coupled receptors (GPCRs) has been sporadically reported, whereas other endogenous GPCR partners of the MRAP protein family are largely unknown. METHODS: Here, we performed single-cell transcriptome analysis and drew a fine GPCR blueprint and MRAPs-associated network of two major endocrine organs, the hypothalamus and adrenal gland at single-cell resolution. We also integrated multiple bulk RNA-seq profiles and single-cell datasets of human and mouse tissues, and narrowed down a list of 48 GPCRs with strong endogenous co-expression correlation with MRAPs. RESULTS: 36 and 46 metabolic-related GPCRs were consequently identified as novel interacting partners of MRAP1 or MRAP2, respectively. MRAPs exhibited protein-protein interactions and varying pharmacological properties on the surface translocation, constitutive activities and ligand-stimulated downstream signalling of these GPCRs. Knockdown of MRAP2 expression by hypothalamic administration of adeno-associated virus (AAV) packed shRNA stimulated body weight gain in mouse model. Co-injection of corticotropinreleasing factor (CRF), the agonist of corticotropin releasing hormone receptor 1 (CRHR1), suppressed feeding behaviour in a MRAP2-dependent manner. CONCLUSIONS: Collectively, our study has comprehensively elucidated the complex GPCR networks in two major endocrine organs and redefined the MRAP protein family as broad-spectrum GPCR modulators. MRAP proteins not only serve as a vital endocrine pivot on the regulation of global GPCR activities in vivo that could explain the composite physiological phenotypes of the MRAP2 null murine model but also provide us with new insights of the phenotyping investigation of GPCR-MRAP functional complexes.


Subject(s)
Carrier Proteins , Receptors, Melanocortin , Animals , Humans , Mice , Receptors, Melanocortin/genetics , Receptors, Melanocortin/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Melanocortins/metabolism , Adrenal Glands/metabolism , Hypothalamus/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
8.
Biochem Biophys Res Commun ; 630: 101-111, 2022 11 19.
Article in English | MEDLINE | ID: mdl-36152347

ABSTRACT

Postmenopausal women have an increased risk of obesity, but the underlying cause is not clear. We unexpectedly found that excess dietary zinc induced severe obesity and a Cushing's-like syndrome without increased food intake in ovariectomized (Ovx) but not in sham-operated mice. Zinc accumulated in the adrenal glands and inhibited adrenal 17,20-lyase activity and steroid synthesis. As adrenal steroids are the only source of estrogen in Ovx mice, estrogen deficiency induced adrenal hyperplasia, glucocorticoid overproduction, and consequent development of a Cushing's-like syndrome. Adrenal steroid supplementation prevented the effects of zinc. Plasma zinc was positively correlated with cortisol level and negatively correlated with the levels of adrenal steroids and estrogen in obese postmenopausal women. The finding of a link between dietary zinc, estrogen deficiency, and postmenopausal obesity, implies that postmenopausal obesity might be prevented by supplementation with a adrenal steroid and avoiding excess dietary zinc.


Subject(s)
Cushing Syndrome , Adrenal Glands , Animals , Cushing Syndrome/etiology , Estrogens/pharmacology , Female , Glucocorticoids/pharmacology , Hydrocortisone , Mice , Obesity/complications , Postmenopause , Steroid 17-alpha-Hydroxylase , Steroids/pharmacology , Zinc/pharmacology
9.
Nutrients ; 13(11)2021 Nov 04.
Article in English | MEDLINE | ID: mdl-34836194

ABSTRACT

(1) Background: Prolonged feeding with a high-fat diet (HFD) acts as a stressor by activating the functions of the hypothalamic-pituitary-adrenal gland (HPA) stress axis, accompanied of hypertension by inducing the renin-angiotensin-aldosterone system. Angiotensinases enzymes are regulatory aminopeptidases of angiotensin metabolism, which together with the dipeptidyl peptidase IV (DPP-IV), pyroglutamyl- and tyrosyl-aminopeptidase (pGluAP, TyrAP), participate in cognitive, stress, metabolic and cardiovascular functions. These functions appear to be modulated by the type of fat used in the diet. (2) Methods: To analyze a possible coordinated response of aminopeptidases, their activities were simultaneously determined in the hypothalamus, adenohypophysis and adrenal gland of adult male rats fed diets enriched with monounsaturated (standard diet (S diet) supplemented with 20% virgin olive oil; VOO diet) or saturated fatty acids (diet S supplemented with 20% butter and 0.1% cholesterol; Bch diet). Aminopeptidase activities were measured by fluorimetry using 2-Naphthylamine as substrates. (3) Results: the hypothalamus did not show differences in any of the experimental diets. In the pituitary, the Bch diet stimulated the renin-angiotensin system (RAS) by increasing certain angiotensinase activities (alanyl-, arginyl- and cystinyl-aminopeptidase) with respect to the S and VOO diets. DPP-IV activity was increased with the Bch diet, and TyrAP activity decrease with the VOO diet, having both a crucial role on stress and eating behavior. In the adrenal gland, both HFDs showed an increase in angiotensinase aspartyl-aminopeptidase. The interrelation of angiotensinases activities in the tissues were depending on the type of diet. In addition, correlations were shown between angiotensinases and aminopeptidases that regulate stress and eating behavior. (4) Conclusions: Taken together, these results support that the source of fat in the diet affects several peptidases activities in the HPA axis, which could be related to alterations in RAS, stress and feeding behavior.


Subject(s)
Aminopeptidases/drug effects , Dietary Fats/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Pituitary-Adrenal System/drug effects , Renin-Angiotensin System/drug effects , Adrenal Glands/metabolism , Animals , Diet, High-Fat/adverse effects , Endopeptidases/drug effects , Fatty Acids/pharmacology , Feeding Behavior/drug effects , Hypothalamus/metabolism , Male , Olive Oil/pharmacology , Pituitary Gland, Anterior/metabolism , Rats , Rats, Wistar , Stress, Physiological/drug effects
10.
Nature ; 598(7882): 641-645, 2021 10.
Article in English | MEDLINE | ID: mdl-34646018

ABSTRACT

Somatosensory autonomic reflexes allow electroacupuncture stimulation (ES) to modulate body physiology at distant sites1-6 (for example, suppressing severe systemic inflammation6-9). Since the 1970s, an emerging organizational rule about these reflexes has been the presence of body-region specificity1-6. For example, ES at the hindlimb ST36 acupoint but not the abdominal ST25 acupoint can drive the vagal-adrenal anti-inflammatory axis in mice10,11. The neuroanatomical basis of this somatotopic organization is, however, unknown. Here we show that PROKR2Cre-marked sensory neurons, which innervate the deep hindlimb fascia (for example, the periosteum) but not abdominal fascia (for example, the peritoneum), are crucial for driving the vagal-adrenal axis. Low-intensity ES at the ST36 site in mice with ablated PROKR2Cre-marked sensory neurons failed to activate hindbrain vagal efferent neurons or to drive catecholamine release from adrenal glands. As a result, ES no longer suppressed systemic inflammation induced by bacterial endotoxins. By contrast, spinal sympathetic reflexes evoked by high-intensity ES at both ST25 and ST36 sites were unaffected. We also show that optogenetic stimulation of PROKR2Cre-marked nerve terminals through the ST36 site is sufficient to drive the vagal-adrenal axis but not sympathetic reflexes. Furthermore, the distribution patterns of PROKR2Cre nerve fibres can retrospectively predict body regions at which low-intensity ES will or will not effectively produce anti-inflammatory effects. Our studies provide a neuroanatomical basis for the selectivity and specificity of acupoints in driving specific autonomic pathways.


Subject(s)
Adrenal Glands/physiology , Autonomic Nervous System , Electroacupuncture , Vagus Nerve/physiology , Acupuncture Points , Animals , Hindlimb/innervation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Reflex
11.
Medicina (Kaunas) ; 57(10)2021 Oct 11.
Article in English | MEDLINE | ID: mdl-34684123

ABSTRACT

Background: A significant number of patients with COVID-19 experience prolonged symptoms, known as Long COVID. The most frequent symptoms are fatigue and cognitive dysfunction. We describe a patient suffering from Long COVID in whom adrenal involvement was highlighted. Methods: The patient described Long COVID symptoms that persist 3 months after the negativization of the molecular swab test. The main symptoms were weakness, brain fog, dizziness, and muscular and joint pain. All routine lab panels for inflammation, anemia, and thyroid and liver function were conducted. Moreover, salivary cortisol and DHEA-S determinations were used to compute the adrenal stress index (ASI). Results: All tests were negative, except the ASI that showed very low levels of free cortisol. The patient started hydrocortisone acetate supplementation. Conclusion: Long COVID symptoms could be explained by an adrenal involvement, due to a COVID-19 action on adrenal glands and by a iatrogenic side effect of high glucocorticoid therapy during the COVID-19 infection. Salivary cortisol determination is effective for establishing a correct recovery plan.


Subject(s)
COVID-19 , Adrenal Glands , COVID-19/complications , Dehydroepiandrosterone Sulfate , Humans , Hydrocortisone/therapeutic use , SARS-CoV-2 , Post-Acute COVID-19 Syndrome
12.
Biomed Pharmacother ; 143: 112141, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34509822

ABSTRACT

AIM: Modified Suanzaoren Decoction (MSZRD) is obtained by improving Suanzaoren Decoction (SZRT), a traditional Chinese herbal prescription that has been used to treat insomnia for more than thousands of years. Our previous study showed that MSZRD can improve the gastrointestinal discomfort related insomnia by regulating Orexin-A. This study is the first study to evaluate the effects and possible mechanisms of MSZRD in mice with insomnia caused by p-chlorophenylalanine (PCPA) combined with multifactor random stimulation. METHODS: After 14 days of multifactor stimulation to ICR mice, a PCPA suspension (30 mg/mL) was injected intraperitoneally for two consecutive days to establish an insomnia model. Three different doses of MSZRD (3.6, 7.2, and 14.4 g/kg/day) were given to ICR mice for 24 days. The food intake and back temperature were measured, and behavioral tests and pentobarbital sodium-induced sleep tests were conducted. The levels of Orexin-A, corticotropin-releasing hormone (CRH), adrenocorticotropic hormone (ACTH), and adrenocortical hormones (CORT) in the serum and 5-hydroxytryptamine (5-HT), dopamine (DA), and norepinephrine (NE) in hypothalamus were measured using enzyme-linked immunosorbent assay (ELISA) kits. The levels of γ-aminobutyric acid (GABA) and glutamic acid (Glu) were measured by high-performance liquid chromatography (HPLC). The expression of 5HT1A receptor (5-HTRIA) and orexin receptor 2 antibody (OX2R) was measured by Western blot (WB) and immunohistochemical staining (ICH). Hematoxylin and eosin (H&E) staining and Nissl staining were used to assess the histological changes in hypothalamus tissue. RESULTS: Of note, MSZRD can shorten the sleep latency of insomnia mice (P < 0.05, 0.01), prolonged the sleep duration of mice (P < 0.05, 0.01), and improve the circadian rhythm disorder relative to placebo-treated animals. Furthermore, MSZRD effectively increased the content of 5-HT and 5-HTR1A protein in the hypothalamus of insomnia mice (P < 0.05, 0.01), while downregulated the content of DA and NE (P < 0.05, 0.01). Importantly, serum GABA concentration was increased by treatment with MSZRD (P < 0.05), as reflected by a decreased Glu/GABA ratio (P < 0.05). Moreover, MSZRD decreased the levels of CORT, ACTH, and CRH related hormones in HPA axis (P < 0.05, 0.01). At the same time, MSZRD significantly downregulated the serum Orexin-A content in insomnia mice (P < 0.05), as well as hypothalamic OX2R expression (P < 0.05). In addition, MSZRD also improved the histopathological changes in hypothalamus in insomnia mice. CONCLUSION: MSZRD has sleep-improvement effect in mice model of insomnia. The mechanism may be that regulating the expression of Orexin-A affects the homeostasis of HPA axis and the release of related neurotransmitters in mice with insomnia.


Subject(s)
Adrenal Glands/drug effects , Behavior, Animal/drug effects , Drugs, Chinese Herbal/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Orexins/metabolism , Sleep Aids, Pharmaceutical/pharmacology , Sleep Initiation and Maintenance Disorders/drug therapy , Sleep/drug effects , Adrenal Glands/metabolism , Adrenal Glands/physiopathology , Animals , Disease Models, Animal , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiopathology , Male , Mice, Inbred ICR , Neurotransmitter Agents/metabolism , Orexin Receptors/metabolism , Signal Transduction , Sleep Initiation and Maintenance Disorders/metabolism , Sleep Initiation and Maintenance Disorders/physiopathology
13.
Metallomics ; 13(7)2021 07 02.
Article in English | MEDLINE | ID: mdl-34132350

ABSTRACT

Selenium (Se) is an important trace element to maintain the body's dynamic balance. Lack of Se can cause inflammation. Studies have shown that inflammation often leads to disorders of the hypothalamic-pituitary-adrenal axis, but the mechanism by which Se deficiency causes inflammation of the porcine adrenal glands is still unclear. In order to study the effect of Se deficiency on the adrenal glands of pigs, we obtained Se-deficient pig adrenal glands through a low-Se diet. The results of mass spectrometry showed that the Se content in the Se-deficient group was only one-tenth of the control group. We detected the expression of the toll-like receptor 4 (TLR4) and downstream factors by qRT-PCR and Western blotting, and found that the lack of Se affected the TLR4/NF-κB pathway. It is known that miR-155-3p, miR-30d-R_1, and miR-146b have all been verified for targeting relationship with TLR4. We confirmed by qRT-PCR that miR-30d-R_1 decreased most significantly in the Se-deficient pig model. Then we tested 25 selenoproteins and some indicators of oxidative stress. It is confirmed that Se deficiency reduces the antioxidant capacity and induces oxidative stress in pig adrenal tissue. In short, a diet lacking Se induces oxidative stress in pig adrenal tissues and leads to inflammation through the miR-30d-R_1/TLR4 pathway. This study provides a reference for the prevention of adrenal inflammation in pigs from a nutritional point of view.


Subject(s)
Adrenal Glands/pathology , Diet/veterinary , Inflammation/pathology , MicroRNAs/genetics , NF-kappa B/metabolism , Selenium/deficiency , Toll-Like Receptor 4/metabolism , Adrenal Glands/metabolism , Animals , Antioxidants , Inflammation/etiology , Inflammation/metabolism , NF-kappa B/genetics , Oxidative Stress , Swine , Toll-Like Receptor 4/genetics
14.
Bioelectrochemistry ; 140: 107830, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33965669

ABSTRACT

Previously we reported that adrenal chromaffin cells exposed to a 5 ns, 5 MV/m pulse release the catecholamines norepinephrine (NE) and epinephrine (EPI) in a Ca2+-dependent manner. Here we determined that NE and EPI release increased with pulse number (one versus five and ten pulses at 1 Hz), established that release occurs by exocytosis, and characterized the exocytotic response in real-time. Evidence of an exocytotic mechanism was the appearance of dopamine-ß-hydroxylase on the plasma membrane, and the demonstration by total internal reflection fluorescence microscopy studies that a train of five or ten pulses at 1 Hz triggered the release of the fluorescent dye acridine orange from secretory granules. Release events were Ca2+-dependent, longer-lived relative to those evoked by nicotinic receptor stimulation, and occurred with a delay of several seconds despite an immediate rise in Ca2+. In complementary studies, cells labeled with the plasma membrane fluorescent dye FM 1-43 and exposed to a train of ten pulses at 1 Hz underwent Ca2+-dependent increases in FM 1-43 fluorescence indicative of granule fusion with the plasma membrane due to exocytosis. These results demonstrate the effectiveness of ultrashort electric pulses for stimulating catecholamine release, signifying their promise as a novel electrostimulation modality for neurosecretion.


Subject(s)
Adrenal Glands/cytology , Calcium/metabolism , Catecholamines/metabolism , Chromaffin Cells/metabolism , Electricity , Exocytosis
15.
Aging (Albany NY) ; 13(9): 13087-13107, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33971622

ABSTRACT

Mimecan encodes a secretory protein that is secreted into the human serum as two mature proteins with molecular masses of 25 and 12 kDa. We found 12-kDa mimecan to be a novel satiety hormone mediated by the upregulation of the expression of interleukin (IL)-1ß and IL-6 in the hypothalamus. Mimecan was found to be expressed in human pituitary corticotroph cells and was up-regulated by glucocorticoids, while the secretion of adrenocorticotropic hormone (ACTH) in pituitary corticotroph AtT-20 cells was induced by mimecan. However, the effects of mimecan in adrenal tissue on the hypothalamic-pituitary-adrenal (HPA) axis functions remain unknown. We demonstrated that the expression of mimecan in adrenal tissues is significantly downregulated by hypoglycemia and scalded stress. It was down-regulated by ACTH, but upregulated by glucocorticoids through in vivo and in vitro studies. We further found that 12-kDa mimecan fused protein increased the corticosterone secretion of adrenal cells in vivo and in vitro. Interestingly, compared to litter-mate mice, the diurnal rhythm of corticosterone secretion was disrupted under basal conditions, and the response to restraint stress was stronger in mimecan knockout mice. These findings suggest that mimecan stimulates corticosterone secretion in the adrenal tissues under basal conditions; however, the down-regulated expression of mimecan by increased ACTH secretion after stress in adrenal tissues might play a role in maintaining the homeostasis of an organism's responses to stress.


Subject(s)
Gene Expression/physiology , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Stress, Physiological/physiology , Adrenal Glands/metabolism , Adrenocorticotropic Hormone/blood , Animals , Glucocorticoids/metabolism , Hypothalamus/metabolism , Mice , Mice, Knockout , Pituitary Gland/metabolism
16.
J Feline Med Surg ; 23(1): 33-49, 2021 01.
Article in English | MEDLINE | ID: mdl-33403910

ABSTRACT

PRACTICAL RELEVANCE: Abdominal ultrasound plays a vital role in the diagnostic work-up of many cats presenting to general and specialist practitioners. Ultrasound examination of the adrenal glands can provide important information pertaining to several conditions including hyperaldosteronism and hyperadrenocorticism. CLINICAL CHALLENGES: Despite ultrasonography being a commonly used modality, many practitioners are not comfortable performing an ultrasound examination or interpreting the resulting images. Even for the experienced ultrasonographer, differentiating between incidental findings, such as adrenal mineralisation, and clinically significant pathological changes can be challenging. AIM: This review, part of an occasional series on feline abdominal ultrasonography, discusses the ultrasonographic examination of the normal and diseased adrenal glands. Aimed at general practitioners who wish to improve their knowledge of and confidence in feline abdominal ultrasound, this review is accompanied by high-resolution images and videos available online as supplementary material. EQUIPMENT: Ultrasound facilities are readily available to most practitioners, although the use of ultrasonography as a diagnostic tool is highly dependent on operator experience. EVIDENCE BASE: Information provided in this article is drawn from the published literature and the author's own clinical experience.


Subject(s)
Adrenal Gland Diseases/veterinary , Adrenal Glands/diagnostic imaging , Cat Diseases/diagnostic imaging , Adrenal Gland Diseases/diagnostic imaging , Animals , Cats , Ultrasonography/veterinary
17.
J Clin Endocrinol Metab ; 106(3): 843-857, 2021 03 08.
Article in English | MEDLINE | ID: mdl-33212489

ABSTRACT

CONTEXT: Disorders affecting adrenal steroidogenesis promote an imbalance in the normally tightly controlled secretion of mineralocorticoids, glucocorticoids, and androgens. This may lead to differences/disorders of sex development in the fetus, as seen in virilized girls with congenital adrenal hyperplasia (CAH). Despite the important endocrine function of human fetal adrenals, neither normal nor dysregulated adrenal steroidogenesis is understood in detail. OBJECTIVE: Due to significant differences in adrenal steroidogenesis between human and model species (except higher primates), we aimed to establish a human fetal adrenal model that enables examination of both de novo and manipulated adrenal steroidogenesis. DESIGN AND SETTING: Human adrenal tissue from 54 1st trimester fetuses were cultured ex vivo as intact tissue fragments for 7 or 14 days. MAIN OUTCOME MEASURES: Model validation included examination of postculture tissue morphology, viability, apoptosis, and quantification of steroid hormones secreted to the culture media measured by liquid chromatography-tandem mass spectrometry. RESULTS: The culture approach maintained cell viability, preserved cell populations of all fetal adrenal zones, and recapitulated de novo adrenal steroidogenesis based on continued secretion of steroidogenic intermediates, glucocorticoids, and androgens. Adrenocorticotropic hormone and ketoconazole treatment of ex vivo cultured human fetal adrenal tissue resulted in the stimulation of steroidogenesis and inhibition of androgen secretion, respectively, demonstrating a treatment-specific response. CONCLUSIONS: Together, these data indicate that ex vivo culture of human fetal adrenal tissue constitutes a novel approach to investigate local effects of pharmaceutical exposures or emerging therapeutic options targeting imbalanced steroidogenesis in adrenal disorders, including CAH.


Subject(s)
Adrenal Glands/cytology , Drug Evaluation, Preclinical/methods , Fetus/cytology , Primary Cell Culture/methods , Steroids/biosynthesis , Adrenal Glands/drug effects , Adrenal Glands/embryology , Adrenal Glands/metabolism , Adrenal Hyperplasia, Congenital/drug therapy , Adrenal Hyperplasia, Congenital/metabolism , Adrenal Hyperplasia, Congenital/pathology , Adrenocorticotropic Hormone/pharmacology , Androgens/metabolism , Cell Survival , Culture Media/chemistry , Female , Glucocorticoids/pharmacology , Humans , Ketoconazole/pharmacology , Metabolic Networks and Pathways/drug effects , Models, Biological , Pregnancy , Steroids/analysis , Steroids/metabolism
18.
Sci Rep ; 10(1): 11281, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32647204

ABSTRACT

Studies have shown that acupuncture is very effective in treating chronic stress depression. However, little is known about the therapeutic mechanism of electro-acupuncture. Metabolomics, on the other hand, is a technology that determines the metabolic changes of organisms caused by various interventions as a whole and is related to the overall effect of electro-acupuncture (EA). 1HNMR, serum sample analysis, and histopathology and molecular biology analysis were used to evaluate the effects of EA. The results show that electro-acupuncture points can regulate the heat pain threshold of chronic stress model rats and change the morphology of adrenal cortex cells Structure, and regulate the contents of corticotropin-releasing hormone, Corticosterone (CORT), glucose, alanine and valine in the samples. These findings help to clarify the therapeutic mechanism of electro-acupuncture on heterologous chronic stress model rats. The effect of electro-acupuncture on improving chronic stress is likely to be achieved by regulating glucose metabolism, which can provide a reference for clinical acupuncture treatment of chronic stress depression.


Subject(s)
Blood Glucose/metabolism , Electroacupuncture , Stress, Physiological , Adrenal Cortex/metabolism , Adrenal Glands/cytology , Alanine/chemistry , Animals , Behavior, Animal , Body Weight , Corticosterone/chemistry , Magnetic Resonance Spectroscopy , Male , Pain Threshold , Rats , Rats, Sprague-Dawley , Valine/chemistry
19.
Free Radic Biol Med ; 156: 137-143, 2020 08 20.
Article in English | MEDLINE | ID: mdl-32574682

ABSTRACT

We previously demonstrated that oral supplementation with antioxidants induced hyperactivity of hypothalamus-pituitary-adrenal (HPA) axis, attested by hypercorticoidism, through an up-regulation of adrenocorticotrophic hormone (ACTH) receptors (MC2R) in adrenal. This study analyzed the role of peroxisome proliferator-activated receptor (PPAR)-γ on HPA axis hyperactivity induced by N-acetyl-cysteine (NAC). Male Swiss-Webster mice were orally treated with NAC for 1, 3, 5, 10, 15, or 18 consecutive days. The PPAR-γ agonist rosiglitazone and/or antagonist GW9662 were daily-injected i.p. for 5 consecutive days, starting concomitantly with NAC treatment. Rosiglitazone treatment inhibited NAC-induced adrenal hypertrophy and hypercorticoidism. Rosiglitazone also significantly reversed the NAC-induced increase in the MC2R expression in adrenal, but not steroidogenic acute regulatory protein (StAR). NAC treatment reduces the expression of PPARγ in the adrenals, but rosiglitazone did not restore the expression of this cytoprotective gene. In addition, GW9662 blocked the ability of rosiglitazone to decrease plasma corticosterone levels in NAC-treated mice. In conclusion, our findings showed that antioxidant supplementation induced a state of hypercorticoidism through down-regulation of PPARγ expression in the adrenals, in a mechanism probably related to a down-regulation of ACTH receptor expression.


Subject(s)
PPAR gamma , Thiazolidinediones , Acetylcysteine/pharmacology , Adrenal Glands/metabolism , Animals , Hypothalamo-Hypophyseal System/metabolism , Male , Mice , PPAR gamma/genetics , PPAR gamma/metabolism , Pituitary-Adrenal System/metabolism , Receptors, Corticotropin , Thiazolidinediones/pharmacology
20.
Domest Anim Endocrinol ; 72: 106473, 2020 07.
Article in English | MEDLINE | ID: mdl-32361423

ABSTRACT

The action of glucocorticoids on target tissues is regulated by the glucocorticoid and mineralocorticoid receptors (codified by the NR3C1 and NR3C2 gene, respectively). Moreover, the prereceptor system, represented by the hydroxysteroid 11-beta dehydrogenases (HSD11Bs), catalyzes the interconversion from active glucocorticoids into inactive compounds. This study aimed to determine whether the expression of the prereceptor system, the corticosteroid receptors, and the molecules regulating their intracellular trafficking (FKBP prolyl isomerase 4 and FKBP prolyl isomerase 5) could be regulated in the hypothalamic-pituitary-adrenal axis and in different type of adipose tissue of calves by the administration of dexamethasone in combination with estradiol or prednisolone. Research about the glucocorticoid effects on bovine target tissues may allow development of new diagnostic methods that use potential molecular biomarkers of glucocorticoid treatment. The administration of dexamethasone in combination with estradiol increased the gene expression of HSD11B1 (P < 0.01), HSD11B2 (P < 0.05), NR3C1 (P < 0.01), and NR3C2 (P < 0.01) in the adrenal glands; NR3C2 in the intramuscular adipose tissue (P < 0.01), and HSD11B1 in the subcutaneous adipose tissue (P < 0.01). Prednisolone administration increased the gene expression of HSD11B1 (P < 0.01), NR3C1 (P < 0.05), and NR3C2 (P < 0.05) in the adrenal glands and HSD11B1 (P < 0.01) in the subcutaneous adipose tissue. Interestingly, most of the examined tissues/organs showed a significant variation of FKBP5 gene expression after the administration of dexamethasone in combination with estradiol. So, these changes suggest that the FKBP5 gene expression could be a possible biomarker of the illegal dexamethasone administration in calves.


Subject(s)
Adipose Tissue/metabolism , Cattle/physiology , Hypothalamo-Hypophyseal System/metabolism , Molecular Chaperones/metabolism , Pituitary-Adrenal System/metabolism , Receptors, Steroid/metabolism , Adrenal Glands/drug effects , Adrenal Glands/metabolism , Animals , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Estradiol/administration & dosage , Estradiol/analogs & derivatives , Estradiol/pharmacology , Gene Expression Regulation/drug effects , Glucocorticoids/pharmacology , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Pituitary Gland/drug effects , Pituitary Gland/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL