Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 15.243
Filter
1.
PeerJ ; 12: e17539, 2024.
Article in English | MEDLINE | ID: mdl-38952964

ABSTRACT

The association between sleep and the immune-endocrine system is well recognized, but the nature of that relationship is not well understood. Sleep fragmentation induces a pro-inflammatory response in peripheral tissues and brain, but it also activates the hypothalamic-pituitary-adrenal (HPA) axis, releasing glucocorticoids (GCs) (cortisol in humans and corticosterone in mice). It is unclear whether this rapid release of glucocorticoids acts to potentiate or dampen the inflammatory response in the short term. The purpose of this study was to determine whether blocking or suppressing glucocorticoid activity will affect the inflammatory response from acute sleep fragmentation (ASF). Male C57BL/6J mice were injected i.p. with either 0.9% NaCl (vehicle 1), metyrapone (a glucocorticoid synthesis inhibitor, dissolved in vehicle 1), 2% ethanol in polyethylene glycol (vehicle 2), or mifepristone (a glucocorticoid receptor antagonist, dissolved in vehicle 2) 10 min before the start of ASF or no sleep fragmentation (NSF). After 24 h, samples were collected from brain (prefrontal cortex, hypothalamus, hippocampus) and periphery (liver, spleen, heart, and epididymal white adipose tissue (EWAT)). Proinflammatory gene expression (TNF-α and IL-1ß) was measured, followed by gene expression analysis. Metyrapone treatment affected pro-inflammatory cytokine gene expression during ASF in some peripheral tissues, but not in the brain. More specifically, metyrapone treatment suppressed IL-1ß expression in EWAT during ASF, which implies a pro-inflammatory effect of GCs. However, in cardiac tissue, metyrapone treatment increased TNF-α expression in ASF mice, suggesting an anti-inflammatory effect of GCs. Mifepristone treatment yielded more significant results than metyrapone, reducing TNF-α expression in liver (only NSF mice) and cardiac tissue during ASF, indicating a pro-inflammatory role. Conversely, in the spleen of ASF-mice, mifepristone increased pro-inflammatory cytokines (TNF-α and IL-1ß), demonstrating an anti-inflammatory role. Furthermore, irrespective of sleep fragmentation, mifepristone increased pro-inflammatory cytokine gene expression in heart (IL-1ß), pre-frontal cortex (IL-1ß), and hypothalamus (IL-1ß). The results provide mixed evidence for pro- and anti-inflammatory functions of corticosterone to regulate inflammatory responses to acute sleep loss.


Subject(s)
Glucocorticoids , Metyrapone , Mice, Inbred C57BL , Mifepristone , Sleep Deprivation , Animals , Male , Metyrapone/pharmacology , Sleep Deprivation/metabolism , Sleep Deprivation/drug therapy , Mice , Mifepristone/pharmacology , Glucocorticoids/pharmacology , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Inflammation/metabolism , Inflammation/drug therapy , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/genetics , Corticosterone/blood , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Brain/metabolism , Brain/drug effects , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/genetics
2.
Acta Neuropathol Commun ; 12(1): 100, 2024 06 17.
Article in English | MEDLINE | ID: mdl-38886854

ABSTRACT

A link between chronic stress and Parkinson's disease (PD) pathogenesis is emerging. Ample evidence demonstrates that the presynaptic neuronal protein alpha-synuclein (asyn) is closely tied to PD pathogenesis. However, it is not known whether stress system dysfunction is present in PD, if asyn is involved, and if, together, they contribute to neurodegeneration. To address these questions, we assess stress axis function in transgenic rats overexpressing full-length wildtype human asyn (asyn BAC rats) and perform multi-level stress and PD phenotyping following chronic corticosterone administration. Stress signaling, namely corticotropin-releasing factor, glucocorticoid and mineralocorticoid receptor gene expression, is also examined in post-mortem PD patient brains. Overexpression of human wildtype asyn leads to HPA axis dysregulation in rats, while chronic corticosterone administration significantly aggravates nigrostriatal degeneration, serine129 phosphorylated asyn (pS129) expression and neuroinflammation, leading to phenoconversion from a prodromal to an overt motor PD phenotype. Interestingly, chronic corticosterone in asyn BAC rats induces a robust, twofold increase in pS129 expression in the hypothalamus, the master regulator of the stress response, while the hippocampus, both a regulator and a target of the stress response, also demonstrates elevated pS129 asyn levels and altered markers of stress signalling. Finally, defective hippocampal stress signalling is mirrored in human PD brains and correlates with asyn expression levels. Taken together, our results link brain stress system dysregulation with asyn and provide evidence that elevated circulating glucocorticoids can contribute to asyn-induced neurodegeneration, ultimately triggering phenoconversion from prodromal to overt PD.


Subject(s)
Corticosterone , Parkinson Disease , Rats, Transgenic , Stress, Psychological , alpha-Synuclein , alpha-Synuclein/metabolism , alpha-Synuclein/genetics , Animals , Parkinson Disease/metabolism , Parkinson Disease/pathology , Humans , Rats , Stress, Psychological/metabolism , Stress, Psychological/pathology , Male , Corticosterone/blood , Brain/metabolism , Brain/pathology , Hypothalamo-Hypophyseal System/metabolism , Female , Pituitary-Adrenal System/metabolism
3.
In Vivo ; 38(4): 1677-1689, 2024.
Article in English | MEDLINE | ID: mdl-38936893

ABSTRACT

BACKGROUND/AIM: Depression is associated with metabolic disorders, including non-alcoholic fatty liver disease (NAFLD). However, the mechanisms underlying the interaction between them are still poorly known. MATERIALS AND METHODS: In this study, mice on a choline deficiency, L-amino acid-defined, high-fat diet (CDAHFD) developing steatosis were challenged with chronic restraint stress (CRS), a protocol widely used to induce depression. The development of depression and steatosis was evaluated using histopathology analysis, ELISA, q-PCR and Western Blot. RESULTS: The contribution of the activated HPA axis to hepatic steatosis progress was fully established, which was validated using a hepatocyte model. Histopathological and biochemical analysis indicated that steatosis was exacerbated by CRS challenge, and behavioral tests indicated that the mice developed depression. Among the screened endocrinal pathways, the hypothalamic-pituitary-adrenal (HPA) axis was significantly activated and the synergistic effect of CDAHFD and CRS in activating the HPA axis was observed. In the hypothalamus, expression of corticotropin-releasing hormone (CRH) was increased by 86% and the protein levels of hypothalamic CRH were upregulated by 25% to 33% by CRS treatment. Plasma CRH levels were elevated by 45-56% and plasma adrenocorticotropic hormone (ACTH) levels were elevated by 29-58% by CRS treatment. In the liver, target genes of the HPA axis were activated, accompanied by disruption of the lipid metabolism and progression of steatohepatitis. The lipid metabolism in the Hepa1-6 cell line treated with endogenous corticosterone (CORT) was in accordance with the aforementioned in vivo responses. CONCLUSION: Depression aggravated hepatic steatosis in CDAHFD-fed mice by activating the HPA axis. The risk of NAFLD development should be fully considered in depressive patients and improvement of psychotic disorders could be an etiological treatment strategy for them.


Subject(s)
Depression , Disease Models, Animal , Hypothalamo-Hypophyseal System , Non-alcoholic Fatty Liver Disease , Pituitary-Adrenal System , Animals , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Mice , Depression/metabolism , Depression/etiology , Depression/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/etiology , Male , Mice, Inbred C57BL , Corticotropin-Releasing Hormone/metabolism , Corticotropin-Releasing Hormone/genetics , Diet, High-Fat/adverse effects , Adrenocorticotropic Hormone/blood , Liver/metabolism , Liver/pathology , Fatty Liver/metabolism , Fatty Liver/etiology , Fatty Liver/pathology , Corticosterone/blood
4.
J Endocrinol ; 262(2)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38829241

ABSTRACT

Glucocorticoids modulate glucose homeostasis, acting on metabolically active tissues such as liver, skeletal muscle, and adipose tissue. Intracellular regulation of glucocorticoid action in adipose tissue impacts metabolic responses to obesity. ATP-binding cassette family C member 1 (ABCC1) is a transmembrane glucocorticoid transporter known to limit the accumulation of exogenously administered corticosterone in adipose tissue. However, the role of ABCC1 in the regulation of endogenous glucocorticoid action and its impact on fuel metabolism has not been studied. Here, we investigate the impact of Abcc1 deficiency on glucocorticoid action and high-fat-diet (HFD)-induced obesity. In lean male mice, deficiency of Abcc1 increased endogenous corticosterone levels in skeletal muscle and adipose tissue but did not impact insulin sensitivity. In contrast, Abcc1-deficient male mice on HFD displayed impaired glucose and insulin tolerance, and fasting hyperinsulinaemia, without alterations in tissue corticosterone levels. Proteomics and bulk RNA sequencing revealed that Abcc1 deficiency amplified the transcriptional response to an obesogenic diet in adipose tissue but not in skeletal muscle. Moreover, Abcc1 deficiency impairs key signalling pathways related to glucose metabolism in both skeletal muscle and adipose tissue, in particular those related to OXPHOS machinery and Glut4. Together, our results highlight a role for ABCC1 in regulating glucose homeostasis, demonstrating diet-dependent effects that are not associated with altered tissue glucocorticoid concentrations.


Subject(s)
Adipose Tissue , Corticosterone , Diet, High-Fat , Insulin Resistance , Multidrug Resistance-Associated Proteins , Muscle, Skeletal , Obesity , Animals , Male , Diet, High-Fat/adverse effects , Mice , Obesity/metabolism , Obesity/genetics , Obesity/etiology , Adipose Tissue/metabolism , Insulin Resistance/physiology , Corticosterone/blood , Corticosterone/metabolism , Muscle, Skeletal/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Multidrug Resistance-Associated Proteins/genetics , Mice, Knockout , Mice, Inbred C57BL , Glucose/metabolism
5.
Endocrinol Diabetes Metab ; 7(4): e487, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38867382

ABSTRACT

INTRODUCTION: High-fat diet (HFD) consumption and being exposed to daily psychological stress, common environmental factors in modern lifestyle, play an important role on metabolic disorders such as glucose homeostasis impairment. The aim of this study was to investigate the effects of high-fat diet (HFD) and psychological stress combination on metabolic response to chronic psychological stress in male rats. METHOD: Male Wistar rats were divided into HFD, and normal diet (ND) groups and then into stress and nonstress subgroups. The diets were applied for 5 weeks, and psychological stress was induced for 7 consecutive days. Then, blood samples were taken to measure glucose, insulin, free fatty acids (FFA), and leptin and corticosterone concentrations. Subsequently, glucose-stimulated insulin release from pancreatic isolated islets was assessed. RESULTS: HFD did not significantly change fasting plasma glucose, insulin and corticosterone levels, whereas increased plasma leptin (7.05 ± 0.33) and FFA (p < 0.01) levels and impaired glucose tolerance. Additionally, HFD and stress combination induced more profound glucose intolerance associated with increased plasma corticosterone (p < 0.01) and leptin (8.63 ± 0.38) levels. However, insulin secretion from isolated islets did not change in the presence of high-fat diet and/or stress. CONCLUSION: HFD should be considered as an intensified factor of metabolic impairments caused by chronic psychological stress.


Subject(s)
Blood Glucose , Corticosterone , Diet, High-Fat , Insulin , Leptin , Rats, Wistar , Stress, Psychological , Animals , Male , Stress, Psychological/metabolism , Diet, High-Fat/adverse effects , Rats , Corticosterone/blood , Insulin/blood , Leptin/blood , Blood Glucose/metabolism , Fatty Acids, Nonesterified/blood , Islets of Langerhans/metabolism , Glucose Intolerance/etiology , Glucose Intolerance/metabolism
6.
Cell Mol Biol (Noisy-le-grand) ; 70(6): 48-53, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38836677

ABSTRACT

The study aimed to determine the effects of probiotic consumption during pregnancy and lactation and post-weaning on acute stress-induced anxiety and gut beneficial microbiota of the female offspring mice.The female offspring mice were divided into several groups: intact, control (only stressed), PBS/dam (dams gavaged with PBS), PRO/dam (dams gavaged with probiotics), PRO/dam+off (both dams and offspring gavaged with probiotics), and PBS/dam+off (both dams and offspring gavaged with PBS)The probiotics chosen are mainly L. rhamnosus, B.breve, and B. longum (108 CFU/ml). Foot shock stress will be applied for one hour on the 43rd day after birth. Behavioral tests were conducted using the open field and elevated plus-maze. Corticosterone was measured by ELISA kit, and intestinal microflora with qPCR.The data showed that PRO/dam+off had more entries into open arms compared to the control group and decreased move distance and time spent in closed arms compared to the control group. However, there was no significant difference between the PRO/dam group and the control group. In the open field test, the control group spent less time in the inner zone compared to the intact group and in PRO/dam+off group. Corticosterone hormone was increased in the control group and was decreased in the PRO/dam+off. Bifidobacteria and Lactobacilli decreased in the control group in comparison to the intact group, and in the PRO/dam+off group increased compared with other groups. Maternal and filial supplementation with a multi-strain probiotic mixture increased levels of beneficial bacteria and reduced stress-induced anxiety in mice.


Subject(s)
Anxiety , Corticosterone , Gastrointestinal Microbiome , Probiotics , Stress, Psychological , Probiotics/administration & dosage , Probiotics/pharmacology , Animals , Gastrointestinal Microbiome/drug effects , Female , Pregnancy , Mice , Stress, Psychological/complications , Corticosterone/blood , Lactation , Behavior, Animal/drug effects
7.
PLoS One ; 19(6): e0305091, 2024.
Article in English | MEDLINE | ID: mdl-38900819

ABSTRACT

Short and long-term sound-induced stress on daily basis can affect the physiology of avian individuals because they are more susceptible to sound stress in an open environment. OBJECTIVES: An ex-situ study was carried out to determine the impact of noise on physiology and ptilochronology of non-breeding male domesticated quail birds. METHODOLOGY: During 60-days long trial, male quail birds, aged 5-weeks, weighing (c.100gm) were used. Out of 72 experimental birds, 18 birds were assigned to the Control Group (G1) while remaining 54 birds were divided equally into 3 treatment groups: Road Traffic noise (G2), Military activity noise (G3) and Human Activities noise (G4). Birds were housed in standard-sized separate cages (20 ×45 × 20 cm), every bird was kept apart in separate cage in open laboratory under maintained environmental conditions. Millet seeds and water were provided to all the experimental birds ad libitum. Noise originated from several sources of recorded high-intensity music (1125 Hz/ 90 dB), was administered for 5-6 hours per day. Observations were recorded in the morning and afternoon. The experiment was conducted during the non-breeding season from August to October in triplicate. Blood sampling was done after 60 days. RESULTS: According to the current study, noise stress significantly (p<0.05) increased the concentrations of creatinine, aspartate aminotransferase (AST), alanine aminotransferase (ALT), alkaline phosphatase (ALP), bilirubin, uric acid, cholesterol, triglycerides, total protein, and glucose while a decline in the levels of albumin was seen in treatment birds of G3. While in terms of hematology, total white blood cells count (TWBC), total red blood cells count (TRBC), mean cell volume (MCV) & packed cell volume (PCV) concentrations were raised in blood of treatment birds of G3. In terms of hormones, noise stress significantly (p<0.05) increased the serum concentrations of Corticosterone in G3 while a significant (p<0.05) decline was observed in the concentrations of luteinizing hormone (LH), thyroid stimulating hormone (TSH), and follicle stimulating hormone (FSH) in the same group. Moreover, fault bar formation in G3 was more prominent than others. CONCLUSION: Noise stress can significantly affect serology, hematology, hormonal physiology and ptilochronology in quail birds.


Subject(s)
Noise , Animals , Male , Noise/adverse effects , Stress, Physiological , Quail/physiology , Corticosterone/blood
8.
Clinics (Sao Paulo) ; 79: 100411, 2024.
Article in English | MEDLINE | ID: mdl-38901134

ABSTRACT

INTRODUCTION: Despite strong evidences supporting the protective role of exercise against stress-induced repercussions, the literature remains inconclusive regarding metabolic aspects. Therefore, this study aimed to evaluate the effect of Physical Training (PT) by swimming on the metabolic parameters of rats subjected to restraint stress. METHODS: Wistar rats (n = 40) were divided into four groups: Control (C), Trained (T), Stressed (S), and Trained/Stressed (TS). The restraint stress protocol involved confining the animals in PVC pipes for 60 minutes/day for 12 weeks. Concurrently, the swimming PT protocol was performed without additional load in entailed sessions of 60 minutes conducted five days a week for the same duration. The following parameters were analyzed: fitness progression assessed by the physical capacity test, body mass, serum level of glucose, triglyceride, cholesterol and corticosterone, as well as glycemic tolerance test, evaluated after glucose administration (2 g/kg, i.p.). RESULTS: Trained groups (T and TS) exhibited enhanced physical capacity (169 ± 21 and 162 ± 22% increase, respectively) compared to untrained groups (C: 9 ± 5 and S: 11 ± 13% increase). Corticosterone levels were significantly higher in the S group (335 ± 9 nmoL/L) compared to C (141 ± 3 nmoL/L), T (174 ± 3 nmoL/L) and TS (231 ± 7 nmoL/L), which did not differ from each other. There were no significant changes in serum glucose, cholesterol, and triglyceride levels among the groups. However, the glycemic curve after glucose loading revealed increased glycemia in the S group (area under curve 913 ± 30 AU) but the TS group exhibited values (673 ± 12 AU) similar to the groups C (644 ± 10 AU) and T (649 ± 9 AU). CONCLUSION: Swimming-based training attenuated stress-induced corticosterone release and prevented glucose intolerance in rats, reinforcing the importance of exercise as a potential strategy to mitigate the pathophysiological effects of stress.


Subject(s)
Blood Glucose , Corticosterone , Physical Conditioning, Animal , Rats, Wistar , Restraint, Physical , Stress, Psychological , Swimming , Animals , Physical Conditioning, Animal/physiology , Male , Corticosterone/blood , Blood Glucose/analysis , Swimming/physiology , Stress, Psychological/metabolism , Cholesterol/blood , Rats , Triglycerides/blood , Time Factors , Glucose Tolerance Test , Random Allocation , Metabolome/physiology
9.
Int J Mol Sci ; 25(11)2024 May 28.
Article in English | MEDLINE | ID: mdl-38892044

ABSTRACT

Anxiety is a common comorbidity of obesity, resulting from prescribing long-term caloric restriction diets (CRDs); patients with a reduced food intake lose weight but present anxious behaviors, poor treatment adherence, and weight regain in the subsequent 5 years. Intermittent fasting (IF) restricts feeding time to 8 h during the activity phase, reducing patients' weight even with no caloric restriction; it is unknown whether an IF regime with ad libitum feeding avoids stress and anxiety development. We compared the corticosterone blood concentration between male Wistar rats fed ad libitum or calorie-restricted with all-day or IF food access after 4 weeks, along with their anxiety parameters when performing the elevated plus maze (EPM). As the amygdalar thyrotropin-releasing hormone (TRH) is believed to have anxiolytic properties, we evaluated its expression changes in association with anxiety levels. The groups formed were the following: a control which was offered food ad libitum (C-adlib) or 30% of C-adlib's energy requirements (C-CRD) all day, and IF groups provided food ad libitum (IF-adlib) or 30% of C-adlib's requirements (IF-CRD) with access from 9:00 to 17:00 h. On day 28, the rats performed the EPM and, after 30 min, were decapitated to analyze their amygdalar TRH mRNA expression by in situ hybridization and corticosterone serum levels. Interestingly, circadian feeding synchronization reduced the body weight, food intake, and animal anxiety levels in both IF groups, with ad libitum (IF-adlib) or restricted (IF-CRD) food access. The anxiety levels of the experimental groups resulted to be negatively associated with TRH expression, which supported its anxiolytic role. Therefore, the low anxiety levels induced by synchronizing feeding with the activity phase would help patients who are dieting to improve their diet therapy adherence.


Subject(s)
Amygdala , Anxiety , Caloric Restriction , Circadian Rhythm , Corticosterone , Rats, Wistar , Thyrotropin-Releasing Hormone , Animals , Anxiety/metabolism , Rats , Male , Amygdala/metabolism , Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/genetics , Caloric Restriction/methods , Corticosterone/blood , Down-Regulation , Feeding Behavior , Fasting , Eating , Body Weight
10.
Int J Mol Sci ; 25(10)2024 May 07.
Article in English | MEDLINE | ID: mdl-38791102

ABSTRACT

Congenital Adrenal Hyperplasia (CAH) is an autosomal recessive disorder impairing cortisol synthesis due to reduced enzymatic activity. This leads to persistent adrenocortical overstimulation and the accumulation of precursors before the blocked enzymatic step. The predominant form of CAH arises from mutations in CYP21A2, causing 21-hydroxylase deficiency (21-OHD). Despite emerging treatment options for CAH, it is not always possible to physiologically replace cortisol levels and counteract hyperandrogenism. Moreover, there is a notable absence of an effective in vivo model for pre-clinical testing. In this work, we developed an animal model for CAH with the clinically relevant point mutation p.R484Q in the previously humanized CYP21A2 mouse strain. Mutant mice showed hyperplastic adrenals and exhibited reduced levels of corticosterone and 11-deoxycorticosterone and an increase in progesterone. Female mutants presented with higher aldosterone concentrations, but blood pressure remained similar between wildtype and mutant mice in both sexes. Male mutant mice have normal fertility with a typical testicular appearance, whereas female mutants are infertile, exhibit an abnormal ovarian structure, and remain in a consistent diestrus phase. Conclusively, we show that the animal model has the potential to contribute to testing new treatment options and to prevent comorbidities that result from hormone-related derangements and treatment-related side effects in CAH patients.


Subject(s)
Adrenal Hyperplasia, Congenital , Disease Models, Animal , Steroid 21-Hydroxylase , Animals , Adrenal Hyperplasia, Congenital/genetics , Adrenal Hyperplasia, Congenital/pathology , Adrenal Hyperplasia, Congenital/metabolism , Steroid 21-Hydroxylase/genetics , Steroid 21-Hydroxylase/metabolism , Mice , Female , Male , Humans , Corticosterone/metabolism , Corticosterone/blood , Aldosterone/metabolism , Adrenal Glands/metabolism , Adrenal Glands/pathology , Mutation , Progesterone/metabolism
11.
Exp Gerontol ; 193: 112466, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38821324

ABSTRACT

Chronic stress (CS) is critically involved in the Alzheimer's disease (AD) pathogenesis resulting in cognitive disturbance. Also, amyloid precursor protein (APP) related gens, pro-inflammatory cytokines, and stress increases AD-related pathogenesis through increasing APP, all are important players in the development of AD. Herein, we explore the possible neuroprotective and anti-amnestic effect of quercetin (QUER) on cognitive deficits induced by scopolamine (SCOP) in stressed rats. Stress induction was performed by exposed of rats to 2-h chronic restraint stress for 10 days. Then rats were supplemented with QUER (25 mg/kg/day oral gavage, for 1 month). Ratswere submitted to intraperitoneal (i.p.) injection of SCOP (1 mg/kg) during the final 9 days of QUER supplementation to induce dementia like condition. Following the interventions, behavioral tests [elevated plus maze (EPM) and novel object recognition memory (NORM)] was examined to analysis the cognitive functions. Meanwhile, prefrontal cortex (PFC) and hippocampus of brain were used for gene expression and biochemical studies. Also, the plasma corticosterone (CORT) level was measured. We established that administration of QUER ameliorated the SCOP-related memory impairment. Also, QUER decreased stress related anxiety like behaviors in the EPM. QUER also altered the interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) levels in both PFC and hippocampus of SCOP treated rats in stress and non-stress conditions. We found that QUER increased APP and amyloid precursor-like protein 2 (APLP2) mRNA expression in both non-stress and stressed rats. Also, our findings imply that QUER suppress the effect of SCOP on cognitive functions. Moreover, decreased APP mRNA expression in the hippocampus were observed following pretreatment of rats with QUER in both stress and non-stress groups. Given that decreased amyloid beta (Aß) expression in the hippocampus of stressed rats, it can be proposed that elevations in APP mRNA expression by QUER activates non-amyloidogenic pathways leading to reduction in Aß levels. However, our findings indicate that QUER can be a therapeutic candidate, which exerts an anti-amnesic property against SCOP-induced memory decline. On the other hand, prior QUER administration in stress condition could be a promising approach against AD prevention.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Cytokines , Disease Models, Animal , Hippocampus , Quercetin , Rats, Wistar , Animals , Quercetin/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Male , Rats , Cytokines/metabolism , Hippocampus/metabolism , Hippocampus/drug effects , Amyloid beta-Protein Precursor/genetics , Amyloid beta-Protein Precursor/metabolism , Stress, Psychological/complications , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/metabolism , Scopolamine , Neuroprotective Agents/pharmacology , Corticosterone/blood , Prefrontal Cortex/metabolism , Prefrontal Cortex/drug effects , Cognition/drug effects
12.
Hum Cell ; 37(4): 1008-1023, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38753278

ABSTRACT

Reproductive aging is associated with altered stress response and many other menopausal symptoms. Little is known about the adrenal expression of the anti-aging protein Klotho or how it is modulated by estrogen in ovariectomized stressed rats. Fifty-six Wistar female rats were assigned into seven equal groups. Sham-operated (Sham), sham stressed (Sham/STS), ovariectomized (OVR), ovariectomized stressed (OVR/STS), ovariectomized stressed rosiglitazone-treated (OVR/STS/R), ovariectomized stressed estrogen-treated (OVR/STS/E), and ovariectomized stressed estrogen/GW9662 co-treated (OVR/STS/E/GW) groups. All stressed rats were subjected daily to a one-hour restraint stress test for 19 days. At the end of the experiment, blood was collected for serum corticosterone (CORT) analysis. Adrenal tissues were obtained and prepared for polymerase chain reaction (PCR) assay, hematoxylin and eosin (H&E), immunohistochemistry-based identification of Klotho and PPAR-γ, and Oil Red O (ORO) staining. The rise in serum CORT was negligible in the OVR/STS group, in contrast to the Sham/STS group. The limited CORT response in the former group was restored by estrogen and rosiglitazone and blocked by estrogen/GW9226 co-administration. ORO-staining revealed a more evident reduction in the adrenal fat in the OVR/STS group, which was reversed by estrogen and counteracted by GW. Also, there was a comparable expression pattern of Klotho and PPAR-γ in the adrenals. The adrenal Klotho decreased in the OVR/STS group, but was reversed by estrogen treatment. GW9226/estrogen co-treatment interfered with the regulatory effect of estrogen on Klotho. The study suggested modulation of the adrenal Kotho expression by estrogen, in the ovariectomized rats subjected to a restraint stress test. This estrogen-provided adrenal protection might be mediated by PPAR-γ activation.


Subject(s)
Adrenal Cortex , Estrogens , Glucuronidase , Klotho Proteins , Ovariectomy , PPAR gamma , Rats, Wistar , Animals , Female , Glucuronidase/metabolism , Glucuronidase/genetics , Adrenal Cortex/metabolism , Adrenal Cortex/drug effects , PPAR gamma/metabolism , PPAR gamma/genetics , Rats , Restraint, Physical , Gene Expression/drug effects , Gene Expression/genetics , Corticosterone/blood , Stress, Psychological/metabolism , Stress, Physiological , Rosiglitazone/pharmacology , Disease Models, Animal , Aging/metabolism , Models, Animal
13.
J Med Chem ; 67(11): 9495-9515, 2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38780432

ABSTRACT

We describe the discovery of a thioester-containing glucocorticoid receptor modulator (GRM) payload and the corresponding antibody-drug conjugate (ADC). Payload 6 was designed for rapid hepatic inactivation to minimize systemic exposure of nonconjugated GRM. Mouse PK indicated that 6 is cleared 10-fold more rapidly than a first-generation GRM payload, resulting in 10-fold lower exposure and 3-fold decrease in Cmax. The anti-mTNF conjugate ADC5 fully inhibited inflammation in mouse contact hypersensitivity with minimal effects on corticosterone, a biomarker for systemic GRM effects, at doses up to and including 100 mg/kg. Concomitant inhibition of P1NP suggests potential delivery to cells involved in the remodeling of bone, which may be a consequence of TNF-targeting or bystander payload effects. Furthermore, ADC5 fully suppressed inflammation in collagen-induced arthritis mouse model after one 10 mg/kg dose for 21 days. The properties of the anti-hTNF conjugate were suitable for liquid formulation and may enable subcutaneous dosing.


Subject(s)
Arthritis, Experimental , Corticosterone , Immunoconjugates , Tumor Necrosis Factor-alpha , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/metabolism , Mice , Immunoconjugates/pharmacology , Immunoconjugates/chemistry , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Corticosterone/blood , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/antagonists & inhibitors , Inflammation/drug therapy , Inflammation/metabolism , Glucocorticoids/pharmacology , Humans , Male , Disease Models, Animal
14.
Gen Comp Endocrinol ; 355: 114545, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38701975

ABSTRACT

In birds, patterns of development of the adrenocortical response to stressors vary among individuals, types of stressors, and species. Since there are benefits and costs of exposure to elevated glucocorticoids, this variation is presumably a product of selection such that animals modulate glucocorticoid secretion in contexts where doing so increases their fitness. In this study, we evaluated hypothalamo-pituitary-adrenal (HPA) activity in first-hatched free-living seabird nestlings that engage in intense sibling competition and facultative siblicide (black-legged kittiwakes, Rissa tridactyla). We sampled 5 day old chicks (of the ∼45 day nestling period), a critical early age when food availability drives establishment of important parent-offspring and intra-brood dynamics. We experimentally supplemented parents with food ("supplemented") and measured chick baseline corticosterone secretion and capacity to rapidly increase corticosterone in response to an acute challenge (handling and 15 min of restraint in a bag). We also used topical administration of corticosterone to evaluate the ability of chicks to downregulate physiologically relevant corticosterone levels on a short time scale (minutes). We found that 5 day old chicks are not hypo-responsive but release corticosterone in proportion to the magnitude of the challenge, showing differences in baseline between parental feeding treatments (supplemented vs non-supplemented), moderate increases in response to handling, and a larger response to restraint (comparable to adults) that also differed between chicks from supplemented and control nests. Topical application of exogenous corticosterone increased circulating levels nearly to restraint-induced levels and induced downregulation of HPA responsiveness to the acute challenge of handling. Parental supplemental feeding did not affect absorbance/clearance or negative feedback. Thus, while endogenous secretion of corticosterone in young chicks is sensitive to environmental context, other aspects of the HPA function, such as rapid negative feedback and/or the ability to clear acute elevations in corticosterone, are not. We conclude that 5 day old kittiwake chicks are capable of robust adrenocortical responses to novel challenges, and are sensitive to parental food availability, which may be transduced behaviorally, nutritionally, or via maternal effects. Questions remain about the function of such rapid, large acute stress-induced increases in corticosterone in very young chicks.


Subject(s)
Charadriiformes , Corticosterone , Animals , Corticosterone/metabolism , Corticosterone/blood , Charadriiformes/physiology , Charadriiformes/metabolism , Hypothalamo-Hypophyseal System/metabolism , Stress, Physiological , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/drug effects , Female , Male
15.
Front Endocrinol (Lausanne) ; 15: 1363468, 2024.
Article in English | MEDLINE | ID: mdl-38808110

ABSTRACT

Social support is vital for mental and physical health and is linked to lower rates of disease and early mortality. Conversely, anti-social behavior can increase mortality risks, both for the initiator and target of the behavior. Chronic stress, which also can increase mortality, may serve as an important link between social behavior and healthy lifespan. There is a growing body of literature in both humans, and model organisms, that chronic social stress can result in more rapid telomere shortening, a measure of biological aging. Here we examine the role of anti-social behavior and social support on physiological markers of stress and aging in the social Japanese quail, Coturnix Japonica. Birds were maintained in groups for their entire lifespan, and longitudinal measures of antisocial behavior (aggressive agonistic behavior), social support (affiliative behavior), baseline corticosterone, change in telomere length, and lifespan were measured. We found quail in affiliative relationships both committed less and were the targets of less aggression compared to birds who were not in these relationships. In addition, birds displaying affiliative behavior had longer telomeres, and longer lifespans. Our work suggests a novel pathway by which social support may buffer against damage at the cellular level resulting in telomere protection and subsequent longer lifespans.


Subject(s)
Aging , Coturnix , Longevity , Social Behavior , Telomere , Animals , Coturnix/physiology , Female , Aging/physiology , Behavior, Animal , Feathers , Telomere Shortening , Aggression/physiology , Corticosterone/blood
16.
Exp Gerontol ; 192: 112453, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38723916

ABSTRACT

Social isolation (SI) after stroke reduces recovery. The aim of this study was to evaluate the effects of SI on corticosterone release and recovery after stroke in aged rats. A total of 64 male Wistar rats (aged 24 months) were used in the present study. All rats were housed in pairs for two weeks. After two weeks, rats were randomly assigned to one of four groups: (1) rats underwent sham surgery and kept socially isolated (control/social isolated (CO/SI) group); (2) rats underwent sham surgery and kept pair housed (control/pair housed (CO/PH) group); (3) rats underwent middle cerebral artery occlusion (MCAO) surgery and kept socially isolated (stroke/isolated (ST/SI) group); (4) rats underwent MCAO surgery and kept pair housed (stroke/pair housed (ST/PH)) group. Behaviors were assessed using the adhesive removal test, rotarod test and social interaction test at 1st, 7th, 14th and 21st days after stroke. Serum biochemical analysis was also performed on the behavioral testing days. Results showed THAT serum corticosterone and MDA levels in CO/PH group were significantly lower than CO/SI group. Serum BDNF levels in CO/PH group was significantly higher than CO/SI group. Serum corticosterone and MDA levels in ST/PH group were lower than ST/SI group. In ST/PH group, serum Total antioxidant capacity (TAC) and BDNF levels were significantly higher than ST/SI group. Biochemical analysis of certain regions of the brain (hippocampus, striatum and cerebral cortex) was performed on 21st day after stroke. In the hippocampus of CO/PH group, BDNF and TAC levels were significantly higher than CO/SI group. The hippocampal MDA level of CO/PH group were significantly lower than CO/SI group. BDNF and TAC levels in the hippocampus, striatum and cerebral cortex of ST/PH group were significantly higher and MDA level was significantly lower as compared with ST/SI group. Both ischemic groups showed sensorimotor recovery over a 21-day period, but recovery of ST/PH group was significantly greater than ST/SI group. Total social interaction time in ST/PH group was significantly longer than ST/SI group. Based on the results of this study, social interaction after stroke enhances histologic and sensorimotor recovery through reduction of HPA activity and corticosterone release, leading to increased TAC and BDNF levels.


Subject(s)
Behavior, Animal , Brain-Derived Neurotrophic Factor , Corticosterone , Infarction, Middle Cerebral Artery , Rats, Wistar , Social Isolation , Animals , Social Isolation/psychology , Corticosterone/blood , Male , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/blood , Infarction, Middle Cerebral Artery/metabolism , Rats , Recovery of Function , Stroke/metabolism , Stroke/psychology , Malondialdehyde/metabolism , Disease Models, Animal , Aging/physiology , Aging/metabolism , Oxidative Stress
17.
Sci Rep ; 14(1): 11519, 2024 05 21.
Article in English | MEDLINE | ID: mdl-38769131

ABSTRACT

Ulcerative colitis (UC) is a refractory inflammatory bowel disease, which is known to cause psychiatric disorders such as anxiety and depression at a high rate in addition to peripheral inflammatory symptoms. However, the pathogenesis of these psychiatric disorders remains mostly unknown. While prior research revealed that the Enterococcus faecalis 2001 (EF-2001) suppressed UC-like symptoms and accompanying depressive-like behaviors, observed in a UC model using dextran sulfate sodium (DSS), whether it has an anxiolytic effect remains unclear. Therefore, we examined whether EF-2001 attenuates DSS-induced anxiety-like behaviors. Treatment with 2% DSS for seven days induced UC-like symptoms and anxiety-like behavior through the hole-board test, increased serum lipopolysaccharide (LPS) and corticosterone concentration, and p-glucocorticoid receptor (GR) in the prefrontal cortex (PFC), and decreased N-methyl-D-aspartate receptor subunit (NR) 2A and NR2B expression levels in the PFC. Interestingly, these changes were reversed by EF-2001 administration. Further, EF-2001 administration enhanced CAMKII/CREB/BDNF-Drebrin pathways in the PFC of DSS-treated mice, and labeling of p-GR, p-CAMKII, and p-CREB showed colocalization with neurons. EF-2001 attenuated anxiety-like behavior by reducing serum LPS and corticosterone levels linked to the improvement of UC symptoms and by facilitating the CAMKII/CREB/BDNF-Drebrin pathways in the PFC. Our findings suggest a close relationship between UC and anxiety.


Subject(s)
Anti-Anxiety Agents , Dextran Sulfate , Disease Models, Animal , Enterococcus faecalis , Animals , Mice , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Dextran Sulfate/toxicity , Male , Anxiety/drug therapy , Lipopolysaccharides , Corticosterone/blood , Prefrontal Cortex/metabolism , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/microbiology , Mice, Inbred C57BL
18.
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
19.
Steroids ; 207: 109438, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38723842

ABSTRACT

To elucidate the effect of cyclooxygenase-2 (COX-2) inhibition on corticosterone release, mice were divided into a group receiving NS398, a selective COX-2 inhibitor at a dose of 3 mg/kg for seven days, and a group receiving NS398 for fourteen days. After this time, the mice were sacrificed, and blood serum was collected. An ELISA protocol was used to analyze serum corticosterone levels. Short-term COX-2 inhibition increased corticosterone levels, while long-term inhibition lowered them. The exact schedule of experiments was repeated after the lipopolysaccharide (LPS) Escherichia coli challenge in mice to check the influence of stress stimuli on the tested parameters. In this case, we observed increases in corticosterone levels, significant in a seven-day pattern. These results indicate that corticosterone levels are regulated through a COX-2-dependent mechanism in mice.


Subject(s)
Corticosterone , Cyclooxygenase 2 Inhibitors , Cyclooxygenase 2 , Lipopolysaccharides , Nitrobenzenes , Sulfonamides , Animals , Mice , Corticosterone/blood , Cyclooxygenase 2 Inhibitors/pharmacology , Nitrobenzenes/pharmacology , Sulfonamides/pharmacology , Lipopolysaccharides/pharmacology , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/blood , Male , Time Factors
20.
Nutrients ; 16(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38794729

ABSTRACT

Polymethoxyflavonoids, such as nobiletin (abundant in Citrus depressa), have been reported to have antioxidant, anti-inflammatory, anticancer, and anti-dementia effects, and are also a circadian clock modulator through retinoic acid receptor-related orphan receptor (ROR) α/γ. However, the optimal timing of nobiletin intake has not yet been determined. Here, we explored the time-dependent treatment effects of nobiletin and a possible novel mechanistic idea for nobiletin-induced circadian clock regulation in mice. In vivo imaging showed that the PER2::LUC rhythm in the peripheral organs was altered in accordance with the timing of nobiletin administration (100 mg/kg). Administration at ZT4 (middle of the light period) caused an advance in the peripheral clock, whereas administration at ZT16 (middle of the dark period) caused an increase in amplitude. In addition, the intraperitoneal injection of nobiletin significantly and potently stimulated corticosterone and adrenaline secretion and caused an increase in Per1 expression in the peripheral tissues. Nobiletin inhibited phosphodiesterase (PDE) 4A1A, 4B1, and 10A2. Nobiletin or rolipram (PDE4 inhibitor) injection, but not SR1078 (RORα/γ agonist), caused acute Per1 expression in the peripheral tissues. Thus, the present study demonstrated a novel function of nobiletin and the regulation of the peripheral circadian clock.


Subject(s)
Circadian Clocks , Corticosterone , Flavones , Animals , Flavones/pharmacology , Circadian Clocks/drug effects , Mice , Male , Corticosterone/blood , Period Circadian Proteins/metabolism , Period Circadian Proteins/genetics , Epinephrine , Mice, Inbred C57BL , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Circadian Rhythm/drug effects , Circadian Rhythm/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...