Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.029
Filter
1.
Neurosci Lett ; 839: 137931, 2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39128819

ABSTRACT

Food deprivation is used in many experimental models and is becoming increasingly prevalent in human diets. The impact of food deprivation on specific brain regions, including the nucleus of the tractus solitarius (NTS), a region that is involved in hunger and satiety sensing, remains to be determined. The NTS is a heterogeneous nucleus that includes corticotropin releasing factor receptor 1 (CRF1) neurons. CRF1 is implicated in both stress and appetite regulation, but the effects of food deprivation on CRF1 NTS neurons are unclear. We used immunofluorescence to examine the effects of 24-hour food deprivation on NTS activity in male and female Sprague-Dawley (SD) rats and CRF1-cre rats using cFos, an immediate early gene and neuronal marker of activation. NTS activity was increased in food deprived male but not female SD rats. In food deprived CRF1-cre rats, males had an increased proportion of active CRF1 + neurons with no change in females. In CRF1-cre rats, increased global NTS activity was observed in food deprived and refed males. Activation of CRF1 + neurons was also increased after deprivation but was reduced by refeeding. In females, food deprivation decreased global NTS activity that was then increased by refeeding, while CRF1 activity was unchanged. Collectively, these data suggest the NTS is differentially activated after food deprivation in a sex-specific manner, whereby males are more sensitive than females. These results provide insight into the role of brainstem stress circuitry in changes associated with conditions including intermittent fasting and eating disorders like anorexia.


Subject(s)
Food Deprivation , Neurons , Rats, Sprague-Dawley , Sex Characteristics , Solitary Nucleus , Animals , Solitary Nucleus/metabolism , Male , Female , Food Deprivation/physiology , Neurons/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Corticotropin-Releasing Hormone/genetics , Rats , Stress, Physiological/physiology
2.
J Therm Biol ; 124: 103959, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39180919

ABSTRACT

Heat stress is the most critical factor affecting animal feeding in summer. This experiment was conducted to investigate the effects of heat stress on the feeding preference of yellow-feathered broilers and its possible mechanism. As a result, the preference of yellow-feathered broilers for Tenebrio molitor was significantly decreased, and the fear response and serum corticosterone of broilers were significantly increased when the ambient temperatures are 35 °C (P < 0.05). In the central nervous system, consistent with the change in feeding preference, decreased dopamine in the nucleus accumbens (NAc) and increased mRNA levels of MAO-B in the ventral tegmental area (VTA) and NAc were found in yellow-feathered broilers (P < 0.05). In addition, we found significantly increased mRNA levels of corticotropin-releasing hormone receptor 1, corticotropin-releasing hormone receptor 2 and glucocorticoid receptor in the VTA and NAc of female broilers (P < 0.05). However, no similar change was found in male broilers. On the other hand, the serum levels of insulin and glucagon-like peptide-1 were increased only in male broilers (P < 0.05). Accordingly, the mRNA levels of insulin receptor and glucagon-like peptide-1 receptor in the VTA and the phosphorylation of mTOR and PI3K were increased only in male broilers (P < 0.05). In summary, the preference of yellow-feathered broilers for Tenebrio molitor feed decreased under heat stress conditions, and hedonic feeding behavior was significantly inhibited. However, the mechanism by which heat stress affects hedonic feeding behavior may contain gender differences. The insulin signaling pathway may participate in the regulation of heat stress on the male broiler reward system, while stress hormone-related receptors in the midbrain may play an important role in the effect of heat stress on the reward system of female broilers.


Subject(s)
Chickens , Corticosterone , Heat-Shock Response , Tenebrio , Animals , Chickens/physiology , Chickens/genetics , Chickens/metabolism , Male , Female , Tenebrio/genetics , Tenebrio/metabolism , Corticosterone/blood , Dopamine/metabolism , Nucleus Accumbens/metabolism , Nucleus Accumbens/physiology , Ventral Tegmental Area/metabolism , Ventral Tegmental Area/physiology , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/genetics , Feeding Behavior , Insulin/blood , Insulin/metabolism , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Food Preferences , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism
3.
J Affect Disord ; 363: 249-257, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39029702

ABSTRACT

BACKGROUND: Major depressive disorder (MDD) is progressively recognized as a stress-related disorder characterized by aberrant brain network dynamics, encompassing both structural and functional domains. Yet, the intricate interplay between these dynamic networks and their molecular underpinnings remains predominantly unexplored. METHODS: Both structural and functional networks were constructed using multimodal neuroimaging data from 183 MDD patients and 300 age- and gender-matched healthy controls (HC). structural-functional connectivity (SC-FC) coupling was evaluated at both the connectome- and nodal-levels. Methylation data of five HPA axis key genes, including NR3C1, FKBP5, CRHBP, CRHR1, and CRHR2, were analyzed using Illumina Infinium Methylation EPIC BeadChip. RESULTS: We observed a significant reduction in SC-FC coupling at the connectome-level in patients with MDD compared to HC. At the nodal level, we found an imbalance in SC-FC coupling, with reduced coupling in cortical regions and increased coupling in subcortical regions. Furthermore, we identified 23 differentially methylated CpG sites on the HPA axis, following adjustment for multiple comparisons and control of age, gender, and medication status. Notably, three CpG sites on NR3C1 (cg01294526, cg19457823, and cg23430507), one CpG site on FKBP5 (cg25563198), one CpG site on CRHR1 (cg26656751), and one CpG site on CRHR2 (cg18351440) exhibited significant associations with SC-FC coupling in MDD patients. CONCLUSIONS: These findings provide valuable insights into the connection between micro-scale epigenetic changes in the HPA axis and SC-FC coupling at macro-scale connectomes. They unveil the mechanisms underlying increased susceptibility to MDD resulting from chronic stress and may suggest potential pharmacological targets within the HPA-axis for MDD treatment.


Subject(s)
Brain , Connectome , DNA Methylation , Depressive Disorder, Major , Epigenesis, Genetic , Humans , Depressive Disorder, Major/physiopathology , Depressive Disorder, Major/genetics , Depressive Disorder, Major/diagnostic imaging , Female , Male , Adult , Brain/physiopathology , Brain/diagnostic imaging , Middle Aged , Tacrolimus Binding Proteins/genetics , Hypothalamo-Hypophyseal System/physiopathology , Hypothalamo-Hypophyseal System/metabolism , Receptors, Corticotropin-Releasing Hormone/genetics , Magnetic Resonance Imaging , Pituitary-Adrenal System/physiopathology , Receptors, Glucocorticoid/genetics , Case-Control Studies
4.
J Exp Biol ; 227(15)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39022893

ABSTRACT

Social status directly affects the health of humans and other animals. Low status individuals receive more antagonistic encounters, have fewer supportive relationships and have worse health outcomes. However, the physiological and cellular processes that mediate the relationship between the social environment and health are incompletely known. Epigenetic regulation of the hypothalamic-pituitary-adrenal (HPA) axis, the neuroendocrine pathway that activates in response to stressors, may be one process that is sensitive to the social environment. Here, we experimentally manipulated plumage, a key social signal in female tree swallows (Tachycineta bicolor) and quantified methylation of four genes in the HPA axis before and after treatment. We found that dulling the white breast plumage affected methylation in one gene, CRHR1; however, the effect depended on the original brightness of the bird. Methylation in this gene was correlated with baseline corticosterone levels, suggesting that DNA methylation of CRHR1 helps regulate glucocorticoid production in this species. Methylation in two other genes, FKBP5 and GR, changed over the course of the experiment, independent of treatment. These results show that methylation of these genes is labile into adulthood and suggest that epigenetic regulation of the HPA axis could help birds respond to current environmental conditions.


Subject(s)
DNA Methylation , Feathers , Hypothalamo-Hypophyseal System , Receptors, Corticotropin-Releasing Hormone , Swallows , Animals , Female , Feathers/physiology , Swallows/genetics , Swallows/physiology , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/physiology , Tacrolimus Binding Proteins/genetics , Tacrolimus Binding Proteins/metabolism , Corticosterone/blood , Corticosterone/metabolism , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/physiology , Epigenesis, Genetic , Stress, Physiological/genetics , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Avian Proteins/genetics , Avian Proteins/metabolism
5.
Sci Adv ; 10(19): eadk7636, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38728397

ABSTRACT

Corticotropin releasing factor (CRF) network in the oval nucleus of bed nuclei of the stria terminalis (ovBNST) is generally indicated in stress, but its role in female-biased susceptibility to anxiety is unknown. Here, we established a female-biased stress paradigm. We found that the CRF release in ovBNST during stress showed female-biased pattern, and ovBNST CRF neurons were more prone to be hyperexcited in female mice during stress in both in vitro and in vivo studies. Moreover, optogenetic modulation to exchange the activation pattern of ovBNST CRF neurons during stress between female and male mice could reverse their susceptibility to anxiety. Last, CRF receptor type 1 (CRFR1) mediated the CRF-induced excitation of ovBNST CRF neurons and showed female-biased expression. Specific knockdown of the CRFR1 level in ovBNST CRF neurons in female or overexpression that in male could reverse their susceptibility to anxiety. Therefore, we identify that CRFR1-mediated hyperexcitation of ovBNST CRF neurons in female mice encode the female-biased susceptibility to anxiety.


Subject(s)
Anxiety , Corticotropin-Releasing Hormone , Neurons , Receptors, Corticotropin-Releasing Hormone , Animals , Female , Male , Mice , Anxiety/metabolism , Avoidance Learning/physiology , Behavior, Animal , Corticotropin-Releasing Hormone/metabolism , Neurons/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Corticotropin-Releasing Hormone/genetics , Septal Nuclei/metabolism , Stress, Psychological/metabolism
6.
Zhen Ci Yan Jiu ; 49(5): 472-479, 2024 May 25.
Article in English, Chinese | MEDLINE | ID: mdl-38764118

ABSTRACT

OBJECTIVES: To investigate the effect of Peitu Yimu(strengthening spleen and soothing liver) acupuncture on intestinal mucosal barrier function and corticotropin-releasing factor (CRF)/CRF receptor 1 (CRFR1) pathway in rats with diarrhea-predominant irritable bowel syndrome (IBS-D), so as to explore its underlying mechanism in alleviating IBS-D. METHODS: Forty female SD rats were randomly divided into blank, model, electroacupuncture (EA), and agonist groups, with 10 rats in each group. Except for the blank group, rats in the other groups were given folium sennae infusion by gavage combined with chronic unpredictable mild stress to establish IBS-D model. Rats in the EA group received acupuncture at "Tianshu"(ST25) and EA at "Zusanli"(ST36) and "Taichong"(LR3) (2 Hz/15 Hz) on one side for 20 min, with the side chosen alternately every other day, for 14 days after modeling. Rats in the agonist group received acupuncture 30 min after intravenous injection of CRFR1 agonist urocortin, with the same manipulation method and time as the EA group. Before and after intervention, visceral pain threshold and stool Bristol scores were measured. Elevated plus maze test and open field test were used to detect anxiety and depression like behavior of rats. ELISA was used to detect the contents of CRF and CRFR1 in rats serum. Immunohistochemistry was used to detect the positive expressions of CRF, CRFR1, zonula occludens protein 1(ZO-1), occlusal protein(Occludin), and closure protein 1 (Claudin-1) in colon tissue. RESULTS: Compared with the blank group, the visceral pain threshold, open arm time percentage (OT%), total distance of movement in the open field test, and positive expression of ZO-1, Occludin, and Claudin-1 in colon were decreased (P<0.01, P<0.05), while Bristol stool scores, serum CRF and CRFR1 contents, and positive expressions of CRF and CRFR1 in colon were increased (P<0.01) in the model group. After intervention and compared with the model group, the visceral pain threshold, OT%, total distance of movement in the open field test, and positive expressions of ZO-1, Occludin, and Claudin-1 in colon were increased (P<0.05, P<0.01), while Bristol stool scores, serum CRF and CRFR1 contents, and positive expressions of CRF and CRFR1 in colon were decreased (P<0.01) in the EA group;the Bristol stool scores, serum CRF content, and CRF positive expression in colon were significantly decreased in the agonist group (P<0.01). CONCLUSIONS: Peitu Yimu acupuncture can significantly improve visceral hypersensitivity and anxiety-depression state in IBS-D rats. Its mechanism may be related to the inhibition of CRF/CRFR1 pathway and restoration of intestinal tight junction protein expressions.


Subject(s)
Acupuncture Therapy , Diarrhea , Intestinal Mucosa , Irritable Bowel Syndrome , Receptors, Corticotropin-Releasing Hormone , Animals , Female , Humans , Rats , Acupuncture Points , Claudin-1/metabolism , Claudin-1/genetics , Corticotropin-Releasing Hormone/metabolism , Corticotropin-Releasing Hormone/genetics , Diarrhea/therapy , Diarrhea/metabolism , Diarrhea/genetics , Disease Models, Animal , Intestinal Mucosa/metabolism , Irritable Bowel Syndrome/therapy , Irritable Bowel Syndrome/metabolism , Irritable Bowel Syndrome/genetics , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Corticotropin-Releasing Hormone/genetics , Zonula Occludens-1 Protein/metabolism , Zonula Occludens-1 Protein/genetics
7.
Int J Mol Sci ; 25(8)2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38673790

ABSTRACT

Cognitive behavioral therapy is based on the view that maladaptive thinking is the causal mechanism of mental disorders. While this view is supported by extensive evidence, very limited work has addressed the factors that contribute to the development of maladaptive thinking. The present study aimed to uncover interactions between childhood maltreatment and multiple genetic differences in irrational beliefs. Childhood maltreatment and irrational beliefs were assessed using multiple self-report instruments in a sample of healthy volunteers (N = 452). Eighteen single-nucleotide polymorphisms were genotyped in six candidate genes related to neurotransmitter function (COMT; SLC6A4; OXTR), neurotrophic factors (BDNF), and the hypothalamic-pituitary-adrenal axis (NR3C1; CRHR1). Gene-environment interactions (G×E) were first explored in models that employed one measure of childhood maltreatment and one measure of irrational beliefs. These effects were then followed up in models in which either the childhood maltreatment measure, the irrational belief measure, or both were substituted by parallel measures. Consistent results across models indicated that childhood maltreatment was positively associated with irrational beliefs, and these relations were significantly influenced by COMT rs165774 and OXTR rs53576. These results remain preliminary until independent replication, but they represent the best available evidence to date on G×E in a fundamental mechanism of psychopathology.


Subject(s)
Gene-Environment Interaction , Polymorphism, Single Nucleotide , Receptors, Glucocorticoid , Receptors, Oxytocin , Humans , Female , Male , Adult , Receptors, Oxytocin/genetics , Receptors, Corticotropin-Releasing Hormone/genetics , Child Abuse/psychology , Middle Aged , Adverse Childhood Experiences/psychology , Serotonin Plasma Membrane Transport Proteins/genetics , Dopamine Plasma Membrane Transport Proteins/genetics , Young Adult , Child
8.
J Youth Adolesc ; 53(2): 273-283, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37891393

ABSTRACT

Corticotropin-releasing hormone receptor 1 (CRHR1), a hormone receptor essential to the activation of HPA axis and the subsequent release of cortisol, plays critical roles in emotional and behavioral responses relevant to attachment. However, the specific roles of CRHR1 polymorphisms in attachment remain unclear. To further clarify these genetic effects, this research conducted a three-wave study to investigate whether the CRHR1 polymorphisms (i.e., rs110402 and rs242924) are associated with the stability and variability of attachment by using a sample of freshmen (N = 604; Mage = 18.57 years, SD = 1.90; 68.8% girls). The results showed that rs110402 and rs242924 were associated with the stability of closeness-dependence. The G alleles of the both polymorphisms were found not to be related to lower attachment stability. However, these polymorphisms were not associated with the variability of attachment. Overall, these findings provide evidence for the contribution of CRHR1 to attachment stability.


Subject(s)
Hypothalamo-Hypophyseal System , Polymorphism, Single Nucleotide , Adolescent , Female , Humans , Male , Emotions , Genotype , Pituitary-Adrenal System , Receptors, Corticotropin-Releasing Hormone/genetics
9.
Reproduction ; 167(1)2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37947291

ABSTRACT

In brief: Corticotropin-releasing hormone binding protein (CRHBP) is fundamental to the stress response and plays an important role in parturition during pregnancy. This study shows that abnormal CRHBP expression could be an early warning sign of recurrent pregnancy loss and that CRHBP knockdown could suppress HTR8/SVneo cell invasion by the PKC signaling pathway via interacting with CRH receptor 2. Abstract: Trophoblast invasion is critical for placentation and pregnancy success. Trophoblast dysfunction results in many pregnancy complications, including recurrent pregnancy loss (RPL). Corticotropin-releasing hormone binding protein (CRHBP) is fundamental to the stress response and plays an important role in parturition during pregnancy via binding with CRH. To further characterize its function in early pregnancy, we explored the expression of CRHBP in villi during early pregnancy. Compared with normal pregnant women, we demonstrated that the expression of CRHBP decreased in the trophoblasts and villi in RPL patients and that knockdown of CRHBP expression could suppress HTR8/SVneo cell invasion significantly. Our further exploration indicated that the capacity of CRHBP for regulating trophoblast invasion was associated with the PKC signaling pathway via interacting with CRH receptor 2. These findings might provide a new fundamental mechanism for successful pregnancy and a new diagnostic and therapeutic target for RPL.


Subject(s)
Abortion, Habitual , Receptors, Corticotropin-Releasing Hormone , Pregnancy , Humans , Female , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Down-Regulation , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Cell Line , Trophoblasts/metabolism , Abortion, Habitual/metabolism , Cell Movement
10.
Peptides ; 170: 171112, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37918484

ABSTRACT

Growth differentiation factor-15 (GDF15) is a stress-activated cytokine that regulates cell growth and inflammatory and stress responses. We previously reported the role and regulation of GDF15 in pituitary corticotrophs. Dexamethasone increases Gdf15 gene expression levels and production. GDF15 suppresses adrenocorticotropic hormone synthesis in pituitary corticotrophs and subsequently mediates the negative feedback effect of glucocorticoids. Here, we analyzed corticotropin-releasing factor (Crf) promoter activity in hypothalamic 4B cells transfected with promoter-driven luciferase reporter constructs. The effects of time and GDF15 concentration on Crf mRNA levels were analyzed using quantitative real-time polymerase chain reaction. Glial cell-derived neurotrophic factor family receptor α-like (GFRAL) protein is expressed in 4B cells. GDF15 increased Crf promoter activity and Crf mRNA levels in 4B cells. The protein kinase A and C pathways also contributed to the GDF15-induced increase in Crf gene expression. GDF15 stimulates GFRAL, subsequently increasing the phosphorylation of AKT, an extracellular signal-related kinase, and the cAMP response element-binding protein. Therefore, GDF15-dependent pathways may be involved in regulating Crf expression under stressful conditions in hypothalamic cells.


Subject(s)
Corticotropin-Releasing Hormone , Growth Differentiation Factor 15 , Hypothalamus , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Growth Differentiation Factors/genetics , Growth Differentiation Factors/metabolism , Growth Differentiation Factors/pharmacology , Hypothalamus/drug effects , Hypothalamus/metabolism , Promoter Regions, Genetic , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , RNA, Messenger/metabolism , Animals , Rats , Growth Differentiation Factor 15/metabolism , Growth Differentiation Factor 15/pharmacology , Humans
11.
Front Endocrinol (Lausanne) ; 14: 1266081, 2023.
Article in English | MEDLINE | ID: mdl-37900150

ABSTRACT

The hypothalamic type 2 corticotropin releasing hormone receptor (CRH-R2) plays critical roles in homeostatic regulation, particularly in fine tuning stress recovery. During acute stress, the CRH-R2 ligands CRH and urocortins promote adaptive responses and feeding inhibition. However, in rodent models of chronic stress, over-exposure of hypothalamic CRH-R2 to its cognate agonists is associated with urocortin 2 (Ucn2) resistance; attenuated cAMP-response element binding protein (CREB) phosphorylation and increased food intake. The molecular mechanisms involved in these altered CRH-R2 signalling responses are not well described. In the present study, we used the adult mouse hypothalamus-derived cell line mHypoA-2/30 to investigate CRH-R2 signalling characteristics focusing on gene expression of molecules involved in feeding and circadian regulation given the role of clock genes in metabolic control. We identified functional CRH-R2 receptors expressed in mHypoA-2/30 cells that differentially regulate CREB and AMP-activated protein kinase (AMPK) phosphorylation and downstream expression of the appetite-regulatory genes proopiomelanocortin (Pomc) and neuropeptide Y (Npy) in accordance with an anorexigenic effect. We studied for the first time the effects of Ucn2 on clock genes in native and in a circadian bioluminescence reporter expressing mHypoA-2/30 cells, detecting enhancing effects of Ucn2 on mRNA levels and rhythm amplitude of the circadian regulator Aryl hydrocarbon receptor nuclear translocator-like protein 1 (Bmal1), which could facilitate anorexic responses in the activity circadian phase. These data uncover novel aspects of CRH-R2 hypothalamic signalling that might be important in regulation of circadian feeding during stress responses.


Subject(s)
Corticotropin-Releasing Hormone , Receptors, Corticotropin-Releasing Hormone , Mice , Animals , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Urocortins/genetics , Urocortins/metabolism , Gene Expression , Neurons/metabolism
12.
Sheng Li Xue Bao ; 75(4): 487-496, 2023 Aug 25.
Article in English | MEDLINE | ID: mdl-37583035

ABSTRACT

It is well established that increased excitability of the presympathetic neurons in the hypothalamic paraventricular nucleus (PVN) during hypertension leads to heightened sympathetic outflow and hypertension. However, the mechanism underlying the overactivation of PVN presympathetic neurons remains unclear. This study aimed to investigate the role of endogenous corticotropin-releasing factor (CRF) on the excitability of presympathetic neurons in PVN using Western blot, arterial blood pressure (ABP) and renal sympathetic nerve activity (RSNA) recording, CRISPR/Cas9 technique and patch-clamp technique. The results showed that CRF protein expression in PVN was significantly upregulated in spontaneously hypertensive rats (SHRs) compared with normotensive Wistar-Kyoto (WKY) rats. Besides, PVN administration of exogenous CRF significantly increased RSNA, heart rate and ABP in WKY rats. In contrast, knockdown of upregulated CRF in PVN of SHRs inhibited CRF expression, led to membrane potential hyperpolarization, and decreased the frequency of current-evoked firings of PVN presympathetic neurons, which were reversed by incubation of exogenous CRF. Perfusion of rat brain slices with artificial cerebrospinal fluid containing CRF receptor 1 (CRFR1) blocker, NBI-35965, or CRF receptor 2 (CRFR2) blocker, Antisauvagine-30, showed that blocking CRFR1, but not CRFR2, hyperpolarized the membrane potential and inhibited the current-evoked firing of PVN presympathetic neurons in SHRs. However, blocking CRFR1 or CRFR2 did not affect the membrane potential and current-evoked firing of presympathetic neurons in WKY rats. Overall, these findings indicate that increased endogenous CRF release from PVN CRF neurons enhances the excitability of presympathetic neurons via activation of CRFR1 in SHRs.


Subject(s)
Hypertension , Paraventricular Hypothalamic Nucleus , Rats , Animals , Rats, Inbred SHR , Paraventricular Hypothalamic Nucleus/physiology , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Rats, Inbred WKY , Corticotropin-Releasing Hormone/pharmacology , Corticotropin-Releasing Hormone/metabolism , Neurons/physiology , Sympathetic Nervous System
13.
J Ovarian Res ; 16(1): 155, 2023 Aug 05.
Article in English | MEDLINE | ID: mdl-37543650

ABSTRACT

BACKGROUND: Women with polycystic ovarian syndrome (PCOS) have increased hypothalamic-pituitary-adrenal (HPA) axis activation, pro-inflammatory mediators, and psychological distress in response to stressors. In women with PCOS, the corticotropin-releasing hormone (CRH) induces an exaggerated HPA response, possibly mediated by one of the CRH receptors (CRHR1 or CRHR2). Both CRHR1 and CRHR2 are implicated in insulin secretion, and variants in CRHR1 and CRHR2 genes may predispose to the mental-metabolic risk for PCOS. METHODS: We phenotyped 212 Italian families with type 2 diabetes (T2D) for PCOS following the Rotterdam diagnostic criteria. We analyzed within CRHR1 and CRHR2 genes, respectively, 36 and 18 microarray-variants for parametric linkage to and/or linkage disequilibrium (LD) with PCOS under the recessive with complete penetrance (R1) and dominant with complete penetrance (D1) models. Subsequentially, we ran a secondary analysis under the models dominant with incomplete penetrance (D2) and recessive with incomplete penetrance (R2). RESULTS: We detected 22 variants in CRHR1 and 1 variant in CRHR2 significantly (p < 0.05) linked to or in LD with PCOS across different inheritance models. CONCLUSIONS: This is the first study to report CRHR1 and CRHR2 as novel risk genes in PCOS. In silico analysis predicted that the detected CRHR1 and CRHR2 risk variants promote negative chromatin activation of their related genes in the ovaries, potentially affecting the female cycle and ovulation. However, CRHR1- and CRHR2-risk variants might also lead to hypercortisolism and confer mental-metabolic pleiotropic effects. Functional studies are needed to confirm the pathogenicity of genes and related variants.


Subject(s)
Diabetes Mellitus, Type 2 , Polycystic Ovary Syndrome , Female , Humans , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Corticotropin-Releasing Hormone/pharmacology , Hypothalamo-Hypophyseal System/metabolism , Polycystic Ovary Syndrome/genetics , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism
14.
Nat Commun ; 14(1): 3953, 2023 07 04.
Article in English | MEDLINE | ID: mdl-37402735

ABSTRACT

Urocortin 2 (UCN2) acts as a ligand for the G protein-coupled receptor corticotropin-releasing hormone receptor 2 (CRHR2). UCN2 has been reported to improve or worsen insulin sensitivity and glucose tolerance in vivo. Here we show that acute dosing of UCN2 induces systemic insulin resistance in male mice and skeletal muscle. Inversely, chronic elevation of UCN2 by injection with adenovirus encoding UCN2 resolves metabolic complications, improving glucose tolerance. CRHR2 recruits Gs in response to low concentrations of UCN2, as well as Gi and ß-Arrestin at high concentrations of UCN2. Pre-treating cells and skeletal muscle ex vivo with UCN2 leads to internalization of CRHR2, dampened ligand-dependent increases in cAMP, and blunted reductions in insulin signaling. These results provide mechanistic insights into how UCN2 regulates insulin sensitivity and glucose metabolism in skeletal muscle and in vivo. Importantly, a working model was derived from these results that unifies the contradictory metabolic effects of UCN2.


Subject(s)
Insulin Resistance , Animals , Male , Mice , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Glucose/metabolism , Insulin , Ligands , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Urocortins/genetics , Urocortins/metabolism
15.
J Comp Neurol ; 531(14): 1389-1424, 2023 10.
Article in English | MEDLINE | ID: mdl-37393534

ABSTRACT

Understanding the neural mechanisms that regulate the stress response is critical to know how animals adapt to a changing world and is one of the key factors to be considered for improving animal welfare. Corticotropin-releasing factor (CRF) is crucial for regulating physiological and endocrine responses, triggering the activation of the sympathetic nervous system and the hypothalamo-pituitary-adrenal axis (HPA) during stress. In mammals, several telencephalic areas, such as the amygdala and the hippocampus, regulate the autonomic system and the HPA responses. These centers include subpopulations of CRF containing neurons that, by way of CRF receptors, play modulatory roles in the emotional and cognitive aspects of stress. CRF binding protein also plays a role, buffering extracellular CRF and regulating its availability. CRF role in activation of the HPA is evolutionary conserved in vertebrates, highlighting the relevance of this system to help animals cope with adversity. However, knowledge on CRF systems in the avian telencephalon is very limited, and no information exists on detailed expression of CRF receptors and binding protein. Knowing that the stress response changes with age, with important variations during the first week posthatching, the aim of this study was to analyze mRNA expression of CRF, CRF receptors 1 and 2, and CRF binding protein in chicken telencephalon throughout embryonic and early posthatching development, using in situ hybridization. Our results demonstrate an early expression of CRF and its receptors in pallial areas regulating sensory processing, sensorimotor integration and cognition, and a late expression in subpallial areas regulating the stress response. However, CRF buffering system develops earlier in the subpallium than in the pallium. These results help to understand the mechanisms underlying the negative effects of noise and light during prehatching stages in chicken, and suggest that stress regulation becomes more sophisticated with age.


Subject(s)
Chickens , Corticotropin-Releasing Hormone , Animals , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Chickens/metabolism , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , RNA, Messenger/metabolism , Hypothalamo-Hypophyseal System/metabolism , Hippocampus/metabolism , Pituitary-Adrenal System/physiology , Mammals
16.
EMBO Rep ; 24(2): e55313, 2023 02 06.
Article in English | MEDLINE | ID: mdl-36413000

ABSTRACT

Growing evidence suggests that the corticotropin-releasing hormone (CRH) signaling pathway, mainly known as a critical initiator of humoral stress responses, has a role in normal neuronal physiology. However, despite the evidence of CRH receptor (CRHR) expression in the embryonic ventricular zone, the exact functions of CRH signaling in embryonic brain development have not yet been fully determined. In this study, we show that CRHR1 is required for the maintenance of neural stem cell properties, as assessed by in vitro neurosphere assays and cell distribution in the embryonic cortical layers following in utero electroporation. Identifying the underlying molecular mechanisms of CRHR1 action, we find that CRHR1 functions are accomplished through the increasing expression of the master transcription factor REST. Furthermore, luciferase reporter and chromatin immunoprecipitation assays reveal that CRHR1-induced CREB activity is responsible for increased REST expression at the transcriptional level. Taken together, these findings indicate that the CRHR1/CREB/REST signaling cascade plays an important role downstream of CRH in the regulation of neural stem cells during embryonic brain development.


Subject(s)
Corticotropin-Releasing Hormone , Neural Stem Cells , Animals , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Neurons/metabolism , Signal Transduction , Neural Stem Cells/metabolism , Mammals/metabolism
17.
Comb Chem High Throughput Screen ; 26(9): 1755-1765, 2023.
Article in English | MEDLINE | ID: mdl-36173042

ABSTRACT

BACKGROUND: Depression is a typical outcome of the repair of posttraumatic stress disorder (PTSD). Based on network pharmacology and neuropharmacology experiments, this study aimed to explore how gastrodin (GAS) reverses depressive symptoms in traumatically stressed rats. METHODS: GAS-related targets were predicted by SwissTargetPrediction; depression-related targets were collected from GeneCards and therapeutic target database (TTD); protein-protein interaction (PPI) network was constructed with its action mechanism being predicted by gene ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment. The animal model of PTSD was replicated by single prolonged stress (SPS). The antidepressant effect of GAS was investigated by the forced swim test (FST) and tail suspension test (TST). The levels of tyrosine hydroxylase (TH) and corticotropin-releasing factor type I receptor (CRF1) in locus ceruleus (LC) and the expression of corticotropin-releasing factor (CRF) in the paraventricular nucleus of the hypothalamus (PVN) and central amygdala (CeA) were measured by immunofluorescence. RESULTS: GAS significantly shortened the tail suspension and swimming immobility in SPS rats in TST and FST experiments (p < 0.05 or p < 0.01). The network analysis showed that the critical antidepressant targets of GAS were 86 targets such as GAPDH, CASP3 MMP9, HRAS, DPP4, and TH, which were significantly enriched in the pathways such as pathways neuroactive ligandreceptor interaction. High doses of GAS could significantly reduce the level of TH and CRF in CEA in the brain of rats with depressive symptoms (p < 0.01) and, at the same time, lower the expression of CRF in PVN (p < 0.05). CONCLUSION: The effect of GAS on depressive symptoms in SPS rats may be closely related to its reduction of CRF expression in PVN and CeA and inhibition of neuron (NE) synthesis in LC.


Subject(s)
Corticotropin-Releasing Hormone , Depression , Rats , Animals , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Depression/drug therapy , Depression/metabolism , Network Pharmacology , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Antidepressive Agents/pharmacology , Antidepressive Agents/therapeutic use
18.
Biosci Biotechnol Biochem ; 87(3): 308-313, 2023 Feb 24.
Article in English | MEDLINE | ID: mdl-36477851

ABSTRACT

Corticotropin-releasing hormone (Crh) and its receptors (Crhr) mediate stress-induced gastrointestinal dysfunctions. Neonatal maternal separation (MS) increased ileal Crhr1 transcript quantities in young rat offspring. Exposure to either MS or adulthood restraint stress increased ileal Crhr1 and Crhr2 transcript quantities only in adult female offspring. Maternal probiotic intervention reversed Crhr overexpression, suggesting a potential early prophylaxis against stress-induced gut dysfunctions.


Subject(s)
Probiotics , Receptors, Corticotropin-Releasing Hormone , Rats , Animals , Female , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Maternal Deprivation , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Gene Expression
19.
Int J Mol Sci ; 23(17)2022 Aug 29.
Article in English | MEDLINE | ID: mdl-36077219

ABSTRACT

The corticotropin-releasing hormone receptor 2 (CRHR2) gene encodes CRHR2, contributing to the hypothalamic-pituitary-adrenal stress response and to hyperglycemia and insulin resistance. CRHR2-/- mice are hypersensitive to stress, and the CRHR2 locus has been linked to type 2 diabetes and depression. While CRHR2 variants confer risk for mood disorders, MDD, and type 2 diabetes, they have not been investigated in familial T2D and MDD. In 212 Italian families with type 2 diabetes and depression, we tested 17 CRHR2 single nucleotide polymorphisms (SNPs), using two-point parametric-linkage and linkage-disequilibrium (i.e., association) analysis (models: dominant-complete-penetrance-D1, dominant-incomplete-penetrance-D2, recessive-complete-penetrance-R1, recessive-incomplete-penetrance-R2). We detected novel linkage/linkage-disequilibrium/association to/with depression (3 SNPs/D1, 2 SNPs/D2, 3 SNPs/R1, 3 SNPs/R2) and type 2 diabetes (3 SNPs/D1, 2 SNPs/D2, 2 SNPs/R1, 1 SNP/R2). All detected risk variants are novel. Two depression-risk variants within one linkage-disequilibrium block replicate each other. Two independent novel SNPs were comorbid while the most significant conferred either depression- or type 2 diabetes-risk. Although the families were primarily ascertained for type 2 diabetes, depression-risk variants showed higher significance than type 2 diabetes-risk variants, implying CRHR2 has a stronger role in depression-risk than type 2 diabetes-risk. In silico analysis predicted variants' dysfunction. CRHR2 is for the first time linked to/in linkage-disequilibrium/association with depression-type 2 diabetes comorbidity and may underlie the shared genetic pathogenesis via pleiotropy.


Subject(s)
Depression/genetics , Diabetes Mellitus, Type 2 , Receptors, Corticotropin-Releasing Hormone/genetics , Animals , Comorbidity , Diabetes Mellitus, Type 2/genetics , Genetic Predisposition to Disease , Linkage Disequilibrium , Mice , Polymorphism, Single Nucleotide
20.
Front Endocrinol (Lausanne) ; 13: 893238, 2022.
Article in English | MEDLINE | ID: mdl-36147561

ABSTRACT

Hypothalamus-pituitary-adrenal (HPA) axis plays critical roles in stress responses under challenging conditions such as hypoxia, via regulating gene expression and integrating activities of hypothalamus-pituitary-targets cells. However, the transcriptional regulatory mechanisms and signaling pathways of hypoxic stress in the pituitary remain to be defined. Here, we report that hypoxia induced dynamic changes in the transcription factors, hormones, and their receptors in the adult rat pituitary. Hypoxia-inducible factors (HIFs), oxidative phosphorylation, and cAMP signaling pathways were all differentially enriched in genes induced by hypoxic stress. In the pituitary gene network, hypoxia activated c-Fos and HIFs with specific pituitary transcription factors (Prop1), targeting the promoters of hormones and their receptors. HIF and its related signaling pathways can be a promising biomarker during acute or constant hypoxia. Hypoxia stimulated the transcription of marker genes for microglia, chemokines, and cytokine receptors of the inflammatory response. Corticotropin-releasing hormone receptor 1 (CRHR1) mediated the transcription of Pomc, Sstr2, and Hif2a, and regulated the function of HPA axis. Together with HIF, c-Fos initiated and modulated dynamic changes in the transcription of hormones and their receptors. The receptors were also implicated in the regulation of functions of target cells in the pituitary network under hypoxic stress. CRHR1 played an integrative role in the hypothalamus-pituitary-target axes. This study provides new evidence for CRHR1 involved changes of hormones, receptors, signaling molecules and pathways in the pituitary induced by hypoxia.


Subject(s)
Hypothalamo-Hypophyseal System , Receptors, Corticotropin-Releasing Hormone , Animals , Hormones/metabolism , Hypothalamo-Hypophyseal System/metabolism , Hypoxia/genetics , Hypoxia/metabolism , Pituitary-Adrenal System/metabolism , Pro-Opiomelanocortin/genetics , RNA, Messenger/genetics , Rats , Receptors, Corticotropin-Releasing Hormone/genetics , Receptors, Corticotropin-Releasing Hormone/metabolism , Receptors, Cytokine/metabolism , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL