Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.442
Filter
1.
Sci Rep ; 14(1): 17056, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39048594

ABSTRACT

Corticotropin-releasing factor (CRF) is mainly secreted from the hypothalamic paraventricular nuclei and plays a crucial role in stress-related responses. Recent studies have reported that CRF is a neuromodulator in the central nervous system. In the cerebellum, CRF is essential for the induction of long-term depression (LTD) at the parallel fiber-Purkinje cell synapses. Given that LTD is thought to be one of the fundamental mechanisms of motor learning, CRF may affect motor learning. However, the role of CRF in motor learning in vivo remains unclear. In this study, we aimed to examine the role of CRF in motor learning. This was achieved through a series of behavioral experiments involving the in vivo administration of CRF and its antagonists. Rats injected with CRF directly into the cerebellum exhibited superior performance on the rotarod test, especially during initial training phases, compared to control subjects. Conversely, rats receiving a CRF receptor antagonist demonstrated reduced endurance on the rotating rod compared to controls. Notably, CRF mRNA expression levels in the cerebellum did not show significant variance between the CRF-injected and control groups. These findings imply a critical role of endogenous CRF in cerebellar motor learning and suggest that exogenous CRF can augment this process. (199 words).


Subject(s)
Cerebellum , Corticotropin-Releasing Hormone , Learning , Animals , Corticotropin-Releasing Hormone/metabolism , Male , Rats , Learning/physiology , Learning/drug effects , Cerebellum/metabolism , Cerebellum/drug effects , Cerebellum/physiology , Motor Activity/drug effects , Receptors, Corticotropin-Releasing Hormone/metabolism , Rats, Sprague-Dawley
2.
Nat Commun ; 15(1): 5522, 2024 Jun 29.
Article in English | MEDLINE | ID: mdl-38951506

ABSTRACT

Failure to appropriately predict and titrate reactivity to threat is a core feature of fear and anxiety-related disorders and is common following early life adversity (ELA). A population of neurons in the lateral central amygdala (CeAL) expressing corticotropin releasing factor (CRF) have been proposed to be key in processing threat of different intensities to mediate active fear expression. Here, we use in vivo fiber photometry to show that ELA results in sex-specific changes in the activity of CeAL CRF+ neurons, yielding divergent mechanisms underlying the augmented startle in ELA mice, a translationally relevant behavior indicative of heightened threat reactivity and hypervigilance. Further, chemogenic inhibition of CeAL CRF+ neurons selectively diminishes startle and produces a long-lasting suppression of threat reactivity. These findings identify a mechanism for sex-differences in susceptibility for anxiety following ELA and have broad implications for understanding the neural circuitry that encodes and gates the behavioral expression of fear.


Subject(s)
Anxiety , Central Amygdaloid Nucleus , Corticotropin-Releasing Hormone , Fear , Neurons , Reflex, Startle , Animals , Corticotropin-Releasing Hormone/metabolism , Fear/physiology , Neurons/metabolism , Neurons/physiology , Mice , Female , Male , Anxiety/physiopathology , Central Amygdaloid Nucleus/metabolism , Reflex, Startle/physiology , Mice, Inbred C57BL , Behavior, Animal/physiology , Stress, Psychological
3.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article in English | MEDLINE | ID: mdl-39000501

ABSTRACT

A large body of evidence indicates that vasopressin (AVP) and steroid hormones are frequently secreted together and closely cooperate in the regulation of blood pressure, metabolism, water-electrolyte balance, and behavior, thereby securing survival and the comfort of life. Vasopressin cooperates with hormones of the hypothalamo-pituitary-adrenal axis (HPA) at several levels through regulation of the release of corticotropin-releasing hormone (CRH), adrenocorticotropic hormone (ACTH), and multiple steroid hormones, as well as through interactions with steroids in the target organs. These interactions are facilitated by positive and negative feedback between specific components of the HPA. Altogether, AVP and the HPA cooperate closely as a coordinated functional AVP-HPA system. It has been shown that cooperation between AVP and steroid hormones may be affected by cellular stress combined with hypoxia, and by metabolic, cardiovascular, and respiratory disorders; neurogenic stress; and inflammation. Growing evidence indicates that central and peripheral interactions between AVP and steroid hormones are reprogrammed in cardiovascular and metabolic diseases and that these rearrangements exert either beneficial or harmful effects. The present review highlights specific mechanisms of the interactions between AVP and steroids at cellular and systemic levels and analyses the consequences of the inappropriate cooperation of various components of the AVP-HPA system for the pathogenesis of cardiovascular and metabolic diseases.


Subject(s)
Cardiovascular Diseases , Hypothalamo-Hypophyseal System , Metabolic Diseases , Pituitary-Adrenal System , Vasopressins , Humans , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Vasopressins/metabolism , Cardiovascular Diseases/metabolism , Animals , Metabolic Diseases/metabolism , Corticotropin-Releasing Hormone/metabolism , Adrenocorticotropic Hormone/metabolism
4.
Exp Neurol ; 378: 114822, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38823676

ABSTRACT

Post-stroke depression (PSD) is a complication of cerebrovascular disease, which can increase mortality after stroke. CRH is one of the main signaling peptides released after activation of the hypothalamic-pituitary-adrenal (HPA) axis in response to stress. It affects synaptic plasticity by regulating inflammation, oxidative stress and autophagy in the central nervous system. And the loss of spines exacerbates depression-like behavior. Therefore, synaptic deficits induced by CRH may be related to post-stroke depression. However, the underlying mechanism remains unclear. The Keap1-Nrf2 complex is one of the core components of the antioxidant response. As an autophagy associated protein, p62 participates in the Keap1-NrF2 pathway through its Keap1 interaction domain. Oxidative stress is involved in the feedback regulation between Keap1-Nrf2 pathway and p62.However, whether the relationship between CRH and the Keap1-Nrf2-p62 pathway is involved in PSD remains unknown. This study found that serum levels of CRH in 22 patients with PSD were higher than those in healthy subjects. We used MCAO combined with CUMS single-cage SD rats to establish an animal model of PSD. Animal experiments showed that CRHR1 antagonist prevented synaptic loss in the hippocampus of PSD rats and alleviated depression-like behavior. CRH induced p62 accumulation in the prefrontal cortex of PSD rats through CRHR1. CRHR1 antagonist inhibited Keap1-Nrf2-p62 pathway by attenuating oxidative stress. In addition, we found that abnormal accumulation of p62 induces PSD. It alleviates depression-like behavior by inhibiting the expression of p62 and promoting the clearance of p62 in PSD rats. These findings can help explore the pathogenesis of PSD and design targeted treatments for PSD.


Subject(s)
Depression , Rats, Sprague-Dawley , Receptors, Corticotropin-Releasing Hormone , Stroke , Animals , Rats , Male , Depression/etiology , Depression/drug therapy , Depression/metabolism , Stroke/complications , Stroke/drug therapy , Stroke/psychology , Stroke/metabolism , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/metabolism , Humans , Down-Regulation/drug effects , Middle Aged , Disease Models, Animal , Female , Aged , Sequestosome-1 Protein/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , Kelch-Like ECH-Associated Protein 1/antagonists & inhibitors , NF-E2-Related Factor 2/metabolism , Corticotropin-Releasing Hormone/metabolism
5.
Int J Mol Sci ; 25(11)2024 May 28.
Article in English | MEDLINE | ID: mdl-38892090

ABSTRACT

Fetal hypoxia and maternal stress frequently culminate in neuropsychiatric afflictions in life. To replicate this condition, we employed a model of prenatal severe hypoxia (PSH) during days 14-16 of rat gestation. Subsequently, both control and PSH rats at 3 months old were subjected to episodes of inescapable stress to induce learned helplessness (LH). The results of the open field test revealed an inclination towards depressive-like behavior in PSH rats. Following LH episodes, control (but not PSH) rats displayed significant anxiety. LH induced an increase in glucocorticoid receptor (GR) levels in extrahypothalamic brain structures, with enhanced nuclear translocation in the hippocampus (HPC) observed both in control and PSH rats. However, only control rats showed an increase in GR nuclear translocation in the amygdala (AMG). The decreased GR levels in the HPC of PSH rats correlated with elevated levels of hypothalamic corticotropin-releasing hormone (CRH) compared with the controls. However, LH resulted in a reduction of the CRH levels in PSH rats, aligning them with those of control rats, without affecting the latter. This study presents evidence that PSH leads to depressive-like behavior in rats, associated with alterations in the glucocorticoid system. Notably, these impairments also contribute to increased resistance to severe stressors.


Subject(s)
Anxiety , Depression , Glucocorticoids , Prenatal Exposure Delayed Effects , Receptors, Glucocorticoid , Animals , Rats , Female , Anxiety/metabolism , Pregnancy , Glucocorticoids/metabolism , Depression/metabolism , Depression/etiology , Receptors, Glucocorticoid/metabolism , Prenatal Exposure Delayed Effects/metabolism , Stress, Psychological/metabolism , Male , Corticotropin-Releasing Hormone/metabolism , Hippocampus/metabolism , Hypoxia/metabolism , Phenotype , Behavior, Animal , Helplessness, Learned , Disease Models, Animal , Amygdala/metabolism , Fetal Hypoxia/metabolism , Fetal Hypoxia/complications
6.
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
7.
Neurosci Biobehav Rev ; 163: 105748, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38857667

ABSTRACT

Corticotropin-releasing factor (CRF, corticoliberin) is a neuromodulatory peptide activating the hypothalamic-pituitary-adrenal (HPA) axis, widely distributed in the central nervous system (CNS) in mammals. In addition to its neuroendocrine effects, CRF is essential in regulating many functions under physiological and pathophysiological conditions through CRF1 and CRF2 receptors (CRF1R, CRF2R). This review aims to present selected examples of the diverse and sometimes opposite effects of CRF and its receptor ligands in various pathophysiological states, including stress/anxiety, depression, and processes associated with brain injury. It seems interesting to draw particular attention to the fact that CRF and its receptor ligands exert different effects depending on the brain structures or subregions, likely stemming from the varied distribution of CRFRs in these regions and interactions with other neurotransmitters. CRFR-mediated region-specific effects might also be related to brain site-specific ligand binding and the associated activated signaling pathways. Intriguingly, different types of CRF molecules can also influence the diverse actions of CRF in the CNS.


Subject(s)
Anxiety , Corticotropin-Releasing Hormone , Receptors, Corticotropin-Releasing Hormone , Receptors, Corticotropin-Releasing Hormone/metabolism , Humans , Animals , Corticotropin-Releasing Hormone/metabolism , Anxiety/metabolism , Anxiety/physiopathology , Brain Injuries/metabolism , Brain Injuries/physiopathology , Stress, Psychological/metabolism , Stress, Psychological/physiopathology , Depression/metabolism , Depression/physiopathology , Brain/metabolism , Brain/physiopathology
8.
eNeuro ; 11(7)2024 Jul.
Article in English | MEDLINE | ID: mdl-38914464

ABSTRACT

Epilepsy is often comorbid with psychiatric illnesses, including anxiety and depression. Despite the high incidence of psychiatric comorbidities in people with epilepsy, few studies address the underlying mechanisms. Stress can trigger epilepsy and depression. Evidence from human and animal studies supports that hypothalamic-pituitary-adrenal (HPA) axis dysfunction may contribute to both disorders and their comorbidity ( Kanner, 2003). Here, we investigate if HPA axis dysfunction may influence epilepsy outcomes and psychiatric comorbidities. We generated a novel mouse model (Kcc2/Crh KO mice) lacking the K+/Cl- cotransporter, KCC2, in corticotropin-releasing hormone (CRH) neurons, which exhibit stress- and seizure-induced HPA axis hyperactivation ( Melon et al., 2018). We used the Kcc2/Crh KO mice to examine the impact on epilepsy outcomes, including seizure frequency/burden, comorbid behavioral deficits, and sudden unexpected death in epilepsy (SUDEP) risk. We found sex differences in HPA axis dysfunction's effect on chronically epileptic KCC2/Crh KO mice seizure burden, vulnerability to comorbid behavioral deficits, and SUDEP. Suppressing HPA axis hyperexcitability in this model using pharmacological or chemogenetic approaches decreased SUDEP incidence, suggesting that HPA axis dysfunction may contribute to SUDEP. Altered neuroendocrine markers were present in SUDEP cases compared with people with epilepsy or individuals without epilepsy. Together, these findings implicate HPA axis dysfunction in the pathophysiological mechanisms contributing to psychiatric comorbidities in epilepsy and SUDEP.


Subject(s)
Corticotropin-Releasing Hormone , Hypothalamo-Hypophyseal System , Mice, Knockout , Pituitary-Adrenal System , Sudden Unexpected Death in Epilepsy , Animals , Hypothalamo-Hypophyseal System/metabolism , Pituitary-Adrenal System/metabolism , Male , Female , Mice , Corticotropin-Releasing Hormone/metabolism , Sex Characteristics , Epilepsy/metabolism , Epilepsy/physiopathology , K Cl- Cotransporters , Symporters/metabolism , Disease Models, Animal , Mice, Inbred C57BL , Humans , Sex Factors
9.
Pituitary ; 27(4): 345-359, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38888685

ABSTRACT

CONTEXT: Once hypercortisolemia is confirmed, differential diagnosis between Cushing's syndrome (CS) due to neoplastic endogenous hypercortisolism and non-neoplastic hypercortisolism (NNH, pseudo-Cushing's syndrome) is crucial. Due to worldwide corticotropin-releasing hormone (CRH) unavailability, accuracy of alternative tests to dexamethasone (Dex)-CRH, is clearly needed. OBJECTIVE: Assess the diagnostic accuracy of Dex-CRH test, desmopressin stimulation test, midnight serum cortisol (MSC), and late-night salivary cortisol (LNSC) levels to distinguish CS from NNH. METHODS: Articles through March 2022 were identified from Scopus, Web of Science, MEDLINE, EMBASE, and PubMed. All steps through the systematic review were performed independently and in duplicate and strictly adhered to the updated PRISMA-DTA checklist. DATA SYNTHESIS: A total of 24 articles (1900 patients) were included. Dex-CRH had a pooled sensitivity and specificity of 91% (95%CI 87-94%; I2 0%) and 82% (73-88%; I2 50%), desmopressin test 86% (81-90%; I2 28%) and 90% (84-94%; I2 15%), MSC 91% (85-94%; I2 66%) and 81% (70-89%; I2 71%), and LNSC 80% (67-89%; I2 57%) and 90% (84-93%; I2 21%), respectively. Summary receiver operating characteristics areas under the curve were Dex-CRH 0.949, desmopressin test 0.936, MSC 0.942, and LNSC 0.950 without visual or statistical significance. The overall risk of studies bias was moderate. CONCLUSION: Dex-CRH, the desmopressin stimulation test, and MSC have similar diagnostic accuracy, with Dex-CRH and MSC having slightly higher sensitivity, and the desmopressin test being more specific. LNSC was the least accurate, probably due to high heterogeneity, intrinsic variability, different assays, and lack of consistent reported cutoffs. When facing this challenging differential diagnosis, the results presented here should increase clinicians' confidence when deciding which test to perform.


Subject(s)
Cushing Syndrome , Humans , Cushing Syndrome/diagnosis , Hydrocortisone/blood , Hydrocortisone/metabolism , Diagnosis, Differential , Corticotropin-Releasing Hormone/metabolism , Dexamethasone , Deamino Arginine Vasopressin
10.
Biochem Biophys Res Commun ; 725: 150219, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-38941883

ABSTRACT

BACKGROUND: Neonates undergo numerous painful procedures throughout their hospitalization. Repeated procedural pain may cause adverse long-term effects. Glucose as a non-pharmacological analgesia, is used for neonate pain management. In this study, potential mechanism of attenuate pain induced by glucose in neurodevelopment effect of neonate pain stimulus was investigated. METHODS: Neonatal rats to perform a repetitive injury model and glucose intervention model in the postnatal day 0-7(P0-7). Pain thresholds were measured by von Frey test weekly. The puberty behavioral outcome, tissue loss and protein expression in hippocampus were analyzed. RESULTS: Oral administration of glucose after repeated pain stimulation can maintain the hippocampal structure in, and reduce the expressions of corticotropin releasing factor (CFR) and glucocorticoid receptor (GR), therefore, resulted in long-term threshold of pain and cognitive improvement. CONCLUSION: Exposure to neonatal repeated procedural pain causes persistent mechanical hypersensitivity and the dysfunction of spatial memory retention at puberty. In addition, glucose can relieve these adverse effects, possibly via decreasing CRF/GR levels to change the hypothalamus-pituitary-adrenal (HPA) axis.


Subject(s)
Animals, Newborn , Corticotropin-Releasing Hormone , Glucose , Hippocampus , Pain , Rats, Sprague-Dawley , Receptors, Glucocorticoid , Animals , Glucose/metabolism , Corticotropin-Releasing Hormone/metabolism , Receptors, Glucocorticoid/metabolism , Pain/metabolism , Pain/etiology , Rats , Hippocampus/metabolism , Hippocampus/drug effects , Male , Pain Threshold/drug effects , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/drug effects , Pituitary-Adrenal System/metabolism , Pituitary-Adrenal System/drug effects , Female
11.
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
12.
Mol Brain ; 17(1): 22, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702738

ABSTRACT

We previously reported that enhanced corticotropin-releasing factor (CRF) signaling in the bed nucleus of the stria terminalis (BNST) caused the aversive responses during acute pain and suppressed the brain reward system during chronic pain. However, it remains to be examined whether chronic pain alters the excitability of CRF neurons in the BNST. In this study we investigated the chronic pain-induced changes in excitability of CRF-expressing neurons in the oval part of the BNST (ovBNSTCRF neurons) by whole-cell patch-clamp electrophysiology. CRF-Cre; Ai14 mice were used to visualize CRF neurons by tdTomato. Electrophysiological recordings from brain slices prepared from a mouse model of neuropathic pain revealed that rheobase and firing threshold were significantly decreased in the chronic pain group compared with the sham-operated control group. Firing rate of the chronic pain group was higher than that of the control group. These data indicate that chronic pain elevated neuronal excitability of ovBNSTCRF neurons.


Subject(s)
Chronic Pain , Corticotropin-Releasing Hormone , Neurons , Septal Nuclei , Animals , Septal Nuclei/metabolism , Corticotropin-Releasing Hormone/metabolism , Neurons/metabolism , Chronic Pain/physiopathology , Chronic Pain/metabolism , Male , Action Potentials/physiology , Mice, Inbred C57BL , Mice
13.
Lab Anim (NY) ; 53(6): 148-159, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38806681

ABSTRACT

Researchers have advocated elevating mouse housing temperatures from the conventional ~22 °C to the mouse thermoneutral point of 30 °C to enhance translational research. However, the impact of environmental temperature on mouse gastrointestinal physiology remains largely unexplored. Here we show that mice raised at 22 °C exhibit whole gut transit speed nearly twice as fast as those raised at 30 °C, primarily driven by a threefold increase in colon transit speed. Furthermore, gut microbiota composition differs between the two temperatures but does not dictate temperature-dependent differences in gut motility. Notably, increased stress signals from the hypothalamic-pituitary-adrenal axis at 22 °C have a pivotal role in mediating temperature-dependent differences in gut motility. Pharmacological and genetic depletion of the stress hormone corticotropin-releasing hormone slows gut motility in stressed 22 °C mice but has no comparable effect in relatively unstressed 30 °C mice. In conclusion, our findings highlight that colder mouse facility temperatures significantly increase gut motility through hormonal stress pathways.


Subject(s)
Gastrointestinal Motility , Mice, Inbred C57BL , Stress, Physiological , Animals , Mice , Male , Temperature , Hypothalamo-Hypophyseal System/physiology , Gastrointestinal Microbiome , Pituitary-Adrenal System/physiology , Corticotropin-Releasing Hormone/metabolism
14.
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
15.
Neuroscience ; 549: 110-120, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38723837

ABSTRACT

The brown rat (Rattus norvegicus) is known to show three types of behavioral responses to novel objects. Whereas some rats are indifferent to novel objects, neophobic and neophilic rats show avoidance and approach behavior, respectively. Here, we compared the dopaminergic, serotonergic, and noradrenergic systems immunohistochemically among these rats. Trapped wild rats and laboratory rats were first individually exposed to the novel objects in their home cage. Wild rats were divided into neophobic and indifferent rats depending on their behavioral responses. Similarly, laboratory rats were divided into neophilic and indifferent rats. Consistent with the behavioral differences, in the paraventricular nucleus of the hypothalamus, Fos expression in corticotropin-releasing hormone-containing neurons was higher in the neophobic rats than in the indifferent rats. In the anterior basal amygdala, the neophobic rats showed higher Fos expression than the indifferent rats. In the posterior basal amygdala, the neophobic and neophilic rats showed lower and higher Fos expressions than the indifferent rats, respectively. When we compared the neuromodulatory systems, in the dorsal raphe, the number of serotonergic neurons and Fos expression in serotonergic neurons increased linearly from neophobic to indifferent to neophilic rats. In the ventral tegmental area, Fos expression in dopaminergic neurons was higher in the neophilic rats than in the indifferent rats. These results demonstrate that approach/avoidance behavior to novel objects is correlated with the serotonergic and dopaminergic systems in the brown rat. We propose that the serotonergic system suppresses avoidance behavior while the dopaminergic system enhances approach behavior to novel objects.


Subject(s)
Avoidance Learning , Animals , Male , Rats , Avoidance Learning/physiology , Serotonergic Neurons/metabolism , Serotonergic Neurons/physiology , Dopaminergic Neurons/metabolism , Dopamine/metabolism , Serotonin/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Brain/metabolism , Exploratory Behavior/physiology , Behavior, Animal/physiology , Corticotropin-Releasing Hormone/metabolism
16.
J Neurosci ; 44(27)2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38789262

ABSTRACT

We previously showed that orexin neurons are activated by hypoxia and facilitate the peripheral chemoreflex (PCR)-mediated hypoxic ventilatory response (HVR), mostly by promoting the respiratory frequency response. Orexin neurons project to the nucleus of the solitary tract (nTS) and the paraventricular nucleus of the hypothalamus (PVN). The PVN contributes significantly to the PCR and contains nTS-projecting corticotropin-releasing hormone (CRH) neurons. We hypothesized that in male rats, orexin neurons contribute to the PCR by activating nTS-projecting CRH neurons. We used neuronal tract tracing and immunohistochemistry (IHC) to quantify the degree that hypoxia activates PVN-projecting orexin neurons. We coupled this with orexin receptor (OxR) blockade with suvorexant (Suvo, 20 mg/kg, i.p.) to assess the degree that orexin facilitates the hypoxia-induced activation of CRH neurons in the PVN, including those projecting to the nTS. In separate groups of rats, we measured the PCR following systemic orexin 1 receptor (Ox1R) blockade (SB-334867; 1 mg/kg) and specific Ox1R knockdown in PVN. OxR blockade with Suvo reduced the number of nTS and PVN neurons activated by hypoxia, including those CRH neurons projecting to nTS. Hypoxia increased the number of activated PVN-projecting orexin neurons but had no effect on the number of activated nTS-projecting orexin neurons. Global Ox1R blockade and partial Ox1R knockdown in the PVN significantly reduced the PCR. Ox1R knockdown also reduced the number of activated PVN neurons and the number of activated tyrosine hydroxylase neurons in the nTS. Our findings suggest orexin facilitates the PCR via nTS-projecting CRH neurons expressing Ox1R.


Subject(s)
Corticotropin-Releasing Hormone , Neurons , Orexin Receptor Antagonists , Orexin Receptors , Orexins , Rats, Sprague-Dawley , Solitary Nucleus , Animals , Male , Corticotropin-Releasing Hormone/metabolism , Orexins/metabolism , Rats , Neurons/metabolism , Neurons/physiology , Neurons/drug effects , Solitary Nucleus/metabolism , Solitary Nucleus/physiology , Solitary Nucleus/drug effects , Orexin Receptor Antagonists/pharmacology , Orexin Receptors/metabolism , Hypoxia/metabolism , Triazoles/pharmacology , Azepines/pharmacology , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/physiology
17.
Obes Rev ; 25(8): e13763, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38699883

ABSTRACT

Obesity is among the most common chronic disorders, worldwide. It is a complex disease that reflects the interactions between environmental influences, multiple genetic allelic variants, and behavioral factors. Recent developments have also shown that biological conditions in utero play an important role in the programming of energy homeostasis systems and might have an impact on obesity and metabolic disease risk. The corticotropin-releasing hormone (CRH) family of neuropeptides, as a central element of energy homeostasis, has been evaluated for its role in the pathophysiology of obesity. This review aims to summarize the relevance and effects of the CRH family of peptides in the pathophysiology of obesity spanning from fetal life to adulthood.


Subject(s)
Corticotropin-Releasing Hormone , Obesity , Humans , Corticotropin-Releasing Hormone/metabolism , Obesity/metabolism , Pregnancy , Female , Energy Metabolism/physiology , Adult , Prenatal Exposure Delayed Effects , Homeostasis/physiology
18.
J Physiol ; 602(14): 3375-3400, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38698722

ABSTRACT

Chronic intermittent hypoxia (CIH) in rodents mimics the hypoxia-induced elevation of blood pressure seen in individuals experiencing episodic breathing. The brainstem nucleus tractus solitarii (nTS) is the first site of visceral sensory afferent integration, and thus is critical for cardiorespiratory homeostasis and its adaptation during a variety of stressors. In addition, the paraventricular nucleus of the hypothalamus (PVN), in part through its nTS projections that contain oxytocin (OT) and/or corticotropin-releasing hormone (CRH), contributes to cardiorespiratory regulation. Within the nTS, these PVN-derived neuropeptides alter nTS activity and the cardiorespiratory response to hypoxia. Nevertheless, their contribution to nTS activity after CIH is not fully understood. We hypothesized that OT and CRH would increase nTS activity to a greater extent following CIH, and co-activation of OT+CRH receptors would further magnify nTS activity. Our data show that compared to their normoxic controls, 10 days' CIH exaggerated nTS discharge, excitatory synaptic currents and Ca2+ influx in response to CRH, which were further enhanced by the addition of OT. CIH increased the tonic functional contribution of CRH receptors, which occurred with elevation of mRNA and protein. Together, our data demonstrate that intermittent hypoxia exaggerates the expression and function of neuropeptides on nTS activity. KEY POINTS: Episodic breathing and chronic intermittent hypoxia (CIH) are associated with autonomic dysregulation, including elevated sympathetic nervous system activity. Altered nucleus tractus solitarii (nTS) activity contributes to this response. Neurons originating in the paraventricular nucleus (PVN), including those containing oxytocin (OT) and corticotropin-releasing hormone (CRH), project to the nTS, and modulate the cardiorespiratory system. Their role in CIH is unknown. In this study, we focused on OT and CRH individually and together on nTS activity from rats exposed to either CIH or normoxia control. We show that after CIH, CRH alone and with OT increased to a greater extent overall nTS discharge, neuronal calcium influx, synaptic transmission to second-order nTS neurons, and OT and CRH receptor expression. These results provide insights into the underlying circuits and mechanisms contributing to autonomic dysfunction during periods of episodic breathing.


Subject(s)
Corticotropin-Releasing Hormone , Hypoxia , Neurons , Oxytocin , Rats, Sprague-Dawley , Solitary Nucleus , Animals , Solitary Nucleus/metabolism , Solitary Nucleus/physiology , Corticotropin-Releasing Hormone/metabolism , Oxytocin/metabolism , Hypoxia/physiopathology , Hypoxia/metabolism , Male , Neurons/physiology , Neurons/metabolism , Neurons/drug effects , Rats , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/physiology , Receptors, Corticotropin-Releasing Hormone/metabolism
19.
Psychopharmacology (Berl) ; 241(8): 1565-1575, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38630316

ABSTRACT

RATIONALE: Corticotropin-releasing factor (CRF), the apical stress-inducing hormone, exacerbates stress and addictive behaviors. TCAP-1 is a peptide that directly inhibits both CRF-mediated stress and addiction-related behaviors; however, the direct action of TCAP-1 on morphine withdrawal-associated behaviors has not previously been examined. OBJECTIVE: To determine whether TCAP-1 administration attenuates behavioral and physiological consequences of morphine withdrawal in mice. METHODS: Mice were administered via subcutaneous route TCAP-1 either before or after initial morphine exposure, after which jumping behavior was quantified to assess the effects of TCAP-1 on naloxone-precipitated morphine withdrawal. As a comparison, mice were treated with nonpeptide CRF1 receptor antagonist CP-154,526. In one experiment, plasma corticosterone (CORT) was also measured as a physiological stress indicator. RESULTS: Pretreatment with TCAP-1 (10-250 nmol/kg) before morphine treatment significantly inhibited the development of naloxone-precipitated withdrawal. TCAP-1 (250-500 nmol/kg) treatment administered after morphine treatment attenuated the behavioral expression of naloxone-precipitated withdrawal. TCAP-1 (250 nmol/kg) treatment during morphine treatment was more effective than the optimal dosing of CP-154,526 (20 mg/kg) at suppressing the behavioral expression of naloxone-precipitated withdrawal, despite similar reduction of withdrawal-induced plasma CORT level increases. CONCLUSIONS: These findings establish TCAP-1 as a potential therapeutic candidate for the prevention and treatment of morphine withdrawal.


Subject(s)
Corticosterone , Morphine , Naloxone , Narcotic Antagonists , Substance Withdrawal Syndrome , Animals , Male , Substance Withdrawal Syndrome/drug therapy , Substance Withdrawal Syndrome/metabolism , Naloxone/pharmacology , Naloxone/administration & dosage , Mice , Narcotic Antagonists/pharmacology , Narcotic Antagonists/administration & dosage , Morphine/administration & dosage , Morphine/pharmacology , Corticosterone/blood , Corticosterone/administration & dosage , Dose-Response Relationship, Drug , Pyrroles/pharmacology , Pyrroles/administration & dosage , Nerve Tissue Proteins/metabolism , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Receptors, Corticotropin-Releasing Hormone/metabolism , Behavior, Animal/drug effects , Morphine Dependence/metabolism , Morphine Dependence/drug therapy , Morphine Dependence/prevention & control , Pyrrolidines/pharmacology , Pyrrolidines/administration & dosage , Injections, Subcutaneous , Corticotropin-Releasing Hormone/metabolism , Corticotropin-Releasing Hormone/pharmacology , Pyrimidines
20.
PLoS One ; 19(4): e0299376, 2024.
Article in English | MEDLINE | ID: mdl-38630738

ABSTRACT

AIM OF THE STUDY: To evaluate the therapeutic effect of SYNC in diarrhea irritable bowel syndrome (IBS-D) and explore its underlying mechanism through transcriptomic sequencing (RNA-Seq). MATERIALS AND METHODS: A rat model of IBS-D was constructed to elucidate the effects of SYNC. Abdominal withdrawal reflex (AWR), fecal water content (FWC), and recording body weight were calculated to assess visceral sensitivity in rats. Histopathological changes in the colon and alterations in mast cell (MC) count were determined. Immunohistochemistry was employed to assess mast cell tryptase (MCT) expression in rat colons. Serum levels of corticotropin-releasing Hormone (CRH), interleukin-6 (IL-6), calcitonin gene-related peptide (CGRP), and 5-hydroxytryptamine (5-HT) were quantified using ELISA. RNA-Seq of colon tissue was performed, followed by Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. Western blot analysis was conducted to quantify the expression levels of key proteins in the Nr4a3 pathway in the colon and hypothalamus tissues of rats. RESULTS: SYNC alleviated visceral hypersensitivity and mood disorders in rats with IBS-D. Moreover, it was positively correlated with its dosage and the observed effects, such as the enhancement of the colon's mucosal lining condition and reduction in the number and activation of MCs within the model group. SYNC reduced the expression levels of factors related to the brain-gut axis and inflammatory markers in the bloodstream. RNA-Seq analysis indicated that SYNC down-regulated the expression of Nr4a3 and PI3K. These SYNC-targeted genes primarily played roles in immune regulation and inflammatory responses, correlating with the modulation of Nr4a3 and the PI3K/AKT pathway. Western blot analysis further confirmed SYNC's influence on inflammation-related MC activation by downregulating key proteins in the Nr4a3/PI3K pathway. CONCLUSIONS: SYNC inhibited mast cell activation and attenuated visceral hypersensitivity in the colon tissues of IBS-D rats. These effects were mediated by the Nr4a3/PI3K signaling pathway.


Subject(s)
Irritable Bowel Syndrome , Rats , Animals , Irritable Bowel Syndrome/pathology , Rats, Sprague-Dawley , Phosphatidylinositol 3-Kinases , Diarrhea , Corticotropin-Releasing Hormone/metabolism , DNA-Binding Proteins , Nerve Tissue Proteins
SELECTION OF CITATIONS
SEARCH DETAIL