Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 12.662
Filtrar
1.
Nat Commun ; 15(1): 8632, 2024 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-39366937

RESUMO

Rapid glucose supply is crucial for animal survival during stress response. How the timescale of stress-induced glucose release precisely controlled by hypothalamic corticotropin-releasing hormone (CRH) neurons remains unclear. Here, we show that stress-induced hyperglycemia can be divided into at least two stages in male mice: the first fast stage is mediated by hypothalamus (paraventricular to ventromedial hypothalamus)-sympathetic (raphe pallidus nucleus to intermediolateral nucleus)-liver (HSL) axis activity; the second delayed stage is mediated by adrenal activity. Blocking the activity of HSL axis impairs predatory evoked flight responses, indicating that the HSL pathway activity is necessary for stress coping. We further reveal the intracellular signal cascade for CRH signal in the hypothalamus, which is mediated by GABAA receptor ß3 subunit phosphorylation at S408/409, results in prevention of GABAA receptor membrane recruitment. Thus, we uncovered the precise timescale of glucose supply during stress which is mediated by adrenal independent HSL and adrenal dependent pathway respectively.


Assuntos
Hormônio Liberador da Corticotropina , Hiperglicemia , Hipotálamo , Fígado , Receptores de GABA-A , Animais , Masculino , Hiperglicemia/metabolismo , Fígado/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Camundongos , Hipotálamo/metabolismo , Receptores de GABA-A/metabolismo , Estresse Fisiológico , Glucose/metabolismo , Transdução de Sinais , Camundongos Endogâmicos C57BL , Fosforilação , Neurônios/metabolismo , Glândulas Suprarrenais/metabolismo , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia
2.
Exp Dermatol ; 33(10): e15182, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39367575

RESUMO

Stress may play a key role in alopecia areata (AA), though the exact interactions of stress with AA remain undefined. Corticotropin-releasing hormone (CRH), the proximal regulator of the stress axis, has been recognized as an immunomodulatory factor in tissues and peripheral blood mononuclear cells (PBMCs). We used multicolour flow cytometry to identify receptor CRHR1 expression on PBMC subsets in AA patients (n = 54) and controls (n = 66). We found that CRHR1 was primarily expressed by circulating monocytes. CRHR1 expression on monocytes was enhanced in AA compared with controls (3.17% vs. 1.44%, p = 0.002, chi-squared test). AA incidence was correlated to elevated CD14+ monocyte numbers (R = 0.092, p = 0.036) and markedly independently correlated with increased CRHR1 expression (R = 0.215, p = 0.027). High CRHR1 expression was significantly related to chronic AA (disease duration >1 year; p = 0.003, chi-squared test), and large lesion area (AA area >25%; p = 0.049, chi-squared test). We also observed enhanced percentages of active monocytes and reduced CD16+ CD3- NK cell numbers in AA patients' PBMCs (p = 0.010; 0.025, respectively). In vitro CRH treatment of PBMCs and human monocyte cell line THP-1 promoted CD86 upregulation. The findings imply that stress-related factors CRH and CRHR1 contribute to AA development and progression where higher CRHR1 expression is associated with chronic AA and larger lesions.


Assuntos
Alopecia em Áreas , Hormônio Liberador da Corticotropina , Monócitos , Receptores de Hormônio Liberador da Corticotropina , Humanos , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Monócitos/metabolismo , Adulto , Masculino , Feminino , Pessoa de Meia-Idade , Alopecia em Áreas/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Adulto Jovem , Estudos de Casos e Controles , Citometria de Fluxo , Receptores de IgG/metabolismo , Células Matadoras Naturais/metabolismo
3.
Biol Sex Differ ; 15(1): 75, 2024 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-39327618

RESUMO

BACKGROUND: Adolescent stress and alcohol exposure increase the risk of maladaptive behaviors and mental disorders in adulthood, with distinct sex-specific differences. Understanding the mechanisms underlying these early events is crucial for developing targeted prevention and treatment strategies. METHODS: Male and female Wistar rats were exposed to acute restraint stress and intermittent alcohol during adolescence. We assessed lasting effects on plasma corticosterone (CORT) and adrenocorticotropic hormone (ACTH) levels, and mRNA expression of genes related to corticotropin releasing hormone (CRH), neuropeptide Y (NPY), corticoid, opioid, and arginine vasopressin systems in the amygdala and hypothalamus. RESULTS: The main findings are as follows: (1) blood alcohol concentrations (BAC) increased after the final alcohol administration, but stressed males had lower BAC than non-stressed males; (2) Males gained significantly more weight than females; (3) Stressed females showed higher ACTH levels than non-stressed females, with no changes in males; (4) Stress increased CORT levels in males, while stressed, alcohol-treated females had lower CORT levels than non-stressed females; (5) CRH: Females had lower Crhr1 levels in the amygdala, while alcohol reduced Crhr2 levels in males but not females. Significant interactions among sex, stress, and alcohol were found in the hypothalamus, with distinct patterns between sexes; (6) NPY: In the amygdala, stress reduced Npy and Npy1r levels in males but increased them in females. Alcohol decreased Npy2r levels in males, with varied effects in females. Similar sex-specific patterns were observed in the hypothalamus; (7) Corticoid system: Stress and alcohol had complex, sex-dependent effects on Pomc, Nr3c1, and Nr3c2 in both brain regions; (8) Opioid receptors: Stress and alcohol blunted the elevated expression of Oprm1, Oprd1, and Oprk1 in the amygdala of males and the hypothalamus of females; (8) Vasopressin: Stress and alcohol interacted significantly to affect Avp and Avpr1a expression in the amygdala, with stronger effects in females. In the hypothalamus, alcohol increased Avp levels in females. CONCLUSIONS: This study demonstrates that adolescent acute stress and alcohol exposure induce lasting, sex-specific alterations in systems involved in reward and stress responses. These findings emphasize the importance of considering sex differences in the prevention and management of HPA dysfunction and psychiatric disorders.


Assuntos
Etanol , RNA Mensageiro , Ratos Wistar , Recompensa , Caracteres Sexuais , Estresse Psicológico , Animais , Masculino , Feminino , Estresse Psicológico/metabolismo , RNA Mensageiro/metabolismo , Etanol/administração & dosagem , Etanol/farmacologia , Encéfalo/metabolismo , Encéfalo/efeitos dos fármacos , Neuropeptídeo Y/metabolismo , Neuropeptídeo Y/genética , Corticosterona/sangue , Hormônio Adrenocorticotrópico/sangue , Ratos , Transdução de Sinais , Hormônio Liberador da Corticotropina/metabolismo , Hormônio Liberador da Corticotropina/genética
4.
J Affect Disord ; 367: 148-156, 2024 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-39233237

RESUMO

BACKGROUND: Many patients with bipolar disorder (BD) do not respond to or have difficulties tolerating lithium and/or other mood stabilizing agents. There is a need for personalized treatments based on biomarkers in guiding treatment options. The calcium voltage-gated channel CACNA1C is a promising candidate for developing personalized treatments. CACNA1C is implicated in BD by genome-wide association studies and several lines of evidence suggest that targeting L-type calcium channels could be an effective treatment strategy. However, before such individualized treatments can be pursued, biomarkers predicting treatment response need to be developed. METHODS: As a first step in testing the hypothesis that CACNA1C genotype is associated with serum levels of CACNA1C, we conducted ELISA measures on serum samples from 100 subjects with BD and 100 control subjects. RESULTS: We observed significantly higher CACNA1C (p < 0.01) protein levels in subjects with BD. The risk single nucleotide polymorpshism (SNP) (rs11062170) showed functional significance as subjects homozygous for the risk allele (CC) had significantly greater CACNA1C protein levels compared to subjects with one (p = 0.013) or no copies (p = 0.009). We observed higher somatostatin (SST) (p < 0.003) protein levels and lower levels of the clock protein aryl hydrocarbon receptor nuclear translocator-like (ARTNL) (p < 0.03) and stress signaling factor corticotrophin releasing hormone (CRH) (p < 0.001) in BD. SST and period 2 (PER2) protein levels were associated with both alcohol dependence and lithium response. CONCLUSIONS: Our findings represent the first evidence for increased serum levels of CACNA1C in BD. Along with altered levels of SST, ARNTL, and CRH our findings suggest CACNA1C is associated with circadian rhythm and stress response disturbances in BD.


Assuntos
Transtorno Bipolar , Canais de Cálcio Tipo L , Ritmo Circadiano , Polimorfismo de Nucleotídeo Único , Humanos , Transtorno Bipolar/sangue , Transtorno Bipolar/genética , Canais de Cálcio Tipo L/genética , Feminino , Masculino , Adulto , Pessoa de Meia-Idade , Ritmo Circadiano/fisiologia , Genótipo , Hormônio Liberador da Corticotropina/sangue , Hormônio Liberador da Corticotropina/genética , Fatores de Transcrição ARNTL/genética , Fatores de Transcrição ARNTL/sangue , Biomarcadores/sangue , Estudos de Casos e Controles , Somatostatina/sangue , Alelos
5.
NPJ Biofilms Microbiomes ; 10(1): 93, 2024 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-39349483

RESUMO

Stress can lead to gut dysbiosis in brain-gut axis disordered diseases as irritable bowel syndrome (IBS), yet the mechanisms how stress transfer from the brain to the gut and disrupt gut microbiota remain elusive. Here we describe a stress-responsive brain-to-gut axis which impairs colonocytes' mitochondria to trigger gut dysbiosis. Patients with IBS exhibit significantly increased facultative anaerobes and decreased obligate anaerobes, related to increased serum corticotropin-releasing hormone (CRH) level and defected colonocytes' mitochondria ultrastructure. Mice exposed to acute stress experienced enhanced CRH-CRH receptor type 1 (CRHR1) signaling, which impaired mitochondria and epithelium hypoxia in the colon, subsequently triggered gut dysbiosis. Antagonizing CRHR1 expression to inhibit cAMP/Ras/MAPK signaling or activating mitochondria respiration conferred resilience against stress-induced mitochondria damaging and epithelium hypoxia impairment, ultimately improving gut dysbiosis. These results suggest that the CRH-CRHR1-mitochondria pathway plays a pivotal role in stress-induced gut dysbiosis that could be therapeutically targeted for stress-induced gastrointestinal diseases. Yiming Zhang et.al report that psychological stress activated Corticotropin-releasing hormone (CRH)-CRH receptor type 1 (CRHR1)-mitochondria pathway to trigger gut dysbiosis and reveal CRHR1 upregulation damages mitochondria via cAMP/Ras/MAPK signaling in colonocytes.


Assuntos
Hormônio Liberador da Corticotropina , Disbiose , Microbioma Gastrointestinal , Mitocôndrias , Receptores de Hormônio Liberador da Corticotropina , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Animais , Mitocôndrias/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Camundongos , Humanos , Transdução de Sinais , Síndrome do Intestino Irritável/metabolismo , Síndrome do Intestino Irritável/microbiologia , Colo/microbiologia , Colo/patologia , Estresse Psicológico , Eixo Encéfalo-Intestino/fisiologia
6.
CNS Neurosci Ther ; 30(9): e70046, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39295107

RESUMO

BACKGROUND: Drug addiction, characterized by compulsive drug use and high relapse rates, arises from complex interactions between reward and aversion systems in the brain. The paraventricular nucleus (PVN), located in the anterior hypothalamus, serves as a neuroendocrine center and is a key component of the hypothalamic-pituitary-adrenal axis. OBJECTIVE: This review aimed to explore how the PVN impacts reward and aversion in drug addiction through stress responses and emotional regulation and to evaluate the potential of PVN as a therapeutic target for drug addiction. METHODS: We review the current literature, focusing on three main neuron types in the PVN-corticotropin-releasing factor, oxytocin, and arginine vasopressin neurons-as well as other related neurons, to understand their roles in modulating addiction. RESULTS: Existing studies highlight the PVN as a key mediator in addiction, playing a dual role in reward and aversion systems. These findings are crucial for understanding addiction mechanisms and developing targeted therapies. CONCLUSION: The role of PVN in stress response and emotional regulation suggests its potential as a therapeutic target in drug addiction, offering new insights for addiction treatment.


Assuntos
Núcleo Hipotalâmico Paraventricular , Recompensa , Transtornos Relacionados ao Uso de Substâncias , Núcleo Hipotalâmico Paraventricular/metabolismo , Núcleo Hipotalâmico Paraventricular/efeitos dos fármacos , Humanos , Animais , Transtornos Relacionados ao Uso de Substâncias/psicologia , Transtornos Relacionados ao Uso de Substâncias/metabolismo , Transtornos Relacionados ao Uso de Substâncias/fisiopatologia , Hormônio Liberador da Corticotropina/metabolismo , Comportamento Aditivo/psicologia , Estresse Psicológico/psicologia , Estresse Psicológico/metabolismo , Aprendizagem da Esquiva/fisiologia , Aprendizagem da Esquiva/efeitos dos fármacos , Ocitocina/metabolismo
7.
Obesity (Silver Spring) ; 32(10): 1885-1896, 2024 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-39315404

RESUMO

OBJECTIVE: Here, we aimed to investigate the role of glucocorticoid and mineralocorticoid receptors (GRs and MRs, respectively) in the regulation of energy homeostasis. METHODS: We used three mouse models with simultaneous deletion of GRs and MRs in either forebrain neurons, the paraventricular nucleus, or corticotropin-releasing hormone (CRH) neurons and compared them with wild-type controls or isolated knockout groups. In addition to body weight, food intake, energy expenditure, insulin sensitivity, fat/lean mass distribution, and plasma corticosterone levels, we also performed transcriptomic analysis of CRH neurons and assessed their response to melanocortinergic stimulation. RESULTS: Similar to global double-knockout models, deletion of GRs and MRs specifically in mature CRH neurons resulted in obesity. Importantly, the latter was accompanied by insulin resistance, but not increased plasma corticosterone levels. Transcriptomic analysis of these neurons revealed upregulation of several genes involved in postsynaptic signal transduction, including the Ptk2b gene, which encodes proline-rich tyrosine kinase 2. Knockout of both nuclear receptors leads to upregulation of Ptk2b in CRH neurons, which results in their diminished responsiveness to melanocortinergic stimulation. CONCLUSIONS: Our data demonstrate the functional redundancy of GRs and MRs in CRH neurons to maintain energy homeostasis and prevent obesity. Simultaneous targeting of both receptors might represent an unprecedented approach to counteract obesity.


Assuntos
Corticosterona , Hormônio Liberador da Corticotropina , Metabolismo Energético , Camundongos Knockout , Neurônios , Obesidade , Receptores de Glucocorticoides , Receptores de Mineralocorticoides , Animais , Masculino , Camundongos , Corticosterona/sangue , Corticosterona/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Resistência à Insulina , Camundongos Endogâmicos C57BL , Neurônios/metabolismo , Obesidade/metabolismo , Núcleo Hipotalâmico Paraventricular/metabolismo , Receptores de Glucocorticoides/metabolismo , Receptores de Glucocorticoides/genética , Receptores de Mineralocorticoides/metabolismo , Receptores de Mineralocorticoides/genética , Feminino
8.
Behav Brain Res ; 475: 115223, 2024 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-39214423

RESUMO

Excessive stress can exceed the adjustment ability of body and cause injury and dysfunction, while elucidation of the mechanism and prevention measures of stress-related injury are still insufficient. The present study was to observe the effect of glucocorticoid (GC) on stress-induced hypothalamic nerve injury and elucidate the potential mechanism. The present study intended to establish a chronic restraint stress rat model for follow-up study. Open field test and elevated plus maze test were used to observe behavioral changes of stress rats; Enzyme-linked immunosorbent assay (ELISA) was used to detect changes in the levels of hypothalamus-pituitary-adrenal (HPA) axis-related hormones and inflammatory factors in hypothalamus; toluidine blue staining was used to observe pathological changes of hypothalamus. The results showed that stress rats showed obvious anxiety-like behaviors, the levels of HPA axis-related hormones and inflammatory factors showed abnormal fluctuations, and morphological results showed significant nerve injury in hypothalamus. Low-dose GC treatment significantly improved behavioral changes, alleviated hypothalamic nerve injury, and restored hypothalamic levels of inflammatory factors, serum levels of GC, corticotropin-releasing hormone (CRH), and adrenocorticotropic hormone (ACTH) and GC level in adrenal cortex of stressed rats, while GC receptor (GR) inhibitor, CRH receptor inhibitor, and adrenalectomy reversed the ameliorative effects of low-dose GC. Our study clarified that low-dose GC can restore stress coping ability by reshaping the homeostasis of the HPA axis, thus alleviating behavioral abnormalities and hypothalamic nerve injury in stressed rats.


Assuntos
Hormônio Adrenocorticotrópico , Glucocorticoides , Homeostase , Sistema Hipotálamo-Hipofisário , Hipotálamo , Sistema Hipófise-Suprarrenal , Ratos Sprague-Dawley , Estresse Psicológico , Animais , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/efeitos dos fármacos , Sistema Hipófise-Suprarrenal/metabolismo , Masculino , Ratos , Glucocorticoides/farmacologia , Estresse Psicológico/tratamento farmacológico , Estresse Psicológico/metabolismo , Homeostase/efeitos dos fármacos , Homeostase/fisiologia , Hormônio Adrenocorticotrópico/sangue , Hipotálamo/efeitos dos fármacos , Hipotálamo/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Hormônio Liberador da Corticotropina/farmacologia , Modelos Animais de Doenças , Comportamento Animal/efeitos dos fármacos
9.
Molecules ; 29(15)2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-39125051

RESUMO

Corticotropin-releasing factor (CRF) is a key neuropeptide hormone that is secreted from the hypothalamus. It is the master hormone of the HPA axis, which orchestrates the physiological and behavioral responses to stress. Many disorders, including anxiety, depression, addiction relapse, and others, are related to over-activation of this system. Thus, new molecules that may interfere with CRF receptor binding may be of value to treat neuropsychiatric stress-related disorders. Also, CRF1R antagonists have recently emerged as potential treatment options for congenital adrenal hyperplasia. Previously, several series of CRF1 receptor antagonists were developed by our group. In continuation of our efforts in this direction, herein we report the synthesis and biological evaluation of a new series of CRF1R antagonists. Representative compounds were evaluated for their binding affinities compared to antalarmin. Four compounds (2, 5, 20, and 21) showed log IC50 values of -8.22, -7.95, -8.04, and -7.88, respectively, compared to -7.78 for antalarmin. This result indicates that these four compounds are superior to antalarmin by 2.5, 1.4, 1.7, and 1.25 times, respectively. It is worth mentioning that compound 2, in terms of IC50, is among the best CRF1R antagonists ever developed in the last 40 years. The in silico physicochemical properties of the lead compounds showed good drug-like properties. Thus, further research in this direction may lead to better and safer CRF receptor antagonists that may have clinical applications, particularly for stress-related disorders and the treatment of congenital adrenal hyperplasia.


Assuntos
Hiperplasia Suprarrenal Congênita , Desenho de Fármacos , Pirimidinas , Receptores de Hormônio Liberador da Corticotropina , Receptores de Hormônio Liberador da Corticotropina/antagonistas & inibidores , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Pirimidinas/química , Pirimidinas/farmacologia , Pirimidinas/síntese química , Humanos , Hiperplasia Suprarrenal Congênita/tratamento farmacológico , Hiperplasia Suprarrenal Congênita/metabolismo , Pirróis/química , Pirróis/síntese química , Pirróis/farmacologia , Hormônio Liberador da Corticotropina/metabolismo , Estresse Psicológico/tratamento farmacológico , Simulação de Acoplamento Molecular
10.
J Neuroimmunol ; 394: 578419, 2024 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-39088908

RESUMO

Oxytocin can regulate immunological activity directly or indirectly; however, immunological functions and mechanisms of oxytocin actions under chronic stress like cesarean delivery (CD) are poorly understood. Our study found that abnormal oxytocin production and secretion in CD rats caused atrophy of thymic tissues. Neurotoxin kainic acid microinjected into the dorsolateral supraoptic nucleus in male rats selectively reduced hypothalamic oxytocin levels, increased corticotrophin-releasing hormone and plasma interleukin-1ß while reducing plasma oxytocin, thyroxine and testosterone levels and causing atrophy of immune tissues. Thus, plasma oxytocin is essential for immunological homeostasis, which involves oxytocin facilitation of thyroid hormone and sex steroid secretion.


Assuntos
Sistema Hipotálamo-Hipofisário , Ocitocina , Animais , Ocitocina/sangue , Ocitocina/metabolismo , Ratos , Masculino , Sistema Hipotálamo-Hipofisário/efeitos dos fármacos , Sistema Hipotálamo-Hipofisário/metabolismo , Feminino , Ratos Sprague-Dawley , Ácido Caínico/toxicidade , Interleucina-1beta/metabolismo , Interleucina-1beta/sangue , Testosterona/sangue , Hormônio Liberador da Corticotropina/metabolismo , Timo/efeitos dos fármacos , Timo/metabolismo
11.
J Neurophysiol ; 132(2): 403-417, 2024 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-39106208

RESUMO

Cholinergic interneurons (ChIs) provide the main source of acetylcholine in the striatum and have emerged as a critical modulator of behavioral flexibility, motivation, and associative learning. In the dorsal striatum (DS), ChIs display heterogeneous firing patterns. Here, we investigated the spontaneous firing patterns of ChIs in the nucleus accumbens (NAc) shell, a region of the ventral striatum. We identified four distinct ChI firing signatures: regular single-spiking, irregular single-spiking, rhythmic bursting, and a mixed-mode pattern composed of bursting activity and regular single spiking. ChIs from females had lower firing rates compared with males and had both a higher proportion of mixed-mode firing patterns and a lower proportion of regular single-spiking neurons compared with males. We further observed that across the estrous cycle, the diestrus phase was characterized by higher proportions of irregular ChI firing patterns compared with other phases. Using pooled data from males and females, we examined how the stress-associated neuropeptide corticotropin releasing factor (CRF) impacts these firing patterns. ChI firing patterns showed differential sensitivity to CRF. This translated into differential ChI sensitivity to CRF across the estrous cycle. Furthermore, CRF shifted the proportion of ChI firing patterns toward more regular spiking activity over bursting patterns. Finally, we found that repeated stressor exposure altered ChI firing patterns and sensitivity to CRF in the NAc core, but not the NAc shell. These findings highlight the heterogeneous nature of ChI firing patterns, which may have implications for accumbal-dependent motivated behaviors.NEW & NOTEWORTHY Cholinergic interneurons (ChIs) within the dorsal and ventral striatum can exert a major influence on network output and motivated behaviors. However, the firing patterns and neuromodulation of ChIs within the ventral striatum, specifically the nucleus accumbens (NAc) shell, are understudied. Here, we report that NAc shell ChIs have heterogeneous ChI firing patterns that are labile and can be modulated by the stress-linked neuropeptide corticotropin releasing factor (CRF) and by the estrous cycle.


Assuntos
Neurônios Colinérgicos , Hormônio Liberador da Corticotropina , Interneurônios , Núcleo Accumbens , Animais , Hormônio Liberador da Corticotropina/metabolismo , Hormônio Liberador da Corticotropina/farmacologia , Feminino , Masculino , Interneurônios/fisiologia , Interneurônios/metabolismo , Núcleo Accumbens/fisiologia , Núcleo Accumbens/metabolismo , Núcleo Accumbens/citologia , Neurônios Colinérgicos/fisiologia , Neurônios Colinérgicos/metabolismo , Ciclo Estral/fisiologia , Potenciais de Ação/fisiologia , Camundongos
12.
Cell Biol Int ; 48(11): 1743-1754, 2024 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-39090812

RESUMO

Corticotropin-releasing hormone (CRH) has been well documented playing a role in the regulation of cellular processes, immune responses, and inflammatory processes that can influence the occurrence and development of tumors. Supervillin (SVIL) is a membrane-associated and actin-binding protein, which is actively involved in the proliferation, spread, and migration of cancer cells. This work investigated CRH's influence on bladder cancer cells' migration and relevant mechanisms. By using human bladder cancer cells T24 and RT4 in wound healing experiments and transwell assay, we found that the migration ability of the T24 cells was significantly increased after CRH treatment. In vivo experiments showed that CRH significantly promoted the metastases of T24 cells in cell line-derived xenograft (CDX) mouse model. Interestingly, downregulation of SVIL by SVIL-specifc small hairpin RNAs significantly reduced the promoting effect of CRH on bladder cancer cell migration. Furthermore, CRH significantly increased SVIL messenger RNA and protein expression in T24 cells, accompanied with AKT and ERK phosphorylation in T24 cells. Pretreatment with AKT inhibitor (MK2206) blocked the CRH-induced SVIL expression and ERK phosphorylation. Also, inhibition of ERK signaling pathway by U0126 significantly reduced the CRH-induced SVIL expression and AKT phosphorylation. It suggested that cross-talking between AKT and ERK pathways was involved in the effect of CRH on SVIL. Taken together, we demonstrated that CRH induced migration of bladder cancer cells, in which AKT and ERK pathways -SVIL played a key role.


Assuntos
Movimento Celular , Hormônio Liberador da Corticotropina , Sistema de Sinalização das MAP Quinases , Proteínas dos Microfilamentos , Proteínas Proto-Oncogênicas c-akt , Neoplasias da Bexiga Urinária , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia , Humanos , Movimento Celular/efeitos dos fármacos , Animais , Proteínas Proto-Oncogênicas c-akt/metabolismo , Linhagem Celular Tumoral , Camundongos , Hormônio Liberador da Corticotropina/metabolismo , Hormônio Liberador da Corticotropina/farmacologia , Proteínas dos Microfilamentos/metabolismo , Proteínas dos Microfilamentos/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Proteínas de Membrana/metabolismo , Proteínas de Membrana/genética , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Compostos Heterocíclicos com 3 Anéis
13.
Neurobiol Dis ; 200: 106649, 2024 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-39187210

RESUMO

Chronic predator stress (CPS) is an important and ecologically relevant tool for inducing anhedonia in animals, but the neural circuits underlying the associated neurobiological changes remain to be identified. Using cell-type-specific manipulations, we found that corticotropin-releasing hormone (CRH) neurons in the medial subthalamic nucleus (mSTN) enhance struggle behaviors in inescapable situations and lead to anhedonia, predominately through projections to the external globus pallidus (GPe). Recordings of in vivo neuronal activity revealed that CPS distorted mSTN-CRH neuronal responsivity to negative and positive stimuli, which may underlie CPS-induced behavioral despair and anhedonia. Furthermore, we discovered presynaptic inputs from the bed nucleus of the stria terminalis (BNST) to mSTN-CRH neurons projecting to the GPe that were enhanced following CPS, and these inputs may mediate such behaviors. This study identifies a neurocircuitry that co-regulates escape response and anhedonia in response to predator stress. This new understanding of the neural basis of defensive behavior in response to predator stress will likely benefit our understanding of neuropsychiatric diseases.


Assuntos
Anedonia , Hormônio Liberador da Corticotropina , Neurônios , Estresse Psicológico , Núcleo Subtalâmico , Animais , Hormônio Liberador da Corticotropina/metabolismo , Estresse Psicológico/fisiopatologia , Estresse Psicológico/metabolismo , Neurônios/fisiologia , Núcleo Subtalâmico/fisiologia , Anedonia/fisiologia , Camundongos , Masculino , Camundongos Endogâmicos C57BL , Reação de Fuga/fisiologia , Vias Neurais/fisiologia , Núcleos Septais/fisiologia , Núcleos Septais/metabolismo , Globo Pálido/fisiologia
14.
Eur J Neurosci ; 60(5): 4937-4953, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39080914

RESUMO

Cholinergic interneurons (ChIs) act as master regulators of striatal output, finely tuning neurotransmission to control motivated behaviours. ChIs are a cellular target of many peptide and hormonal neuromodulators, including corticotropin-releasing factor, opioids, insulin and leptin, which can influence an animal's behaviour by signalling stress, pleasure, pain and nutritional status. However, little is known about how sex hormones via estrogen receptors influence the function of these other neuromodulators. Here, we performed in situ hybridisation on mouse striatal tissue to characterise the effect of sex and sex hormones on choline acetyltransferase (Chat), estrogen receptor alpha (Esr1) and corticotropin-releasing factor type 1 receptor (Crhr1) expression. Although we did not detect sex differences in ChAT protein levels in the dorsal striatum or nucleus accumbens, we found that female mice have more Chat mRNA-expressing neurons than males in both the dorsal striatum and nucleus accumbens. At the population level, we observed a sexually dimorphic distribution of Esr1- and Crhr1-expressing ChIs in the ventral striatum that was negatively correlated in intact females, which was abolished by ovariectomy and not present in males. Only in the NAc did we find a significant population of ChIs that co-express Crhr1 and Esr1 in females and to a lesser extent in males. At the cellular level, Crhr1 and Esr1 transcript levels were negatively correlated only during the estrus phase in females, indicating that changes in sex hormone levels can modulate the interaction between Crhr1 and Esr1 mRNA levels.


Assuntos
Neurônios Colinérgicos , Hormônio Liberador da Corticotropina , Receptor alfa de Estrogênio , Estrogênios , Interneurônios , Núcleo Accumbens , Receptores de Hormônio Liberador da Corticotropina , Animais , Masculino , Núcleo Accumbens/metabolismo , Feminino , Hormônio Liberador da Corticotropina/metabolismo , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Interneurônios/metabolismo , Receptor alfa de Estrogênio/metabolismo , Camundongos , Neurônios Colinérgicos/metabolismo , Estrogênios/metabolismo , Caracteres Sexuais , Camundongos Endogâmicos C57BL , Colina O-Acetiltransferase/metabolismo , Ovariectomia
15.
Brain Res ; 1842: 149112, 2024 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-38969083

RESUMO

It has been reported that the clinical symptoms of functional dyspepsia (FD) exacerbate upon stress while the gender-related factors have been incompletely understood. This study aims to investigate the role of sex in chronic heterotypic stress (CHS)-induced autonomic and gastric motor dysfunction. For CHS, the rats were exposed to the combination of different stressors for 7 consecutive days. Subsequently, electrocardiography was recorded in anesthetized rats to evaluate heart rate variability (HRV) for the determination of autonomic outflow and sympathovagal balance. Solid gastric emptying (GE) was measured in control and CHS-loaded male and female rats. The immunoreactivities of catecholaminergic cell marker tyrosine hydroxylase (TH), choline acetyltransferase (ChAT), corticotropin releasing factor (CRF), and estrogen receptor (ER-α/ß) were evaluated in medullary and pontine brainstem sections by immunohistochemistry. Compared with the controls, CHS significantly delayed GE in males but not in females. There was no significant sex-related difference in parasympathetic indicator HF under either control or CHS conditions. Sympathetic indicator LF was significantly higher in control females compared to the males. The higher sympathetic output in females was found to be attenuated upon CHS; in contrast, the elevated sympathetic output was detected in CHS-loaded males. No sex- or stress-related effect was observed on ChAT immunoreactivity in the dorsal motor nucleus of N.vagus (DMV). In males, greater number of TH-ir cells was observed in the caudal locus coeruleus (LC), while they were more densely detected in the rostral LC of females. Regardless of sex, CHS elevated immunoreactivity of TH throughout the LC. Under basal conditions, greater number of TH-ir cells was detected in the rostral ventrolateral medulla (RVLM) of females. In contrast, CHS remarkably increased the number of TH-ir cells in the RVLM of males which was found to be decreased in females. There was no sex-related alteration in TH immunoreactivity in the nucleus tractus solitarius (NTS) of control rats, while CHS affected both sexes in a similar manner. Compared with females, CRF immunoreactivity was prominently observed in control males, while both of which were stimulated by CHS. ER-α/ß was found to be co-expressed with TH in the NTS and LC which exhibit no alteration related to either sex or stress status. These results indicate a sexual dimorphism in the catecholaminergic and the CRF system in brainstem which might be involved in the CHS-induced autonomic and visceral dysfunction occurred in males.


Assuntos
Ratos Sprague-Dawley , Caracteres Sexuais , Estresse Psicológico , Animais , Masculino , Feminino , Estresse Psicológico/metabolismo , Estresse Psicológico/fisiopatologia , Ratos , Rombencéfalo/metabolismo , Motilidade Gastrointestinal/fisiologia , Catecolaminas/metabolismo , Tirosina 3-Mono-Oxigenase/metabolismo , Sistema Nervoso Autônomo/fisiopatologia , Sistema Nervoso Autônomo/metabolismo , Frequência Cardíaca/fisiologia , Hormônio Liberador da Corticotropina/metabolismo , Esvaziamento Gástrico/fisiologia , Colina O-Acetiltransferase/metabolismo
17.
Nat Commun ; 15(1): 5522, 2024 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-38951506

RESUMO

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.


Assuntos
Ansiedade , Núcleo Central da Amígdala , Hormônio Liberador da Corticotropina , Medo , Neurônios , Reflexo de Sobressalto , Animais , Hormônio Liberador da Corticotropina/metabolismo , Medo/fisiologia , Neurônios/metabolismo , Neurônios/fisiologia , Camundongos , Feminino , Masculino , Ansiedade/fisiopatologia , Núcleo Central da Amígdala/metabolismo , Reflexo de Sobressalto/fisiologia , Camundongos Endogâmicos C57BL , Comportamento Animal/fisiologia , Estresse Psicológico
18.
Behav Brain Res ; 472: 115139, 2024 08 24.
Artigo em Inglês | MEDLINE | ID: mdl-38969017

RESUMO

Numerous studies have demonstrated that chronic stress during pregnancy (CSDP) can induce depression and hippocampal damage in offspring. It has also been observed that high levels of corticotropin-releasing hormone (CRH) can damage hippocampal neurons, and intraperitoneal injection of a corticotropin releasing hormone receptor 1 (CRHR1) antagonist decreases depression-like behavior and hippocampal neuronal damage in a mouse depression model. However, whether CSDP causes hippocampal damage and depression in offspring through the interaction of CRH and hippocampal CRHR1 remains unknown and warrants further investigation. Therefore, hippocampal Crhr1 conditional gene knockout mice and C57/BL6J mice were used to study these questions. Depression-related indexs in male offspring mice were examined using the forced swim test (FST), sucrose preference test (SPT), tail suspension test (TST) and open field test (OFT). Serum CRH levels were measured by enzyme-linked immunosorbent assay (ELISA). Golgi-Cox staining was used to examine the morphological changes of hippocampal neuronal dendrites. Neuronal apoptosis in the hippocampal CA3 regions was detected by terminal deoxynucleotidy transferase dUTP nick end labeling (TUNEL) staining. The levels of mammalian target of rapamycin (mTOR), phosphorylated mTOR (p-mTOR) and protein kinase B (AKT) proteins were measured by Western blot analysis. This study showed that CSDP induces depression-like behavior, hippocampal neuronal dendrite damage and apoptosis in male offspring mice. Conditional gene knockout of hippocampal Crhr1 in mice reduced CSDP-induced depression-like behavior, hippocampal neuronal dendrite damage and apoptosis in male offspring, and counteracted the CSDP-induced decreased expression of p-Akt and mTOR activity in male offspring hippocampus. These findings demonstrated that CSDP might inhibit the Akt/mTOR pathway by increasing the levels of CRH, leading to increased CRH-mediated activation of hippocampal CRHR1, thereby inducing synaptic impairment and apoptosis in hippocampal neurons, which in turn leads to depression-like behavior in offspring.


Assuntos
Hormônio Liberador da Corticotropina , Depressão , Hipocampo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Efeitos Tardios da Exposição Pré-Natal , Receptores de Hormônio Liberador da Corticotropina , Estresse Psicológico , Animais , Receptores de Hormônio Liberador da Corticotropina/metabolismo , Feminino , Masculino , Hipocampo/metabolismo , Hipocampo/patologia , Gravidez , Estresse Psicológico/metabolismo , Depressão/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Apoptose/fisiologia , Modelos Animais de Doenças , Comportamento Animal/fisiologia , Serina-Treonina Quinases TOR/metabolismo
19.
Stress ; 27(1): 2377272, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-39020286

RESUMO

Aberrant functioning of the hypothalamic-pituitary-adrenal (HPA) axis is a hallmark of conditions such as depression, anxiety disorders, and post-traumatic stress disorder. Early-life adversity and genetic variation can interaction to disrupt HPA axis regulation, potentially contributing to certain forms of psychopathology. This study employs a rhesus macaque model to investigate how early parental neglect interacts with a single nucleotide polymorphism within the promoter region of the corticotropin-releasing hormone (CRH-248) gene, impacting the development of the HPA axis. For the initial six months of life, 307 rhesus monkey infants (n = 146 females, n = 161 males) were either reared with their mothers (MR) in conditions emulating the natural environment (control group) or raised without maternal care in groups with constant or 3-hours daily access to same-aged peers (NR). Blood samples collected on days 30, 60, 90, and 120 of life under stressful conditions were assayed for plasma cortisol and adrenocorticotropic hormone (ACTH) concentrations. Findings revealed that NR subjects exhibited a significant blunting of both ACTH and cortisol concentrations. Notably, there was a gene-by-environment interaction observed for ACTH and cortisol levels, with NR subjects with the polymorphism displaying higher ACTH concentrations and lower cortisol concentrations. To the extent that these results generalize to humans, they suggest that early parental neglect may render individuals vulnerable to HPA axis dysfunction, a susceptibility that is modulated by CRH-248 genotype-a gene-by-environment interaction that leaves a lasting developmental signature.


Assuntos
Hormônio Liberador da Corticotropina , Hidrocortisona , Sistema Hipotálamo-Hipofisário , Macaca mulatta , Sistema Hipófise-Suprarrenal , Polimorfismo de Nucleotídeo Único , Regiões Promotoras Genéticas , Animais , Sistema Hipotálamo-Hipofisário/metabolismo , Feminino , Hormônio Liberador da Corticotropina/genética , Masculino , Hidrocortisona/sangue , Genótipo , Estresse Psicológico/genética , Interação Gene-Ambiente , Privação Materna , Hormônio Adrenocorticotrópico/sangue
20.
Int J Mol Sci ; 25(13)2024 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-39000501

RESUMO

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.


Assuntos
Doenças Cardiovasculares , Sistema Hipotálamo-Hipofisário , Doenças Metabólicas , Sistema Hipófise-Suprarrenal , Vasopressinas , Humanos , Sistema Hipotálamo-Hipofisário/metabolismo , Sistema Hipófise-Suprarrenal/metabolismo , Vasopressinas/metabolismo , Doenças Cardiovasculares/metabolismo , Animais , Doenças Metabólicas/metabolismo , Hormônio Liberador da Corticotropina/metabolismo , Hormônio Adrenocorticotrópico/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA