Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.284
Filter
1.
Nat Commun ; 15(1): 8575, 2024 Oct 03.
Article in English | MEDLINE | ID: mdl-39362860

ABSTRACT

Dysregulation of brain homeostasis is associated with neuropsychiatric conditions such as major depressive disorder. However, underlying neural-circuit mechanisms remain not well-understood. We show in mice that chronic restraint stress (CRS) and social defeat stress (SDS) are both associated with disruption of excitation (E)-inhibition (I) balance, with increased E/I ratios, in medial preoptic area (MPOA) circuits, but through affecting different neuronal types. CRS results in elevated activity in glutamatergic neurons, and their suppression mitigates CRS-induced depressive-like behaviors. Paraventricular hypothalamic input to these neurons contributes to induction but not expression of depressive-like behaviors. Their projections to ventral tegmental area and periaqueductal gray/dorsal raphe suppress midbrain dopaminergic and serotonergic activity, respectively, and mediate expression of divergent depressive-like symptoms. By contrast, SDS results in reduced activity of GABAergic neurons, and their activation alleviates SDS-induced depressive-like behaviors. Thus, E/I imbalance with relatively increased excitation in MPOA circuits may be a general mechanism underlying depression caused by different etiological factors.


Subject(s)
Depression , GABAergic Neurons , Preoptic Area , Stress, Psychological , Animals , Preoptic Area/metabolism , Stress, Psychological/complications , Stress, Psychological/physiopathology , Depression/etiology , Depression/metabolism , Depression/physiopathology , Mice , Male , GABAergic Neurons/metabolism , Mice, Inbred C57BL , Ventral Tegmental Area/physiopathology , Social Defeat , Behavior, Animal , Neurons/metabolism , Restraint, Physical , Disease Models, Animal , Neural Pathways/physiopathology , Paraventricular Hypothalamic Nucleus/metabolism
2.
PeerJ ; 12: e18166, 2024.
Article in English | MEDLINE | ID: mdl-39346076

ABSTRACT

Background: Fibrosis after nephrotoxic injury is common. Activation of the paraventricular nucleus (PVN) renin-angiotensin system (RAS) and sympathetic nervous system (SNS) are common mechanism of renal fibrosis. However, there have limited knowledge about which brain regions are most affected by Angiotensin II (Ang II) after nephrotoxic injury, what role does Angiotensin II type 1a receptors (AT1R) signaling play and how this affects the outcomes of the kidneys. Methods: In nephrotoxic folic acid-induced chronic kidney disease (FA-CKD) mouse models, we have integrated retrograde tracer techniques with studies on AT1afl/fl mice to pinpoint an excessively active central pathway that connects the paraventricular nucleus (PVN) to the rostral ventrolateral medulla (RVLM). This pathway plays a pivotal role in determining the kidney's fibrotic response following injury induced by folic acid. Results: FA-CKD (vs sham) had increased in the kidney SNS activity and Ang II expression in the central PVN. The activation of Ang II in the PVN triggers the activation of the PVN-RVLM pathway, amplifies SNS output, thus facilitating fibrosis development in FA-CKD mouse. Blocking sympathetic traffic or deleting AT1a in the PVN alleviated renal fibrosis in FA-CKD mice. Conclusions: The FA-CKD mice have increased the expression of Ang II in PVN, thereby activating AT1a-positive PVN neurons project to the RVLM, where SNS activity is engaged to initiate fibrotic processes. The Ang II in PVN may contribute to the development of kidney fibrosis after nephrotoxic folic acid-induced kidney injury.


Subject(s)
Disease Models, Animal , Fibrosis , Folic Acid , Kidney , Receptor, Angiotensin, Type 1 , Renal Insufficiency, Chronic , Sympathetic Nervous System , Animals , Receptor, Angiotensin, Type 1/metabolism , Receptor, Angiotensin, Type 1/genetics , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/chemically induced , Renal Insufficiency, Chronic/metabolism , Mice , Folic Acid/pharmacology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/pathology , Kidney/pathology , Kidney/drug effects , Kidney/metabolism , Kidney/innervation , Angiotensin II/pharmacology , Male , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/pathology , Mice, Inbred C57BL
3.
J Headache Pain ; 25(1): 152, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39289629

ABSTRACT

BACKGROUND: Migraine is among the most prevalent and burdensome neurological disorders in the United States based on disability-adjusted life years. Cortical spreading depolarization (SD) is the most likely electrophysiological cause of migraine aura and may be linked to trigeminal nociception. We previously demonstrated, using a minimally invasive optogenetic approach of SD induction (opto-SD), that opto-SD triggers acute periorbital mechanical allodynia that is reversed by 5HT1B/1D receptor agonists, supporting SD-induced activation of migraine-relevant trigeminal pain pathways in mice. Recent data highlight hypothalamic neural circuits in migraine, and SD may activate hypothalamic neurons. Furthermore, neuroanatomical, electrophysiological, and behavioral data suggest a homeostatic analgesic function of hypothalamic neuropeptide hormone, oxytocin. We, therefore, examined the role of hypothalamic paraventricular nucleus (PVN) and oxytocinergic (OXT) signaling in opto-SD-induced trigeminal pain behavior. METHODS: We induced a single opto-SD in adult male and female Thy1-ChR2-YFP transgenic mice and quantified fos immunolabeling in the PVN and supraoptic nucleus (SON) compared with sham controls. Oxytocin expression was also measured in fos-positive neurons in the PVN. Periorbital mechanical allodynia was tested after treatment with selective OXT receptor antagonist L-368,899 (5 to 25 mg/kg i.p.) or vehicle at 1, 2, and 4 h after opto-SD or sham stimulation using von Frey monofilaments. RESULTS: Opto-SD significantly increased the number of fos immunoreactive cells in the PVN and SON as compared to sham stimulation (p < 0.001, p = 0.018, respectively). A subpopulation of fos-positive neurons also stained positive for oxytocin. Opto-SD evoked periorbital mechanical allodynia 1 h after SD (p = 0.001 vs. sham), which recovered quickly within 2 h (p = 0.638). OXT receptor antagonist L-368,899 dose-dependently prolonged SD-induced periorbital allodynia (p < 0.001). L-368,899 did not affect mechanical thresholds in the absence of opto-SD. CONCLUSIONS: These data support an SD-induced activation of PVN neurons and a role for endogenous OXT in alleviating acute SD-induced trigeminal allodynia by shortening its duration.


Subject(s)
Hyperalgesia , Mice, Transgenic , Oxytocin , Animals , Oxytocin/metabolism , Male , Female , Mice , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Cortical Spreading Depression/physiology , Cortical Spreading Depression/drug effects , Receptors, Oxytocin/metabolism , Supraoptic Nucleus/metabolism , Supraoptic Nucleus/drug effects , Disease Models, Animal , Camphanes , Piperazines
4.
Front Endocrinol (Lausanne) ; 15: 1449326, 2024.
Article in English | MEDLINE | ID: mdl-39286269

ABSTRACT

Background: The part played by oxytocin and oxytocin neurons in the regulation of food intake is controversial. There is much pharmacological data to support a role for oxytocin notably in regulating sugar consumption, however, several recent experiments have questioned the importance of oxytocin neurons themselves. Methods: Here we use a combination of histological and chemogenetic techniques to investigate the selective activation or inhibition of oxytocin neurons in the hypothalamic paraventricular nucleus (OxtPVH). We then identify a pathway from OxtPVH neurons to the bed nucleus of the stria terminalis using the cell-selective expression of channel rhodopsin. Results: OxtPVH neurons increase their expression of cFos after both physiological (fast-induced re-feeding or oral lipid) and pharmacological (systemic administration of cholecystokinin or lithium chloride) anorectic signals. Chemogenetic activation of OxtPVH neurons is sufficient to decrease free-feeding in Oxt Cre:hM3Dq mice, while inhibition in Oxt Cre:hM4Di mice attenuates the response to administration of cholecystokinin. Activation of OxtPVH neurons also increases energy expenditure and core-body temperature, without a significant effect on locomotor activity. Finally, the selective, optogenetic stimulation of a pathway from OxtPVH neurons to the bed nucleus of the stria terminalis reduces the consumption of sucrose. Conclusion: Our results support a role for oxytocin neurons in the regulation of whole-body metabolism, including a modulatory action on food intake and energy expenditure. Furthermore, we demonstrate that the pathway from OxtPVH neurons to the bed nucleus of the stria terminalis can regulate sugar consumption.


Subject(s)
Energy Intake , Energy Metabolism , Neurons , Oxytocin , Paraventricular Hypothalamic Nucleus , Septal Nuclei , Animals , Oxytocin/pharmacology , Oxytocin/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Septal Nuclei/metabolism , Septal Nuclei/physiology , Mice , Neurons/metabolism , Male , Sucrose/pharmacology , Mice, Transgenic , Mice, Inbred C57BL , Eating/physiology
5.
CNS Neurosci Ther ; 30(9): e70046, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39295107

ABSTRACT

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.


Subject(s)
Paraventricular Hypothalamic Nucleus , Reward , Substance-Related Disorders , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Humans , Animals , Substance-Related Disorders/psychology , Substance-Related Disorders/metabolism , Substance-Related Disorders/physiopathology , Corticotropin-Releasing Hormone/metabolism , Behavior, Addictive/psychology , Stress, Psychological/psychology , Stress, Psychological/metabolism , Avoidance Learning/physiology , Avoidance Learning/drug effects , Oxytocin/metabolism
6.
Immunity ; 57(9): 2000-2002, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39260350

ABSTRACT

Our brain is not an immune-privileged island isolated from peripheries, but how non-neuronal brain cells interact with the peripheral system is not well understood. Wei et al. report that microglia in the hypothalamic paraventricular nucleus (PVN) with unique vasculature can detect ATP derived from hemodynamic disturbance. These microglia in the PVN regulate the response to hypertension via ATP-P2Y12-C/EBPß signaling.


Subject(s)
Blood Pressure , Brain , Microglia , Paraventricular Hypothalamic Nucleus , Microglia/immunology , Microglia/physiology , Microglia/metabolism , Animals , Humans , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/immunology , Paraventricular Hypothalamic Nucleus/physiology , Blood Pressure/physiology , Brain/immunology , Adenosine Triphosphate/metabolism , Signal Transduction , Hypertension/immunology , Hypertension/physiopathology , CCAAT-Enhancer-Binding Protein-beta/metabolism
7.
Commun Biol ; 7(1): 1193, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333722

ABSTRACT

Relationships between adult females are fundamental to understanding diversity in animal social systems. While cooperative relationships between kin are known to promote fitness benefits, the proximate mechanisms underlying this are not well understood. Here we show that when related female house mice (Mus musculus domesticus) cooperate to rear young communally, those with higher endogenous oxytocin levels have more egalitarian and successful cooperative relationships. Sisters with higher oxytocin concentrations in the paraventricular nucleus (PVN) of the hypothalamus weaned significantly more offspring, had lower reproductive skew and spent more equal proportions of time in the nest. By contrast, PVN oxytocin was unrelated to the number of weaned offspring produced in the absence of cooperation, and did not vary in response to manipulation of nest site availability or social cues of outgroup competition. By linking fitness consequences of cooperation with oxytocin, our findings have broad implications for understanding the evolution of egalitarian social relationships.


Subject(s)
Cooperative Behavior , Oxytocin , Social Behavior , Animals , Oxytocin/metabolism , Mice , Female , Paraventricular Hypothalamic Nucleus/metabolism , Reproduction , Behavior, Animal
8.
Obesity (Silver Spring) ; 32(10): 1885-1896, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39315404

ABSTRACT

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.


Subject(s)
Corticosterone , Corticotropin-Releasing Hormone , Energy Metabolism , Mice, Knockout , Neurons , Obesity , Receptors, Glucocorticoid , Receptors, Mineralocorticoid , Animals , Male , Mice , Corticosterone/blood , Corticosterone/metabolism , Corticotropin-Releasing Hormone/metabolism , Insulin Resistance , Mice, Inbred C57BL , Neurons/metabolism , Obesity/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/genetics , Receptors, Mineralocorticoid/metabolism , Receptors, Mineralocorticoid/genetics , Female
9.
Clin Exp Hypertens ; 46(1): 2402260, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-39305040

ABSTRACT

BACKGROUND: Gestational diabetes can lead to increased blood pressure in offspring, accompanied by impaired renal sodium excretion function and vasoconstriction and diastole dysfunction. However, there are few studies on whether it is accompanied by increased sympathetic nerve activity. METHODS: Pregnant C57BL/6 mice were intraperitoneally injected with streptozotocin (35 mg/kg) or citrate buffer at day 0 of gestation. The mice of control mother offspring (CMO) and diabetic mother offspring (DMO) at 16 weeks of age were infused with vehicle (artificial cerebrospinal fluid, aCSF, 0.4 µL/h) or tempol (1 mmol/L, 0.4 µL/h) into the bilateral paraventricular nucleus (PVN) of mice for 4 weeks, respectively. RESULTS: Compared with CMO group, SBP and peripheral sympathetic nerve activity (increased heart rate, LF/HF and plasma norepinephrine and decreased SDNN and RMSSD) were increased in DMO group, which was accompanied by increased angiotensin II type-1 receptor (AT1R) expression and function in PVN. The increase in AT1R expression levels was attributed to a decrease in the methylation level of the AT1R promoter region, resulting in an increase in AT1R mRNA levels in PVN of DMO. Moreover, compared with CMO group, the levels of oxidative stress were increased and DNMT1 expression was decreased in PVN of DMO. Bilateral PVN infusion of tempol attenuated oxidative stress increased the level of DNMT1 expression and the binding of DNMT1 to the AT1R promoter region, which reduced mRNA and protein expression level of AT1R, heart rate and SBP in DMO, but not in CMO. CONCLUSIONS: The present study provides evidence for overactive sympathetic nervous systems in the pathogenesis of gestational diabetes-induced hypertension in offspring. Central antioxidant intervention in the PVN may be an important treatment strategy for fetal-programmed hypertension.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes, Gestational , Hypertension , Mice, Inbred C57BL , Sympathetic Nervous System , Animals , Pregnancy , Sympathetic Nervous System/physiopathology , Female , Mice , Diabetes, Gestational/physiopathology , Hypertension/physiopathology , Hypertension/etiology , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Experimental/complications , Cyclic N-Oxides/pharmacology , Prenatal Exposure Delayed Effects/physiopathology , Spin Labels , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/physiopathology , Blood Pressure/physiology , Receptor, Angiotensin, Type 1/genetics , Male , Heart Rate/physiology , Oxidative Stress
10.
Genes Brain Behav ; 23(5): e12907, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39246030

ABSTRACT

Avian brood parasitism is an evolutionarily derived behavior for which the neurobiological mechanisms are mostly unexplored. We aimed to identify brain regions that have diverged in the brood-parasitic brain using relative transcript abundance of social neuropeptides and receptors. We compared behavioral responses and transcript abundance in three brain regions in the brown-headed cowbird (BHCO), a brood parasite, and a closely related parental species, the red-winged blackbird (RWBL). Females of both species were treated with mesotocin (MT; avian homolog of oxytocin) or saline prior to exposure to nest stimuli. Results reveal that MT promotes approach toward nests with eggs rather than nests with begging nestlings in both species. We also examined relative transcript abundance of the five social neuropeptides and receptors in the brain regions examined: preoptic area (POA), paraventricular nucleus (PVN) and bed nucleus of the stria terminalis (BST). We found that MT-treated cowbirds but not blackbirds exhibited lower transcript abundance for two receptors, corticotropin-releasing factor 2 (CRFR2) and prolactin receptor (PRLR) in BST. Additionally, MT-treated cowbirds had higher PRLR in POA, comparable to those found in blackbirds, regardless of treatment. No other transcripts of interest exhibited significant differences as a result of MT treatment, but we found a significant effect of species in the three regions. Together, these results indicate that POA, PVN, and BST represent neural nodes that have diverged in avian brood parasites and may serve as neural substrates of brood-parasitic behavior.


Subject(s)
Nesting Behavior , Oxytocin , Animals , Oxytocin/metabolism , Oxytocin/genetics , Oxytocin/pharmacology , Oxytocin/analogs & derivatives , Female , Songbirds/genetics , Brain/metabolism , Species Specificity , Paraventricular Hypothalamic Nucleus/metabolism , Septal Nuclei/metabolism , Preoptic Area/metabolism
11.
Immunity ; 57(9): 2030-2042.e8, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39116878

ABSTRACT

Hypertension is usually accompanied by elevated sympathetic tonicity, but how sympathetic hyperactivity is triggered is not clear. Recent advances revealed that microglia-centered neuroinflammation contributes to sympathetic excitation in hypertension. In this study, we performed a temporospatial analysis of microglia at both morphological and transcriptomic levels and found that microglia in the hypothalamic paraventricular nucleus (PVN), a sympathetic center, were early responders to hypertensive challenges. Vasculature analyses revealed that the PVN was characterized by high capillary density, thin vessel diameter, and complex vascular topology relative to other brain regions. As such, the PVN was susceptible to the penetration of ATP released from the vasculature in response to hemodynamic disturbance after blood pressure increase. Mechanistically, ATP ligation to microglial P2Y12 receptor was responsible for microglial inflammatory activation and the eventual sympathetic overflow. Together, these findings identified a distinct vasculature pattern rendering vulnerability of PVN pre-sympathetic neurons to hypertension-associated microglia-mediated inflammatory insults.


Subject(s)
Hemodynamics , Hypertension , Microglia , Paraventricular Hypothalamic Nucleus , Sympathetic Nervous System , Paraventricular Hypothalamic Nucleus/metabolism , Animals , Microglia/metabolism , Hypertension/physiopathology , Mice , Sympathetic Nervous System/physiopathology , Male , Mice, Inbred C57BL , Adenosine Triphosphate/metabolism , Receptors, Purinergic P2Y12/metabolism , Inflammation/immunology , Blood Pressure , Neurons/metabolism
12.
Behav Brain Res ; 474: 115175, 2024 10 02.
Article in English | MEDLINE | ID: mdl-39098399

ABSTRACT

Stress-related disorders are becoming increasingly common and are often associated with cognitive impairments. Within this context, the endocannabinoid (ECB) system, particularly the type 1 cannabinoid (CB1) receptor, seems to play a decisive role in restoring body homeostasis. There is consistent evidence in the literature that disrupted CB1-mediated neurotransmission can ultimately contribute to stress-related diseases. Therefore, the present study aimed to evaluate the participation of CB1 receptors in the integrity of stress-induced peripheral and behavioral responses. For this purpose, male adult Wistar rats underwent physical restraint (1 h/day, for 7 days), followed by a single administration of rimonabant (CB1 receptor antagonist, 3 mg/Kg, intraperitonial) at the end of stress protocol. Animals were then subjected to evaluation of neuroendocrine responses, behavioral tests and quantification of Iba-1 (microglial) immunoreactivity in the parvocellular subdivisions of the paraventricular nucleus of the hypothalamus (PVN). No effects of restraint stress or rimonabant administration were detected on body mass variation. However, stress significantly increased adrenal relative mass and corticosterone secretion, and reduced thymus relative size. The stress effects on adrenal size and corticosterone plasma levels were absent in rimonabant-treated rats, but the thymus size was further reduced in the restraint-rimonabant group. Restraint stress also induced anhedonia, a depression-like behavior, and reduced object recognition index, indicating memory recovery impairment. Treatment with the CB1 antagonist significantly reversed stress-induced anhedonia and memory deficit. In the PVN, restraint stress reduced the number of Iba-1 positive cells in the medial parvocellular region of vehicle- but not rimonabant-treated animals. Taken together, these results indicate that the acute inhibition of the CB1-mediated endogenous pathway restores stress-induced depression-like behaviors and memory loss, suggesting a role for endocannabinoids in the neuro-immune-endocrine interplay at both peripheral and hypothalamic levels.


Subject(s)
Anhedonia , Cannabinoid Receptor Antagonists , Corticosterone , Memory Disorders , Rats, Wistar , Receptor, Cannabinoid, CB1 , Restraint, Physical , Rimonabant , Stress, Psychological , Animals , Rimonabant/pharmacology , Male , Stress, Psychological/metabolism , Anhedonia/drug effects , Anhedonia/physiology , Rats , Memory Disorders/drug therapy , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Corticosterone/blood , Cannabinoid Receptor Antagonists/pharmacology , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Disease Models, Animal , Behavior, Animal/drug effects , Piperidines/pharmacology , Pyrazoles/pharmacology
13.
Nat Commun ; 15(1): 6941, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138162

ABSTRACT

Glucagon-like peptide 1 (GLP-1) stimulates insulin secretion and holds significant pharmacological potential. Nevertheless, the regulation of energy homeostasis by centrally-produced GLP-1 remains partially understood. Preproglucagon cells, known to release GLP-1, are found in the olfactory bulb (OB). We show that activating GLP-1 receptors (GLP-1R) in the OB stimulates insulin secretion in response to oral glucose in lean and diet-induced obese male mice. This is associated with reduced noradrenaline content in the pancreas and blocked by an α2-adrenergic receptor agonist, implicating functional involvement of the sympathetic nervous system (SNS). Inhibiting GABAA receptors in the paraventricular nucleus of the hypothalamus (PVN), the control centre of the SNS, abolishes the enhancing effect on insulin secretion induced by OB GLP-1R. Therefore, OB GLP-1-dependent regulation of insulin secretion relies on a relay within the PVN. This study provides evidence that OB GLP-1 signalling engages a top-down neural mechanism to control insulin secretion via the SNS.


Subject(s)
Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Insulin Secretion , Mice, Inbred C57BL , Olfactory Bulb , Paraventricular Hypothalamic Nucleus , Animals , Glucagon-Like Peptide 1/metabolism , Male , Olfactory Bulb/metabolism , Olfactory Bulb/drug effects , Insulin Secretion/drug effects , Glucagon-Like Peptide-1 Receptor/metabolism , Mice , Paraventricular Hypothalamic Nucleus/metabolism , Insulin/metabolism , Obesity/metabolism , Sympathetic Nervous System/metabolism , Neurons/metabolism , Signal Transduction , Norepinephrine/metabolism , Glucose/metabolism
14.
Sci Rep ; 14(1): 20043, 2024 08 29.
Article in English | MEDLINE | ID: mdl-39209992

ABSTRACT

Within the brain, the connections between neurons are constantly changing in response to environmental stimuli. A prime environmental regulator of neuronal activity is diet, and previous work has highlighted changes in hypothalamic connections in response to diets high in dietary fat and elevated sucrose. We sought to determine if the change in hypothalamic neuronal connections was driven primarily by an elevation in dietary fat alone. Analysis was performed in both male and female animals. We measured Agouti-related peptide (AgRP) neuropeptide and Synaptophysin markers in the paraventricular nucleus of the hypothalamus (PVH) in response to an acute 48 h high fat diet challenge. Using two image analysis methods described in previous studies, an effect of a high fat diet on AgRP neuronal projections in the PVH of male or female mice was not identified. These results suggest that it may not be dietary fat alone that is responsible for the previously published alterations in hypothalamic connections. Future work should focus on deciphering the role of individual macronutrients on neuroanatomical and functional changes.


Subject(s)
Agouti-Related Protein , Diet, High-Fat , Paraventricular Hypothalamic Nucleus , Animals , Agouti-Related Protein/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Male , Female , Mice , Diet, High-Fat/adverse effects , Dietary Fats/pharmacology , Dietary Fats/metabolism , Neurons/metabolism , Mice, Inbred C57BL , Synaptophysin/metabolism
15.
Nutrients ; 16(16)2024 Aug 06.
Article in English | MEDLINE | ID: mdl-39203718

ABSTRACT

BACKGROUND: Puerarin is an isoflavone compound isolated from the roots of a leguminous plant, the wild kudzu. Various functional activities of this compound in multiple diseases have been reported. However, the effect and mechanism of puerarin in improving blood pressure remain non-elucidated. PURPOSE: The current study was designed to assess the preventive effects of puerarin on the onset and progression of hypertension and to verify the hypothesis that puerarin alleviates blood pressure by inhibiting the ROS/TLR4/NLRP3 inflammasome signaling pathway in the hypothalamic paraventricular nucleus (PVN) of salt-induced prehypertensive rats. METHODS: Male Dahl salt-sensitive rats were fed low NaCl salt (3% in drinking water) for the control (NS) group or 8% (HS) to induce prehypertension. Each batch was divided into two group and treated by bilateral PVN microinjection with either artificial cerebrospinal fluid or puerarin through a micro-osmotic pump for 6 weeks. The mean arterial pressure (MAP) was recorded, and samples were collected and analyzed. RESULTS: We concluded that puerarin significantly prevented the elevation of blood pressure and effectively alleviated the increase in heart rate caused by high salt. Norepinephrine (NE) in the plasma of salt-induced prehypertensive rats also decreased upon puerarin chronic infusion. Additionally, analysis of the PVN sample revealed that puerarin pretreatment decreased the positive cells and gene level of TLR4 (Toll-like receptor 4), NLRP3, Caspase-1 p10, NOX2, MyD88, NOX4, and proinflammatory cytokines in the PVN. Puerarin pretreatment also decreased NF-κBp65 activity, inhibited oxidative stress, and alleviated inflammatory responses in the PVN. CONCLUSION: We conclude that puerarin alleviated blood pressure via inhibition of the ROS/TLR4/NLRP3 inflammasome signaling pathway in the PVN, suggesting the therapeutic potential of puerarin in the prevention of hypertension.


Subject(s)
Blood Pressure , Inflammasomes , Isoflavones , NLR Family, Pyrin Domain-Containing 3 Protein , Paraventricular Hypothalamic Nucleus , Reactive Oxygen Species , Signal Transduction , Toll-Like Receptor 4 , Animals , Male , Rats , Blood Pressure/drug effects , Disease Models, Animal , Hypertension/chemically induced , Hypertension/drug therapy , Inflammasomes/metabolism , Inflammasomes/drug effects , Isoflavones/pharmacology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Prehypertension/drug therapy , Rats, Inbred Dahl , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Sodium Chloride, Dietary , Toll-Like Receptor 4/metabolism
16.
Neurochem Int ; 179: 105825, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39097233

ABSTRACT

There is evidence that tumor necrosis factor alpha (TNFα) influences autonomic processes coordinated within the hypothalamic paraventricular nucleus (PVN), however, the signaling mechanisms subserving TNFα's actions in this brain area are unclear. In non-neuronal cell types, TNFα has been shown to play an important role in canonical NADPH oxidase (NOX2)-mediated production of reactive oxygen species (ROS), molecules also known to be critically involved in hypertension. However, little is known about the role of TNFα in NOX2-dependent ROS production in the PVN within the context of hypertension. Using dual labeling immunoelectron microscopy and dihydroethidium (DHE) microfluorography, we provide structural and functional evidence for interactions between TNFα and NOX2 in the PVN. The TNFα type 1 receptor (TNFR1), the major mediator of TNFα signaling in the PVN, was commonly co-localized with the catalytic gp91phox subunit of NOX2 in postsynaptic sites of PVN neurons. Additionally, there was an increase in dual labeled dendritic profiles following fourteen-day slow-pressor angiotensin II (AngII) infusion. Using DHE microfluorography, it was also shown that TNFα application resulted in a NOX2-dependent increase in ROS in isolated PVN neurons projecting to the spinal cord. Further, TNFα-mediated ROS production was heightened after AngII infusion. The finding that TNFR1 and gp91phox are positioned for rapid interactions, particularly in PVN-spinal cord projection neurons, provides a molecular substrate by which inflammatory signaling and oxidative stress may jointly contribute to AngII hypertension.


Subject(s)
Angiotensin II , NADPH Oxidase 2 , Neurons , Paraventricular Hypothalamic Nucleus , Rats, Sprague-Dawley , Reactive Oxygen Species , Tumor Necrosis Factor-alpha , Animals , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Reactive Oxygen Species/metabolism , Angiotensin II/pharmacology , Angiotensin II/metabolism , NADPH Oxidase 2/metabolism , Tumor Necrosis Factor-alpha/metabolism , Neurons/metabolism , Neurons/drug effects , Male , NADPH Oxidases/metabolism , Rats , Membrane Glycoproteins/metabolism
17.
Neuropharmacology ; 260: 110129, 2024 Dec 01.
Article in English | MEDLINE | ID: mdl-39179173

ABSTRACT

Hyperactivity of the hypothalamic-pituitary-adrenal (HPA) axis during chronic stress is essential for the pathogenesis of depression, and increased activity of cAMP response element binding protein (CREB)-regulated transcription co-activator 1 (CRTC1) in the paraventricular nucleus (PVN) plays a critical role. As a well-investigated microRNA (miRNA), miR-184 has two forms, miR-184-3p and miR-184-5p. Recently, miRNAs target genes predictive analysis and dual-luciferase reporter assays identified an inhibitory role of miR-184-3p on CRTC1 expression. Therefore, we speculated that miR-184-3p regulation was responsible for the effects of chronic stress on CRTC1 in the PVN. Various methods, including the chronic social defeat stress (CSDS) model of depression, behavioral tests, Western blotting, co-immunoprecipitation (Co-IP), quantitative real-time reverse transcription PCR (qRT-PCR), immunofluorescence, and adeno-associated virus (AAV)-mediated gene transfer, were used. CSDS evidently downregulated the level of miR-184-3p, but not miR-184-5p, in the PVN. Genetic knockdown and pharmacological inhibition of miR-184-3p in the PVN induced various depressive-like symptoms (e.g., abnormal behaviors, HPA hyperactivity, enhanced CRTC1 function in PVN neurons, downregulation of hippocampal neurogenesis, and decreased brain-derived neurotrophic factor (BDNF) signaling) in naïve male C57BL/6J mice. In contrast, genetic overexpression and pharmacological activation of miR-184-3p in the PVN produced significant beneficial effects against CSDS. MiR-184-3p in the PVN was necessary for the antidepressant actions of two well-known SSRIs, fluoxetine and paroxetine. Collectively. miR-184-3p was also implicated in the neurobiology of depression and may be a viable target for novel antidepressants.


Subject(s)
Depression , Hypothalamo-Hypophyseal System , Mice, Inbred C57BL , MicroRNAs , Paraventricular Hypothalamic Nucleus , Pituitary-Adrenal System , Stress, Psychological , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , Paraventricular Hypothalamic Nucleus/metabolism , Male , Mice , Hypothalamo-Hypophyseal System/metabolism , Depression/metabolism , Depression/genetics , Pituitary-Adrenal System/metabolism , Stress, Psychological/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Social Defeat
18.
Toxicology ; 508: 153935, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39182713

ABSTRACT

Polychlorinated biphenyls (PCBs) are industrial pollutants that act as endocrine disruptors and alter thyroid function. However, it is still unclear whether PCBs can affect hypothalamic thyrotropin releasing hormone (Trh) mRNA expression through TH signaling disruption. As salt-loading dehydration induces tertiary hypothyroidism in the hypothalamic parvocellular paraventricular nuclei (paPVN), and perinatal exposure to Aroclor 1254 (A1254) disrupts the hydric balance in rats, we hypothesized that TRH synthesis could be altered during dehydration in TRH neurons that control the hypothalamic-pituitary-thyroid (HPT) axis activity in rats perinatally exposed to A1254. We examined Trh mRNA expression in the paPVN and the response to salt-loading dehydration (hyperosmotic (hyper) stress) in the progeny of Wistar pregnant rats receiving 0 mg/kg BW (control) or 30 mg/kg BW A1254 daily from gestational days 10-19. Three-month-old offspring were subjected to normosmotic or hyper conditions and Trh mRNA, glucocorticoid receptor (GR) mRNA expression were measured in the PVN by RT-PCR. TRH mRNA and TRH+ neurons were measured in the paPVN by fluorescent in situ hybridization (FISH). As expected, Trh mRNA levels were decreased in the paPVN of male and female rats in the hyper group. Basal Trh mRNA expression and serum TSH were decreased in male rats in the A1254 group. Notably, Trh mRNA levels were further decreased in the paPVN of male and female A1254 + hyper rats, in which the GR mRNA expression was significantly decreased. These results support the hypothesis that perinatal exposure to A1254 results in inadequate adaptive response of the HPT axis in adulthood and contributes to dysregulation of the HPT axis response to salt-loading dehydration.


Subject(s)
Paraventricular Hypothalamic Nucleus , Prenatal Exposure Delayed Effects , RNA, Messenger , Rats, Wistar , Thyrotropin-Releasing Hormone , Animals , Female , Thyrotropin-Releasing Hormone/metabolism , Thyrotropin-Releasing Hormone/genetics , Male , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Pregnancy , RNA, Messenger/metabolism , RNA, Messenger/genetics , Prenatal Exposure Delayed Effects/chemically induced , Rats , Receptors, Glucocorticoid/metabolism , Receptors, Glucocorticoid/genetics , Endocrine Disruptors/toxicity
19.
Neurosci Lett ; 841: 137948, 2024 Oct 15.
Article in English | MEDLINE | ID: mdl-39179131

ABSTRACT

The paraventricular hypothalamic nucleus (PVH) is an important neuroendocrine center involved in pain regulation, but the nociceptive afferent routes for the nucleus are still unclear. We examined the profile of PVH receiving injurious information by a combination of retrograde tracing with Fluoro-Gold (FG) and FOS expression induced by formalin stimuli. The result showed that formalin injection induced significantly increased expression of FOS in the PVH, among which oxytocin containing neurons are one neuronal phenotype. Immunofluorescent staining of FG and FOS revealed that double labeled neurons were strikingly distributed in the area 2 of the cingulate cortex (Cg2), the lateral septal nucleus (LS), the periaqueductal gray (PAG), the posterior hypothalamic area (PH), and the lateral parabrachial nucleus (LPB). In the five regions, LPB had the biggest number and the highest ratio of FOS expression in FG labeled neurons, with main subnuclei distribution in the external, superior, dorsal, and central parts. Further immunofluorescent triple staining disclosed that about one third of FG and FOS double labeled neurons in the LPB were immunoreactive for calcitonin gene related peptide (CGRP). In conclusion, the present study demonstrates the nociceptive input profile of the PVH area under inflammatory pain and suggests that neurons in the LPB may play essential roles in transmitting noxious information to the PVH.


Subject(s)
Formaldehyde , Paraventricular Hypothalamic Nucleus , Animals , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Formaldehyde/toxicity , Male , Calcitonin Gene-Related Peptide/metabolism , Mice , Nociception/drug effects , Nociception/physiology , Proto-Oncogene Proteins c-fos/metabolism , Neurons/metabolism , Neurons/drug effects , Oxytocin/metabolism , Pain/metabolism , Pain/chemically induced , Parabrachial Nucleus/metabolism , Parabrachial Nucleus/drug effects
20.
J Cardiovasc Pharmacol ; 84(4): 468-478, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39115898

ABSTRACT

ABSTRACT: The hypothalamic paraventricular nucleus (PVN) plays a central role in regulating cardiovascular activity and blood pressure. We administered hydroxylamine hydrochloride (HA), a cystathionine-ß-synthase inhibitor, into the PVN to suppress endogenous hydrogen sulfide and investigate its effects on the mitogen-activated protein kinase (MAPK) pathway in high salt (HS)-induced hypertension. We randomly divided 40 male Dahl salt-sensitive rats into 4 groups: the normal salt (NS) + PVN vehicle group, the NS + PVN HA group, the HS + PVN vehicle group, and the HS + PVN HA group, with 10 rats in each group. The rats in the NS groups were fed a NS diet containing 0.3% NaCl, while the HS groups were fed a HS diet containing 8% NaCl. The mean arterial pressure was calculated after noninvasive measurement using an automatic sphygmomanometer to occlude the tail cuff once a week. HA or vehicle was infused into the bilateral PVN using Alzet osmotic mini pumps for 6 weeks after the hypertension model was successfully established. We measured the levels of H 2 S in the PVN and plasma norepinephrine using enzyme linked immunosorbent assay. In addition, we assessed the parameters of the MAPK pathway, inflammation, and oxidative stress through western blotting, immunohistochemical analysis, or real-time polymerase chain reaction. In this study, we discovered that decreased levels of endogenous hydrogen sulfide in the PVN contributed to the onset of HS-induced hypertension. This was linked to the activation of the MAPK signaling pathway, proinflammatory cytokines, and oxidative stress in the PVN, as well as the activation of the sympathetic nervous system.


Subject(s)
Disease Models, Animal , Hydrogen Sulfide , Hypertension , Paraventricular Hypothalamic Nucleus , Rats, Inbred Dahl , Sodium Chloride, Dietary , Animals , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/enzymology , Paraventricular Hypothalamic Nucleus/physiopathology , Male , Hydrogen Sulfide/metabolism , Hypertension/chemically induced , Hypertension/physiopathology , Hypertension/metabolism , Norepinephrine/metabolism , Hydroxylamine/pharmacology , Arterial Pressure/drug effects , MAP Kinase Signaling System/drug effects , Rats , Oxidative Stress/drug effects , Enzyme Inhibitors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL