Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.378
Filter
1.
PLoS One ; 19(8): e0300081, 2024.
Article in English | MEDLINE | ID: mdl-39163355

ABSTRACT

Pain is a major non-motor symptom of Parkinson's disease (PD). Alterations in the descending pain inhibitory system (DPIS) have been reported to trigger hyperalgesia in PD patients. However, the underlying mechanisms remain unclear. In the current study, dopaminergic nigrostriatal lesions were induced in rats by injecting 6-hydroxydopamine (6-OHDA) into their medial forebrain bundle. The neural mechanisms underlying changes in nociception in the orofacial region of 6-OHDA-lesioned rats was examined by injecting formalin into the vibrissa pad. The 6-OHDA-lesioned rats were seen to exhibit increased frequency of face-rubbing and more c-Fos immunoreactive (c-Fos-IR) cells in the trigeminal spinal subnucleus caudalis (Vc), confirming hyperalgesia. Examination of the number of c-Fos-IR cells in the DPIS nuclei [including the midbrain ventrolateral periaqueductal gray, the locus coeruleus, the nucleus raphe magnus, and paraventricular nucleus (PVN)] showed that 6-OHDA-lesioned rats exhibited a significantly lower number of c-Fos-IR cells in the magnocellular division of the PVN (mPVN) after formalin injection compared to sham-operated rats. Moreover, the 6-OHDA-lesioned rats also exhibited significantly lower plasma oxytocin (OT) concentration and percentage of oxytocin-immunoreactive (OT-IR) neurons expressing c-Fos protein in the mPVN and dorsal parvocellular division of the PVN (dpPVN), which secrete the analgesic hormone OT upon activation by nociceptive stimuli, when compared to the sham-operated rats. The effect of OT on hyperalgesia in 6-OHDA-lesioned rats was examined by injecting formalin into the vibrissa pad after intracisternal administration of OT, and the findings showed a decrease in the frequency of face rubbing and the number of c-Fos-IR cells in the Vc. In conclusion, these findings confirm presence of hyperalgesia in PD rats, potentially due to suppression of the analgesic effects of OT originating from the PVN.


Subject(s)
Disease Models, Animal , Hyperalgesia , Oxidopamine , Oxytocin , Parkinson Disease , Proto-Oncogene Proteins c-fos , Animals , Hyperalgesia/metabolism , Hyperalgesia/drug therapy , Oxytocin/pharmacology , Rats , Male , Proto-Oncogene Proteins c-fos/metabolism , Parkinson Disease/metabolism , Parkinson Disease/drug therapy , Rats, Sprague-Dawley , Analgesics/pharmacology , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/drug effects
2.
Mol Brain ; 17(1): 49, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090731

ABSTRACT

Neurexin-3 (Nrxn3) has been genetically associated with obesity, but the underlying neural mechanisms remain poorly understood. This study aimed to investigate the role of Nrxn3 in the paraventricular nucleus of the hypothalamus (PVN) in regulating energy balance and glucose homeostasis. We found that Nrxn3 expression in the PVN was upregulated in response to metabolic stressors, including cold exposure and fasting. Using Cre-loxP technology, we selectively ablated Nrxn3 in CaMKIIα-expressing neurons of the PVN in male mice. This genetic manipulation resulted in marked weight gain attributable to increased adiposity and impaired glucose tolerance, without affecting food intake. Our findings identify PVN CaMKIIα-expressing neurons as a critical locus where Nrxn3 modulates energy balance by regulating adipogenesis and glucose metabolism, independently of appetite. These results reveal a novel neural mechanism potentially linking Nrxn3 dysfunction to obesity pathogenesis, suggesting that targeting PVN Nrxn3-dependent neural pathways may inform new therapeutic approaches for obesity prevention and treatment.


Subject(s)
Body Weight , Eating , Glucose , Homeostasis , Nerve Tissue Proteins , Paraventricular Hypothalamic Nucleus , Animals , Male , Mice , Cell Adhesion Molecules, Neuronal/metabolism , Eating/physiology , Energy Metabolism , Glucose/metabolism , Mice, Inbred C57BL , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism
3.
Life Sci Alliance ; 7(10)2024 Oct.
Article in English | MEDLINE | ID: mdl-39107065

ABSTRACT

The brain controls energy homeostasis by regulating food intake through signaling within the melanocortin system. Whilst we understand the role of the hypothalamus within this system, how extra-hypothalamic brain regions are involved in controlling energy balance remains unclear. Here we show that the melanocortin 3 receptor (MC3R) is expressed in the paraventricular nucleus of the thalamus (PVT). We tested whether fasting would change the activity of MC3R neurons in this region by assessing the levels of c-Fos and pCREB as neuronal activity markers. We determined that overnight fasting causes a significant reduction in pCREB levels within PVT-MC3R neurons. We then questioned whether perturbation of MC3R signaling, during fasting, would result in altered refeeding. Using chemogenetic approaches, we show that modulation of MC3R activity, during the fasting period, does not impact body weight regain or total food intake in the refeeding period. However, we did observe significant differences in the pattern of feeding-related behavior. These findings suggest that the PVT is a region where MC3R neurons respond to energy deprivation and modulate refeeding behavior.


Subject(s)
Fasting , Neurons , Paraventricular Hypothalamic Nucleus , Receptor, Melanocortin, Type 3 , Animals , Fasting/physiology , Neurons/metabolism , Neurons/physiology , Receptor, Melanocortin, Type 3/metabolism , Receptor, Melanocortin, Type 3/genetics , Mice , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/physiology , Male , Feeding Behavior/physiology , Eating/physiology , Midline Thalamic Nuclei/physiology , Midline Thalamic Nuclei/metabolism , Energy Metabolism , Mice, Inbred C57BL , Signal Transduction
4.
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
5.
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
6.
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
7.
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
8.
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
9.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39000096

ABSTRACT

The arginine vasopressin (AVP)-magnocellular neurosecretory system (AVPMNS) in the hypothalamus plays a critical role in homeostatic regulation as well as in allostatic motivational behaviors. However, it remains unclear whether adult neurogenesis exists in the AVPMNS. By using immunoreaction against AVP, neurophysin II, glial fibrillar acidic protein (GFAP), cell division marker (Ki67), migrating neuroblast markers (doublecortin, DCX), microglial marker (Ionized calcium binding adaptor molecule 1, Iba1), and 5'-bromo-2'-deoxyuridine (BrdU), we report morphological evidence that low-rate neurogenesis and migration occur in adult AVPMNS in the rat hypothalamus. Tangential AVP/GFAP migration routes and AVP/DCX neuronal chains as well as ascending AVP axonal scaffolds were observed. Chronic water deprivation significantly increased the BrdU+ nuclei within both the supraaoptic (SON) and paraventricular (PVN) nuclei. These findings raise new questions about AVPMNS's potential hormonal role for brain physiological adaptation across the lifespan, with possible involvement in coping with homeostatic adversities.


Subject(s)
Cell Movement , Doublecortin Protein , Neurogenesis , Neurons , Animals , Rats , Neurons/metabolism , Neurons/cytology , Male , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Hypothalamus/metabolism , Hypothalamus/cytology , Arginine Vasopressin/metabolism
10.
Pharmacol Biochem Behav ; 243: 173839, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39079561

ABSTRACT

Puberty is a critical period of emotional development and neuroplasticity. However, most studies have focused on early development, with limited research on puberty, particularly the parental presence. In this study, four groups were established, and pubertal maternal presence (PMP) was assessed until postnatal days 21 (PD21), 28 (PD28), 35 (PD35), and 42 (PD42), respectively. The social interaction and anxiety behaviors, as well as the expression of oxytocin (OT) in the paraventricular nucleus (PVN) and supraoptic nucleus (SON), and the number of new generated neurons and the expression of estrogen receptor alpha (ERα) in the dentate gyrus (DG) were assessed. The results suggest that there is a lot of physical contact between the mother and offspring from 21 to 42 days of age, which reduces anxiety in both female and male offspring in adulthood; for example, the PMP increased the amount of time mice spent in the center area in the open field experiment and in the bright area in the light-dark box experiment. PMP increased OT expression in the PVN and SON and the number of newly generated neurons in the DG. However, there was a sexual difference in ERα, with ERα increasing in females but decreasing in males. In conclusion, PMP reduces the anxiety of offspring in adulthood, increases OT in the PVN and SON, and adult neurogenesis; ERα in the DG may be involved in this process.


Subject(s)
Anxiety , Dentate Gyrus , Estrogen Receptor alpha , Neurogenesis , Oxytocin , Paraventricular Hypothalamic Nucleus , Animals , Anxiety/metabolism , Mice , Male , Female , Estrogen Receptor alpha/metabolism , Oxytocin/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Dentate Gyrus/metabolism , Sexual Maturation , Supraoptic Nucleus/metabolism , Maternal Behavior/physiology , Behavior, Animal , Social Interaction
11.
Cell ; 187(16): 4176-4192.e17, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-38959890

ABSTRACT

Hypothalamic neural circuits regulate instinctive behaviors such as food seeking, the fight/flight response, socialization, and maternal care. Here, we identified microdeletions on chromosome Xq23 disrupting the brain-expressed transient receptor potential (TRP) channel 5 (TRPC5). This family of channels detects sensory stimuli and converts them into electrical signals interpretable by the brain. Male TRPC5 deletion carriers exhibited food seeking, obesity, anxiety, and autism, which were recapitulated in knockin male mice harboring a human loss-of-function TRPC5 mutation. Women carrying TRPC5 deletions had severe postpartum depression. As mothers, female knockin mice exhibited anhedonia and depression-like behavior with impaired care of offspring. Deletion of Trpc5 from oxytocin neurons in the hypothalamic paraventricular nucleus caused obesity in both sexes and postpartum depressive behavior in females, while Trpc5 overexpression in oxytocin neurons in knock-in mice reversed these phenotypes. We demonstrate that TRPC5 plays a pivotal role in mediating innate human behaviors fundamental to survival, including food seeking and maternal care.


Subject(s)
Depression, Postpartum , Neurons , Obesity , TRPC Cation Channels , Animals , Female , Mice , Obesity/metabolism , Obesity/genetics , Male , Humans , TRPC Cation Channels/metabolism , TRPC Cation Channels/genetics , Depression, Postpartum/metabolism , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Mice, Inbred C57BL , Oxytocin/metabolism , Maternal Behavior
12.
Neurochem Res ; 49(10): 2926-2939, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39078522

ABSTRACT

Dexmedetomidine (DEX) is a highly selective α2-adrenoceptor agonist with sedative effects on sleep homeostasis. Oxytocin-expressing (OXT) neurons in the paraventricular nucleus (PVN) of the hypothalamus (PVNOXT) regulate sexual reproduction, drinking, sleep-wakefulness, and other instinctive behaviors. To investigate the effect of DEX on the activity and signal transmission of PVNOXT in regulating the sleep-wakefulness cycle. Here, we employed OXT-cre mice to selectively target and express the designer receptors exclusively activated by designer drugs (DREADD)-based chemogenetic tool hM3D(Gq) in PVNOXT neurons. Combining chemogenetic methods with electroencephalogram (EEG) /electromyogram (EMG) recordings, we found that cannula injection of DEX in PVN significantly increased the duration of non-rapid eye movement (NREM) sleep in mice. Furthermore, the chemogenetic activation of PVNOXT neurons using i.p. injection of clozapine N-oxide (CNO) after cannula injection of DEX to PVN led to a substantial increase in wakefulness. Electrophysiological results showed that DEX decreased the frequency of action potential (AP) and the spontaneous excitatory postsynaptic current (sEPSC) of PVNOXT neurons through α2-adrenoceptors. Therefore, these results identify that DEX promotes sleep and maintains sleep homeostasis by inhibiting PVNOXT neurons through the α2-adrenoceptor.


Subject(s)
Dexmedetomidine , Neurons , Oxytocin , Paraventricular Hypothalamic Nucleus , Animals , Dexmedetomidine/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Oxytocin/pharmacology , Neurons/drug effects , Neurons/metabolism , Mice , Male , Wakefulness/drug effects , Wakefulness/physiology , Adrenergic alpha-2 Receptor Agonists/pharmacology , Mice, Transgenic , Mice, Inbred C57BL , Hypnotics and Sedatives/pharmacology , Sleep/drug effects , Sleep/physiology
13.
Hypertens Res ; 47(9): 2363-2376, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38969805

ABSTRACT

The renin-angiotensin system (RAS) and the sympathetic nervous system (SNS) are two major blood pressure-regulating systems. The link between the renal and cerebral RAS axes was provided by reflex activation of renal afferents and efferent sympathetic nerves. There is a self-sustaining enhancement of the brain and the intrarenal RAS. In this study, prenatal exposure to lipopolysaccharide (LPS) led to increased RAS activity in the paraventricular nucleus (PVN) and overactivation of sympathetic outflow, accompanied by increased production of reactive oxygen species (ROS) and disturbances between inhibitory and excitatory neurons in PVN. The AT1 receptor blocker losartan and α2 adrenergic receptor agonist clonidine in the PVN significantly decreased renal sympathetic nerve activity (RSNA) and synchronously reduced systolic blood pressure. Prenatal LPS stimulation caused H3 acetylation at H3K9 and H3K14 in the PVN, which suggested that epigenetic changes are involved in transmitting the prenatal adverse stimulative information to the next generation. Additionally, melatonin treatment during pregnancy reduced RAS activity and ROS levels in the PVN; balanced the activity of inhibitory and excitatory neurons in the PVN; increased urine sodium secretion; reduced RSNA and blood pressure. In conclusion, prenatal LPS leads to increased RAS expression within the PVN and overactivation of the sympathetic outflow, thereby contributing to hypertension in offspring rats. Melatonin is expected to be a promising agent for preventing prenatal LPS exposure-induced hypertension.


Subject(s)
Blood Pressure , Lipopolysaccharides , Paraventricular Hypothalamic Nucleus , Prenatal Exposure Delayed Effects , Renin-Angiotensin System , Sympathetic Nervous System , Animals , Pregnancy , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/physiology , Sympathetic Nervous System/drug effects , Female , Rats , Blood Pressure/drug effects , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Male
14.
Hypertens Res ; 47(9): 2393-2404, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39039283

ABSTRACT

Chronic hypertensive pregnancy (CHP) is a growing health issue with unknown etiology. Vasopressin (VP), a nonapeptide synthesized in paraventricular (PVN) and supraoptic nucleus (SON), is a well-known neuroendocrine and autonomic modulator of the cardiovascular system, related to hypertension development. We quantified gene expression of VP and its receptors, V1aR and V1bR, within the PVN and SON in CHP and normal pregnancy, and assessed levels of secreted plasma VP. Also, we evaluated autonomic cardiovascular adaptations to CHP using spectral indices of blood pressure (BPV) and heart rate (HRV) short-term variability, and spontaneous baroreflex sensitivity (BRS). Experiments were performed in female spontaneously hypertensive rats (SHRs) and in normotensive Wistar rats (WRs). Animals were equipped with a radiotelemetry probe for continuous hemodynamic recordings before and during pregnancy. BPV, HRV and BRS were assessed using spectral analysis and the sequence method, respectively. Plasma VP was determined by ELISA whilst VP, V1aR, and V1bR gene expression was analyzed by real-time-quantitative PCR (RT-qPCR). The results show that non-pregnant SHRs exhibit greater VP, V1aR, and V1bR gene expression in both PVN and SON respectively, compared to Wistar dams. Pregnancy decreased VP gene expression in the SON of SHRs but increased it in the PVN and SON of WRs. Pregnant SHRs exhibited a marked drop in plasma VP concentration associated with BP normalization. This triggered marked tachycardia, heart rate variability increase, and BRS increase in pregnant SHRs. It follows that regardless of BP normalization in late pregnancy, SHRs exhibit cardiovascular vulnerability and compensate by recruiting vagal mechanisms. Pregnant SHR dams have reduced expression of VP in SON associated with increased V1bR expression, lower plasma VP, normal BP during late pregnancy and marked signs of enhanced sympathetic cardiac stimulation (increased HR and LFHR variability) and recruitment of vagal mechanisms (enhancement of BRS and HFHR variability).


Subject(s)
Baroreflex , Blood Pressure , Heart Rate , Rats, Inbred SHR , Rats, Wistar , Vasopressins , Animals , Female , Pregnancy , Vasopressins/blood , Vasopressins/metabolism , Rats , Baroreflex/physiology , Heart Rate/physiology , Blood Pressure/physiology , Hypertension, Pregnancy-Induced/physiopathology , Autonomic Nervous System/physiopathology , Receptors, Vasopressin/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/physiopathology , Supraoptic Nucleus/metabolism
15.
Cardiovasc Toxicol ; 24(9): 904-917, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39008239

ABSTRACT

Hypertension is a globally prevalent disease, but the pathogenesis remains largely unclear. AMP-activated protein kinase (AMPK) is a nutrition-sensitive signal of cellular energy metabolism, which has a certain influence on the development of hypertension. Previously, we found a down-regulation of the phosphorylated (p-) form of AMPK, and the up-regulation of the angiotensin II type 1 receptor (AT1-R) and that of p-ERK1/2 in the hypothalamic paraventricular nucleus (PVN) of hypertensive rats. However, the exact mechanism underlying the relationship between AMPK and AT1-R in the PVN during hypertension remains unclear. Thus, we hypothesized that AMPK modulates AT1-R through the ERK1/2-NF-κB pathway in the PVN, thereby inhibiting sympathetic nerve activity and improving hypertension. To examine this hypothesis, we employed a renovascular hypertensive animal model developed via two-kidney, one-clip (2K1C) and sham-operated (SHAM). Artificial cerebrospinal fluid (aCSF), used as vehicle, or 5-amino-1-ß-D-ribofuranosyl-imidazole-4-carboxamide (AICAR, an AMPK activator, 60 µg/day) was microinjected bilaterally in the PVN of these rats for 4 weeks. In 2K1C rats, there an increase in systolic blood pressure (SBP) and circulating norepinephrine (NE). Also, the hypertensive rats had lowered expression of p-AMPK and p-AMPK/AMPK, elevated expression of p-ERK1/2, p-ERK1/2/ERK1/2 and AT1-R, increased NF-κB p65 activity in the PVN compared with the levels of these biomarkers in SHAM rats. Four weeks of bilateral PVN injection of AMPK activator AICAR, attenuated the NE level and SBP, increased the expression of p-AMPK and p-AMPK/AMPK, lessened the NF-κB p65 activity, decreased the expression of p-ERK1/2, p-ERK1/2/ERK1/2 and AT1-R in the PVN of 2K1C rats. Data from this study imply that the activation of AMPK within the PVN suppressed AT1-R expression through inhibiting the ERK1/2-NF-κB pathway, decreased the activity of the sympathetic nervous system, improved hypertension.


Subject(s)
AMP-Activated Protein Kinases , Disease Models, Animal , Enzyme Activation , Hypertension, Renovascular , Mitogen-Activated Protein Kinase 3 , Paraventricular Hypothalamic Nucleus , Rats, Sprague-Dawley , Receptor, Angiotensin, Type 1 , Animals , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/enzymology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/physiopathology , Hypertension, Renovascular/physiopathology , Hypertension, Renovascular/enzymology , Hypertension, Renovascular/metabolism , Hypertension, Renovascular/drug therapy , Male , AMP-Activated Protein Kinases/metabolism , Phosphorylation , Receptor, Angiotensin, Type 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Transcription Factor RelA/metabolism , Ribonucleotides/pharmacology , Mitogen-Activated Protein Kinase 1/metabolism , Sympathetic Nervous System/physiopathology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , NF-kappa B/metabolism , Signal Transduction , Antihypertensive Agents/pharmacology , Rats
16.
Eur J Neurosci ; 60(5): 4861-4876, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39054660

ABSTRACT

Accumulating evidence suggests that electroacupuncture (EA) has obvious therapeutic effects and unique advantages in alleviating myocardial ischemia-reperfusion injury (MIRI), while the underlying neuromolecular mechanisms of EA intervention for MIRI have not been fully elucidated. The aim of the study is to investigate the role of the neural pathway of hypothalamic paraventricular nucleus (PVN) neurons projecting to the rostral ventrolateral medulla (RVLM) in the alleviation of MIRI rats by EA preconditioning. MIRI models were established by ligating the left anterior descending coronary artery for 30 min followed by reperfusion for 2 h. Electrocardiogram recording, chemogenetics, enzyme-linked immunosorbent assay, multichannel physiology recording and haematoxylin-eosin and immunofluorescence staining methods were conducted to demonstrate that the firing frequencies of neurons in the PVN and the expression of c-Fos decreased by EA pretreatment. Meanwhile, EA preconditioning significantly reduced the levels of creatine kinase isoenzymes (CK-MB), cardiac troponin I (cTnI) and lactic dehydrogenase (LDH). Virus tracing showed a projection connection between PVN and RVLM. The inhibition of the PVN-RVLM neural pathway could replicate the protective effect of EA pretreatment on MIRI rats. However, the activation of the pathway weakened the effect of EA preconditioning. EA pretreatment alleviated MIRI by regulating PVN neurons projecting to RVLM. This work provides novel evidence of EA pretreatment for alleviating MIRI.


Subject(s)
Electroacupuncture , Medulla Oblongata , Myocardial Reperfusion Injury , Neurons , Paraventricular Hypothalamic Nucleus , Rats, Sprague-Dawley , Animals , Electroacupuncture/methods , Paraventricular Hypothalamic Nucleus/metabolism , Medulla Oblongata/metabolism , Medulla Oblongata/physiology , Male , Neurons/physiology , Neurons/metabolism , Myocardial Reperfusion Injury/therapy , Myocardial Reperfusion Injury/metabolism , Rats , Neural Pathways/physiology , Neural Pathways/metabolism , Troponin I/metabolism , Proto-Oncogene Proteins c-fos/metabolism
17.
Nat Commun ; 15(1): 5321, 2024 Jun 22.
Article in English | MEDLINE | ID: mdl-38909051

ABSTRACT

Psychedelics have experienced renewed interest following positive clinical effects, however the neurobiological mechanisms underlying effects remain unclear. The paraventricular nucleus of the hypothalamus (PVN) plays an integral role in stress response, autonomic function, social behavior, and other affective processes. We investigated the effect of psilocin, the psychoactive metabolite of psilocybin, on PVN reactivity in Sprague Dawley rats. Psilocin increased stimulus-independent PVN activity as measured by c-Fos expression in male and female rats. Psilocin increased PVN reactivity to an aversive air-puff stimulus in males but not females. Reactivity was restored at 2- and 7-days post-injection with no group differences. Additionally, prior psilocin injection did not affect PVN reactivity following acute restraint stress. Experimental groups sub-classified by baseline threat responding indicate that increased male PVN reactivity is driven by active threat responders. These findings identify the PVN as a significant site of psychedelic drug action with implications for threat responding behavior.


Subject(s)
Hallucinogens , Paraventricular Hypothalamic Nucleus , Psilocybin , Rats, Sprague-Dawley , Animals , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Male , Psilocybin/analogs & derivatives , Psilocybin/pharmacology , Psilocybin/administration & dosage , Female , Rats , Hallucinogens/pharmacology , Hallucinogens/administration & dosage , Proto-Oncogene Proteins c-fos/metabolism , Behavior, Animal/drug effects , Stress, Psychological/physiopathology , Stress, Psychological/drug therapy
18.
Clin Exp Hypertens ; 46(1): 2361671, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38841901

ABSTRACT

BACKGROUND: Fibroblast growth factor 21 (FGF21) has a protective effect against cardiovascular disease. However, the role of FGF21 in hypertension remains elusive. METHODS: Ten-week-old male C57BL/6 mice were randomly divided into normal-salt (NS) group, NS+FGF21 group, deoxycorticosterone acetate-salt (DOCA) group and DOCA+FGF21 group. The mice in NS group underwent uninephrectomy without receiving DOCA and 1% NaCl and the mice in DOCA group were subjected to uninephrectomy and DOCA-salt (DOCA and 1% NaCl) treatment for 6 weeks. At the same time, the mice were infused with vehicle (artificial cerebrospinal fluid, aCSF) or FGF21 (1 mg/kg) into the bilateral paraventricular nucleus (PVN) of mice. RESULTS: Here, we showed that FGF21 treatment lowered DOCA salt-induced inflammation and oxidative stress in the PVN, which reduced sympathetic nerve activity and hypertension. Mechanistically, FGF21 treatment decreased the expression of HNF4α and inhibited the binding activity of HNF4α to the promoter region of ACE2 in the PVN of DOCA salt-treated mice, which further up-regulated ACE2/Ang (1-7) signals in the PVN. In addition, ACE2 deficiency abolished the protective effect of FGF21 in DOCA salt-treated mice, suggesting that FGF21-mediated antihypertensive effect was dependent on ACE2. CONCLUSIONS: The results demonstrate that FGF21 protects against salt-sensitive hypertension via regulating HNF4α/ACE2/Ang (1-7) axis in the PVN of DOCA salt-treated mice via multi-organ crosstalk between liver, brain and blood vessels.


Subject(s)
Angiotensin-Converting Enzyme 2 , Desoxycorticosterone Acetate , Fibroblast Growth Factors , Hepatocyte Nuclear Factor 4 , Hypertension , Mice, Inbred C57BL , Paraventricular Hypothalamic Nucleus , Animals , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Fibroblast Growth Factors/metabolism , Male , Mice , Hypertension/metabolism , Hypertension/physiopathology , Angiotensin-Converting Enzyme 2/metabolism , Hepatocyte Nuclear Factor 4/metabolism , Hepatocyte Nuclear Factor 4/genetics , Oxidative Stress/drug effects , Blood Pressure/drug effects , Sodium Chloride, Dietary
19.
Adv Sci (Weinh) ; 11(31): e2400437, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38885417

ABSTRACT

SH2B1 mutations are associated with obesity, type 2 diabetes, and metabolic dysfunction-associated steatotic liver disease (MASLD) in humans. Global deletion of Sh2b1 results in severe obesity, type 2 diabetes, and MASLD in mice. Neuron-specific restoration of SH2B1 rescues the obesity phenotype of Sh2b1-null mice, indicating that the brain is a main SH2B1 target. However, SH2B1 neurocircuits remain elusive. SH2B1-expressing neurons in the paraventricular hypothalamus (PVHSH2B1) and a PVHSH2B1→dorsal raphe nucleus (DRN) neurocircuit are identified here. PVHSH2B1 axons monosynaptically innervate DRN neurons. Optogenetic stimulation of PVHSH2B1 axonal fibers in the DRN suppresses food intake. Chronic inhibition of PVHSH2B1 neurons causes obesity. In male and female mice, either embryonic-onset or adult-onset deletion of Sh2b1 in PVH neurons causes energy imbalance, obesity, insulin resistance, glucose intolerance, and MASLD. Ablation of Sh2b1 in the DRN-projecting PVHSH2B1 subpopulation also causes energy imbalance, obesity, and metabolic disorders. Conversely, SH2B1 overexpression in either total or DRN-projecting PVHSH2B1 neurons protects against diet-induced obesity. SH2B1 binds to TrkB and enhances brain-derived neurotrophic factor (BDNF) signaling. Ablation of Sh2b1 in PVHSH2B1 neurons induces BDNF resistance in the PVH, contributing to obesity. In conclusion, these results unveil a previously unrecognized PVHSH2B1→DRN neurocircuit through which SH2B1 defends against obesity by enhancing BDNF/TrkB signaling.


Subject(s)
Adaptor Proteins, Signal Transducing , Obesity , Paraventricular Hypothalamic Nucleus , Animals , Obesity/metabolism , Obesity/genetics , Mice , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Male , Female , Paraventricular Hypothalamic Nucleus/metabolism , Disease Models, Animal , Metabolic Diseases/metabolism , Metabolic Diseases/genetics , Energy Metabolism/genetics , Energy Metabolism/physiology , Dorsal Raphe Nucleus/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Neurons/metabolism
20.
Brain Behav Immun ; 120: 499-512, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38944162

ABSTRACT

The gut microbiota and neurological development of neonatal mice are susceptible to environmental factors that may lead to altered behavior into adulthood. However, the role that changed gut microbiota and neurodevelopment early in life play in this needs to be clarified. In this study, by modeling early-life environmental changes by cross-fostering BALB/c mice, we revealed the effects of the environment during the critical period of postnatal development on adult social behavior and their relationship with the gut microbiota and the nervous system. The neural projections exist between the ascending colon and oxytocin neurons in the paraventricular nuclei (PVN), peripheral oxytocin levels and PVN neuron numbers decreased after cross-fostering, and sex-specific alteration in gut microbiota and its metabolites may be involved in social impairments and immune imbalances brought by cross-fostering via the gut-brain axis. Our findings also suggest that social cognitive impairment may result from a combination of PVN oxytocinergic neurons, gut microbiota, and metabolites.


Subject(s)
Brain-Gut Axis , Gastrointestinal Microbiome , Mice, Inbred BALB C , Neurons , Oxytocin , Paraventricular Hypothalamic Nucleus , Social Behavior , Animals , Gastrointestinal Microbiome/physiology , Mice , Oxytocin/metabolism , Male , Female , Paraventricular Hypothalamic Nucleus/metabolism , Brain-Gut Axis/physiology , Neurons/metabolism , Brain/metabolism , Behavior, Animal/physiology , Colon/metabolism , Colon/microbiology , Animals, Newborn
SELECTION OF CITATIONS
SEARCH DETAIL