Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
Neurosci Lett ; 773: 136518, 2022 03 16.
Article in English | MEDLINE | ID: mdl-35150776

ABSTRACT

In normal hormonal conditions, increased neuronal activity in the ventromedial hypothalamus (VMH) induces lordosis whereas activation of the preoptic area (POA) exerts an opposite effect. In the present work, we explored the effect of bilateral infusion of different doses of the apelin-13 (0.37, 0.75, 1.5, and 15 µg) in both brain areas on the expression of lordosis behavior. Lordosis quotient and lordosis reflex score were performed at 30, 120, and 240 min. Weak lordosis was observed following the 0.37 µg dose of apelin-13 at 30 min in the VMH of EB-primed rats; however, the rest of the doses induced significant lordosis relative to the control group. At 120 min, all doses induced lordosis behavior, while at 240 min, the highest dose of 15 µg did not induce significant differences. Interestingly, only the 0.75 µg infusion of apelin in the POA induced significant lordosis at 120 and 240 min. These results indicate that apelin-13 acts preferably in HVM and slightly in POA to initiate lordosis behavior in estrogen-primed rats.


Subject(s)
Intercellular Signaling Peptides and Proteins , Lordosis , Preoptic Area , Animals , Estradiol/pharmacology , Estrogens/pharmacology , Hypothalamus/drug effects , Hypothalamus/pathology , Intercellular Signaling Peptides and Proteins/pharmacology , Lordosis/chemically induced , Preoptic Area/drug effects , Preoptic Area/pathology , Progesterone/pharmacology , Rats , Sexual Behavior, Animal/drug effects , Ventromedial Hypothalamic Nucleus/drug effects , Ventromedial Hypothalamic Nucleus/pathology
2.
Neural Plast ; 2021: 7806370, 2021.
Article in English | MEDLINE | ID: mdl-34306063

ABSTRACT

Chronic stress is one of the main risk factors of bone loss. While the neurons and neural circuits of the ventromedial hypothalamus (VMH) mediate bone loss induced by chronic stress, the detailed intrinsic mechanisms within the VMH nucleus still need to be explored. Astrocytes in brain regions play important roles in the regulation of metabolism and anxiety-like behavior through interactions with surrounding neurons. However, whether astrocytes in the VMH affect neuronal activity and therefore regulate chronic stress-induced anxiety and bone loss remain elusive. In this study, we found that VMH astrocytes were activated during chronic stress-induced anxiety and bone loss. Pharmacogenetic activation of the Gi and Gq pathways in VMH astrocytes reduced and increased the levels of anxiety and bone loss, respectively. Furthermore, activation of VMH astrocytes by optogenetics induced depolarization in neighboring steroidogenic factor-1 (SF-1) neurons, which was diminished by administration of N-methyl-D-aspartic acid (NMDA) receptor blocker but not by alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor blocker. These results suggest that there may be a functional "glial-neuron microcircuit" in VMH nuclei that mediates anxiety and bone loss induced by chronic stress. This study not only advances our understanding of glial cell function but also provides a potential intervention target for chronic stress-induced anxiety and bone loss therapy.


Subject(s)
Anxiety Disorders/physiopathology , Astrocytes/physiology , Bone Resorption/physiopathology , Stress, Psychological/complications , Ventromedial Hypothalamic Nucleus/pathology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Bone Density , Bone Resorption/etiology , Bone Resorption/prevention & control , Chronic Disease , Clozapine/pharmacology , Clozapine/therapeutic use , Elevated Plus Maze Test , Emotions , GTP-Binding Protein alpha Subunits, Gi-Go/agonists , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Genes, Reporter , Male , Mice , Mice, Inbred C57BL , Neurons/physiology , Open Field Test , Optogenetics , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-fos/genetics , Random Allocation , Receptors, N-Methyl-D-Aspartate/physiology , Ventromedial Hypothalamic Nucleus/physiopathology
3.
J Chem Neuroanat ; 113: 101919, 2021 04.
Article in English | MEDLINE | ID: mdl-33497806

ABSTRACT

The ventromedial hypothalamic nucleus (VMH) is located in the tuberal region of the hypothalamus and is traditionally considered the satiety center. In obese Zucker rats, which express a mutation in the leptin receptor gene and exhibit obesity from the first weeks of life, the morphology of VMH neurons is unknown. In the present study, we found that the dendritic length of VMH neurons in obese Zucker rats was significantly shorter than that in Long Evans rats. This finding allows us to suggest that obese Zucker rats exhibit both neuronal metabolic alterations related to leptin and a reduction in the flow of information within the neuronal circuits in which the VMH nucleus participates to regulate foraging.


Subject(s)
Dendrites/pathology , Neurons/pathology , Obesity/pathology , Ventromedial Hypothalamic Nucleus/pathology , Animals , Cell Shape/physiology , Rats , Rats, Zucker
4.
Neuroendocrinology ; 110(3-4): 271-281, 2020.
Article in English | MEDLINE | ID: mdl-31167202

ABSTRACT

Leptin signaling pathways, stemming primarily from the hypothalamus, are necessary for maintaining normal energy homeostasis and body weight. In both rodents and humans, dysregulation of leptin signaling leads to morbid obesity and diabetes. Since leptin resistance is considered a primary factor underlying obesity, understanding the regulation of leptin signaling could lead to therapeutic tools and provide insights into the causality of obesity. While leptin actions in some hypothalamic regions such as the arcuate nuclei have been characterized, less is known about leptin activity in the hypothalamic ventromedial nuclei (VMN). Recently, pituitary adenylate cyclase-activating polypeptide (PACAP) has been shown to reduce feeding behavior and alter metabolism when administered into the VMN in a pattern similar to that of leptin. In the current study, we examined whether leptin and PACAP actions in the VMN share overlapping pathways in the regulation of energy balance. Interestingly, PACAP administration into the VMN increased STAT3 phosphorylation and SOCS3 mRNA expression, both of which are hallmarks of leptin receptor activation. In addition, BDNF mRNA expression in the VMN was increased by both leptin and PACAP administration. Moreover, antagonizing PACAP receptors fully reversed the behavioral and cellular effects of leptin injections into the VMN. Electrophysiological studies further illustrated that leptin-induced effects on VMN neurons were blocked by antagonizing PACAP receptors. We conclude that leptin dependency on PACAP signaling in the VMN suggests a potential common signaling cascade, allowing a tonically and systemically secreted neuropeptide to be more precisely regulated by central neuropeptides.


Subject(s)
Behavior, Animal/physiology , Body Temperature Regulation/physiology , Eating/physiology , Leptin/metabolism , Pituitary Adenylate Cyclase-Activating Polypeptide/metabolism , Signal Transduction/physiology , Ventromedial Hypothalamic Nucleus/pathology , Animals , Brain-Derived Neurotrophic Factor/metabolism , Male , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/metabolism
5.
Cell Rep ; 28(5): 1195-1205.e3, 2019 07 30.
Article in English | MEDLINE | ID: mdl-31365864

ABSTRACT

Impulsively aggressive individuals may suddenly attack others when under stress, but the neural circuitry underlying stress-provoked aggression is poorly understood. Here, we report that acute stress activates ventral hippocampus (vHip) neurons to induce attack behavior in post-weaning socially isolated mice. Chemogenetic inhibition of vHip neural activity blunts stress-provoked attack behavior, whereas chemogenetic activation promotes it. The activation of cell bodies in vHip neurons projecting into the ventromedial hypothalamus (VMH) induces attack behavior, suggesting that the vHip-VMH projection contributes to impulsive aggression. Furthermore, optogenetic inhibition of vHip glutamatergic neurons blocks stress-provoked attacks, whereas optogenetic activation of vHip glutamatergic neurons drives attack behavior. These results show direct evidence that vHip-VMH neural circuitry modulates attack behavior in socially isolated mice.


Subject(s)
Aggression , Hippocampus , Stress, Psychological , Ventromedial Hypothalamic Nucleus , Animals , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Mice , Mice, Inbred BALB C , Stress, Psychological/metabolism , Stress, Psychological/pathology , Stress, Psychological/physiopathology , Ventromedial Hypothalamic Nucleus/metabolism , Ventromedial Hypothalamic Nucleus/pathology , Ventromedial Hypothalamic Nucleus/physiopathology
6.
Sci Rep ; 8(1): 10170, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29977067

ABSTRACT

Previous studies showed that restraint water-immersion stress (RWIS) increases the expression of Fos protein in the ventromedial hypothalamic nucleus (VMH), indicating the VMH involving in the stress-induced gastric mucosal injury (SGMI). The present study was designed to investigate its possible neuro-regulatory mechanisms in rats receiving either VMH lesions or sham surgery. The model for SGMI was developed by restraint and water (21 ± 1 °C) immersion for 2 h. Gastric mucosal injury index, gastric motility, gastric acid secretion and Fos expression in the hypothalamus and brainstem were examined on the 15th postoperative day in RWIS rats. Gastric mucosal injury in VMH-lesioned rats was obviously aggravated compared to the control. Gastric acidity under RWIS was obviously higher in VMH-lesioned rats than that in sham rats. Meantime, the VMH-lesioned rats exhibited marked increases in the amplitude of gastric motility in the VMH lesions group after RWIS. In VMH-lesioned rats, Fos expression significantly increased in the dorsal motor nucleus of the vagus (DMV), the nucleus of the solitary tract (NTS), the area postrema (AP), the paraventricular nucleus (PVN) and the supraoptic nucleus (SON) in response to RWIS. These results indicate that VMH lesions can aggravate the stress-induced gastric mucosal injury through the VMH-dorsal vagal complex (DVC)-vagal nerve pathway.


Subject(s)
Gastric Mucosa/injuries , Gastric Mucosa/pathology , Stress, Psychological/complications , Ventromedial Hypothalamic Nucleus/pathology , Animals , Body Weight , Gastric Acid/metabolism , Immersion , Male , Neurons/pathology , Proto-Oncogene Proteins c-fos/metabolism , Rats, Wistar , Restraint, Physical , Ventromedial Hypothalamic Nucleus/metabolism , Water
7.
Reprod Sci ; 25(6): 867-872, 2018 06.
Article in English | MEDLINE | ID: mdl-28359185

ABSTRACT

BACKGROUND AND OBJECTIVE: The most important alkaloid of opium family, morphine, may show unfavorable effect on the reproductive organs. This research investigated the effect of microinjection of morphine into the rat's ventromedial hypothalamus (VMH) on cystic genesis in the ovary and the health of neurons of VMH. MATERIALS AND METHODS: Female rats (Wistar, weighing 200-250 g) kept under standard conditions were cannulated under anesthesia by Stoelting stereotaxic instrument at the coordinates anterior-posterior: -1.92, ventral: 9, lateral: 0.5. After being recovered, they were microinjected single morphine (0.1-0.4 µg/rat, once intra-VMH) and/or naloxone hydrochloride (0.1-0.4 µg/rat, once intra-VMH) using a 5-µL Hamilton syringe with the polyethylene tubing. The control group solely given physiological saline (1 µL/rat, intra-VMH). Three days after the experiment, both ovary and brain samples were collected from the control and the experimental groups, and they were studied histopathologically. The brain samples were checked out with the aid of the cresyl violet, and the ovaries were stained by the hematoxylin and eosin. The samples were also biometrically examined to compare the ovaries' cystic formations. Also, the number of healthy or injured neurons in the nuclei was compared. RESULTS: The ovaries of morphine-treated rats showed polycystic characteristics in comparison with the control samples. In addition, the brain slices of the morphine-treated rats illustrated a significant decrease in intact neurons. Both mal effects were resolved in the presence of naloxone. CONCLUSION: These results may show that the morphine induces anovulatory infertility probably by hypothalamus-pituitary-ovary axis dysfunction.


Subject(s)
Morphine/administration & dosage , Narcotics/administration & dosage , Ovary/drug effects , Ovary/pathology , Ventromedial Hypothalamic Nucleus/drug effects , Ventromedial Hypothalamic Nucleus/pathology , Animals , Female , Menstrual Cycle , Naloxone/administration & dosage , Narcotic Antagonists/administration & dosage , Neurons/drug effects , Neurons/pathology , Polycystic Ovary Syndrome/chemically induced , Rats, Wistar
8.
Hypertension ; 68(2): 491-500, 2016 08.
Article in English | MEDLINE | ID: mdl-27296999

ABSTRACT

High fat diet (HFD)-induced hypertension in rabbits is neurogenic and caused by the central action of leptin, which is thought to be dependent on activation of α-melanocortin-stimulating hormone (α-MSH) and neuropeptide Y-positive neurons projecting to the dorsomedial hypothalamus (DMH) and ventromedial hypothalamus (VMH). However, leptin may act directly in these nuclei. Here, we assessed the contribution of leptin, α-MSH, and neuropeptide Y signaling in the DMH and VMH to diet-induced hypertension. Male New Zealand white rabbits were instrumented with a cannula for drug injections into the DMH or VMH and a renal sympathetic nerve activity (RSNA) electrode. After 3 weeks of an HFD (13.3% fat; n=19), rabbits exhibited higher RSNA, mean arterial pressure (MAP), and heart rate compared with control diet-fed animals (4.2% fat; n=15). Intra-VMH injections of a leptin receptor antagonist or SHU9119, a melanocortin 3/4 receptor antagonist, decreased MAP, heart rate, and RSNA compared with vehicle in HFD rabbits (P<0.05) but not in control diet-fed animals. By contrast, α-MSH or neuropeptide Y injected into the VMH had no effect on MAP but produced sympathoexcitation in HFD rabbits (P<0.05) but not in control diet-fed rabbits. The effects of the leptin antagonist, α-MSH, or neuropeptide Y injections into the DMH on MAP or RSNA of HFD rabbits were not different from those after vehicle injection. α-MSH into the DMH of control diet-fed animals did increase MAP, heart rate, and RSNA. We conclude that the VMH is the likely origin of leptin-mediated sympathoexcitation and α-MSH hypersensitivity that contribute to obesity-related hypertension.


Subject(s)
Dorsomedial Hypothalamic Nucleus/metabolism , Hypertension , Melanocyte-Stimulating Hormones/pharmacology , Neuropeptide Y , Obesity , Sympathetic Nervous System/drug effects , Ventromedial Hypothalamic Nucleus/metabolism , alpha-MSH , Animals , Blood Pressure/drug effects , Blood Pressure/physiology , Diet, High-Fat/adverse effects , Dorsomedial Hypothalamic Nucleus/pathology , Hypertension/etiology , Hypertension/metabolism , Hypertension/physiopathology , Leptin/metabolism , Male , Neuropeptide Y/metabolism , Neuropeptide Y/pharmacology , Obesity/etiology , Obesity/metabolism , Obesity/physiopathology , Rabbits , Receptors, Leptin/antagonists & inhibitors , Receptors, Melanocortin/antagonists & inhibitors , Signal Transduction/drug effects , Sympathetic Nervous System/metabolism , Sympathetic Nervous System/physiopathology , Ventromedial Hypothalamic Nucleus/pathology , alpha-MSH/metabolism , alpha-MSH/pharmacology
10.
Cell Rep ; 10(1): 62-74, 2015 Jan 06.
Article in English | MEDLINE | ID: mdl-25543145

ABSTRACT

Estrogen-receptor alpha (ERα) neurons in the ventrolateral region of the ventromedial hypothalamus (VMHVL) control an array of sex-specific responses to maximize reproductive success. In females, these VMHVL neurons are believed to coordinate metabolism and reproduction. However, it remains unknown whether specific neuronal populations control distinct components of this physiological repertoire. Here, we identify a subset of ERα VMHVL neurons that promotes hormone-dependent female locomotion. Activating Nkx2-1-expressing VMHVL neurons via pharmacogenetics elicits a female-specific burst of spontaneous movement, which requires ERα and Tac1 signaling. Disrupting the development of Nkx2-1(+) VMHVL neurons results in female-specific obesity, inactivity, and loss of VMHVL neurons coexpressing ERα and Tac1. Unexpectedly, two responses controlled by ERα(+) neurons, fertility and brown adipose tissue thermogenesis, are unaffected. We conclude that a dedicated subset of VMHVL neurons marked by ERα, NKX2-1, and Tac1 regulates estrogen-dependent fluctuations in physical activity and constitutes one of several neuroendocrine modules that drive sex-specific responses.


Subject(s)
Estrogen Receptor alpha/metabolism , Locomotion/genetics , Nuclear Proteins/biosynthesis , Obesity/metabolism , Tachykinins/genetics , Transcription Factors/biosynthesis , Animals , Estrogen Receptor alpha/genetics , Estrogens/metabolism , Female , Mice , Neurons/metabolism , Neurons/pathology , Nuclear Proteins/genetics , Obesity/genetics , Obesity/physiopathology , Sex Characteristics , Tachykinins/metabolism , Thyroid Nuclear Factor 1 , Transcription Factors/genetics , Ventromedial Hypothalamic Nucleus/metabolism , Ventromedial Hypothalamic Nucleus/pathology
11.
J Neuroendocrinol ; 26(4): 258-66, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24629021

ABSTRACT

To determine the relationship between the right and left sides of the ventrolateral ventromedial hypothalamic nucleus (vlVMN) in regulating the expression of oestrogen receptor (ER)α, the unilateral vlVMN was lesioned and the number of ERα-immunoreactive cells and the ERα mRNA level in the intact side of the vlVMN and arcuate nucleus (ARC) were measured in ovariectomised rats. Twenty-four hours after lesioning, brain samples were collected for analysis of ERα expression by immunohistochemistry and the real-time reverse transcriptase-polymerase chain reaction. The number of ERα-immunoreactive cells in the intact side of the vlVMN but not the ARC in the unilateral lesioned group was significantly higher than that in the control or sham-lesioned group. Expression levels of ERα mRNA in the intact side of the vlVMN but not the ARC in unilateral lesioned rats were significantly higher than those in the sham-lesioned group. Of transcript variants with alternative 5'-untranslated regions (0S, 0N, 0, 0T and E1), the ERα 0 transcript level was significantly increased. These results indicate that unilateral damage of vlVMN induces an increase in ERα in the intact side by increasing ERα transcription in a promoter-specific manner. The findings also suggest the existence of new neuroendocrine control system between the right and left sides for the expression of ERα in the vlVMN.


Subject(s)
Estrogen Receptor alpha/metabolism , Ovariectomy , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Base Sequence , DNA Primers , Female , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Ventromedial Hypothalamic Nucleus/pathology
12.
Endocrinology ; 155(5): 1806-16, 2014 May.
Article in English | MEDLINE | ID: mdl-24564394

ABSTRACT

Adiponectin, an adipocyte-derived hormone, regulates glucose and lipid metabolism. It is also antiinflammatory. During obesity, adiponectin levels and sensitivity are reduced. Whereas the action of adiponectin in the periphery is well established the neuroendocrine role of adiponectin is largely unknown. To address this we analyzed the expression of adiponectin and the 2 adiponectin receptors (AdipoR1 and AdipoR2) in response to fasting and to diet-induced and genetic obesity. We also investigated the acute impact of adiponectin on central regulation of glucose homeostasis. Adiponectin (1 µg) was injected intracerebroventricularly (ICV), and glucose tolerance tests were performed in dietary and genetic obese mice. Finally, the influence of ICV adiponectin administration on central signaling cascades regulating glucose homeostasis and on markers of hypothalamic inflammation was assessed. Gene expression of adiponectin was down-regulated whereas AdipoR1 was up-regulated in the arcuate nucleus of fasted mice. High-fat (HF) feeding increased AdipoR1 and AdipoR2 gene expression in this region. In mice on a HF diet and in leptin-deficient mice acute ICV adiponectin improved glucose tolerance 60 minutes after injection, whereas normoglycemia in control mice was unaffected. ICV adiponectin increased pAKT, decreased phospho-AMP-activated protein kinase, and did not change phospho-signal transducer and activator of transcription 3 immunoreactivity. In HF-fed mice, ICV adiponectin reversed parameters of hypothalamic inflammation and insulin resistance as determined by the number of phospho-glycogen synthase kinase 3 ß(Ser9) and phospho-c-Jun N-terminal kinase (Thr183/Tyr185) immunoreactive cells in the arcuate nucleus and ventromedial hypothalamus. This study demonstrates that the insulin-sensitizing properties of adiponectin are at least partially based on a neuroendocrine mechanism that involves centrally synthesized adiponectin.


Subject(s)
Adiponectin/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Glucose Intolerance/prevention & control , Hypoglycemic Agents/therapeutic use , Insulin Resistance , Obesity/physiopathology , Adiponectin/administration & dosage , Adiponectin/adverse effects , Adiponectin/antagonists & inhibitors , Adiponectin/genetics , Adiponectin/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/pathology , Diet, High-Fat/adverse effects , Glucose Intolerance/etiology , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/adverse effects , Injections, Intraventricular , Leptin/genetics , Leptin/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Receptors, Adiponectin/biosynthesis , Receptors, Adiponectin/genetics , Receptors, Adiponectin/metabolism , Signal Transduction/drug effects , Ventromedial Hypothalamic Nucleus/drug effects , Ventromedial Hypothalamic Nucleus/immunology , Ventromedial Hypothalamic Nucleus/metabolism , Ventromedial Hypothalamic Nucleus/pathology
13.
Curr Diab Rep ; 14(3): 465, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24510608

ABSTRACT

It is now widely accepted that the brain makes important contributions to the dysregulated glucose metabolism, altered feeding behaviors, and the obesity often seen in type 2 diabetes (T2D). Although studies focusing on genetic, cellular, and molecular regulatory elements in pancreas, liver, adipose tissue etc provide a good understanding of how these processes relate to T2D, our knowledge of how brain wiring patterns are organized is much less developed. This article discusses animal studies that illustrate the importance of understanding the network organization of those brain regions most closely implicated in T2D. It will describe the brain networks, as well as the methodologies used to explore them. To illustrate some of the gaps in our knowledge, we will discuss the connectional network of the ventromedial nucleus and its adjacent cell groups in the hypothalamus; structures that are widely recognized as key elements in the brain's ability to control glycemia, feeding, and body weight.


Subject(s)
Brain/physiopathology , Diabetes Mellitus, Type 2/physiopathology , Nerve Net/physiopathology , Animals , Brain/metabolism , Diabetes Mellitus, Type 2/metabolism , Humans , Nerve Net/metabolism , Nerve Net/pathology , Ventromedial Hypothalamic Nucleus/metabolism , Ventromedial Hypothalamic Nucleus/pathology , Ventromedial Hypothalamic Nucleus/physiopathology
14.
Horm Behav ; 65(3): 254-63, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24333848

ABSTRACT

Behaviors associated with breeding are seasonally modulated in a variety of species. These changes in behavior are mediated by sex steroids, levels of which likewise vary with season. The effects of androgens on behaviors associated with breeding may in turn be partly mediated by the nonapeptides vasopressin (VP) and oxytocin (OT) in mammals, and vasotocin (VT) in birds. The effects of testosterone (T) on production of these neuropeptides have been well-studied; however, the regulation of VT receptors by T is not well understood. In this study, we investigated steroid-dependent regulation of VT receptor (VTR) mRNA in a seasonally breeding songbird, the white-throated sparrow (Zonotrichia albicollis). We focused on VTR subtypes that have been most strongly implicated in social behavior: V1a and oxytocin-like receptor (OTR). Using in situ hybridization, we show that T-treatment of non-breeding males altered V1a and OTR mRNA expression in several regions associated with seasonal reproductive behaviors. For example, T-treatment increased V1a mRNA expression in the medial preoptic area, bed nucleus of the stria terminalis, and ventromedial hypothalamus. T-treatment also affected both V1a and OTR mRNA expression in nuclei of the song system; some of these effects depended on the presence or absence of a chromosomal rearrangement that affects singing behavior, plasma T, and VT immunolabeling in this species. Overall, our results strengthen evidence that VT helps mediate the behavioral effects of T in songbirds, and suggest that the chromosomal rearrangement in this species may affect the sensitivity of the VT system to seasonal changes in T.


Subject(s)
Preoptic Area/metabolism , Receptors, Vasopressin/metabolism , Septal Nuclei/metabolism , Sparrows/physiology , Testosterone/physiology , Ventromedial Hypothalamic Nucleus/metabolism , Vocalization, Animal/physiology , Animals , Breeding , Male , Preoptic Area/pathology , RNA, Messenger/metabolism , Receptors, Oxytocin/metabolism , Seasons , Septal Nuclei/pathology , Sexual Behavior, Animal/physiology , Testosterone/blood , Ventromedial Hypothalamic Nucleus/pathology
15.
Endocrinology ; 154(9): 3067-76, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23782941

ABSTRACT

In rodent models of type 1 diabetes, leptin administration into brain ventricles normalizes blood glucose at doses that have no effect when given peripherally. The ventromedial nucleus of the hypothalamus (VMN) is a potential target for leptin's antidiabetic effects because leptin-sensitive neurons in this brain area are implicated in glucose homeostasis. To test this hypothesis, we injected leptin directly into the bilateral VMN of rats with streptozotocin-induced uncontrolled diabetes mellitus. This intervention completely normalized both hyperglycemia and the elevated rates of hepatic glucose production and plasma glucagon levels but had no effect on tissue glucose uptake in the skeletal muscle or brown adipose tissue as measured using tracer dilution techniques during a basal clamp. To determine whether VMN leptin signaling is required for leptin-mediated normalization of diabetic hyperglycemia, we studied mice in which the leptin receptor gene was deleted in VMN steroidogenic factor 1 neurons using cre-loxP technology. Our findings indicate leptin action within these neurons is not required for the correction of diabetic hyperglycemia by central leptin infusion. We conclude that leptin signaling in the VMN is sufficient to mediate leptin's antidiabetic action but may not be necessary for this effect. Leptin action within a distributed neuronal network may mediate its effects on glucose homeostasis.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , Hyperglycemia/prevention & control , Leptin/metabolism , Neurons/metabolism , Receptors, Leptin/metabolism , Signal Transduction , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Diabetes Mellitus, Type 1/blood , Diabetes Mellitus, Type 1/drug therapy , Diabetes Mellitus, Type 1/pathology , Glucagon/blood , Glucagon/metabolism , Glucagon-Secreting Cells/drug effects , Glucagon-Secreting Cells/metabolism , Gluconeogenesis/drug effects , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/therapeutic use , Infusions, Intraventricular , Injections, Intraventricular , Leptin/administration & dosage , Leptin/genetics , Leptin/therapeutic use , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/agonists , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/pathology , Rats , Rats, Wistar , Receptors, Leptin/agonists , Receptors, Leptin/genetics , Recombinant Proteins/administration & dosage , Recombinant Proteins/metabolism , Recombinant Proteins/therapeutic use , Signal Transduction/drug effects , Ventromedial Hypothalamic Nucleus/drug effects , Ventromedial Hypothalamic Nucleus/pathology
16.
Neuropharmacology ; 71: 164-73, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23603200

ABSTRACT

The ventromedial nucleus of the hypothalamus (VMH) plays an important role in feeding and energy homeostasis. Electroconvulsive seizure (ECS) therapy is highly effective in the treatment of several psychiatric diseases, including depression, but may also have beneficial effects in other neurological diseases. Although it has been reported that the neurons of the VMH are strongly activated by ECS stimulation, the specific effects of ECS in this hypothalamic subnucleus remain unknown. To address this issue, we investigated the changes in gene expression in microdissected-VMH samples in response to ECS in mice, and examined the behavioral effects of ECS on feeding behavior. ECS significantly induced the expression of immediate-early genes such as Fos, Fosb, and Jun, as well as Bdnf, Adcyap1, Hrh1, and Crhr2 in the VMH. Given that signals of these gene products are suggested to have anorexigenic roles in the VMH, we also examined the effect of ECS on food intake and body weight. Repeated ECS had a suppressive effect on food intake and body weight gain under both regular and high-fat diet conditions. Furthermore, gold-thioglucose-induced hypothalamic lesions, including the VMH and the arcuate nucleus, abolished the anorexigenic effects of ECS, indicating the requirement for the activation of the hypothalamus. Our data show an effect of ECS on increased expression of anorexigenic factors in the VMH, and suggest a role in the regulation of energy homeostasis by ECS.


Subject(s)
Appetite Regulation , Electroconvulsive Therapy , Nerve Tissue Proteins/biosynthesis , Neurons/metabolism , Obesity/therapy , Up-Regulation , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Appetite Regulation/drug effects , Appetitive Behavior/drug effects , Aurothioglucose/toxicity , Behavior, Animal/drug effects , Diet, High-Fat/adverse effects , Feeding Behavior/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Microdissection , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/pathology , Neurotoxicity Syndromes/complications , Obesity/complications , Obesity/metabolism , Obesity/pathology , Up-Regulation/drug effects , Ventromedial Hypothalamic Nucleus/drug effects , Ventromedial Hypothalamic Nucleus/pathology , Weight Loss/drug effects
17.
J Neuroimmunol ; 257(1-2): 1-6, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23411096

ABSTRACT

Over the past 20 years, the functional autonomy of both the immune and central nervous systems has been successfully challenged. Although the ventromedial hypothalamus (VMH) is one of the centers of parasympathetic nervous system, to date, there has been little reported regarding the role of the hypothalamus in directly changing the expression of immune-related genes. Recently, it has been reported that VMH lesions can directly change the expression of immune-related gene families. The present review focuses on the relationships between the VMH and the expressions of immune-related genes.


Subject(s)
Gene Expression Regulation/immunology , Ventromedial Hypothalamic Nucleus/immunology , Ventromedial Hypothalamic Nucleus/pathology , Animals , Cytokines/biosynthesis , Cytokines/genetics , Humans , Oligonucleotide Array Sequence Analysis/methods , Ventromedial Hypothalamic Nucleus/metabolism
18.
Proc Natl Acad Sci U S A ; 110(11): 4333-8, 2013 Mar 12.
Article in English | MEDLINE | ID: mdl-23440210

ABSTRACT

The nuclear receptor Chicken Ovalbumin Upstream Promoter-Transcription Factor II (COUP-TFII) is an important coordinator of glucose homeostasis through its function in different organs such as the endocrine pancreas, adipose tissue, skeletal muscle, and liver. Recently we have demonstrated that COUP-TFII expression in the hypothalamus is restricted to a subpopulation of neurons expressing the steroidogenic factor 1 transcription factor, known to play a crucial role in glucose homeostasis. To understand the functional significance of COUP-TFII expression in the steroidogenic factor 1 neurons, we generated hypothalamic ventromedial nucleus-specific COUP-TFII KO mice using the cyclization recombination/locus of X-overP1 technology. The heterozygous mutant mice display insulin hypersensitivity and a leaner phenotype associated with increased energy expenditure and similar food intake. These mutant mice also present a defective counterregulation to hypoglycemia with altered glucagon secretion. Moreover, the mutant mice are more likely to develop hypoglycemia-associated autonomic failure in response to recurrent hypoglycemic or glucopenic events. Therefore, COUP-TFII expression levels in the ventromedial nucleus are keys in the ability to resist the onset of hypoglycemia-associated autonomic failure.


Subject(s)
COUP Transcription Factor II/biosynthesis , Glucose/metabolism , Hypoglycemia/metabolism , Nerve Tissue Proteins/biosynthesis , Neurons/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Autonomic Nervous System Diseases/etiology , Autonomic Nervous System Diseases/genetics , Autonomic Nervous System Diseases/metabolism , Autonomic Nervous System Diseases/pathology , COUP Transcription Factor II/genetics , Chickens , Glucose/genetics , Heterozygote , Hypoglycemia/complications , Hypoglycemia/genetics , Hypoglycemia/pathology , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/pathology , Organ Specificity/genetics , Steroidogenic Factor 1/genetics , Steroidogenic Factor 1/metabolism , Ventromedial Hypothalamic Nucleus/pathology
19.
PLoS One ; 7(11): e48733, 2012.
Article in English | MEDLINE | ID: mdl-23139814

ABSTRACT

Infantile neuronal ceroid lipofuscinosis (INCL) is a fatal neurodegenerative disorder caused by a deficiency of palmitoyl-protein thioesterase-1 (PPT1). We have previously shown that children with INCL have increased risk of hypothermia during anesthesia and that PPT1-deficiency in mice is associated with disruption of adaptive energy metabolism, downregulation of peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), and mitochondrial dysfunction. Here we hypothesized that Ppt1-knockout mice, a well-studied model of INCL that shows many of the neurologic manifestations of the disease, would recapitulate the thermoregulation impairment observed in children with INCL. We also hypothesized that when exposed to cold, Ppt1-knockout mice would be unable to maintain body temperature as in mice thermogenesis requires upregulation of Pgc-1α and uncoupling protein 1 (Ucp-1) in brown adipose tissue. We found that the Ppt1-KO mice had lower basal body temperature as they aged and developed hypothermia during cold exposure. Surprisingly, this inability to maintain body temperature during cold exposure in Ppt1-KO mice was associated with an adequate upregulation of Pgc-1α and Ucp-1 but with lower levels of sympathetic neurotransmitters in brown adipose tissue. In addition, during baseline conditions, brown adipose tissue of Ppt1-KO mice had less vacuolization (lipid droplets) compared to wild-type animals. After cold stress, wild-type animals had significant decreases whereas Ppt1-KO had insignificant changes in lipid droplets compared with baseline measurements, thus suggesting that Ppt1-KO had less lipolysis in response to cold stress. These results uncover a previously unknown phenotype associated with PPT1 deficiency, that of altered thermoregulation, which is associated with impaired lipolysis and neurotransmitter release to brown adipose tissue during cold exposure. These findings suggest that INCL should be added to the list of neurodegenerative diseases that are linked to alterations in peripheral metabolic processes. In addition, extrapolating these findings clinically, impaired thermoregulation and hypothermia are potential risks in patients with INCL.


Subject(s)
Adipose Tissue, Brown/abnormalities , Adipose Tissue, Brown/physiopathology , Body Temperature Regulation/physiology , Neuronal Ceroid-Lipofuscinoses/enzymology , Neuronal Ceroid-Lipofuscinoses/physiopathology , Thiolester Hydrolases/deficiency , Adenosine Triphosphate/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Animals , Cold Temperature , Disease Models, Animal , Female , Hot Temperature , Ion Channels/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/metabolism , NAD/metabolism , Neuronal Ceroid-Lipofuscinoses/pathology , Neurotransmitter Agents/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Receptors, Adrenergic, beta/metabolism , Thiolester Hydrolases/metabolism , Uncoupling Protein 1 , Ventromedial Hypothalamic Nucleus/metabolism , Ventromedial Hypothalamic Nucleus/pathology , Ventromedial Hypothalamic Nucleus/physiopathology
20.
Mol Pain ; 8: 78, 2012 Oct 23.
Article in English | MEDLINE | ID: mdl-23092240

ABSTRACT

BACKGROUND: Our previous studies have shown that complete Freund's adjuvant (CFA)-induced masseter inflammation and microinjection of the pro-inflammatory cytokine interleukin-1ß (IL-1ß) into the subnucleus interpolaris/subnucleus caudalis transition zone of the spinal trigeminal nucleus (Vi/Vc) can induce contralateral orofacial hyperalgesia in rat models. We have also shown that contralateral hyperalgesia is attenuated with a lesion of the rostral ventromedial medulla (RVM), a critical site of descending pain modulation. Here we investigated the involvement of the RVM-Vi/Vc circuitry in mediating contralateral orofacial hyperalgesia after an injection of CFA into the masseter muscle. RESULTS: Microinjection of the IL-1 receptor antagonist (5 nmol, n=6) into the ipsilateral Vi/Vc attenuated the CFA-induced contralateral hyperalgesia but not the ipsilateral hyperalgesia. Intra-RVM post-treatment injection of the NK1 receptor antagonists, RP67580 (0.5-11.4 nmol) and L-733,060 (0.5-11.4 nmol), attenuated CFA-induced bilateral hyperalgesia and IL-1ß induced bilateral hyperalgesia. Serotonin depletion in RVM neurons prior to intra-masseter CFA injection prevented the development of contralateral hyperalgesia 1-3 days after CFA injection. Inhibition of 5-HT(3) receptors in the contralateral Vi/Vc with direct microinjection of the select 5-HT(3) receptor antagonist, Y-25130 (2.6-12.9 nmol), attenuated CFA-induced contralateral hyperalgesia. Lesions to the ipsilateral Vc prevented the development of ipsilateral hyperalgesia but did not prevent the development of contralateral hyperalgesia. CONCLUSIONS: These results suggest that the development of CFA-induced contralateral orofacial hyperalgesia is mediated through descending facilitatory mechanisms of the RVM-Vi/Vc circuitry.


Subject(s)
Facial Pain/pathology , Hyperalgesia/pathology , Intralaminar Thalamic Nuclei/pathology , Medulla Oblongata/pathology , Nerve Net/pathology , Trigeminal Caudal Nucleus/pathology , Ventromedial Hypothalamic Nucleus/pathology , Animals , Facial Pain/complications , Facial Pain/metabolism , Freund's Adjuvant , Hyperalgesia/complications , Hyperalgesia/metabolism , Injections , Intralaminar Thalamic Nuclei/metabolism , Male , Medulla Oblongata/metabolism , Models, Biological , Nerve Net/metabolism , Neurokinin-1 Receptor Antagonists , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-1/antagonists & inhibitors , Receptors, Interleukin-1/metabolism , Receptors, Neurokinin-1/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Reproducibility of Results , Serotonin/deficiency , Serotonin/metabolism , Trigeminal Caudal Nucleus/metabolism , Ventromedial Hypothalamic Nucleus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...