Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Elife ; 112022 10 25.
Article in English | MEDLINE | ID: mdl-36281647

ABSTRACT

Decades of studies have revealed molecular and neural circuit bases for body weight homeostasis. Neural hormone oxytocin (Oxt) has received attention in this context because it is produced by neurons in the paraventricular hypothalamic nucleus (PVH), a known output center of hypothalamic regulation of appetite. Oxt has an anorexigenic effect, as shown in human studies, and can mediate satiety signals in rodents. However, the function of Oxt signaling in the physiological regulation of appetite has remained in question, because whole-body knockout (KO) of Oxt or Oxt receptor (Oxtr) has little effect on food intake. We herein show that acute conditional KO (cKO) of Oxt selectively in the adult PVH, but not in the supraoptic nucleus, markedly increases body weight and food intake, with an elevated level of plasma triglyceride and leptin. Intraperitoneal administration of Oxt rescues the hyperphagic phenotype of the PVH Oxt cKO model. Furthermore, we show that cKO of Oxtr selectively in the posterior hypothalamic regions, especially the arcuate hypothalamic nucleus, a primary center for appetite regulations, phenocopies hyperphagic obesity. Collectively, these data reveal that Oxt signaling in the arcuate nucleus suppresses excessive food intake.


Subject(s)
Leptin , Oxytocin , Humans , Mice , Animals , Hyperphagia , Obesity/genetics , Paraventricular Hypothalamic Nucleus , Body Weight , Hypothalamus , Hypothalamus, Posterior , Triglycerides
2.
J Food Drug Anal ; 29(4): 653-675, 2021 12 15.
Article in English | MEDLINE | ID: mdl-35649140

ABSTRACT

Oxytocin (Oxt) is known to regulate social communication, stress and body weight. The activation of Oxt receptors (OTR) has clinical potential to abate stress disorders and metabolic syndrome. Kamikihito (KKT) is a traditional Japanese medicine used to treat psychological stress-related disorders. We investigated the effects of KKT, its ingredients and chemical components on Oxt neurons and OTR. C-Fos expression was examined after oral and peripheral administration of KKT in rats. Electrophysiological change of Oxt neurons and Oxt release upon application of KKT were measured in rat brain slice. The direct effect of KKT, its ingredients and its chemical components were examined by cytosolic Ca2+([Ca2+]i) measurement in Oxt neurons and OTR-expressing HEK293 cells. Both intraperitoneal and oral administration of KKT in rats induced c-Fos expression in neurons of the paraventricular nucleus (PVN) including Oxt neurons. Application of KKT induced activation of Oxt neurons and Oxt release. KKT increased [Ca2+]i in OTR-expressing HEK293 cells, and failed to activate with OTR antagonist. KKT-induced PVN Oxt neuron activation was also attenuated by OTR antagonist. Seven chemical components (rutin, ursolic acid, (Z )-butylidenephtalide, p-cymene, senkunolide, [6]-shogaol, [8]-shogaol) of three ingredients (Zizyphi Fructus, Angelicae Acutilobae Radix, Zingiberis Rhizoma) from KKT had potential to activate OTR. KKT can directly activate PVN Oxt neurons by interacting with OTR. The interaction of seven chemical components from KKT may contribute to activate OTR. Effect of KKT on Oxt neurons and OTR may contribute to the treatment of Oxt related disorders.


Subject(s)
Oxytocin , Receptors, Oxytocin , Animals , HEK293 Cells , Humans , Japan , Medicine, East Asian Traditional , Oxytocin/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rats , Receptors, Oxytocin/genetics , Receptors, Oxytocin/metabolism
3.
Article in English | MEDLINE | ID: mdl-32719656

ABSTRACT

Excessive intake of fat is a major risk factor for lifestyle-related diseases such as heart disease and also affects brain function such as object recognition memory, social recognition, anxiety behavior, and depression-like behavior. Although oxytocin (OXT) has been reported to improve object recognition, social recognition, anxiety behavior, and depression-like behavior in specific conditions, previous studies did not explore the impact of OXT in high-fat diet (HFD)-fed mice. Furthermore, it remains unclear whether intake of HFD affects OXT/oxytocin receptor (OXTR) in the brain. Here, we demonstrated that peripheral OXT administration improves not only social recognition but also object recognition and depressive-like behavior in HFD-fed mice. In contrast, peripheral OXT administration to HFD-fed male mice increased fear and anxiety-related behavior. In addition, we observed that intake of HFD decreased OXTR and c-fos mRNA expression in the hippocampus, specifically. Furthermore, peripheral OXT administration increased OXT mRNA expression in the hypothalamus. Altogether, these findings suggest that OXT has the potential to improve various recognition memory processes via peripheral administration but also has side effects that increase fear-related behavior in males.


Subject(s)
Behavior, Animal/physiology , Memory/physiology , Obesity/physiopathology , Obesity/psychology , Oxytocin/physiology , Animals , Anxiety/physiopathology , Depression/physiopathology , Diet, High-Fat , Fear/physiology , Hypothalamus/drug effects , Hypothalamus/physiology , Male , Memory/drug effects , Mice, Inbred C57BL , Neurons/drug effects , Neurons/physiology , Oxytocin/administration & dosage , Receptors, Oxytocin/physiology , Social Behavior
4.
Nature ; 582(7811): 246-252, 2020 06.
Article in English | MEDLINE | ID: mdl-32499648

ABSTRACT

A wealth of specialized neuroendocrine command systems intercalated within the hypothalamus control the most fundamental physiological needs in vertebrates1,2. Nevertheless, we lack a developmental blueprint that integrates the molecular determinants of neuronal and glial diversity along temporal and spatial scales of hypothalamus development3. Here we combine single-cell RNA sequencing of 51,199 mouse cells of ectodermal origin, gene regulatory network (GRN) screens in conjunction with genome-wide association study-based disease phenotyping, and genetic lineage reconstruction to show that nine glial and thirty-three neuronal subtypes are generated by mid-gestation under the control of distinct GRNs. Combinatorial molecular codes that arise from neurotransmitters, neuropeptides and transcription factors are minimally required to decode the taxonomical hierarchy of hypothalamic neurons. The differentiation of γ-aminobutyric acid (GABA) and dopamine neurons, but not glutamate neurons, relies on quasi-stable intermediate states, with a pool of GABA progenitors giving rise to dopamine cells4. We found an unexpected abundance of chemotropic proliferation and guidance cues that are commonly implicated in dorsal (cortical) patterning5 in the hypothalamus. In particular, loss of SLIT-ROBO signalling impaired both the production and positioning of periventricular dopamine neurons. Overall, we identify molecular principles that shape the developmental architecture of the hypothalamus and show how neuronal heterogeneity is transformed into a multimodal neural unit to provide virtually infinite adaptive potential throughout life.


Subject(s)
Gene Expression Regulation, Developmental , Hypothalamus/cytology , Hypothalamus/embryology , Morphogenesis , Animals , Cell Differentiation , Cell Lineage , Dopamine/metabolism , Dopaminergic Neurons/cytology , Dopaminergic Neurons/metabolism , Ectoderm/cytology , Ectoderm/metabolism , Female , GABAergic Neurons/cytology , GABAergic Neurons/metabolism , Gene Regulatory Networks , Genome-Wide Association Study , Glutamic Acid/metabolism , Hypothalamus/metabolism , Male , Mice , Morphogenesis/genetics , Nerve Tissue Proteins/metabolism , Neuroglia/cytology , Neuroglia/metabolism , Neuropeptides/metabolism , Neurotransmitter Agents/metabolism , Receptors, Immunologic/metabolism , Regulon/genetics , Signal Transduction , Transcription Factors/metabolism , gamma-Aminobutyric Acid/metabolism , Roundabout Proteins
5.
Nat Commun ; 11(1): 1885, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32313029

ABSTRACT

The oxytocin receptor (OTR) plays critical roles in social behavior development. Despite its significance, brain-wide quantitative understanding of OTR expression remains limited in postnatally developing brains. Here, we develop postnatal 3D template brains to register whole brain images with cellular resolution to systematically quantify OTR cell densities. We utilize fluorescent reporter mice (Otrvenus/+) and find that cortical regions show temporally and spatially heterogeneous patterns with transient postnatal OTR expression without cell death. Cortical OTR cells are largely glutamatergic neurons with the exception of cells in layer 6b. Subcortical regions show similar temporal regulation except the hypothalamus and two hypothalamic nuclei display sexually dimorphic OTR expression. Lack of OTR expression correlates with reduced dendritic spine densities in selected cortical regions of developing brains. Lastly, we create a website to visualize our high-resolution imaging data. In summary, our research provides a comprehensive resource for postnatal OTR expression in the mouse brain.


Subject(s)
Brain/growth & development , Brain/metabolism , Receptors, Oxytocin/genetics , Receptors, Oxytocin/metabolism , Animals , Brain/diagnostic imaging , Brain Mapping , Down-Regulation , Female , Gene Expression Regulation, Developmental , Hypothalamus/metabolism , Male , Mice , Mice, Knockout , Mice, Transgenic , Nervous System/growth & development , Neurons/metabolism , Oxytocin/metabolism , Sex Characteristics
6.
J Neuroendocrinol ; 32(4): e12815, 2020 04.
Article in English | MEDLINE | ID: mdl-31770473

ABSTRACT

A monoclonal antibody against oxytocin was generated in 7a5 hybridoma cells derived from myeloma cells and lymphocytes from the spleen of mice immunised with a synthetic oxytocin peptide. The 7a5 monoclonal antibody bound with oxytocin in enzyme-linked immunosorbent assays. 7a5 cell growth medium was diluted up to 5000-fold and used for immunohistochemistry. First, to test the specificity of the 7a5 antibody against oxytocin, we stained brain tissues of oxytocin knockout mice, comprising mice in which the first exon of the oxytocin-neurophysin gene is deleted. No 7a5 immunoreactivity was detected in the paraventricular nucleus (PVN) of the hypothalamus of oxytocin knockout mice; however, this area was strongly stained with the anti-vasopressin polyclonal antibody, HM07. Tissue preparations of the wild-type mouse PVN and supraoptic nucleus (SON) displayed 7a5 immunoreactivity that was indistinguishable from the staining produced with an anti-oxytocin polyclonal antibody, HM06. The immunoreactivity of HM06 in the PVN was similar to that of an anti-oxytocin monoclonal antibody, PS38. We then examined the cross-reactivity of 7a5 with arginine vasopressin. The majority of cell soma and processes stained by 7a5 were not co-stained with the vasopressin antibody in SON and PVN regions. Furthermore, the suprachiasmatic nucleus was stained by the vasopressin antibody but not by 7a5. These results demonstrate that 7a5 is a new anti-oxytocin monoclonal antibody recognising oxytocin and not vasopressin; therefore, 7a5 can be used to investigate the role of oxytocin in the brain.


Subject(s)
Hypothalamus/metabolism , Immunohistochemistry , Neurons/metabolism , Oxytocin/metabolism , Animals , Antibodies, Monoclonal , Mice , Mice, Knockout
7.
PLoS One ; 13(8): e0202476, 2018.
Article in English | MEDLINE | ID: mdl-30106994

ABSTRACT

Mouse Peg3 encodes a DNA-binding protein involved in the milk letdown process. In the current study, we tested whether PEG3 controls the expression of the oxytocin receptor gene. According to the results, PEG3 directly binds to a genomic region within the 3rd exon of Oxtr, which contains a DNA-binding motif for PEG3. In nursing female mice, removal of PEG3 resulted in the increased expression of Oxtr in mammary epithelial cells and also in the hypothalamus. This suggests a repressor role of PEG3 in the expression of Oxtr in these tissues. Overall, this study suggests that Peg3 may function as a direct transcriptional regulator for Oxtr expression that acts to moderate the milk letdown process.


Subject(s)
Gene Expression Regulation/physiology , Hypothalamus/metabolism , Kruppel-Like Transcription Factors/metabolism , Lactation/physiology , Mammary Glands, Animal/metabolism , Receptors, Oxytocin/biosynthesis , Response Elements , Animals , Female , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Mutant Strains , Receptors, Oxytocin/genetics
8.
Anim Sci J ; 89(8): 1169-1177, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29770537

ABSTRACT

Human epigenetic studies suggest that consumption of seaweed prevents mammary cancer, which possibly is explained by iodine daily intake. In this study, we evaluated the efficacy of dietary intake of iodine-enriched eggs on mammary tumor incidence caused by the expression of activated type ErbB2. Female transgenic mice were divided into three groups, and fed a basic diet, a diet supplemented with ordinary eggs, or with iodine-enriched eggs. The number of mammary tumors greater than 5 mm in diameter was recorded in mice at 6 months of age. We report that the average number of mammary tumors per mouse was significantly lower in the iodine-enriched egg-added diet group than in either the basic diet or ordinary egg diet groups. These results indicate that iodine intake through livestock-derived products can reduce the incidence of mammary cancers caused by the expression of activated type ErbB2.


Subject(s)
Animal Feed , Diet/veterinary , Dietary Supplements , Eggs , Gene Expression , Iodine/administration & dosage , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/prevention & control , Receptor, ErbB-2/genetics , Transcriptional Activation , Animals , Female , Incidence , Mice, Transgenic
9.
J Neurosci ; 35(35): 12248-60, 2015 Sep 02.
Article in English | MEDLINE | ID: mdl-26338335

ABSTRACT

The major regulator of the neuroendocrine stress response in the brain is corticotropin releasing factor (CRF), whose transcription is controlled by CREB and its cofactors CRTC2/3 (TORC2/3). Phosphorylated CRTCs are sequestered in the cytoplasm, but rapidly dephosphorylated and translocated into the nucleus following a stressful stimulus. As the stress response is attenuated by oxytocin (OT), we tested whether OT interferes with CRTC translocation and, thereby, Crf expression. OT (1 nmol, i.c.v.) delayed the stress-induced increase of nuclear CRTC3 and Crf hnRNA levels in the paraventricular nucleus of male rats and mice, but did not affect either parameter in the absence of the stressor. The increase in Crf hnRNA levels at later time points was parallel to elevated nuclear CRTC2/3 levels. A direct effect of Thr(4) Gly(7)-OT (TGOT) on CRTC3 translocation and Crf expression was found in rat primary hypothalamic neurons, amygdaloid (Ar-5), hypothalamic (H32), and human neuroblastoma (Be(2)M17) cell lines. CRTC3, but not CRCT2, knockdown using siRNA in Be(2)M17 cells prevented the effect of TGOT on Crf hnRNA levels. Chromatin-immunoprecipitation demonstrated that TGOT reduced CRTC3, but not CRTC2, binding to the Crf promoter after 10 min of forskolin stimulation. Together, the results indicate that OT modulates CRTC3 translocation, the binding of CRTC3 to the Crf promoter and, ultimately, transcription of the Crf gene. SIGNIFICANCE STATEMENT: The neuropeptide oxytocin has been proposed to reduce hypothalamic-pituitary-adrenal (HPA) axis activation during stress. The underlying mechanisms are, however, elusive. In this study we show that activation of the oxytocin receptor in the paraventricular nucleus delays transcription of the gene encoding corticotropin releasing factor (Crf), the main regulator of the stress response. It does so by sequestering the coactivator of the transcription factor CREB, CRTC3, in the cytosol, resulting in reduced binding of CRTC3 to the Crf gene promoter and subsequent Crf gene expression. This novel oxytocin receptor-mediated intracellular mechanism might provide a basis for the treatment of exaggerated stress responses in the future.


Subject(s)
CREB-Binding Protein/metabolism , Corticotropin-Releasing Hormone/metabolism , Gene Expression Regulation , Oxytocin/pharmacology , Stress, Psychological/metabolism , Thromboplastin/metabolism , Animals , Cells, Cultured , Colforsin/pharmacology , Corticotropin-Releasing Hormone/genetics , Disease Models, Animal , Female , Gene Expression Regulation/drug effects , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/physiology , Oxytocics/pharmacology , Oxytocics/therapeutic use , Oxytocin/analogs & derivatives , Oxytocin/therapeutic use , Protein Transport/drug effects , Protein Transport/genetics , Rats , Rats, Wistar , Receptors, Oxytocin/metabolism , Signal Transduction/drug effects , Stress, Psychological/drug therapy , Stress, Psychological/pathology
10.
Clin Nutr ; 33(2): 302-10, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23755843

ABSTRACT

BACKGROUND AND AIMS: Large-scale epidemiological studies have shown that drinking more than two cups of coffee per day reduces the risks of hepatitis and liver cancer. However, the heterogeneity of the human genome requires studies of experimental animal models with defined genetic backgrounds to evaluate the coffee effects on liver diseases. We evaluated the efficacy of coffee consumption with one of experimental animal models for human disease. METHOD: We used the Long Evans Cinnamon (LEC) rat, which onsets severe hepatitis and high incidence of liver cancer, due to the accumulation of copper and iron in livers caused by the genetic mutation in Atp7B gene, and leading to the continuous oxidative stress. We determined the expression of inflammation related genes, and amounts of copper and iron in livers, and incidence of the pre-neoplastic foci in the liver tissue of LEC rats. RESULTS: Coffee administration for 25 weeks delayed the occurrence of hepatitis by two weeks, significantly improved survival, reduced the expression of inflammatory cytokines, and reduced the incidence of small pre-neoplastic liver foci in LEC rats. There was no significant difference in the accumulation of copper and iron in livers, indicating that coffee administration does not affect to the metabolism of these metals. These findings indicate that drinking coffee potentially prevents hepatitis and liver carcinogenesis through its anti-inflammatory effects. CONCLUSION: This study showed the efficacy of coffee in the prevention of hepatitis and liver carcinogenesis in the LEC model.


Subject(s)
Coffee/chemistry , Hepatitis/prevention & control , Inflammation/metabolism , Liver Neoplasms/prevention & control , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Alanine Transaminase/metabolism , Animals , Caffeine/pharmacology , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cinnamates/pharmacology , Copper/adverse effects , Copper/pharmacokinetics , Copper-Transporting ATPases , Gene Expression/drug effects , Glutathione Transferase/metabolism , Interleukin-6/genetics , Interleukin-6/metabolism , Iron/adverse effects , Iron/pharmacokinetics , Liver/drug effects , Liver/metabolism , Liver Neoplasms/chemically induced , Male , Oxidative Stress/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred LEC , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
11.
Endocrinology ; 154(11): 4305-15, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24002032

ABSTRACT

Oxytocin (OXT) and OXT receptor (OXTR) have been implicated in the regulation of energy homeostasis, but the detailed mechanism is still unclear. We recently showed late-onset obesity and impaired cold-induced thermogenesis in male OXTR knockout (Oxtr(-/-)) mice. Here we demonstrate that the OXTR in the hypothalamus has important functions in thermoregulation. Male Oxtr(-/-) mice failed to maintain their body temperatures during exposure to a cold environment. Oxtr(-/-) mice also showed decreased neuronal activation in the thermoregulatory hypothalamic region during cold exposure. Normal cold-induced thermogenesis was recovered in Oxtr(-/-) mice by restoring OXTR to the hypothalamus with an adeno-associated virus-Oxtr vector. In addition, brown adipose tissue (BAT) in Oxtr(-/-) mice contained larger lipid droplets in both 10- and 20-week-old compared with BAT from age-matched Oxtr(+/+) control mice. In BAT, the expression level of ß3-adrenergic receptor at normal temperature was lower in Oxtr(-/-) mice than that in control mice. In contrast, α2A-adrenergic receptor expression level was higher in BAT from Oxtr(-/-) mice in both normal and cold temperatures. Because ß3- and α2A-adrenergic receptors are known to have opposite effects on the thermoregulation, the imbalance of adrenergic receptors is suspected to affect this dysfunction in the thermoregulation. Our study is the first to demonstrate that the central OXT/OXTR system plays important roles in the regulation of body temperature homeostasis.


Subject(s)
Body Temperature Regulation/physiology , Hypothalamus/metabolism , Receptors, Oxytocin/metabolism , Adipose Tissue, Brown , Animals , Cold Temperature , Energy Metabolism , Male , Mice , Mice, Knockout , Oxytocin/genetics , Oxytocin/metabolism , Receptors, Oxytocin/genetics
12.
Proc Natl Acad Sci U S A ; 106(17): 7149-54, 2009 Apr 28.
Article in English | MEDLINE | ID: mdl-19369205

ABSTRACT

We report that oxytocin (OT), a primitive neurohypophyseal hormone, hitherto thought solely to modulate lactation and social bonding, is a direct regulator of bone mass. Deletion of OT or the OT receptor (Oxtr) in male or female mice causes osteoporosis resulting from reduced bone formation. Consistent with low bone formation, OT stimulates the differentiation of osteoblasts to a mineralizing phenotype by causing the up-regulation of BMP-2, which in turn controls Schnurri-2 and 3, Osterix, and ATF-4 expression. In contrast, OT has dual effects on the osteoclast. It stimulates osteoclast formation both directly, by activating NF-kappaB and MAP kinase signaling, and indirectly through the up-regulation of RANK-L. On the other hand, OT inhibits bone resorption by mature osteoclasts by triggering cytosolic Ca(2+) release and NO synthesis. Together, the complementary genetic and pharmacologic approaches reveal OT as a novel anabolic regulator of bone mass, with potential implications for osteoporosis therapy.


Subject(s)
Bone and Bones/metabolism , Oxytocin/metabolism , Animals , Bone and Bones/cytology , Bone and Bones/drug effects , Calcium/metabolism , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Osteogenesis/drug effects , Oxytocin/deficiency , Oxytocin/genetics , Oxytocin/pharmacology
13.
Prog Brain Res ; 170: 79-90, 2008.
Article in English | MEDLINE | ID: mdl-18655874

ABSTRACT

To further define the function of the oxytocin receptor (OXTR) in vivo, we generated mice deficient in the Oxtr gene (Oxtr-/-). Oxtr-/- mice had no obvious deficits in fertility or sexual behaviour, but displayed several aberrations in social behaviours, including male aggression, and mother-offspring interaction. In addition, they showed novel physiological defects including obesity, and dysfunction in body temperature control when exposed to cold. We review here our new findings with Oxtr-/- mice, and introduce newly generated Oxtr-Venus knockin mice as a potential tool for examining molecular physiology of Oxtr-neurons.


Subject(s)
Energy Metabolism , Hypothalamus/physiology , Oxytocin/deficiency , Oxytocin/genetics , Receptors, Oxytocin/deficiency , Receptors, Oxytocin/physiology , Sexual Behavior, Animal , Social Behavior , Animals , Blood Pressure , Body Temperature , Drinking Behavior , Feeding Behavior , Female , Heart Rate , Lactation , Male , Maternal Behavior , Mice , Mice, Knockout , Milk/metabolism , Myometrium/physiology , Oxytocin/physiology , Pregnancy , Receptors, Oxytocin/genetics
14.
Neurosci Lett ; 391(1-2): 22-7, 2005 Dec 31.
Article in English | MEDLINE | ID: mdl-16157455

ABSTRACT

Salt loading reduces neuroendocrine responses to stressful stimuli. Noxious stimuli facilitate noradrenaline release in the hypothalamus and, as a result, activate oxytocin neurones. Here, we examined effects of salt loading upon plasma oxytocin concentrations and noradrenaline release in the hypothalamus after footshocks. Male rats were allowed to drink 2% NaCl for 7 days. Salt loading reduced the footshock-induced increase in plasma oxytocin concentrations and noradrenaline release in the supraoptic nucleus (SON). Acute administration of hypertonic saline also attenuated the footshock-induced noradrenaline increase in the supraoptic nucleus. In contrast, salt loading did not significantly change activation of A1 catecholaminergic neurones in the medulla oblongata, as measured by expression of Fos protein. These data suggest that salt loading presynaptically suppresses noradrenaline release in the hypothalamus and oxytocin release into the blood after footshocks.


Subject(s)
Hypothalamus/metabolism , Norepinephrine/metabolism , Oxytocin/metabolism , Pain/metabolism , Pain/prevention & control , Sodium Chloride/administration & dosage , Administration, Oral , Animals , Electric Stimulation/adverse effects , Hypothalamus/drug effects , Male , Pain/etiology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL