Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters

Database
Language
Affiliation country
Publication year range
1.
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
2.
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
3.
Neuropsychopharmacology ; 37(11): 2522-30, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22781838

ABSTRACT

Prepulse inhibition (PPI) deficits are among the most reproducible phenotypic markers found in schizophrenic patients. We recently reported that nisoxetine, a selective norepinephrine transporter (NET) inhibitor, reversed the PPI deficits that have been identified in dopamine transporter (DAT) knockout (KO) mice. However, the mechanisms underlying nisoxetine-induced PPI recovery in DAT KO mice were unclear in previous experiments. To clarify these mechanisms, PPI was tested after microinjections of nisoxetine into the medial prefrontal cortex (mPFc) or nucleus accumbens (NAc) in wildtype (WT) and DAT KO mice. c-Fos immunohistochemistry provided an indicator of neural activation. Multiple-fluorescent-labeling procedures and the retrograde tracer fluorogold were employed to identify nisoxetine-activated neurons and circuits. Systemic nisoxetine activated the mPFc, the NAc shell, the basolateral amygdala, and the subiculum. Infusions of nisoxetine into the mPFc reversed PPI deficits in DAT KO mice, but produced no changes in WT mice, while infusion of nisoxetine into the NAc had no effect on PPI in both WT and DAT KO mice. Experiments using multiple-fluorescent labeling/fluorogold revealed that nisoxetine activates presumed glutamatergic pyramidal cells that project from the mPFc to the NAc. Activated glutamatergic projections from the mPFc to the NAc appear to have substantial roles in the ability of a NET inhibitor to normalize PPI deficits in DAT KO. Thus, this data suggest that selective NET inhibitors such as nisoxetine might improve information processing deficits in schizophrenia via regulation of cortico-subcortical neuromodulation.


Subject(s)
Brain/metabolism , Dopamine Plasma Membrane Transport Proteins/deficiency , Gene Expression Regulation/genetics , Inhibition, Psychological , Reflex, Startle/genetics , Acoustic Stimulation/adverse effects , Analysis of Variance , Animals , Brain/drug effects , Drug Administration Routes , Fluoxetine/analogs & derivatives , Fluoxetine/pharmacology , Gene Expression Regulation/drug effects , Glutamate Decarboxylase/metabolism , Glutaminase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nerve Tissue Proteins/metabolism , Neurotransmitter Agents/metabolism , Neurotransmitter Uptake Inhibitors/pharmacology , Proto-Oncogene Proteins c-fos/metabolism , Reflex, Startle/drug effects , Sex Factors , Stilbamidines
4.
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
SELECTION OF CITATIONS
SEARCH DETAIL