Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
Behav Brain Res ; 462: 114867, 2024 Mar 28.
Article in English | MEDLINE | ID: mdl-38246394

ABSTRACT

Disruption of the brain serotoninergic (5-HT) system during development induces long-lasting changes in molecular profile, cytoarchitecture, and function of neurons, impacting behavioral regulation throughout life. In male and female rats, we investigate the effect of neonatal tryptophan hydroxylase (TPH) inhibition by using para-chlorophenylalanine (pCPA) on the expression of 5-HTergic system components and neuropeptides related to adolescent social play behavior regulation. We observed sex-dependent 5-HT levels decrease after pCPA-treatment in the dorsal raphe nucleus (DRN) at 17 and 35 days. Neonatal pCPA-treatment increased playing, social and locomotory behaviors assessed in adolescent rats of both sexes. The pCPA-treated rats demonstrated decreased Crh (17 days) and increased Trh (35 days) expression in the hypothalamic paraventricular nucleus (PVN). There was sex dimorphism in Htr2c (17 days) and VGF (35 days) in the prefrontal cortex, with the females expressing higher levels of it than males. Our results indicate that neonatal pCPA-treatment results in a long-lasting and sex-dependent DRN 5-HT synthesis changes, decreased Crh, and increased Trh expression in the PVN, resulting in a hyperactivity-like phenotype during adolescence. The present work demonstrates that the impairment of TPH function leads to neurobehavioral disorders related to hyperactivity and impulsivity, such as attention deficit hyperactivity disorder (ADHD).


Subject(s)
Paraventricular Hypothalamic Nucleus , Serotonin , Rats , Female , Male , Animals , Fenclonine/pharmacology , Paraventricular Hypothalamic Nucleus/metabolism , Serotonin/metabolism , Dorsal Raphe Nucleus/metabolism , Tryptophan Hydroxylase/metabolism
2.
Endocrinology ; 164(11)2023 09 23.
Article in English | MEDLINE | ID: mdl-37823477

ABSTRACT

The hormone ghrelin displays several well-characterized functions, including some with pharmaceutical interest. The receptor for ghrelin, the growth hormone secretagogue receptor (GHSR), is expressed in the hypothalamic paraventricular nucleus (PVH), a critical hub for the integration of metabolic, neuroendocrine, autonomic, and behavioral functions. Here, we performed a neuroanatomical and functional characterization of the neuronal types mediating ghrelin actions in the PVH of male mice. We found that fluorescent ghrelin mainly labels PVH neurons immunoreactive for nitric oxide synthase 1 (NOS1), which catalyze the production of nitric oxide [NO]). Centrally injected ghrelin increases c-Fos in NOS1 PVH neurons and NOS1 phosphorylation in the PVH. We also found that a high dose of systemically injected ghrelin increases the ghrelin level in the cerebrospinal fluid and in the periventricular PVH, and induces c-Fos in NOS1 PVH neurons. Such a high dose of systemically injected ghrelin activates a subset of NOS1 PVH neurons, which do not express oxytocin, via an arcuate nucleus-independent mechanism. Finally, we found that pharmacological inhibition of NO production fully abrogates ghrelin-induced increase of calcium concentration in corticotropin-releasing hormone neurons of the PVH whereas it partially impairs ghrelin-induced increase of plasma glucocorticoid levels. Thus, plasma ghrelin can directly target a subset of NO-producing neurons of the PVH that is involved in ghrelin-induced activation of the hypothalamic-pituitary-adrenal neuroendocrine axis.


Subject(s)
Corticotropin-Releasing Hormone , Ghrelin , Mice , Male , Animals , Corticotropin-Releasing Hormone/metabolism , Ghrelin/pharmacology , Ghrelin/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Hypothalamo-Hypophyseal System/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Neurons/metabolism
3.
J Neuroendocrinol ; 35(11): e13334, 2023 11.
Article in English | MEDLINE | ID: mdl-37667574

ABSTRACT

In addition to being recognised for involvement in cardiovascular control and hydromineral balance, the renin-angiotensin system (RAS) has also been associated with the neuroendocrine control of energy balance. One of the main brain sites for angiotensin II (ANG II)/type 1 receptor (AT1 R) signalling is the subfornical organ (SFO), a circumventricular organ related to the control of autonomic functions, motivated behaviours and energy metabolism. Thus, we hypothesised that circulating ANG II may act on the SFO AT1 R receptors to integrate metabolic and hydromineral balance. We evaluated whether food deprivation can modulate systemic RAS activity and Agrt1a brain expression, and if ANG II/AT1 R signalling influences the hypothalamic expression of mRNAs encoding neuropeptides and food and water ingestion in fed and fasted Wistar rats. We found a significant increase in both ANG I and ANG II plasma levels after 24 and 48 h of fasting. Expression of Agrt1a mRNA in the SFO and paraventricular nucleus (PVN) also increased after food deprivation for 48 h. Treatment of fasted rats with low doses of losartan in drinking water attenuated the decrease in glycemia and meal-associated water intake without changing the expression in PVN or arcuate nucleus of mRNAs encoding selected neuropeptides related to energy homeostasis control. These findings point to a possible role of peripheral ANG II/SFO-AT1 R signalling in the control of refeeding-induced thirst. On the other hand, intracerebroventricular losartan treatment decreased food and water intake over dark time in fed but not in fasted rats.


Subject(s)
Fasting , Subfornical Organ , Animals , Male , Rats , Angiotensin II/pharmacology , Brain/metabolism , Fasting/metabolism , Losartan/pharmacology , Paraventricular Hypothalamic Nucleus/metabolism , Rats, Wistar , Receptor, Angiotensin, Type 1/metabolism , Subfornical Organ/metabolism
4.
J Nutr Biochem ; 119: 109372, 2023 09.
Article in English | MEDLINE | ID: mdl-37169229

ABSTRACT

High-fat diet consumption causes hypothalamic inflammation, dysregulating the leptin pathway, which, in turn, compromises the modulation of hypothalamic neuronal activities and predisposes obesity development. Intermittent fasting (IF) and exercise training (ET) have been demonstrated as efficient interventions to modulate hypothalamic inflammation and neuronal activity. However, no studies have evaluated whether combining these interventions could induce better results in reestablishing hypothalamic homeostasis disrupted by high-fat diet intake. The 8-week-old male C57BL/6 mice were randomly assigned into 2 groups: sedentary mice fed a standard diet (CT), and sedentary mice fed a high-fat diet (HF). After 8 weeks of an HF diet, part of the HF group (now 16 weeks old) was randomly subjected to different interventions for 6 weeks: HF-IF = HF diet mice submitted to IF; HF-T = HF diet mice submitted to ET; HF-IFT = HF diet mice submitted to IF and ET. All interventions decreased the body weight gain induced by high-fat diet intake, associated with reduced calorie consumption in week 14. Only the HF-IFT group presented improved serum insulin, leptin, resistin, and Tnf-alpha levels concomitantly with decreased hypothalamic inflammation. The HF-IFT group also demonstrated increased Pomc mRNA expression associated with enhanced pSTAT3 expression in the hypothalamic arcuate and ventromedial hypothalamic nuclei. Our data indicate that the beneficial effects of the combination of IF and ET on energy homeostasis are associated with increased leptin sensitivity in the hypothalamic arcuate nucleus and ventromedial hypothalamic nucleus, which is likely due to an improvement in hypothalamic inflammatory pathways in these nuclei.


Subject(s)
Diet, High-Fat , Leptin , Male , Mice , Animals , Diet, High-Fat/adverse effects , Intermittent Fasting , Dietary Fats/pharmacology , Mice, Inbred C57BL , Hypothalamus/metabolism , Inflammation/metabolism
5.
Thyroid ; 33(7): 867-876, 2023 07.
Article in English | MEDLINE | ID: mdl-37166378

ABSTRACT

Background: Thyrotropin-releasing hormone (TRH) neurons in the paraventricular nucleus of the hypothalamus (PVN) have been identified as direct regulators of thyrotropin (TSH) and thyroid hormone (TH) levels. They play a significant role in context of negative feedback by TH at the level of TRH gene expression and during fasting when TH levels fall due, in part, to suppression of TRH gene expression. Methods: To test these functions directly for the first time, we used a chemogenetic approach and activated PVN TRH neurons in both fed and fasted mice. Next, to demonstrate the signals that regulate the fasting response in TRH neurons, we activated or inhibited agouti-related protein (AgRP)/neuropeptide Y (NPY) neurons in the arcuate nucleus of the hypothalamus of fed or fasted mice, respectively. To determine if the same TRH neurons responsive to melanocortin signaling mediate negative feedback by TH, we disrupted the thyroid hormone receptor beta (TRß) in all melanocortin 4 receptor (MC4R) neurons in the PVN. Results: Activation of TRH neurons led to increased TSH and TH levels within 2 hours demonstrating the specific role of PVN TRH neurons in the regulation of the hypothalamic-pituitary-thyroid (HPT) axis. Moreover, activation of PVN TRH neurons prevented the fall in TH levels in fasting mice. Stimulation of AgRP/NPY neurons led to a fall in TH levels despite increasing feeding. Inhibition of these same neurons prevented the fall in TH levels during a fast presumably via their ability to directly regulate PVN TRH neurons via, in part, the MC4R. Surprisingly, TH-mediated feedback was not impaired in mice lacking TRß in MC4R neurons. Conclusions: TRH neurons are major regulators of the HPT axis and the fasting-induced suppression of TH levels. The latter relies, at least in part, on the activation of AgRP/NPY neurons in the arcuate nucleus. Interestingly, present data do not support an important role for TRß signaling in regulating MC4R neurons in the PVN. Thus, it remains possible that different subsets of TRH neurons in the PVN mediate responses to energy balance and to TH feedback.


Subject(s)
Thyrotropin-Releasing Hormone , Thyrotropin , Mice , Animals , Thyrotropin-Releasing Hormone/metabolism , Thyrotropin/metabolism , Agouti-Related Protein/genetics , Agouti-Related Protein/metabolism , Thyroid Gland/metabolism , Pituitary Hormone-Releasing Hormones/metabolism , Hypothalamus , Thyroid Hormones/metabolism , Paraventricular Hypothalamic Nucleus , Neurons/metabolism
6.
Reprod Toxicol ; 119: 108410, 2023 08.
Article in English | MEDLINE | ID: mdl-37211340

ABSTRACT

We previously reported that female rats placed on a diet containing refined carbohydrates (HCD) resulted in obesity and reproductive abnormalities, such as high serum LH concentration and abnormal ovarian function. However, the impacts at the hypothalamic-pituitary (HP) function, specifically regarding pathways linked to reproductive axis modulation are unknown. In this study, we assessed whether subacute feeding with HCD results in abnormal reproductive control in the HP axis. Female rats were fed with HCD for 15 days and reproductive HP axis morphophysiology was assessed. HCD reduced hypothalamic mRNA expression (Kiss1, Lepr, and Amhr2) and increased pituitary LHß+ cells. These changes likely contribute to the increase in serum LH concentration observed in HCD. Blunted estrogen negative feedback was observed in HCD, with increased kisspeptin protein expression in the arcuate nucleus of the hypothalamus (ARH), lower LHß+ cells and LH concentration in ovariectomized (OVX)+HCD rats. Thus, these data suggest that HCD feeding led to female abnormal reproductive control of HP axis.


Subject(s)
Hypothalamus , Obesity , Rats , Female , Animals , Hypothalamus/metabolism , Obesity/metabolism , Arcuate Nucleus of Hypothalamus/metabolism , Diet , Carbohydrates , Kisspeptins/genetics , Kisspeptins/metabolism
8.
Cell Biosci ; 12(1): 118, 2022 Jul 28.
Article in English | MEDLINE | ID: mdl-35902915

ABSTRACT

BACKGROUND: The activation of the hypothalamic-pituitary-adrenal (HPA) axis is essential for metabolic adaptation in response to fasting. However, the neurocircuitry connecting changes in the peripheral energy stores to the activity of hypothalamic paraventricular corticotrophin-releasing factor (CRFPVN) neurons, the master controller of the HPA axis activity, is not completely understood. Our main goal was to determine if hypothalamic arcuate nucleus (ARC) POMC and AgRP neurons can communicate fasting-induced changes in peripheral energy stores, associated to a fall in plasma leptin levels, to CRFPVN neurons to modulate the HPA axis activity in mice. RESULTS: We observed increased plasma corticosterone levels associate with increased CRFPVN mRNA expression and increased CRFPVN neuronal activity in 36 h fasted mice. These responses were associated with a fall in plasma leptin levels and changes in the mRNA expression of Agrp and Pomc in the ARC. Fasting-induced decrease in plasma leptin partially modulated these responses through a change in the activity of ARC neurons. The chemogenetic activation of POMCARC by DREADDs did not affect fasting-induced activation of the HPA axis. DREADDs inhibition of AgRPARC neurons reduced the content of CRFPVN and increased its accumulation in the median eminence but had no effect on corticosterone secretion induced by fasting. CONCLUSION: Our data indicate that AgRPARC neurons are part of the neurocircuitry involved in the coupling of PVNCRF activity to changes in peripheral energy stores induced by prolonged fasting.

9.
Int J Mol Sci ; 23(10)2022 May 16.
Article in English | MEDLINE | ID: mdl-35628338

ABSTRACT

Low-grade inflammation of the hypothalamus is associated with the disturbance of energy balance. The endocannabinoid system has been implicated in the development and maintenance of obesity as well as in the control of immune responses. The type 2 cannabinoid receptor (CB2) signaling has been associated with anti-inflammatory effects. Therefore, in high fat diet (HFD)-induced obese mice, we modulated CB2 signaling and investigated its effects on energy homeostasis and hypothalamic microgliosis/astrogliosis. We observed no effect on caloric intake and body weight gain in control diet-fed animals that received prolonged icv infusion of the CB2 receptor agonist HU308. Interestingly, we observed a decrease in glucose tolerance in HFD-fed animals treated with HU308. Prolonged icv infusion of HU308 increases astrogliosis in the ventromedial nucleus (VMH) of obese animals and reduced HFD-induced microgliosis in the hypothalamic arcuate (ARC) but not in the paraventricular (PVN) or VMH nuclei. These data indicate that central CB2 signaling modulates glucose homeostasis and glial reactivity in obesogenic conditions, irrespective of changes in body weight.


Subject(s)
Diet, High-Fat , Gliosis , Animals , Body Weight , Brain , Diet, High-Fat/adverse effects , Glucose , Hypothalamus , Mice , Obesity/etiology
10.
Front Nutr ; 9: 858320, 2022.
Article in English | MEDLINE | ID: mdl-35445066

ABSTRACT

Intermittent fasting (IF) is a popular intervention used to fight overweight/obesity. This condition is accompanied by hypothalamic inflammation, limiting the proper signaling of molecular pathways, with consequent dysregulation of food intake and energy homeostasis. This mini-review explored the therapeutic modulation potential of IF regarding the disruption of these molecular pathways. IF seems to modulate inflammatory pathways in the brain, which may also be correlated with the brain-microbiota axis, improving hypothalamic signaling of leptin and insulin, and inducing the autophagic pathway in hypothalamic neurons, contributing to weight loss in obesity. Evidence also suggests that when an IF protocol is performed without respecting the circadian cycle, it can lead to dysregulation in the expression of circadian cycle regulatory genes, with potential health damage. In conclusion, IF may have the potential to be an adjuvant treatment to improve the reestablishment of hypothalamic responses in obesity.

11.
J Neuroendocrinol ; 33(5): e12976, 2021 Apr 26.
Article in English | MEDLINE | ID: mdl-33900672

ABSTRACT

The present study investigated the hormonal and neural responses to stress in a perimenopause animal model induced by 4-vinylcyclohexene diepoxide (VCD), which induces progressive follicular depletion in rodents, allowing studies on the transition to ovarian failure. Female rats, aged 28 days old, were s.c. injected for 15 consecutive days with corn oil or VCD. At 85 ± 5 days after the onset of treatment, the jugular vein was cannulated in the afternoon of metoestrus and in next morning (dioestrus) at 10.00 am, rats were subjected to 30 minutes of restraint stress. Blood samples were withdrawn before (-5 minutes), during (2, 5, 15 and 30 minutes) and after (45, 60 and 90 minutes) stress and plasma prolactin, progesterone and corticosterone levels were measured. Animals were perfused, brains processed for c-Fos/tyrosine hydroxylase (TH) in the locus coeruleus (LC) and c-Fos/corticotrophin-releasing factor (CRF) in the paraventricular nucleus (PVN). In unstressed rats the density of ß-endorphin fibres was assessed in LC and PVN. In VCD-treated rats, stress-induced prolactin peak was higher, basal and peak progesterone levels were lower, and both levels of corticosterone were similar to controls. However, the recovery period was longer for both adrenal hormones. In VCD-treated rats the number of c-Fos/TH and c-Fos/CRF-immunoreactive neurones was higher whereas the density of ß-endorphin fibres was lower in LC and PVN. We surmise that the hyperactivity of the LC and PVN neurones in VCD-treated rats may be a result of the lower progesterone levels that resulted in the decrease of ß-endorphin content in both nuclei, thus impairing the negative-feedback mechanism in the recovery period.

12.
Mol Neurobiol ; 58(3): 1036-1051, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33083963

ABSTRACT

The serotoninergic system plays an important role in the ontogeny of the mammalian central nervous system, and changes in serotonin production during development may lead to permanent changes in brain cytoarchitecture and function. The present study investigated the programming effects of neonatal serotonin depletion on behavior and molecular components of the serotoninergic system in adult male and female rats. Subcutaneous para-chlorophenylalanine (pCPA) administration (100 mg kg-1) was performed daily on postnatal days 8-16 to deplete brain serotonin content. During adulthood, elevated plus-maze, open field, social interaction, forced swimming, and food, saline, and sucrose intake tests were performed. Relative expression of serotonin neurotransmission components in several brain areas was determined by qPCR. Additionally, serotonin immunofluorescence and neuropeptide mRNA expression were assessed in dorsal raphe (DRN) and paraventricular (PVN) nuclei, respectively. Rat performance in behavioral tests demonstrated a general increase in locomotor activity and active escape behavior as well as decreased anxiety-like behavior after neonatal brain serotonin depletion. The behavioral programming effects due to neonatal serotonin depletion were more pronounced in females than males. At the gene expression level, the mRNA of Tph1 and Tph2 were lower in DRN while Htr2c was higher in the amygdala of pCPA-treated males, while Htr1a, Htr2c, Oxt, Avp, Crh, and Trh were not different in any treatments or sex in PVN. The results indicate that neonatal serotonin depletion has long-term consequences on locomotion and anxiety-like behavior associated with long-lasting molecular changes in the brain serotoninergic system in adult rats.


Subject(s)
Aging/pathology , Anti-Anxiety Agents/metabolism , Serotonin/deficiency , Sex Characteristics , Amygdala/metabolism , Animals , Animals, Newborn , Body Weight , Brain/metabolism , Dorsal Raphe Nucleus/metabolism , Elevated Plus Maze Test , Feeding Behavior , Female , Gene Expression Regulation , Male , Open Field Test , Paraventricular Hypothalamic Nucleus/metabolism , Prefrontal Cortex/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Serotonin/metabolism , Social Interaction , Swimming
13.
J Endocrinol ; 242(2): 125-138, 2019 08.
Article in English | MEDLINE | ID: mdl-31189132

ABSTRACT

Adrenalectomy (ADX) induces hypophagia and glucocorticoids counter-regulate the peripheral metabolic effects of insulin. This study evaluated the effects of ADX on ICV (lateral ventricle) injection of insulin-induced changes on food intake, mRNA expression of hypothalamic neuropeptides (insulin receptor (InsR), proopiomelanocortin, cocaine and amphetamine-regulated transcript (Cart), agouti-related protein, neuropeptide Y (Npy) in the arcuate nucleus of the hypothalamus (ARC), corticotrophin-releasing factor in the paraventricular nucleus of the hypothalamus) and hypothalamic protein content of insulin signaling-related molecules (insulin receptor substrate (IRS) 1, protein kinase B (AKT), extracellular-signal-regulated kinase (ERK1/2), c-Jun N-terminal kinase (JNK), protein tyrosine phosphatase-1B (PTP1B) and T cell protein tyrosine phosphatase (TCPTP)) Compared with sham animals, ADX increased the hypothalamic content of pJNK/JNK, PTP1B and TCPTP, as well as decreased mRNA expression of InsR, and corticosterone (B) treatment reversed these effects. Insulin central injection enhanced hypothalamic content of pAKT/AKT and Cart mRNA expression, decreased Npy mRNA expression and food intake only in sham rats, without effects in ADX and ADX + B rats. Insulin did not alter the hypothalamic phosphorylation of IRS1 and ERK1/2 in the three experimental groups. These data demonstrate that ADX reduces the expression of InsR and increases insulin counter-regulators in the hypothalamus, as well as ADX abolishes hypophagia, activation of hypothalamic AKT pathway and changes in Cart and Npy mRNA expression in the ARC induced by insulin. Thus, the higher levels of insulin counter-regulatory proteins and lower expression of InsR in the hypothalamus are likely to underlie impaired insulin-induced hypophagia and responses in the hypothalamus after ADX.


Subject(s)
Adrenalectomy/methods , Eating/drug effects , Hypothalamus/drug effects , Insulin/pharmacology , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Corticosterone/pharmacology , Gene Expression/drug effects , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/pharmacology , Hypothalamus/metabolism , Injections, Intraventricular , Insulin/administration & dosage , Male , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Rats, Wistar , Receptor, Insulin/genetics , Receptor, Insulin/metabolism
14.
Endocrinology ; 158(11): 3929-3942, 2017 11 01.
Article in English | MEDLINE | ID: mdl-28938405

ABSTRACT

Leptin is a permissive factor for puberty initiation, participating as a metabolic cue in the activation of the kisspeptin (Kiss1)-gonadotropin-releasing hormone neuronal circuitry; however, it has no direct effect on Kiss1 neurons. Leptin acts on hypothalamic cocaine- and amphetamine-regulated transcript (CART) neurons, participating in the regulation of energy homeostasis. We investigated the influence of a short-term high-fat diet (HFD) on the effect of leptin on puberty timing. Kiss1-hrGFP female mice received a HFD or regular diet (RD) after weaning at postnatal day (PN)21 and were studied at PN28 and PN32. The HFD increased body weight and plasma leptin concentrations and decreased the age at vaginal opening (HFD, 32 ± 0.53 days; RD, 38 ± 0.67 days). Similar colocalization of neurokinin B and dynorphin in Kiss1-hrGFP neurons of the arcuate nucleus (ARC) was observed between the HFD and RD groups. The HFD increased CART expression in the ARC and Kiss1 messenger RNA expression in the anteroventral periventricular (AVPV)/anterior periventricular (Pe). The HFD also increased the number of ARC CART neurons expressing leptin-induced phosphorylated STAT3 (signal transducer and activator of transcription 3) at PN32. Close apposition of CART fibers to Kiss1-hrGFP neurons was observed in the ARC of both RD- and HFD-fed mice. In conclusion, these data reinforce the notion that a HFD increases kisspeptin expression in the AVPV/Pe and advances puberty initiation. Furthermore, we have demonstrated that the HFD-induced earlier puberty is associated with an increase in CART expression in the ARC. Therefore, these data indicate that CART neurons in the ARC can mediate the effect of leptin on Kiss1 neurons in early puberty induced by a HFD.


Subject(s)
Diet, High-Fat , Dietary Fats/pharmacology , Leptin/metabolism , Leptin/pharmacology , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Sexual Maturation/drug effects , Animals , Female , Mice , Mice, Transgenic , Neurons/metabolism , Time Factors
15.
Int J Mol Sci ; 18(7)2017 Jul 04.
Article in English | MEDLINE | ID: mdl-28677618

ABSTRACT

Metabolic endotoxemia contributes to low-grade inflammation in obesity, which causes insulin resistance due to the activation of intracellular proinflammatory pathways, such as the c-Jun N-terminal Kinase (JNK) cascade in the hypothalamus and other tissues. However, it remains unclear whether the proinflammatory process precedes insulin resistance or it appears because of the development of obesity. Hypothalamic low-grade inflammation was induced by prolonged lipopolysaccharide (LPS) exposure to investigate if central insulin resistance is induced by an inflammatory stimulus regardless of obesity. Male Wistar rats were treated with single (1 LPS) or repeated injections (6 LPS) of LPS (100 µg/kg, IP) to evaluate the phosphorylation of the insulin receptor substrate-1 (IRS1), Protein kinase B (AKT), and JNK in the hypothalamus. Single LPS increased the expression of pIRS1, pAKT, and pJNK, whereas the repeated LPS treatment failed to recruit pIRS1 and pAKT. The 6 LPS treated rats showed increased total JNK and pJNK. The 6 LPS rats became unresponsive to the hypophagic effect induced by central insulin administration (12 µM/5 µL, ICV). Prolonged exposure to LPS (24 h) impaired the insulin-induced AKT phosphorylation and the translocation of the transcription factor forkhead box protein O1 (FoxO1) from the nucleus to the cytoplasm of the cultured hypothalamic GT1-7 cells. Central administration of the JNK inhibitor (20 µM/5 µL, ICV) restored the ability of insulin to phosphorylate IRS1 and AKT in 6 LPS rats. The present data suggest that an increased JNK activity in the hypothalamus underlies the development of insulin resistance during prolonged exposure to endotoxins. Our study reveals that weight gain is not mandatory for the development of hypothalamic insulin resistance and the blockade of proinflammatory pathways could be useful for restoring the insulin signaling during prolonged low-grade inflammation as seen in obesity.


Subject(s)
Body Weight , Hypothalamus/metabolism , Inflammation/etiology , Inflammation/metabolism , Insulin Resistance , Lipopolysaccharides/adverse effects , Animals , Disease Models, Animal , Endotoxemia , Inflammation/pathology , Insulin/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Male , Neurons/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Rats , Signal Transduction
16.
Horm Behav ; 93: 166-174, 2017 07.
Article in English | MEDLINE | ID: mdl-28576646

ABSTRACT

Acute administration of lipopolysaccharide (LPS) from Gram-negative bacteria induces hypophagia. However, the repeated administration of LPS leads to desensitization of hypophagia, which is associated with increased hypothalamic p-AMPK expression. Because ghrelin and endocannabinoids modulate AMPK activity in the hypothalamus, we hypothesized that these neuromodulators play a role in the reversal of tolerance to hypophagia in rats under long-term exposure to LPS. Male Wistar rats were treated with single (1 LPS, 100µg/kg body weight, ip) or repeated injections of LPS over 6days (6 LPS). Food intake was reduced in the 1 LPS, but not in the 6 LPS group. 6 LPS rats showed an increased serum concentration of acylated ghrelin and reduced ghrelin receptor mRNA expression in the hypothalamus. Ghrelin injection (40µg/kg body weight, ip) increased food intake, body weight gain, p-AMPK hypothalamic expression, neuropeptide Y (NPY) and Agouti related peptide (AgRP) mRNA expression in control animals (Saline). However, in 6 LPS rats, ghrelin did not alter these parameters. Central administration of a CB1R antagonist (AM251, 200ng/µl in 5µl/rat) induced hypophagia in 6 LPS animals, suggesting that the endocannabinoid system contributes to preserved food intake during LPS tolerance. In the presence of AM251, the ability of ghrelin to phosphorylate AMPK in the hypothalamus of 6 LPS group was restored, but not its orexigenic effect. Our data highlight that the orexigenic effects of ghrelin require CB1R signaling downstream of AMPK activation. Moreover, CB1R-mediated pathways contribute to the absence of hypophagia during repeated exposure to endotoxin.


Subject(s)
Adenylate Kinase/metabolism , Ghrelin/metabolism , Hypothalamus/drug effects , Lipopolysaccharides/pharmacology , Receptor, Cannabinoid, CB1/metabolism , Animals , Endocannabinoids/metabolism , Hypothalamus/metabolism , Male , Neuropeptide Y/metabolism , Phosphorylation/drug effects , Piperidines/pharmacology , Pyrazoles/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptors, Ghrelin/metabolism , Signal Transduction/drug effects
17.
Mol Metab ; 5(6): 379-391, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27257598

ABSTRACT

OBJECTIVE: Hypophagia and increased energy expenditure under inflammatory conditions, such as that observed after bacterial lipopolysaccharide (LPS) administration, are associated with leptin secretion. The hypophagic effect of leptin depends in part on the activation of PI3K signaling pathway. However, the role of PI3K in the endotoxemia-induced hypophagia has not been determined. METHODS: In an attempt to examine the functional contribution of the PI3K pathway in hypophagia and weight loss induced by LPS (100 ug/Kg, ip), we performed a central pharmacological PI3K inhibition (LY294002). Additionally, to gain mechanistic insights on the role of the catalytic PI3K p110α subunit in leptin responsive cells, mice expressing Cre-recombinase driven by the Lepr promoter (LepR-Cre) were crossed with mice carrying a loxP-modified p110α allele (Pi3kca gene) (LepR(Δp110α)). As studies have suggested that the PI3K p110ß subunit has a dominant role over p110α in energy homeostasis, we further crossed LepR-Cre mice with loxP-modified p110α and p110ß (Pi3kcb gene) alleles (LepR(Δp110α+ß)). In order to verify the requirement of leptin in PI3K effects on food intake, we also used leptin-deficient ob/ob mice. RESULTS: We found that LPS stimulates PI3K and STAT3 signaling pathways in cells expressing the leptin receptor. Central PI3K inhibition prevented LPS-induced hypophagia and weight loss. Genetic deletion of p110α subunit selectively in LepR cells had no effect on LPS-induced hypophagia and weight loss. However, p110α and p110ß double deletion in LepR cells prevented LPS-induced hypophagia and partially reversed the weight loss. Leptin deficiency blunted LPS-induced acute pAKT and pSTAT3 phosphorylation and the acute suppression of food intake. CONCLUSIONS: Our studies show that the PI3K p110ß subunit in LepR cells is required for acute endotoxemic hypophagia. The data provide promising approaches for PI3K inhibition in preventing low energy balance and cachectic states during inflammatory challenges.

18.
Neurosci Lett ; 606: 123-8, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26320022

ABSTRACT

Adrenalectomy (ADX)-induced hypophagia is associated with increased activation of corticotrophin-releasing factor (CRF) and oxytocin (OT) neurons in the paraventricular nucleus of the hypothalamus (PVN) after refeeding. CRF2- and OT-receptor antagonists abolish the hypophagia and the augmented activation of the nucleus of the solitary tract neurons induced by feeding after ADX. In addition, OT-receptor antagonist reversed CRF-induced anorexia. We evaluated the effect of intracerebroventricular pretreatment with CRF2-receptor antagonist, antisauvagine-30 (AS30), on the activation of OT neurons of the PVN in response to refeeding of sham, adrenalectomized (ADX) and ADX rats replaced with corticosterone (ADX+B). In vehicle-pretreated animals, refeeding increased the number of Fos+OT double labeled neurons in the posterior parvocellular subdivision of the PVN (PaPo) of sham, ADX and ADX+B animals, with higher Fos expression and OT neuronal activation in the ADX group. AS30 reversed refeeding-induced increased activation of OT and non-OT neurons in the PaPo in the ADX group. In the medial parvocellular subdivision of the PVN (PaMP) of vehicle-pretreated animals, the number of Fos- and Fos+OT-immunoreactive neurons was increased after refeeding in ADX group. AS30 in the ADX group attenuated the enhanced Fos expression but not the number of Fos+OT double labeled neurons in the PaMP. In conclusion, CRF2-receptor antagonist reverses the increased activation of OT neurons in the PaPo induced by feeding in ADX animals, suggesting that OT neurons might be downstream mediators of CRF effects on satiety-related responses after ADX.


Subject(s)
Midline Thalamic Nuclei/metabolism , Neurons/metabolism , Oxytocin/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Satiety Response , Adrenalectomy , Animals , Eating , Midline Thalamic Nuclei/cytology , Rats , Rats, Wistar , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors
19.
Brain Res ; 1600: 32-41, 2015 Mar 10.
Article in English | MEDLINE | ID: mdl-25573193

ABSTRACT

We compared glucocorticoid receptor (GR) gene expression in the hippocampus of rats subjected to a low protein, "malnourished" diet (M; 6% protein) or a control, "well-nourished" diet (W; 16% protein), exposed to different environmental stimulation (environmental enrichment, E; no enrichment, N) between postnatal day 8 (P8) and P35. Rats were tested on the elevated plus maze (EPM) on P36. Male Wistar rats were split into two groups at birth according to diet (M or W) and subdivided according to environmental stimulation (E or N). GR expression was determined using real-time polymerase chain reaction and GR immunohistochemistry in the hippocampus. Our results showed that MN rats spent more time and made more entries into the open arms of the EPM compared to W rats. On the other hand, ME rats spent a similar percentage of time, and made a similar number of entries, in the open arms as WN rats. Following the EPM test, GR mRNA expression in the hippocampus was different in MN rats compared to W rats; expression was also different between M and ME rats; mRNA and expression of GR receptors in WN rats was similar to that observed in WE rats. These data also show that the effects of malnutrition on risk assessment in the EPM were reversed by E. Early malnutrition may alter GR expression in the hippocampus and environmental enrichment may exert a neuroprotective effect on malnutrition-induced brain injury.


Subject(s)
Anxiety/metabolism , Environment , Exploratory Behavior , Hippocampus/metabolism , Protein-Energy Malnutrition/metabolism , Receptors, Glucocorticoid/metabolism , Animals , Diet , Male , Rats , Rats, Wistar
20.
Am J Physiol Endocrinol Metab ; 308(1): E40-50, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25352433

ABSTRACT

Leptin resistance is induced by the feedback inhibitors tyrosine phosphatase-1B (PTP1B) and decreased Src homology 2 domain-containing tyrosine phosphatase-2 (SHP-2) signaling. To investigate the participation of PTP1B and SHP-2 in LPS-induced leptin resistance, we injected repeated (6-LPS) intraperitoneal LPS doses (100 µg/kg ip) for comparison with a single (1-LPS) treatment and evaluated the expression of SHP-2, PTP1B, p-ERK1/2, and p-STAT3 in the hypothalamus of male Wistar rats. The single LPS treatment increased the expression of p-STAT3 and PTP1B but not SHP-2. The repeated LPS treatment reduced SHP-2, increased PTP1B, and did not change p-STAT3. We observed that the PTP1B expression induced by the endotoxin was highly colocalized with leptin receptor cells in the hypothalamus of LepRb-IRES-Cre-tdTomato reporter mice. The single, but not the repeated, LPS treatment decreased the food intake and body weight. Leptin had no stimulatory effect on the hypophagia, body weight loss, or pSTAT3 expression in 6-LPS rats, indicating leptin unresponsiveness. Notably, the PTP1B inhibitor (3.0 nmol/rat in 5 µl icv) restored the LPS-induced hypophagia in 6-LPS rats and restored the ability of leptin to reduce food intake and body weight as well as to phosphorylate STAT3 in the arcuate, paraventricular, and ventromedial nuclei of the hypothalamus. The present data suggest that an increased PTP1B expression in the hypothalamus underlies the development of leptin resistance during repeated exposure to LPS. Our findings contribute to understanding the mechanisms involved in leptin resistance during low-grade inflammation as seen in obesity.


Subject(s)
Drug Resistance , Inflammation/metabolism , Leptin/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/physiology , Animals , Drug Resistance/drug effects , Drug Resistance/genetics , Hypothalamus/drug effects , Hypothalamus/metabolism , Lipopolysaccharides , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity/genetics , Obesity/metabolism , Rats , Rats, Wistar , Receptors, Leptin/genetics , Receptors, Leptin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL