Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Neurotoxicology ; 99: 244-253, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37944760

ABSTRACT

Misused volatile solvents typically contain toluene (TOL) as the main psychoactive ingredient. Cyclohexane (CHX) can also be present and is considered a safer alternative. Solvent misuse often occurs at early stages of life, leading to permanent neurobehavioral impairment and growth retardation. However, a comprehensive examination of the effects of TOL and CHX on stress regulation and energy balance is lacking. Here, we compared the effect of a binge-pattern exposure to TOL or CHX (4,000 or 8,000 ppm) on body weight, food intake, the hypothalamus-pituitary-adrenal (HPA) and hypothalamus-pituitary-thyroid (HPT) axes in male adolescent Wistar rats. At 8,000 ppm, TOL decreased body weight gain without affecting food intake. In addition, TOL and CHX altered the HPA and HPT axes' function in a solvent- and concentration-dependent manner. The highest TOL concentration produced HPA axis hyperactivation in animals not subjected to stress, which was evidenced by increased corticotropin-releasing-factor (CRF) release from the median eminence (ME), elevated adrenocorticotropin hormone (ACTH) and corticosterone serum levels, and decreased CRF mRNA levels in the hypothalamic paraventricular nucleus (PVN). TOL (8,000 ppm) also increased triiodothyronine (T3) serum levels, decreased pro-thyrotropin-releasing-hormone (pro-TRH) mRNA transcription in the PVN, pro-TRH content in the ME, and serum thyroid stimulating hormone (TSH) levels. CHX did not affect the HPA axis. We propose that the increased HPT axis activity induced by TOL can be related to the impaired body weight gain associated with inhalant misuse. These findings may contribute to a better understanding of the effects of the misused solvents TOL and CHX.


Subject(s)
Corticotropin-Releasing Hormone , Hypothalamo-Hypophyseal System , Rats , Male , Animals , Rats, Wistar , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Toluene/toxicity , Pituitary-Adrenal System/metabolism , Hypothalamus/metabolism , Body Weight , RNA, Messenger , Solvents/toxicity , Corticosterone
2.
Psychoneuroendocrinology ; 155: 106331, 2023 09.
Article in English | MEDLINE | ID: mdl-37437420

ABSTRACT

Premenstrual Dysphoric Disorder (PMDD) is related to an abrupt drop in progesterone and impairments in the HPA axis that cause anxiety. Suffering persons report higher daily-life stress and anxiety proneness that may contribute to developing PMDD, considered a chronic stress-related disorder. Here, we explored the effect of chronic unpredictable stress (CUS) in rats subjected to progesterone withdrawal (PW) and evaluated gene expression of HPA axis activation in the stress-vulnerable Wistar-Kyoto (WKY) rat strain that is prone to anxiety. Ovariectomized WKY rats were randomly assigned to CUS or Standard-housed conditions (SHC) for 30 days. To induce PW, animals received 2 mg/kg of progesterone on day 25th for 5 days; 24 h later, they were tested using the anxiety-like burying behavior test (BBT). After behavioral completion, rats were euthanized, and brains were extracted to measure Crh (PVN) and Nr3c1 (hippocampus) mRNA. Blood corticosterone and vasopressin levels were determined. Results showed that PW exacerbated anxiety-like behaviors through passive coping in CUS-WKY. PW decreased Crh-PVN mRNA and the Nr3c1-hippocampal mRNA expression in SHC. CUS decreased Crh-PVN mRNA compared to SHC, and no further changes were observed by PW or BBT exposure. CUS reduced Nr3c1-hippocampal gene expression compared to SHC animals, and lower Nr3c1 mRNA was detected due to BBT. The PW increased corticosterone in SHC and CUS rats; however, CUS blunted corticosterone when combined with PW+BBT and similarly occurred in vasopressin concentrations. Chronic stress blunts the response of components of the HPA axis regulation when PW and BBT (systemic and psychogenic stressors, respectively) are presented. This response may facilitate less adaptive behaviors through passive coping in stress-vulnerable subjects in a preclinical model of premenstrual anxiety.


Subject(s)
Premenstrual Dysphoric Disorder , Progesterone , Humans , Rats , Female , Animals , Rats, Inbred WKY , Progesterone/metabolism , Corticosterone , Premenstrual Dysphoric Disorder/metabolism , Hypothalamo-Hypophyseal System/metabolism , Neurobiology , Pituitary-Adrenal System/metabolism , Stress, Psychological/etiology , Vasopressins/metabolism , RNA, Messenger/metabolism
3.
Psychoneuroendocrinology ; 125: 105099, 2021 03.
Article in English | MEDLINE | ID: mdl-33387926

ABSTRACT

Chronic stress exposure impacts negatively in individuals leading to food addiction, overweight or obesity. Stress-genes and their translation products are responsible for the responses of humans to adverse environments. Alterations in stress-genes expression or protein function may induce behaviors as compulsive eating of high-energy containing food, which decreases stress-induced negative feelings. However, chronic stress is not assessed in Mexican population. We analyzed here the association between polymorphisms of CRH, CRHR2 and glucocorticoids (GR, NR3C1) receptor genes with food addiction and obesity and overweight in Mexican patients of a Nutrition Clinic. We recruited 508 individuals of both genders, who accepted to participate in the study at their first visit to the clinic, obtaining their fat mass percentage and a blood sample for the genetic analysis. Participants answered the Yale's food addiction scale and were subjected to a Trier social test, as an acute stressful stimulus. Pre and post-test saliva samples were obtained to evaluate cortisol levels and adrenal axis' response to the acute stress. The 63% of participants classified as stressed (S); 6.5% of normal-weight individuals showed food-addiction, whereas 63% of participants with food-addiction were also stressed. The fat mass percentage was greater in stress-addiction than in stressed non-addiction participants. The best interaction model for obesity development risk comprehended the presence of polymorphisms of the three genes that in combination with food addiction increased the risk for developing obesity 2.8-4-fold. Thus, frequent stress exposure favors food-addiction, which along with genetic susceptibility seems to add up to Mexican obesity/overweight rates.


Subject(s)
Behavior, Addictive , Food Addiction , Behavior, Addictive/genetics , Female , Humans , Male , Obesity/genetics , Overweight/genetics , Polymorphism, Genetic
4.
Neurochem Int ; 141: 104876, 2020 12.
Article in English | MEDLINE | ID: mdl-33049337

ABSTRACT

Women around menopause are vulnerable to present psychiatric and metabolic disorders; thus, therapies that contribute to treat both pathologies are required. Previous reports showed that an aqueous extract of pomegranate (Punica granatum), enriched in ellagitannins, exerts an antidepressant-like effect in ovariectomized rats. We analyze whether this aqueous extract of P. granatum (AE-PG) prevents the anxiety-like behavior induced by a cafeteria diet (CAF) in middle-aged ovariectomized rats at the same time that it prevents an increase in body weight, glucose, lipids, and the changes on mRNA expression of the peroxisome proliferator-activated receptor-gamma (PPAR-γ) in the liver. Also, the effects of AE-PG on the protein levels of PPAR-γphospho-PPAR-γ, extracellular signal-regulated protein kinase (ERK1/2) and phospho-ERK1/2 were measured in the hippocampus and amygdala. CAF induced anxiety-like behavior, augmented lipids and glucose blood levels, body weight, visceral fat, insulin resistance, and decreased mRNA expression of PPAR-γ in the liver. In rats fed with the CAF, AE-PG prevented the anxiety-like behavior, reduced body weight, lowered lipid levels, reduced insulin resistance, and increased PPAR-γ mRNA expression in the liver. In the hippocampus, ERK1/2 but not PPAR-γ protein levels were decreased by CAF, while AE-PG prevented these effects. In the amygdala, CAF increased the phosphorylation of PPARγ, and AE-PG prevented it. In contrast, AE-PG rescued the decreased ERK1/2 protein level in the hippocampus caused by CAF. In conclusion, AE-PG treatment prevented anxiogenic and metabolic effects induced by CAF, and its effects appear to be mediated by ERK1/2 and PPARγ depending on the brain area studied.


Subject(s)
Antidepressive Agents/pharmacology , Anxiety/psychology , Hydrolyzable Tannins/pharmacology , Menopause/metabolism , Menopause/psychology , Metabolism/drug effects , Plant Extracts/pharmacology , Pomegranate/chemistry , Adiposity/drug effects , Animals , Antidepressive Agents/chemistry , Anxiety/prevention & control , Blood Glucose/metabolism , Diet , Female , Hydrolyzable Tannins/chemistry , Lipid Metabolism/drug effects , MAP Kinase Signaling System/drug effects , Ovariectomy , PPAR gamma/metabolism , Plant Extracts/chemistry , Rats
5.
Neurosci Lett ; 739: 135403, 2020 11 20.
Article in English | MEDLINE | ID: mdl-32980456

ABSTRACT

Feeding-regulatory peptides such as thyrotropin-releasing hormone (TRH), α-melanocyte-stimulating hormone (α-MSH) and their receptors are expressed in brain regions involved in the homeostatic and hedonic control of food intake, such as the hypothalamus and the mesolimbic system, respectively. The nucleus accumbens (NAc) is part of the latter, a brain circuit involved in processing reward stimuli and the appetitive motivation of feeding. When TRH or α-MSH are administered in the NAc, both decrease food intake, through activating their respective receptors, TRH-R1 and MC4R. The actions of α-MSH as a homeostatic feeding-regulator involves the increase of hypothalamic TRH expression, thus, we aimed to identify whether TRH signaling in the NAc was also participating in α-MSH-induced reduction of food intake. α-MSH administration in the NAc of 48 h fasted rats reduced their food intake during the 2-h period of refeeding, increased accumbal TRH mRNA expression and decreased that of MC4R. Such downregulated MC4R mRNA levels implied a compensatory decrease of α-MSH actions in the NAc after the previous pathway stimulation. The co-administration of α-MSH along with an antisense oligonucleotide directed against pro-TRH mRNA in the NAc impaired the α-MSH-induced feeding reduction, supporting that the accumbal TRHergic pathway is downstream of α-MSH actions to inhibit feeding. Our results suggested that TRH in the NAc mediates some effects of α-MSH on inhibition of food intake; this supports the role of TRH not only as a homeostatic regulator but also as modulating the motivational aspects of feeding.


Subject(s)
Eating , Nucleus Accumbens/metabolism , Protein Precursors/metabolism , Pyrrolidonecarboxylic Acid/analogs & derivatives , Thyrotropin-Releasing Hormone/metabolism , alpha-MSH/metabolism , Animals , Male , Pyrrolidonecarboxylic Acid/metabolism , RNA, Messenger/metabolism , Rats, Wistar
6.
Peptides ; 127: 170285, 2020 Feb 24.
Article in English | MEDLINE | ID: mdl-32105809

ABSTRACT

Early-life overfeeding (OF) disrupts neuroendocrine systems, energy homeostasis and food intake regulation inducing overeating and overweight in adults. Adult rats raised in small litters during lactation, display hyperphagia and overweight since weaning and exhibit a decrease in thyrotropin-releasing hormone (TRH) mRNA expression in hypothalamic paraventricular nucleus (PVN). This is counterintuitive because TRH expression should increase to activate the hypothalamic-pituitary-thyroid (HPT) axis and promote energy expenditure, thus, HPT axis seems inhibited in OF rats. Leptin, an adipocyte-synthesized hormone that stimulates hypothalamic TRH expression, enhances both TRH anorectic effects and HPT axis-induced metabolic rate. To evaluate hypothalamic resistance to the anorectic and HPT axis stimulatory actions of leptin, we injected leptin i.p. to ad libitum fed and to 48-h fasted adult control (reared in normal litters) and to small-litter reared (OF) male Wistar rats. Findings showed that HPT axis was still responsive to leptin, since PVN TRH mRNA levels, median eminence TRH release and T4 serum concentration increased in both, ad libitum and fasted OF rats after leptin administrations. Leptin was ineffective to reduce feeding of OF animals. By comparing leptin receptor (ObRb) expression changes between arcuate and PVN nuclei, we observed that arcuate ObRb was not modified in response to leptin administrations in OF rats, likely accounting for the differential effects in feeding and HPT axis function. Nevertheless, ObRb expression was modified by leptin in the PVN of OF rats to the same extent as controls; this supports the hormone's role as a therapeutic agent for early onset obesity in adults.

7.
J Neuroendocrinol ; 29(11)2017 11.
Article in English | MEDLINE | ID: mdl-28984394

ABSTRACT

Negative energy balance promotes physiological adaptations that ensure the survival of animals. The hypothalamic-pituitary-thyroid axis regulates basal energy expenditure and its down-regulating adaptation to negative energy balance is well described: in fasting, the serum content of thyrotrophin (TSH) and thyroid hormones (TH) decreases, enhancing the survival odds of individuals. By contrast, dehydration-induced anorexic (DIA) rats present an impaired hypothalamic-pituitary-thyroid (HPT) axis adaptation despite their negative energy balance: increased circulating TSH levels. The implication of cocaine- and amphetamine-regulated transcript (CART), an anorexic peptide, in HPT axis function impairment and food-avoidance behaviour displayed by DIA animals is unknown. Because CART is co-expressed with the peptide that regulates the HPT axis in hypophysiotrophic paraventricular nucleus (PVN) neurones (TSH-releasing hormone), we analysed CART expression and possible implications with respect to high TSH levels of DIA animals. We examined whether changes in CART expression from the lateral hypothalamic area (LHA) and arcuate nucleus (ARC) could participate in food-avoidance of DIA rats. DIA and forced-food restricted (FFR) animals reduced their body weight and food intake. FFR rats had a down-regulation of their HPT axis (reduced serum TH and TSH content), whereas DIA animals had reduced TH but increased TSH levels. CART mRNA expression in the ARC decreased similarly between experimental groups and diminished in anterior, medial PVN and in LHA of FFR animals, whereas DIA animals showed unchanged levels. This impaired CART mRNA expression in the anterior PVN and LHA could be related to the aberrant feeding behaviour of DIA rats but not to their deregulated HPT axis function.


Subject(s)
Amphetamines/pharmacology , Anorexia/genetics , Cocaine/pharmacology , Hypothalamic Area, Lateral/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Transcription, Genetic/drug effects , Animals , Anorexia/etiology , Appetite Regulation/genetics , Arcuate Nucleus of Hypothalamus/metabolism , Arcuate Nucleus of Hypothalamus/physiopathology , Avoidance Learning , Body Weight/genetics , Dehydration/complications , Dehydration/genetics , Feeding Behavior , Hypothalamic Area, Lateral/metabolism , Male , Nerve Tissue Proteins/genetics , Paraventricular Hypothalamic Nucleus/metabolism , Rats , Thyroid Hormones/blood , Thyrotropin/blood
8.
Neuropeptides ; 63: 75-82, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28162848

ABSTRACT

Early-life stress induces endocrine and metabolic alterations that increase food intake and overweight in adulthood. The stress response activates the corticotropin-releasing hormone (CRH) and urocortins' (Ucns) system in the hypothalamic paraventricular nucleus (PVN). These peptides induce anorexic effects through CRH-R2 receptor activation; however, chronic stressed animals develop hyperphagia despite of high PVN CRH expression. We analyzed this paradoxical behavior in adult rats subjected to maternal separation (MS) for 180min/daily during post-natal days 2-14, evaluating their body weight gain, food intake, serum corticosterone and vasopressin concentrations, PVN mRNA expression of CRH-R1, CRH-R2, CRH, Ucn2, Ucn3, vasopressin and CRH-R2 protein levels. MS adults increased their feeding, weight gain as well as circulating corticosterone and vasopressin levels, evincing chronic hyperactivity of the stress system. MS induced higher PVN CRH, Ucn2 and CRH-R2 mRNA expression and protein levels of CRH-R2 showed a tendency to decrease in the cellular membrane fraction. An intra-PVN injection of the CRH-R2 antagonist antisauvagine-30 in control adults increased receptor's mRNA expression, mimicking the observed PVN receptor's up-regulation of early-life MS adults. An injection of Ucn-2 directly into the PVN reduced food intake and increased PVN pCREB/CREB ratio in control animals; in contrast, Ucn-2 was unable to reduce food intake and enhance phosphorylated-CREB levels in PVN of MS rats. In conclusion, the chronic hyperactivity of the stress axis and PVN CRH-R2 resistance to Ucn2 effects, supported impaired receptor functionality in MS animals, probably due to its chronic stimulation by CRH or Ucn2, induced by early-life stress.


Subject(s)
Hyperphagia/metabolism , Maternal Deprivation , Paraventricular Hypothalamic Nucleus/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism , Animals , Body Weight/drug effects , Body Weight/physiology , Corticosterone/blood , Eating/drug effects , Eating/physiology , Paraventricular Hypothalamic Nucleus/drug effects , Rats , Rats, Wistar , Urocortins/pharmacology , Vasopressins/blood
9.
J Endocrinol ; 232(1): 15-28, 2017 01.
Article in English | MEDLINE | ID: mdl-27729464

ABSTRACT

Feeding patterns are important factors in obesity evolvement. Time-restricted feeding schedules (tRF) during resting phase change energy homeostasis regulation, disrupting the circadian release of metabolism-regulating hormones, such as leptin, insulin and corticosterone and promoting body weight gain. Thyroid (HPT) and adrenal (HPA) axes exhibit a circadian regulation and are involved in energy expenditure, thus studying their parameters in tRF paradigms will elucidate their role in energy homeostasis impairments under such conditions. As tRF in young animals is poorly studied, we subjected prepuberal rats to a tRF either in light (LPF) or in darkness phase (DPF) and analyzed HPT and HPA response when they reach adulthood, as well as their arcuate (ARC) and paraventricular (PVN) hypothalamic nuclei neurons' sensitivity to leptin in subsets of 10-week-old animals after fasting and with i.p. leptin treatment. LPF group showed high body weight and food intake, along with increased visceral fat pads, corticosterone, leptin and insulin serum levels, whereas circulating T4 decreased. HPA axis hyperactivity was demonstrated by their high PVN Crf mRNA expression; the blunted activity of HPT axis, by the decreased hypophysiotropic PVN Trh mRNA expression. Trh impaired expression to the positive energy balance in LPF, accounted for their ARC leptin resistance, evinced by an increased Npy and Socs3 mRNA expression. We concluded that the hyperphagia of prepuberal LPF animals could account for the HPA axis hyperactivity and for the HPT blocked function due to the altered ARC leptin signaling and impaired NPY regulation on PVN TRH neurons.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Circadian Rhythm/physiology , Feeding Behavior/physiology , Leptin/pharmacology , Paraventricular Hypothalamic Nucleus/metabolism , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Body Weight/drug effects , Body Weight/physiology , Corticosterone/blood , Corticotropin-Releasing Hormone/metabolism , Eating/drug effects , Eating/physiology , Energy Metabolism/drug effects , Energy Metabolism/physiology , Insulin/blood , Leptin/blood , Neurons/drug effects , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Rats , Thyroxine/blood
10.
Behav Brain Res ; 319: 165-173, 2017 02 15.
Article in English | MEDLINE | ID: mdl-27864049

ABSTRACT

Thyrotropin-releasing hormone (TRH) has anorexigenic and anxiolytic functions when injected intraventricularly. Nucleus accumbens (NAcc) is a possible brain region involved, since it expresses proTRH. TRH from hypothalamic paraventricular nucleus (PVN) has a food intake-regulating role. TRHergic pathways of NAcc and PVN are implicated in anxiety and feeding. Both behaviors depend on cAMP and phosphorylated-cAMP response element binding protein (pCREB) intracellular levels. Intracellular levels of cAMP are controlled by the degrading activity of phosphodiesterases (PDEs). Since TRH transcription is activated by pCREB, a specific inhibitor of PDE7B may regulate TRH-induced effects on anxiety and feeding. We evaluated the effectiveness of an intra-accumbal and intraperitoneal (i.p.) administration of a PDE7 inhibitor (BRL-50481) on rats' anxiety-like behavior and food intake; also on TRH mRNA and protein expression in NAcc and PVN to define its mediating role on the PDE7 inhibitor-induced behavioral changes. Accumbal injection of 4µg/0.3µL of PDE7 inhibitor decreased rats' anxiety. The i.p. injection of 0.2mg/kg of the inhibitor was able to increase the PVN TRH mRNA expression and to decrease feeding but did not change animals' anxiety levels; in contrast, 2mg/kg b.w inhibitor enhanced accumbal TRH mRNA, induced anxiolysis with no change in food intake. PDE7 inhibitor induced anxiolytic and anorexigenic like behavior depending on the dose used. Results supported hypothalamic TRH mediated feeding-reduction effects, and accumbal TRH mediation of inhibitor-induced anxiolysis. Thus, an i.p dose of this inhibitor might be reducing anxiety with no change in feeding, which could be useful for obese patients.


Subject(s)
Anxiety/chemically induced , Cyclic Nucleotide Phosphodiesterases, Type 7/antagonists & inhibitors , Feeding Behavior/drug effects , Nitro Compounds/pharmacology , Nucleus Accumbens/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Sulfonamides/pharmacology , Thyrotropin-Releasing Hormone/metabolism , Animals , Anxiety/drug therapy , Cyclic AMP/metabolism , DNA, Antisense/pharmacology , Dose-Response Relationship, Drug , Eating/drug effects , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Gene Expression Regulation/drug effects , Iodide Peroxidase/genetics , Iodide Peroxidase/metabolism , Male , Maze Learning/drug effects , Nitro Compounds/therapeutic use , Rats , Rats, Wistar , Sulfonamides/therapeutic use , Thyrotropin-Releasing Hormone/genetics , Time Factors , Iodothyronine Deiodinase Type II
11.
Behav Brain Res ; 306: 128-36, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27006143

ABSTRACT

The thyrotropin-releasing hormone (TRH), an anorexigenic factor that reduces food intake in food-restricted animals, may be involved in motivation for food. Injected centrally, TRH impairs acquisition of food-rewarded behavior. Through the TRH-R1 receptors, TRH injected in the nucleus accumbens increases dopamine content-perhaps the mechanism by which the peptide modulates food motivation. This, however, is still to be demonstrated. We sought to evaluate dopamine release by microdialysis after a TRH injection into the nucleus accumbens shell in free-moving fasted rats. In addition, we assessed dopamine content and turnover by HPLC and the relationship with the motivation for food by analyzing the performance of rats during a progressive-ratio (PR) operant-conditioning test. Finally, we determined serum leptin and triiodothyronine (T3) levels in order to evaluate the animals' metabolic response to food restriction and the impact of intra-accumbal TRH administration on circulating hormones. Intra-accumbal injections of TRH reduced food intake in food-restricted rats-compared to counterparts treated with saline-, without further decreasing T3 or leptin levels, which dropped due to their dietary regime. TRH-injected rats had lower breaking points on the PR schedule, which indicated lower motivation to eat. Accordingly, compared to saline-treated animals, dopamine release and turnover increased in the nucleus accumbens of TRH-injected rats, a finding that suggests a relationship between motivation for food and TRH-induced release of dopamine.


Subject(s)
Dopamine/metabolism , Eating/drug effects , Motivation/drug effects , Nucleus Accumbens/drug effects , Thyrotropin-Releasing Hormone/pharmacology , 3,4-Dihydroxyphenylacetic Acid/metabolism , Analysis of Variance , Animals , Conditioning, Operant/drug effects , Food Deprivation , Leptin/blood , Male , Microdialysis , Nucleus Accumbens/metabolism , Rats , Rats, Wistar , Reward , Triiodothyronine/blood
12.
J Endocrinol ; 221(1): 77-88, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24464021

ABSTRACT

Neuroendocrine axes adapt to nutrient availability. During fasting, the function of the hypothalamus-pituitary-thyroid axis (HPT) is reduced, whereas that of the hypothalamus-pituitary-adrenal axis (HPA) is increased. Overfeeding-induced hyperleptinemia during lactation may alter the regulatory set point of neuroendocrine axes and their adaptability to fasting in adulthood. Hyperleptinemia is developed in rodents by litter size reduction during lactation; adult rats from small litters become overweight, but their paraventricular nucleus (PVN) TRH synthesis is unchanged. It is unclear whether peptide expression still responds to nutrient availability. PVN corticotropin-releasing factor (CRF) expression has not been evaluated in this model. We analyzed adaptability of HPT and HPA axes to fasting-induced low leptin levels of reduced-litter adult rats. Offspring litters were reduced to 2-3/dam (early-overfed) or maintained at 8/dam (controls, C). At 10 weeks old, a subset of animals from each group was fasted for 48 h and leptin, corticosterone, and thyroid hormones serum levels were analyzed. In brain, expressions of leptin receptor, NPY and SOCS3, were evaluated in arcuate nucleus, and those of proTRH and proCRF in PVN by real-time PCR. ProTRH expression in anterior and medial PVN subcompartments was assayed by in situ hybridization. Early-overfed adults developed hyperphagia and excessive weight, together with decreased proTRH expression in anterior PVN, supporting the anorexigenic effects of TRH. Early-overfed rats presented low PVN proTRH synthesis, whereas fasting did not induce a further reduction. Fasting-induced stress was unable to increase corticosterone levels, contributing to reduced body weight loss in early-overfed rats. We concluded that early overfeeding impaired the adaptability of HPT and HPA axes to excess weight and fasting in adults.


Subject(s)
Corticotropin-Releasing Hormone/genetics , Overnutrition/genetics , Overnutrition/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Protein Precursors/genetics , Pyrrolidonecarboxylic Acid/analogs & derivatives , Thyrotropin-Releasing Hormone/genetics , Animals , Corticosterone/metabolism , Corticotropin-Releasing Hormone/metabolism , Fasting/metabolism , Female , Humans , Leptin/metabolism , Litter Size , Male , Pituitary-Adrenal System/metabolism , Protein Precursors/metabolism , Pyrrolidonecarboxylic Acid/metabolism , Rats , Rats, Wistar , Thyroid Gland/metabolism , Thyroid Hormones/metabolism , Thyrotropin-Releasing Hormone/metabolism
13.
Behav Brain Res ; 235(2): 189-94, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-22963992

ABSTRACT

Anxiolytic effects of alcohol participate in the reinforcing properties of the drug, in which nucleus accumbens (NAcc) is implicated. The opioidergic system in NAcc is considered a main pathway involved in the emotional responses of animals: rats microinjected with morphine in NAcc and the systemic administration of µ-opioid receptors (MOR) agonists yield low anxiety scores in the elevated plus maze (EPM), a behavioral test of anxiety. However, the specific participation of NAcc MOR in the anxiolytic effect of ethanol has not been studied. AC5, a cAMP-synthezising adenylyl-cyclase, is highly expressed in NAcc; it is negatively coupled to MOR and has been implicated in anxiety levels of animals. We evaluated the anxiolytic effects of an intra-gastric administration of ethanol (2.5 g/kg) in animals subjected to EPM at 1, 4, and 8 h after drug or water exposure. Locomotion was assayed with the open-field test; we also measured accumbal AC5 and MOR mRNA levels by RT-PCR. After 1 h, ethanol-exposed animals showed anxiolytic-like behavior, as well as decreased and increased AC5 and MOR expression in NAcc, respectively. Intra-accumbal injection of ß-funaltrexamine (FNA), a MOR antagonist, did not block ethanol-induced anxiolysis, rather it induced a tendency to increase anxiety levels in the water-exposed group. FNA partially decreased accumbal AC5 expression in ethanol-treated rats. We concluded that AC5 in NAcc is participating in the emotional effects of ethanol; that MOR was not mediating the drug-induced AC5 reduction in NAcc nor the ethanol-induced anxiolysis. MOR only might be involved in basal levels of anxiety of animals.


Subject(s)
Adenylyl Cyclases/metabolism , Anti-Anxiety Agents/therapeutic use , Anxiety/drug therapy , Ethanol/therapeutic use , Gene Expression Regulation, Enzymologic/drug effects , Nucleus Accumbens/drug effects , Adenylyl Cyclases/genetics , Analysis of Variance , Animals , Anti-Anxiety Agents/pharmacology , Anxiety/pathology , Disease Models, Animal , Ethanol/pharmacology , Exploratory Behavior/drug effects , Male , Maze Learning/drug effects , Naltrexone/analogs & derivatives , Naltrexone/pharmacology , Narcotic Antagonists/pharmacology , Nucleus Accumbens/metabolism , Nucleus Accumbens/physiopathology , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , Time Factors
14.
Endocrinology ; 153(8): 4067-76, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22719053

ABSTRACT

TRH synthesized in hypothalamic paraventricular nucleus (PVN) regulates thyroid axis function and is also implicated in anorexigenic effects. Under energy deficit, animals present decreased PVN TRH expression and release, low TSH levels, and increased appetite. Dehydration-induced anorexia (DIA) model allows insight into underlying mechanisms of feeding regulation. Animals drinking a 2.5% NaCl solution for 7 d present body weight reduction; despite their negative energy balance, they avoid food and have increased PVN TRH expression and TSH serum levels. These findings support an inhibiting role of PVN TRH in feeding control. We compared TRH expression by in situ hybridization in PVN subdivisions of 7-d dehydrated male rats to those of a pair-fed group (forced food-restricted) with similar metabolic changes than DIA, but motivated to eat, and to controls. We measured peripheral deiodinase activities, and expression and activity of medial basal hypothalamic type 2 deiodinase and pyroglutamyl-aminopeptidase II, to understand their regulating role in PVN TRH changes between food restriction and anorexia. TRH mRNA levels increased in anterior (aPVN) and medial-caudal subdivisions in DIA rats, whereas it decreased in medial PVN in both experimental groups. We confirmed the nonhypophysiotropic nature of aPVN TRHergic cells by injecting ip fluorogold tracer. Findings support a subspecialization of TRHergic hypophysiotrophic cells that responded differently between anorexic and food-restricted animals; also, that aPVN TRH participates in food intake regulation. Increased type 2 deiodinase activity seemed responsible for low medial PVN TRH synthesis, whereas increased medial basal hypothalamic pyroglutamyl-aminopeptidase II activity in DIA rats might counteract their high TRH release.


Subject(s)
Aminopeptidases/metabolism , Anorexia/metabolism , Iodide Peroxidase/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Pyrrolidonecarboxylic Acid/analogs & derivatives , Thyrotropin-Releasing Hormone/metabolism , Aminopeptidases/genetics , Animals , Anorexia/etiology , Body Weight/physiology , Caloric Restriction , Dehydration , Eating/physiology , In Situ Hybridization , Iodide Peroxidase/genetics , Male , Pyrrolidonecarboxylic Acid/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Thyrotropin-Releasing Hormone/genetics , Iodothyronine Deiodinase Type II
15.
Regul Pept ; 159(1-3): 54-60, 2010 Jan 08.
Article in English | MEDLINE | ID: mdl-19800927

ABSTRACT

Food-restricted animals present metabolic adaptations that facilitate food-seeking behavior and decelerate energy utilization by reducing the hypothalamus-pituitary-thyroid (HPT) axis function. Stress by dehydration induces an anorexic behavior in rats, loss of weight and reduced food intake when compared to ad libitum fed animals, however these alterations are accompanied by HPT axis changes such as increased serum thyrotropin levels and enhanced expression of thyrotropin-releasing hormone (TRH) in the paraventricular nucleus of the hypothalamus, which is considered as anorexigenic peptide. In contrast, a pair-fed group conformed by forced-food-restricted animals (FFR) (eating the exact same amount of food as dehydration-induced anorexic rats--DIA rats) present decreased TRH mRNA levels. NPY synthesis in the arcuate nucleus and orexin-expressing neurons from the lateral hypothalamic area (LHA) are activated during food restriction. These brain structures project into PVN, suggesting that NPY and orexins are possible factors involved in TRHergic neuron activation in DIA rats. Leptin signaling is another likely factor to be involved in TRH differential expression. Therefore, to gain more insight into the regulation of the feeding behavior in the experimental models, we analyzed Y1, Y5, Ox1-R and Ob-R(b) mRNA levels in PVN and prepro-orexin in LHA, since their signaling to the PVN might be altering TRH synthesis and feeding in DIA animals. Prepro-orexinergic cells were activated in FFR animals; Ox1-R and Y1 expression was reduced in FFR vs. controls or DIA group. Compensatory changes in PVN receptor expression of some feeding-related peptides in anorexic rats may alter TRHergic neural response to energy demands.


Subject(s)
Anorexia/metabolism , Dehydration/metabolism , Gene Expression Regulation , Hypothalamo-Hypophyseal System , Intracellular Signaling Peptides and Proteins/metabolism , Neuropeptides/metabolism , Pituitary-Adrenal System , Receptors, G-Protein-Coupled/biosynthesis , Receptors, Neuropeptide/biosynthesis , Animals , Anorexia/etiology , Dehydration/complications , Feeding Behavior , Leptin/metabolism , Male , Malnutrition/metabolism , Neurons/metabolism , Neuropeptide Y/metabolism , Orexin Receptors , Orexins , Paraventricular Hypothalamic Nucleus/metabolism , Rats , Rats, Wistar , Receptors, Neuropeptide Y/biosynthesis , Signal Transduction , Thyrotropin/biosynthesis , Thyrotropin-Releasing Hormone/biosynthesis
16.
Neurochem Int ; 48(1): 31-42, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16213061

ABSTRACT

Subthreshold electrical stimulation of the amygdala (kindling) activates neuronal pathways increasing the expression of several neuropeptides including thyrotropin releasing-hormone (TRH). Partial kindling enhances TRH expression and the activity or its inactivating ectoenzyme; once kindling is established (stage V), TRH and its mRNA levels are further increased but TRH-binding and pyroglutamyl aminopeptidase II (PPII) activity decreased in epileptogenic areas. To determine whether variations in TRH receptor binding or PPII activity are due to regulation of their synthesis, mRNA levels of TRH receptors (R1, R2) and PPII were semi-quantified by RT-PCR in amygdala, frontal cortex and hippocampus of kindled rats sacrificed at stage II or V. Increased mRNA levels of PPII were found at stage II in amygdala and frontal cortex, and of pro-TRH and TRH-R2, in amygdala and hippocampus. At stage V, pro-TRH mRNA levels increased and those of PPII, decreased in the three regions; TRH-R2 mRNA levels diminished in amygdala and frontal cortex and of TRH-R1 only in amygdala. In situ hybridization analyses revealed, at stage II, enhanced TRH-R1 mRNA levels in dentate gyrus and amygdala while decreased in piriform cortex; those of TRH-R2 increased in amygdala, CA2, dentate gyrus, piriform cortex, thalamus and subiculum and of PPII, in CAs and piriform cortex. In contrast, at stage V decreased expression of TRH-R1 occurred in amygdala, CA2/3, dentate gyrus and piriform cortex; of TRH-R2 in CA2, thalamus and piriform cortex, and of PPII in CA2, and amygdala. The magnitude of changes differed between ipsi and contralateral side. These results support a trans-synaptic modulation of all elements involved in TRH transmission in conditions that stimulate the activity of TRHergic neurons. They show that reported changes in PPII activity or TRH-binding caused by kindling relate to regulation of the expression of TRH receptors and degrading enzyme.


Subject(s)
Amygdala/physiology , Gene Expression Regulation/physiology , Kindling, Neurologic , Thyrotropin-Releasing Hormone/physiology , Animals , Base Sequence , DNA Primers , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Thyrotropin-Releasing Hormone/genetics , Reverse Transcriptase Polymerase Chain Reaction
17.
Neurochem Int ; 46(4): 347-56, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15707699

ABSTRACT

Released TRH is inactivated by an ectopeptidase, pyroglutamyl aminopeptidase II (PPII). PPII expression and activity are stringently regulated in adenohypophysis, and in rat brain, during kindling stimulation that activates TRHergic neurons. To gain further insight into the possible regulation of PPII, we studied the effect of an acute intraperitoneal ethanol administration that affects TRH content and expression. PPII activity was determined by a fluorometric assay and PPII mRNA levels by semi-quantitative RT-PCR. Activity decreased in frontal cortex 1 h after ethanol injection and, after 6 h, in hippocampus, amygdala and n. accumbens. PPII mRNA levels decreased at 30 and 60 min in frontal cortex and n. accumbens while increased at longer times in these regions and, in hippocampus and hypothalamus. NMDA and GABA(A) receptors' agonists and antagonists were tested at 1 h (+/-ethanol) on PPII activity and mRNA levels, as well as on TRH content and its mRNA. In n. accumbens, PPII mRNA levels decreased by ethanol, MK-801, and muscimol while picrotoxin or NMDA reversed ethanol's inhibition. Ethanol decreased TRH content and increased TRH mRNA levels as MK-801 or muscimol did (NMDA or picrotoxin reverted the effect of ethanol). In frontal cortex, PPII activity was inhibited by ethanol, NMDA and MK-801 with ethanol; its mRNA levels were reduced by ethanol, MK-801 and muscimol (NMDA and picrotoxin reverted ethanol's inhibition). These results show that PPII expression and activity can be regulated in conditions where TRHergic neurons are modulated. Effects of ethanol on PPII mRNA levels as well as those of TRH and its mRNA may involve GABA or NMDA receptors in n. accumbens. Changes observed in frontal cortex suggest combined effects with stress. The response was region-specific in magnitude, tendency and kinetics. These results give further support for brain PPII regulation in conditions that modulate the activity of TRHergic neurons.


Subject(s)
Aminopeptidases/drug effects , Brain Chemistry/drug effects , Ethanol/pharmacology , Limbic System/drug effects , Pyrrolidonecarboxylic Acid/analogs & derivatives , Thyrotropin-Releasing Hormone/metabolism , Alcohol-Induced Disorders, Nervous System/enzymology , Alcohol-Induced Disorders, Nervous System/genetics , Alcohol-Induced Disorders, Nervous System/physiopathology , Aminopeptidases/genetics , Aminopeptidases/metabolism , Animals , Brain Chemistry/genetics , Down-Regulation/drug effects , Down-Regulation/genetics , Drug Administration Schedule , Excitatory Amino Acid Antagonists/pharmacology , GABA Antagonists/pharmacology , GABA-A Receptor Antagonists , Limbic System/enzymology , Limbic System/physiopathology , Male , Neural Pathways/drug effects , Neural Pathways/enzymology , Neural Pathways/physiopathology , Neurons/drug effects , Neurons/enzymology , Pyrrolidonecarboxylic Acid/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, GABA-A/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Stress, Physiological/metabolism , Stress, Physiological/physiopathology , Thyrotropin-Releasing Hormone/genetics
18.
Regul Pept ; 127(1-3): 141-50, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15680480

ABSTRACT

Thyrotropin-releasing hormone (TRH), its receptors and inactivating enzyme (PPII) are present in limbic regions. Nutritional changes or acute ethanol administration in male rats differentially modulate TRH or PPII expression. Chronic ethanol effect was studied in male (3, 6 and 8 weeks) and female rats (6 weeks) including naive and pair-fed (glucose) groups. Daily solid food and liquid intake, serum TSH and corticosterone, TRH content and PPII activity in limbic regions, were quantified. Gender differences were found in ethanol and total caloric intake and body weight gain, TSH and corticosterone levels. Ethanol consumption decreased TRH content and PPII activity in frontal cortex of male rats after 3-6 weeks. In contrast, glucose ingestion altered, by the third week, TRH content in amygdala, hippocampus, hypothalamus and nucleus accumbens, PPII activity in hippocampus and frontal cortex; by the sixth week, TRH content in amygdala and n. accumbens of male and females. Withdrawal at 24 h after 3-week ethanol ingestion decreased TRH content in amygdala and PPII activity in n. accumbens, while withdrawal from glucose reverted some of the effects produced by chronic glucose ingestion. Variations in TRH content or PPII activity support a region specific involvement of TRH neurons that depend on the treatment.


Subject(s)
Aminopeptidases/metabolism , Ethanol , Glucose , Limbic System/metabolism , Pyrrolidonecarboxylic Acid/analogs & derivatives , Thyrotropin-Releasing Hormone/metabolism , Adrenal Cortex Hormones/blood , Animals , Drinking , Eating , Ethanol/administration & dosage , Ethanol/metabolism , Female , Glucose/administration & dosage , Glucose/metabolism , Limbic System/anatomy & histology , Male , Pyrrolidonecarboxylic Acid/metabolism , Rats , Rats, Wistar , Thyrotropin/blood
19.
Neurochem Int ; 41(4): 237-49, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12106775

ABSTRACT

The effect of chronic ethanol consumption during pregnancy and lactation on thyrotropin releasing hormone (TRH) metabolism was investigated in the hypothalamus and limbic areas of female rats and their weaned pups. Pregnant female rats received ethanol or isocaloric glucose solution during pregnancy either alone, or also during the 3 weeks of lactation. Thyrotropin (TSH) and corticosterone levels were measured in serum; TRH and TRH-gly concentrations were determined in hypothalamus, hippocampus, n.accumbens, frontal cortex and amygdala of dams and pups at 21 days after parturition. Ethanol or glucose consumption during pregnancy and lactation produced a decrease in TSH levels compared with control animals fed at libitum; water replacement during lactation normalized TSH levels only in glucose-fed dams. Pups from ethanol or pair-fed dams showed low weight and increased TSH levels compared with normal rats. Variations in TRH metabolism were detected in limbic areas. Chronic ethanol caused a decrease in the levels of TRH in the hippocampus and frontal cortex of dams. In contrast, glucose chronic ingestion increased TRH content specifically in n.accumbens and amygdala of dams. Most of the variations in TRH content of limbic areas of pups were not specific for glucose or ethanol treatment and correlated with the deleterious effect of the mother's thyroid condition, although some differences were observed depending on pup's gender. These results support the involvement of TRHergic neurons in the limbic system of the female rat exposed to alcohol or glucose during pregnancy and lactation.


Subject(s)
Ethanol/pharmacology , Glucose/pharmacology , Hypothalamus/drug effects , Limbic System/drug effects , Thyrotropin-Releasing Hormone/metabolism , Animals , Body Weight , Ethanol/administration & dosage , Female , Glucose/administration & dosage , Hypothalamus/cytology , Hypothalamus/metabolism , Limbic System/cytology , Limbic System/metabolism , Male , Neurons/metabolism , Pregnancy , Rats , Rats, Wistar
20.
Neurochem Int ; 37(5-6): 483-96, 2000.
Article in English | MEDLINE | ID: mdl-10871700

ABSTRACT

Thyrotropin releasing hormone (TRH) present in several brain areas has been proposed as a neuromodulator. Its administration produces opposite effects to those observed with acute ethanol consumption. Opioid peptides, in contrast, have been proposed to mediate some of the effects of alcohol intoxication. We measured TRH content and the levels of its mRNA in hypothalamic and limbic zones 1-24 h after acute ethanol injection. We report here fast and transient changes in the content of TRH and its mRNA in these areas. The levels of proenkephalin mRNA varied differently from those of proTRH mRNA, depending on the time and region studied. Wistar rats were administered one dose of ethanol (intraperitoneal, 3 g/kg body weight) and brains dissected in hypothalamus, hippocampus, amygdala, n. accumbens and frontal cortex, for TRH quantification by radioimmunoassay or for proTRH mRNA measurement by RT-PCR. After 1 h injection, TRH levels were increased in hippocampus and decreased in n. accumbens; after 4 h, it decreased in the hypothalamus, frontal cortex and amygdala, recovering to control values in all regions at 24 h. ProTRH mRNA levels increased at 1 h post-injection in total hypothalamus and hippocampus, while they decreased in the frontal cortex. The effect of ethanol was also studied in primary culture of hypothalamic cells; a fast and transient increase in proTRH mRNA was observed at 1 h of incubation (0.001% final ethanol concentration). Changes in the mRNA levels of proTRH and proenkephalin were quantified by in situ hybridization in rats administered ethanol intragastrically (2.5 g/kg). Opposite alterations were observed for these two mRNAs in hippocampus and frontal cortex, while in n. accumbens and the paraventricular nucleus of the hypothalamus, both mRNA levels were increased but with different kinetics. These results give support for TRH and enkephalin neurons as targets of ethanol and, as possible mediators of some of its observed behavioral effects.


Subject(s)
Enkephalins/metabolism , Ethanol/pharmacology , Hypothalamus/metabolism , Limbic System/metabolism , Protein Precursors/metabolism , Thyrotropin-Releasing Hormone/metabolism , Animals , Enkephalins/genetics , Injections, Intraperitoneal , Male , Protein Precursors/genetics , Pyrrolidonecarboxylic Acid/analogs & derivatives , RNA, Messenger/metabolism , Rats , Rats, Wistar , Thyrotropin-Releasing Hormone/genetics , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...