Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.089
Filter
1.
Mol Biol Rep ; 51(1): 656, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38740671

ABSTRACT

BACKGROUND: Prokineticin 2 (PROK2), an important neuropeptide that plays a key role in the neuronal migration of gonadotropin-releasing hormone (GnRH) in the hypothalamus, is known to have regulatory effects on the gonads. In the present study, the impact of intracerebroventricular (icv) PROK2 infusion on hypothalamic-pituitary-gonadal axis (HPG) hormones, testicular tissues, and sperm concentration was investigated. METHODS AND RESULTS: Rats were randomly divided into four groups: control, sham, PROK2 1.5 and PROK2 4.5. Rats in the PROK2 1.5 and PROK2 4.5 groups were administered 1.5 nmol and 4.5 nmol PROK2 intracerebroventricularly for 7 days via an osmotic mini pump (1 µl/h), respectively. Rat blood serum follicle stimulating hormone (FSH), luteinizing hormone (LH) and testosterone hormone levels were determined with the ELISA method in the blood samples after 7 days of infusion. GnRH mRNA expression was determined with the RT-PCR in hypothalamus tissues. analyze Sperm concentration was determined, and testicular tissue was examined histologically with the hematoxylin-eosin staining method. It was observed that GnRH mRNA expression increased in both PROK2 infusion groups. Serum FSH, LH and testosterone hormone levels also increased in these groups. Although sperm concentration increased in PROK2 infusion groups when compared to the control and sham, the differences were not statistically significant. Testicular tissue seminiferous epithelial thickness was higher in the PROK2 groups when compared to the control and sham groups. CONCLUSION: The present study findings demonstrated that icv PROK2 infusion induced the HPG axis. It could be suggested that PROK2 could be a potential agent in the treatment of male infertility induced by endocrinological defects.


Subject(s)
Follicle Stimulating Hormone , Gastrointestinal Hormones , Gonadotropin-Releasing Hormone , Luteinizing Hormone , Neuropeptides , Testis , Testosterone , Male , Animals , Rats , Gastrointestinal Hormones/metabolism , Gonadotropin-Releasing Hormone/metabolism , Testosterone/blood , Testosterone/metabolism , Follicle Stimulating Hormone/blood , Follicle Stimulating Hormone/metabolism , Testis/metabolism , Testis/drug effects , Luteinizing Hormone/blood , Luteinizing Hormone/metabolism , Neuropeptides/metabolism , Neuropeptides/pharmacology , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/drug effects , Infusions, Intraventricular , Hypothalamus/metabolism , Hypothalamus/drug effects , Sperm Count , Rats, Sprague-Dawley , Hypothalamic-Pituitary-Gonadal Axis
2.
J Neurosci Res ; 102(5): e25339, 2024 May.
Article in English | MEDLINE | ID: mdl-38741550

ABSTRACT

Diets rich in saturated fats are more detrimental to health than those containing mono- or unsaturated fats. Fatty acids are an important source of energy, but they also relay information regarding nutritional status to hypothalamic metabolic circuits and when in excess can be detrimental to these circuits. Astrocytes are the main site of central fatty acid ß-oxidation, and hypothalamic astrocytes participate in energy homeostasis, in part by modulating hormonal and nutritional signals reaching metabolic neurons, as well as in the inflammatory response to high-fat diets. Thus, we hypothesized that how hypothalamic astrocytes process-specific fatty acids participates in determining the differential metabolic response and that this is sex dependent as males and females respond differently to high-fat diets. Male and female primary hypothalamic astrocyte cultures were treated with oleic acid (OA) or palmitic acid (PA) for 24 h, and an untargeted metabolomics study was performed. A clear predictive model for PA exposure was obtained, while the metabolome after OA exposure was not different from controls. The observed modifications in metabolites, as well as the expression levels of key metabolic enzymes, indicate a reduction in the activity of the Krebs and glutamate/glutamine cycles in response to PA. In addition, there were specific differences between the response of astrocytes from male and female mice, as well as between hypothalamic and cerebral cortical astrocytes. Thus, the response of hypothalamic astrocytes to specific fatty acids could result in differential impacts on surrounding metabolic neurons and resulting in varied systemic metabolic outcomes.


Subject(s)
Astrocytes , Hypothalamus , Oleic Acid , Palmitic Acid , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Oleic Acid/pharmacology , Female , Palmitic Acid/pharmacology , Hypothalamus/metabolism , Hypothalamus/drug effects , Male , Mice , Mice, Inbred C57BL , Sex Characteristics , Cells, Cultured
3.
PLoS One ; 19(5): e0303528, 2024.
Article in English | MEDLINE | ID: mdl-38753618

ABSTRACT

Arsenic has been identified as an environmental toxicant acting through various mechanisms, including the disruption of endocrine pathways. The present study assessed the ability of a single intraperitoneal injection of arsenic, to modify the mRNA expression levels of estrogen- and thyroid hormone receptors (ERα,ß; TRα,ß) and peroxisome proliferator-activated receptor gamma (PPARγ) in hypothalamic tissue homogenates of prepubertal mice in vivo. Mitochondrial respiration (MRR) was also measured, and the corresponding mitochondrial ultrastructure was analyzed. Results show that ERα,ß, and TRα expression was significantly increased by arsenic, in all concentrations examined. In contrast, TRß and PPARγ remained unaffected after arsenic injection. Arsenic-induced dose-dependent changes in state 4 mitochondrial respiration (St4). Mitochondrial morphology was affected by arsenic in that the 5 mg dose increased the size but decreased the number of mitochondria in agouti-related protein- (AgRP), while increasing the size without affecting the number of mitochondria in pro-opiomelanocortin (POMC) neurons. Arsenic also increased the size of the mitochondrial matrix per host mitochondrion. Complex analysis of dose-dependent response patterns between receptor mRNA, mitochondrial morphology, and mitochondrial respiration in the neuroendocrine hypothalamus suggests that instant arsenic effects on receptor mRNAs may not be directly reflected in St3-4 values, however, mitochondrial dynamics is affected, which predicts more pronounced effects in hypothalamus-regulated homeostatic processes after long-term arsenic exposure.


Subject(s)
Arsenic , Hypothalamus , Mitochondria , PPAR gamma , RNA, Messenger , Animals , Hypothalamus/metabolism , Hypothalamus/drug effects , Mice , Mitochondria/metabolism , Mitochondria/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , PPAR gamma/metabolism , PPAR gamma/genetics , Arsenic/toxicity , Receptors, Thyroid Hormone/metabolism , Receptors, Thyroid Hormone/genetics , Male , Receptors, Estrogen/metabolism , Receptors, Estrogen/genetics , Cell Respiration/drug effects , Gene Expression Regulation/drug effects
4.
Stress ; 27(1): 2357330, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38775373

ABSTRACT

Why individuals suffer negative consequences following stress is a complex phenomenon that is dictated by individual factors, the timing of stress within the lifespan, and when in the lifespan the consequences are measured. Women who undergo adverse childhood experiences are at risk for lasting biological consequences, including affective and stress dysregulation. We have shown that pubertal adversity is associated with a blunted hypothalamic-pituitary-adrenal axis glucocorticoid response in peripartum humans and mice. In mice, our prior examination of the paraventricular nucleus (PVN) of the hypothalamus showed that pubertal stress led to an upregulation of baseline mRNA expression of six immediate early genes (IEGs) in the PVN of adult, pregnant mice. Separately, we showed that the pregnancy-associated hormone allopregnanolone is necessary and sufficient to produce the blunted stress response phenotype in pubertally stressed mice. In the current study, we further examined a potential mechanistic role for the IEGs in the PVN. We found that in pubertally stressed adult female, but not male, mice, intra-PVN allopregnanolone was sufficient to recapitulate the baseline IEG mRNA expression profile previously observed in pubertally stressed, pregnant mice. We also examined baseline IEG mRNA expression during adolescence, where we found that IEGs have developmental trajectories that showed sex-specific disruption by pubertal stress. Altogether, these data establish that IEGs may act as a key molecular switch involved in increased vulnerability to negative outcomes in adult, pubertally stressed animals. How the factors that produce vulnerability combine throughout the lifespan is key to our understanding of the etiology of stress-related disorders.


Subject(s)
Paraventricular Hypothalamic Nucleus , Stress, Psychological , Transcriptome , Animals , Female , Male , Mice , Stress, Psychological/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Pregnanolone , Hypothalamus/metabolism , Hypothalamus/drug effects , Pregnancy , Hypothalamo-Hypophyseal System/metabolism , Hypothalamo-Hypophyseal System/drug effects , Sexual Maturation , Genes, Immediate-Early
5.
Proc Natl Acad Sci U S A ; 121(21): e2313207121, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38753512

ABSTRACT

Arginine vasopressin (AVP) neurons of the hypothalamic paraventricular region (AVPPVN) mediate sex-biased social behaviors across most species, including mammals. In mice, neural sex differences are thought to be established during a critical window around birth ( embryonic (E) day 18 to postnatal (P) day 2) whereby circulating testosterone from the fetal testis is converted to estrogen in sex-dimorphic brain regions. Here, we found that AVPPVN neurons are sexually dimorphic by E15.5, prior to this critical window, and that gestational bisphenol A (BPA) exposure permanently masculinized female AVPPVN neuronal numbers, projections, and electrophysiological properties, causing them to display male-like phenotypes into adulthood. Moreover, we showed that nearly twice as many neurons that became AVP+ by P0 were born at E11 in males and BPA-exposed females compared to control females, suggesting that AVPPVN neuronal masculinization occurs between E11 and P0. We further narrowed this sensitive period to around the timing of neurogenesis by demonstrating that exogenous estrogen exposure from E14.5 to E15.5 masculinized female AVPPVN neuronal numbers, whereas a pan-estrogen receptor antagonist exposed from E13.5 to E15.5 blocked masculinization of males. Finally, we showed that restricting BPA exposure to E7.5-E15.5 caused adult females to display increased social dominance over control females, consistent with an acquisition of male-like behaviors. Our study reveals an E11.5 to E15.5 window of estrogen sensitivity impacting AVPPVN sex differentiation, which is impacted by prenatal BPA exposure.


Subject(s)
Benzhydryl Compounds , Neurons , Phenols , Sex Differentiation , Animals , Benzhydryl Compounds/toxicity , Phenols/toxicity , Female , Male , Mice , Sex Differentiation/drug effects , Neurons/drug effects , Neurons/metabolism , Pregnancy , Hypothalamus/metabolism , Hypothalamus/drug effects , Neurogenesis/drug effects , Arginine Vasopressin/metabolism , Vasopressins/metabolism , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Mice, Inbred C57BL , Estrogens/metabolism , Estrogens/pharmacology
6.
Transl Psychiatry ; 14(1): 208, 2024 May 25.
Article in English | MEDLINE | ID: mdl-38796566

ABSTRACT

In clinical settings, tumor compression, trauma, surgical injury, and other types of injury can cause hypothalamic damage, resulting in various types of hypothalamic dysfunction. Impaired release of oxytocin can lead to cognitive impairment and affect prognosis and long-term quality of life after hypothalamic injury. Hypothalamic injury-induced cognitive dysfunction was detected in male animals. Behavioral parameters were measured to assess the characteristics of cognitive dysfunction induced by hypothalamic-pituitary stalk lesions. Brains were collected for high-throughput RNA sequencing and immunostaining to identify pathophysiological changes in hippocampal regions highly associated with cognitive function after injury to corresponding hypothalamic areas. Through transcriptomic analysis, we confirmed the loss of oxytocin neurons after hypothalamic injury and the reversal of hypothalamic-induced cognitive dysfunction after oxytocin supplementation. Furthermore, overactivation of the ERK signaling pathway and ß-amyloid deposition in the hippocampal region after hypothalamic injury were observed, and cognitive function was restored after inhibition of ERK signaling pathway overactivation. Our findings suggest that cognitive dysfunction after hypothalamic injury may be caused by ERK hyperphosphorylation in the hippocampal region resulting from a decrease in the number of oxytocin neurons, which in turn causes ß-amyloid deposition.


Subject(s)
Amyloid beta-Peptides , Cognitive Dysfunction , Hippocampus , Hypothalamus , MAP Kinase Signaling System , Oxytocin , Oxytocin/metabolism , Oxytocin/pharmacology , Animals , Hippocampus/metabolism , Hippocampus/drug effects , Male , Cognitive Dysfunction/etiology , Cognitive Dysfunction/metabolism , Hypothalamus/metabolism , Hypothalamus/drug effects , MAP Kinase Signaling System/drug effects , Amyloid beta-Peptides/metabolism , Neurons/drug effects , Neurons/metabolism , Disease Models, Animal , Mice , Phosphorylation
7.
Neuropharmacology ; 253: 109971, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38705568

ABSTRACT

The impact of environmental enrichment (EE) on natural rewards, including social and appetitive rewards, was investigated in male Swiss mice. EE, known for providing animals with various stimuli, was assessed for its effects on conditioned place preference (CPP) associated with ethanol and social stimuli. We previously demonstrated that EE increased the levels of the prosocial neuropeptide oxytocin (OT) in the hypothalamus and enhanced ethanol rewarding effects via an oxytocinergic mechanism. This study also investigated the impact of EE on social dominance and motivation for rewards, measured OT-mediated phospholipase C (PLC) activity in striatal membranes, and assessed OT expression in the hypothalamus. The role of dopamine in motivating rewards was considered, along with the interaction between OT and D1 receptors (DR) in the nucleus accumbens (NAc). Results showed that EE mice exhibited a preference for ethanol reward over social reward, a pattern replicated by the OT analogue Carbetocin. EE mice demonstrated increased social dominance and reduced motivation for appetitive taste stimuli. Higher OT mRNA levels in the hypothalamus were followed by diminished OT receptor (OTR) signaling activity in the striatum of EE mice. Additionally, EE mice displayed elevated D1R expression, which was attenuated by the OTR antagonist (L-368-889). The findings underscore the reinforcing effect of EE on ethanol and social rewards through an oxytocinergic mechanism. Nonetheless, they suggest that mechanisms other than the prosocial effect of EE may contribute to the ethanol pro-rewarding effect of EE and Carbetocin. They also point towards an OT-dopamine interaction potentially underlying some of these effects.


Subject(s)
Dopamine , Ethanol , Nucleus Accumbens , Oxytocin , Receptors, Dopamine D1 , Receptors, Oxytocin , Reward , Animals , Oxytocin/metabolism , Oxytocin/analogs & derivatives , Male , Ethanol/pharmacology , Ethanol/administration & dosage , Mice , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D1/antagonists & inhibitors , Dopamine/metabolism , Receptors, Oxytocin/metabolism , Receptors, Oxytocin/antagonists & inhibitors , Nucleus Accumbens/metabolism , Nucleus Accumbens/drug effects , Environment , Hypothalamus/metabolism , Hypothalamus/drug effects , Central Nervous System Depressants/pharmacology , Social Dominance , Social Behavior , Motivation/physiology , Motivation/drug effects
8.
Nature ; 629(8014): 1133-1141, 2024 May.
Article in English | MEDLINE | ID: mdl-38750368

ABSTRACT

The N-methyl-D-aspartate (NMDA) receptor is a glutamate-activated cation channel that is critical to many processes in the brain. Genome-wide association studies suggest that glutamatergic neurotransmission and NMDA receptor-mediated synaptic plasticity are important for body weight homeostasis1. Here we report the engineering and preclinical development of a bimodal molecule that integrates NMDA receptor antagonism with glucagon-like peptide-1 (GLP-1) receptor agonism to effectively reverse obesity, hyperglycaemia and dyslipidaemia in rodent models of metabolic disease. GLP-1-directed delivery of the NMDA receptor antagonist MK-801 affects neuroplasticity in the hypothalamus and brainstem. Importantly, targeting of MK-801 to GLP-1 receptor-expressing brain regions circumvents adverse physiological and behavioural effects associated with MK-801 monotherapy. In summary, our approach demonstrates the feasibility of using peptide-mediated targeting to achieve cell-specific ionotropic receptor modulation and highlights the therapeutic potential of unimolecular mixed GLP-1 receptor agonism and NMDA receptor antagonism for safe and effective obesity treatment.


Subject(s)
Dizocilpine Maleate , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor , Obesity , Receptors, N-Methyl-D-Aspartate , Animals , Humans , Male , Mice , Rats , Brain Stem/metabolism , Brain Stem/drug effects , Disease Models, Animal , Dizocilpine Maleate/adverse effects , Dizocilpine Maleate/pharmacology , Dizocilpine Maleate/therapeutic use , Dyslipidemias/drug therapy , Dyslipidemias/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Mice, Inbred C57BL , Neuronal Plasticity/drug effects , Obesity/drug therapy , Obesity/metabolism , Rats, Sprague-Dawley , Rats, Wistar , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors
9.
Biochim Biophys Acta Mol Basis Dis ; 1870(5): 167227, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38733774

ABSTRACT

Olanzapine (OLA) is a highly obesogenic second-generation antipsychotic (SGA). Recently we demonstrated that, contrarily to OLA oral treatment, intraperitoneal (i.p.) administration resulted in weight loss and absence of hepatic steatosis in wild-type (WT) and protein tyrosine phosphatase 1B (PTP1B)-deficient (KO) male mice. This protection relied on two central-peripheral axes connecting hypothalamic AMPK with brown/inguinal white adipose tissue (BAT/iWAT) uncoupling protein-1 (UCP-1) and hypothalamic JNK with hepatic fatty acid synthase (FAS). Herein, we addressed OLA i.p. treatment effects in WT and PTP1B-KO female mice. Contrarily to our previous results in WT females receiving OLA orally, the i.p. treatment did not induce weight gain or hyperphagia. Molecularly, in females OLA failed to diminish hypothalamic phospho-AMPK or elevate BAT UCP-1 and energy expenditure (EE) despite the preservation of iWAT browning. Conversely, OLA i.p. treatment in ovariectomized mice reduced hypothalamic phospho-AMPK, increased BAT/iWAT UCP-1 and EE, and induced weight loss as occurred in males. Pretreatment of hypothalamic neurons with 17ß-estradiol (E2) abolished OLA effects on AMPK. Moreover, neither hypothalamic JNK activation nor hepatic FAS upregulation were found in WT and PTP1B-KO females receiving OLA via i.p. Importantly, this axis was reestablished upon ovariectomy. In this line, E2 prevented OLA-induced phospho-JNK in hypothalamic neurons. These results support the role of estrogens in sex-related dimorphism in OLA treatment. This study evidenced the benefit of OLA i.p. administration in preventing its obesogenic effects in female mice that could offer clinical value.


Subject(s)
Adipose Tissue, Brown , Estrogens , Hypothalamus , Liver , Mice, Knockout , Olanzapine , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Uncoupling Protein 1 , Animals , Female , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Hypothalamus/metabolism , Hypothalamus/drug effects , Mice , Liver/metabolism , Liver/drug effects , Estrogens/metabolism , Estrogens/pharmacology , Olanzapine/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Uncoupling Protein 1/metabolism , Uncoupling Protein 1/genetics , Male , Energy Metabolism/drug effects , Injections, Intraperitoneal , Adipose Tissue, White/metabolism , Adipose Tissue, White/drug effects , Mice, Inbred C57BL , Estradiol/pharmacology , Ovariectomy
10.
Behav Brain Res ; 466: 115001, 2024 May 28.
Article in English | MEDLINE | ID: mdl-38642861

ABSTRACT

INTRODUCTION: Anorexia Nervosa (AN) is a psycho-socio-biological disease characterized by severe weight loss as result of dieting and hyperactivity. Effective treatments are scarce, despite its significant prevalence and mortality. AN patients show lower basal insulin levels and increased metabolic clearance, leading to weight loss, cognitive deficits, and hormonal imbalances. Low-dose polymer insulin could potentially reverse these effects by restoring brain function, reducing fear of weight gain, encouraging food intake, and restoring fat depots. This study evaluates an insulin delivery system designed for sustained release and AN treatment. METHODS: AN-like model was established through dietary restriction (DR). On days 1-25, mice were on DR, and on days 26-31 they were on ad libitum regimen. An insulin-loaded delivery system was administered subcutaneously (1% w/w insulin). The impact of insulin treatment on gene expression in the hippocampus (cognition, regulation of stress, neurogenesis) and hypothalamus (eating behavior, mood) was assessed. Behavioral assays were conducted to evaluate motor activity and cognitive function. RESULTS: The delivery system demonstrated sustained insulin release, maintaining therapeutic plasma levels. Diet restriction mice treated with the insulin delivery system showed body weight restoration. Gene expression analysis revealed enhanced expression of CB1 and CB2 genes associated with improved eating behavior and cognition, while POMC expression was reduced. Insulin-polymer treatment restored cognitive function and decreased hyperactivity in the AN-like model. CONCLUSION: The PSA-RA-based insulin delivery system effectively restores metabolic balance, body weight, and cognitive function in the AN model. Its ability to steadily release insulin makes it a promising candidate for AN treatment."


Subject(s)
Anorexia Nervosa , Body Weight , Disease Models, Animal , Insulin , Animals , Insulin/administration & dosage , Insulin/pharmacology , Mice , Anorexia Nervosa/drug therapy , Anorexia Nervosa/metabolism , Body Weight/drug effects , Cognition/drug effects , Hippocampus/metabolism , Hippocampus/drug effects , Female , Hypothalamus/metabolism , Hypothalamus/drug effects , Mice, Inbred C57BL
11.
Biosci Rep ; 44(4)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38577975

ABSTRACT

Since 1975, the incidence of obesity has increased to epidemic proportions, and the number of patients with obesity has quadrupled. Obesity is a major risk factor for developing other serious diseases, such as type 2 diabetes mellitus, hypertension, and cardiovascular diseases. Recent epidemiologic studies have defined obesity as a risk factor for the development of neurodegenerative diseases, such as Alzheimer's disease (AD) and other types of dementia. Despite all these serious comorbidities associated with obesity, there is still a lack of effective antiobesity treatment. Promising candidates for the treatment of obesity are anorexigenic neuropeptides, which are peptides produced by neurons in brain areas implicated in food intake regulation, such as the hypothalamus or the brainstem. These peptides efficiently reduce food intake and body weight. Moreover, because of the proven interconnection between obesity and the risk of developing AD, the potential neuroprotective effects of these two agents in animal models of neurodegeneration have been examined. The objective of this review was to explore anorexigenic neuropeptides produced and acting within the brain, emphasizing their potential not only for the treatment of obesity but also for the treatment of neurodegenerative disorders.


Subject(s)
Anti-Obesity Agents , Neuropeptides , Neuroprotective Agents , Obesity , Humans , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Animals , Obesity/drug therapy , Obesity/metabolism , Neuropeptides/metabolism , Neuropeptides/pharmacology , Neuropeptides/therapeutic use , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Neurodegenerative Diseases/drug therapy , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/prevention & control , Hypothalamus/drug effects , Hypothalamus/metabolism , Hypothalamus/pathology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Alzheimer Disease/prevention & control , Brain/drug effects , Brain/metabolism , Brain/pathology , Eating/drug effects
12.
PLoS One ; 19(4): e0300544, 2024.
Article in English | MEDLINE | ID: mdl-38656972

ABSTRACT

Obesity is a major global health epidemic that has adverse effects on both the people affected as well as the cost to society. Several anti-obesity drugs that target GLP-1 receptors have recently come to the market. Here, we describe the effects of tesofensine, a novel anti-obesity drug that acts as a triple monoamine neurotransmitter reuptake inhibitor. Using various techniques, we investigated its effects on weight loss and underlying neuronal mechanisms in mice and rats. These include behavioral tasks, DeepLabCut videotaped analysis, electrophysiological ensemble recordings, optogenetic activation, and chemogenetic silencing of GABAergic neurons in the Lateral Hypothalamus (LH). We found that tesofensine induces a greater weight loss in obese rats than lean rats, while differentially modulating the neuronal ensembles and population activity in LH. In Vgat-ChR2 and Vgat-IRES-cre transgenic mice, we found for the first time that tesofensine inhibited a subset of LH GABAergic neurons, reducing their ability to promote feeding behavior, and chemogenetically silencing them enhanced tesofensine's food-suppressing effects. Unlike phentermine, a dopaminergic appetite suppressant, tesofensine causes few, if any, head-weaving stereotypy at therapeutic doses. Most importantly, we found that tesofensine prolonged the weight loss induced by 5-HTP, a serotonin precursor, and blocked the body weight rebound that often occurs after weight loss. Behavioral studies on rats with the tastant sucrose indicated that tesofensine's appetite suppressant effects are independent of taste aversion and do not directly affect the perception of sweetness or palatability of sucrose. In summary, our data provide new insights into the effects of tesofensine on weight loss and the underlying neuronal mechanisms, suggesting that tesofensine may be an effective treatment for obesity and that it may be a valuable adjunct to other appetite suppressants to prevent body weight rebound.


Subject(s)
Anti-Obesity Agents , Bridged Bicyclo Compounds, Heterocyclic , GABAergic Neurons , Obesity , Animals , GABAergic Neurons/drug effects , GABAergic Neurons/metabolism , Rats , Mice , Anti-Obesity Agents/pharmacology , Male , Obesity/drug therapy , Obesity/metabolism , Feeding Behavior/drug effects , Hypothalamic Area, Lateral/drug effects , Hypothalamic Area, Lateral/metabolism , Hypothalamus/drug effects , Hypothalamus/metabolism , Mice, Transgenic , Weight Loss/drug effects , Rats, Sprague-Dawley
13.
Commun Biol ; 7(1): 460, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38649481

ABSTRACT

NGLY1 deficiency is a genetic disease caused by biallelic mutations of the Ngly1 gene. Although epileptic seizure is one of the most severe symptoms in patients with NGLY1 deficiency, preclinical studies have not been conducted due to the lack of animal models for epileptic seizures in NGLY1 deficiency. Here, we observed the behaviors of male and female Ngly1-/- mice by video monitoring and found that these mice exhibit spontaneous seizure-like behaviors. Gene expression analyses and enzyme immunoassay revealed significant decreases in oxytocin, a well-known neuropeptide, in the hypothalamus of Ngly1-/- mice. Seizure-like behaviors in Ngly1-/- mice were transiently suppressed by a single intranasal administration of oxytocin. These findings suggest the therapeutic potential of oxytocin for epileptic seizure in patients with NGLY1 deficiency and contribute to the clarification of the disease mechanism.


Subject(s)
Congenital Disorders of Glycosylation , Oxytocin , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase , Seizures , Animals , Female , Male , Mice , Administration, Intranasal , Behavior, Animal/drug effects , Disease Models, Animal , Hypothalamus/metabolism , Hypothalamus/drug effects , Mice, Inbred C57BL , Mice, Knockout , Oxytocin/administration & dosage , Oxytocin/pharmacology , Seizures/drug therapy , Seizures/etiology , Congenital Disorders of Glycosylation/complications , Congenital Disorders of Glycosylation/drug therapy , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/deficiency
14.
In Vivo ; 38(3): 1133-1142, 2024.
Article in English | MEDLINE | ID: mdl-38688635

ABSTRACT

BACKGROUND/AIM: Cancer-induced bone pain (CIBP) is one of the most common symptoms of bone metastasis of tumor cells. The hypothalamus may play a pivotal role in the regulation of CIBP. However, little is known about the exact mechanisms. MATERIALS AND METHODS: First, we established a CIBP model to explore the relationship among hypothalamic ghrelin, NPY and CIBP. Then, we exogenously administered NPY and NPY receptor antagonists to investigate whether hypothalamic NPY exerted an antinociceptive effect through binding to NPY receptors. Finally, we exogenously administered ghrelin to investigate whether ghrelin alleviated CIBP by inducing the production of hypothalamic NPY through the AMPK-mTOR pathway. Body weight, food intake and behavioral indicators of CIBP were measured every 3 days. Hypothalamic ghrelin, NPY and the AMPK-mTOR pathway were also measured. RESULTS: The expression of hypothalamic ghrelin and NPY was simultaneously decreased in cancer-bearing rats, which was accompanied by CIBP. Intracerebroventricular (i.c.v.) administration of NPY significantly alleviated CIBP in the short term. The antinociceptive effect of NPY was reversed with the i.c.v. administration of the Y1R and Y2R antagonists. The administration of ghrelin activated the AMPK-mTOR pathway and induced hypothalamic NPY production to alleviate CIBP. This effect of ghrelin on NPY and antinociception was reversed with the administration of a GHS-R1α antagonist. CONCLUSION: Ghrelin could induce the production of hypothalamic NPY through the AMPK-mTOR pathway to alleviate CIBP, which can provide a novel therapeutic mechanism for CIBP.


Subject(s)
AMP-Activated Protein Kinases , Bone Neoplasms , Cancer Pain , Disease Models, Animal , Ghrelin , Hypothalamus , Neuropeptide Y , Signal Transduction , TOR Serine-Threonine Kinases , Animals , Ghrelin/pharmacology , Hypothalamus/metabolism , Hypothalamus/drug effects , TOR Serine-Threonine Kinases/metabolism , Neuropeptide Y/metabolism , Rats , Cancer Pain/etiology , Cancer Pain/drug therapy , Cancer Pain/metabolism , Signal Transduction/drug effects , AMP-Activated Protein Kinases/metabolism , Bone Neoplasms/metabolism , Bone Neoplasms/complications , Bone Neoplasms/drug therapy , Male , Cell Line, Tumor , Female
15.
Neurosci Lett ; 830: 137770, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38616004

ABSTRACT

Women are disproportionately affected by stress-related disorders like depression. In our prior research, we discovered that females exhibit lower basal hypothalamic reelin levels, and these levels are differentially influenced by chronic stress induced through repeated corticosterone (CORT) injections. Although epigenetic mechanisms involving DNA methylation and the formation of repressor complexes by DNA methyl-transferases (DNMTs) and Methyl-CpG binding protein 2 (MeCP2) have been recognized as regulators of reelin expression in vitro, there is limited understanding of the impact of stress on the epigenetic regulation of reelin in vivo and whether sex differences exist in these mechanisms. To address these questions, we conducted various biochemical analyses on hypothalamic brain samples obtained from male and female rats previously treated with either 21 days of CORT (40 mg/kg) or vehicle (0.9 % saline) subcutaneous injections. Upon chronic CORT treatment, a reduction in reelin fragment NR2 was noted in males, while the full-length molecule remained unaffected. This decrease paralleled with an elevation in MeCP2 and a reduction in DNMT3a protein levels only in males. Importantly, sex differences in baseline and CORT-induced reelin protein levels were not associated with changes in the methylation status of the Reln promoter. These findings suggest that CORT-induced reelin decreases in the hypothalamus may be a combination of alterations in downstream processes beyond gene transcription. This research brings novel insights into the sexually distinct consequences of chronic stress, an essential aspect to understand, particularly concerning its role in the development of depression.


Subject(s)
Cell Adhesion Molecules, Neuronal , Corticosterone , DNA Methyltransferase 3A , Extracellular Matrix Proteins , Hypothalamus , Methyl-CpG-Binding Protein 2 , Nerve Tissue Proteins , Reelin Protein , Serine Endopeptidases , Animals , Female , Male , Rats , Cell Adhesion Molecules, Neuronal/metabolism , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A/metabolism , Extracellular Matrix Proteins/metabolism , Hypothalamus/metabolism , Hypothalamus/drug effects , Methyl-CpG-Binding Protein 2/metabolism , Methyl-CpG-Binding Protein 2/genetics , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Serine Endopeptidases/metabolism , Sex Characteristics , Rats, Long-Evans
16.
Food Funct ; 15(8): 4627-4641, 2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38592736

ABSTRACT

Diet-induced thermogenesis (DIT) is crucial for maintaining body weight homeostasis, and the role of dietary fatty acids in modulating DIT is essential. However, the underlying mechanism of fatty acid regulated diet-induced thermogenesis remains elusive. Utilizing the diet- and genetic ablation-induced obese mice models, we found that the C16 unsaturated fatty acids, trans-palmitoleic acid (TPA) and cis-palmitoleic acid (CPA), significantly increased the energy expenditure by promoting the thermogenesis of brown adipose tissues and the production of beige cells in white adipose. As a result, there is a significant reduction in the occurrence of obesity, associated hepatic steatosis and hyperglycemia. Notably, TPA exhibited more potent effects on promoting DIT and alleviating obesity than CPA did. Using inhibitor and gene deletion mice models, we unveiled that TPA acted as a signaling molecule to play a biological function, which could be sensed by the hypothalamic FFAR1 to activate the sympathetic nervous system in promoting adipose tissue thermogenesis. Together, these results demonstrate the underlying mechanism of free fatty acids associated-DIT and will provide fresh insights into the roles of trans-fatty acids in the development of obesity.


Subject(s)
Fatty Acids, Monounsaturated , Hypothalamus , Mice, Inbred C57BL , Obesity , Receptors, G-Protein-Coupled , Signal Transduction , Thermogenesis , Animals , Thermogenesis/drug effects , Mice , Obesity/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , Fatty Acids, Monounsaturated/pharmacology , Hypothalamus/metabolism , Hypothalamus/drug effects , Male , Signal Transduction/drug effects , Energy Metabolism/drug effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Adipose Tissue, White/drug effects , Diet, High-Fat
17.
Am J Physiol Endocrinol Metab ; 326(5): E735-E746, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38597830

ABSTRACT

Most studies on fat appetite have focused on long-chain triglycerides (LCTs) due to their obesogenic properties. Medium-chain triglycerides (MCTs), conversely, exhibit antiobesogenic effects; however, the regulation of MCT intake remains elusive. Here, we demonstrate that mice can distinguish between MCTs and LCTs, and the specific appetite for MCTs is governed by hepatic ß-oxidation. We generated liver-specific medium-chain acyl-CoA dehydrogenase (MCAD)-deficient (MCADL-/-) mice and analyzed their preference for MCT and LCT solutions using glyceryl trioctanoate (C8-TG), glyceryl tridecanoate (C10-TG), corn oil, and lard oil in two-bottle choice tests conducted over 8 days. In addition, we used lick microstructure analyses to evaluate the palatability and appetite for MCT and LCT solutions. Finally, we measured the expression levels of genes associated with fat ingestion (Galanin, Qrfp, and Nmu) in the hypothalamus 2 h after oral gavage of fat. Compared with control mice, MCADL-/- mice exhibited a significantly reduced preference for MCT solutions, with no alteration in the preference for LCTs. Lick analysis revealed that MCADL-/- mice displayed a significantly decreased appetite for MCT solutions only while the palatability of both MCT and LCT solutions remained unaffected. Hypothalamic Galanin expression in control mice was elevated by oral gavage of C8-TG but not by LCTs, and this response was abrogated in MCADL-/- mice. In summary, our data suggest that hepatic ß-oxidation is required for MCT-specific appetite but not for LCT-specific appetite. The induction of hypothalamic galanin upon MCT ingestion, dependent on hepatic ß-oxidation, could be involved in the regulation of MCT-specific appetite.NEW & NOTEWORTHY Whether and how medium-chain triglyceride (MCT) intake is regulated remains unknown. Here, we showed that mice can discriminate between MCTs and LCTs. Hepatic ß-oxidation participates in MCT-specific appetite, and hypothalamic galanin may be one of the factors that regulate MCT intake. Because of the antiobesity effects of MCTs, studying MCT-specific appetite may help combat obesity by promoting the intake of MCTs instead of LCTs.


Subject(s)
Acyl-CoA Dehydrogenase , Appetite , Fatty Acids , Liver , Mice, Knockout , Oxidation-Reduction , Triglycerides , Animals , Triglycerides/metabolism , Mice , Oxidation-Reduction/drug effects , Liver/metabolism , Liver/drug effects , Male , Fatty Acids/metabolism , Appetite/drug effects , Appetite/physiology , Acyl-CoA Dehydrogenase/metabolism , Acyl-CoA Dehydrogenase/genetics , Mice, Inbred C57BL , Hypothalamus/metabolism , Hypothalamus/drug effects
18.
Cell Mol Biol (Noisy-le-grand) ; 70(4): 53-60, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38678627

ABSTRACT

Cobalt protoporphyrin (CoPP) is a synthetic heme analog that has been observed to reduce food intake and promote sustained weight loss. While the precise mechanisms responsible for these effects remain elusive, earlier research has hinted at the potential involvement of nitric oxide synthase in the hypothalamus. This study aimed to delve into CoPP's impact on the activities of crucial antioxidant enzymes: superoxide dismutase (SOD), catalase, glutathione peroxidase (GPx), glutathione reductase (GR), and glutathione-S-transferase (GST) across seven distinct brain regions (hippocampus, hypothalamus, prefrontal cortex, motor cortex, striatum, midbrain, and cerebellum), as well as in the liver and kidneys. Female Wistar rats weighing 180 to 200 grams received a single subcutaneous dose of 25 µmol/kg CoPP. After six days, brain tissue was extracted to assess the activities of antioxidant enzymes and quantify malondialdehyde levels. Our findings confirm that CoPP administration triggers the characteristic effects of decreased food intake and reduced body weight. Moreover, it led to an increase in SOD activity in the hypothalamus, a pivotal brain region associated with food intake regulation. Notably, CoPP-treated rats exhibited elevated enzymatic activity of catalase, GR, and GST in the motor cortex without concurrent signs of heightened oxidative stress. These results underscore a strong connection between the antioxidant system and food intake regulation. They also emphasize the need for further investigation into the roles of antioxidant enzymes in modulating food intake and the ensuing weight loss, using CoPP as a valuable research tool.


Subject(s)
Antioxidants , Hypothalamus , Motor Cortex , Protoporphyrins , Animals , Female , Rats , Antioxidants/metabolism , Body Weight/drug effects , Catalase/metabolism , Eating/drug effects , Glutathione Peroxidase/drug effects , Glutathione Peroxidase/metabolism , Glutathione Reductase/drug effects , Glutathione Reductase/metabolism , Glutathione Transferase/drug effects , Glutathione Transferase/metabolism , Hypothalamus/metabolism , Hypothalamus/drug effects , Hypothalamus/enzymology , Malondialdehyde/metabolism , Motor Cortex/drug effects , Motor Cortex/metabolism , Motor Cortex/enzymology , Oxidative Stress/drug effects , Protoporphyrins/pharmacology , Rats, Wistar , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism
19.
Am J Physiol Endocrinol Metab ; 326(5): E681-E695, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38597829

ABSTRACT

Hypothalamic proopiomelanocortin (POMC) neurons are sensors of signals that reflect the energy stored in the body. Inducing mild stress in proopiomelanocortin neurons protects them from the damage promoted by the consumption of a high-fat diet, mitigating the development of obesity; however, the cellular mechanisms behind these effects are unknown. Here, we induced mild stress in a proopiomelanocortin neuron cell line by inhibiting Crif1. In proopiomelanocortin neurons exposed to high levels of palmitate, the partial inhibition of Crif1 reverted the defects in mitochondrial respiration and ATP production; this was accompanied by improved mitochondrial fusion/fission cycling. Furthermore, the partial inhibition of Crif1 resulted in increased reactive oxygen species production, increased fatty acid oxidation, and reduced dependency on glucose for mitochondrial respiration. These changes were dependent on the activity of CPT-1. Thus, we identified a CPT-1-dependent metabolic shift toward greater utilization of fatty acids as substrates for respiration as the mechanism behind the protective effect of mild stress against palmitate-induced damage of proopiomelanocortin neurons.NEW & NOTEWORTHY Saturated fats can damage hypothalamic neurons resulting in positive energy balance, and this is mitigated by mild cellular stress; however, the mechanisms behind this protective effect are unknown. Using a proopiomelanocortin cell line, we show that under exposure to a high concentration of palmitate, the partial inhibition of the mitochondrial protein Crif1 results in protection due to a metabolic shift warranted by the increased expression and activity of the mitochondrial fatty acid transporter CPT-1.


Subject(s)
Carnitine O-Palmitoyltransferase , Cell Cycle Proteins , Fatty Acids , Mitochondria , Animals , Mice , Carnitine O-Palmitoyltransferase/metabolism , Carnitine O-Palmitoyltransferase/genetics , Cell Line , Fatty Acids/metabolism , Hypothalamus/metabolism , Hypothalamus/drug effects , Mitochondria/metabolism , Mitochondria/drug effects , Neurons/drug effects , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Pro-Opiomelanocortin/genetics , Reactive Oxygen Species/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism
20.
Life Sci ; 346: 122649, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38626868

ABSTRACT

AIMS: Leptin irresponsiveness, which is often associated with obesity, can have significant impacts on the hypothalamic proteome of individuals, including those who are lean. While mounting evidence on leptin irresponsiveness has focused on obese individuals, understanding the early molecular and proteomic changes associated with deficient hypothalamic leptin signaling in lean individuals is essential for early intervention and prevention of metabolic disorders. Leptin receptor antagonists block the binding of leptin to its receptors, potentially reducing its effects and used in cases where excessive leptin activity might be harmful. MATERIALS AND METHODS: In this work, we blocked the central actions of leptin in lean male adult Wistar rat by chronically administering intracerebroventricularly the superactive leptin receptor antagonist (SLA) (D23L/L39A/D40A/F41A) and investigated its impact on the hypothalamic proteome using label-free sequential window acquisition of all theoretical fragment ion spectra mass spectrometry (SWATH-MS) for quantitative proteomics. KEY FINDINGS: Our results show an accumulation of proteins involved in mRNA processing, mRNA stability, and translation in the hypothalamus of SLA-treated rats. Conversely, hypothalamic leptin signaling deficiency reduces the representation of proteins implicated in energy metabolism, neural circuitry, and neurotransmitter release. SIGNIFICANCE: The alterations in the adult rat hypothalamic proteome contribute to dysregulate appetite, metabolism, and energy balance, which are key factors in the development and progression of obesity and related metabolic disorders. Additionally, using bioinformatic analysis, we identified a series of transcription factors that are potentially involved in the upstream regulatory mechanisms responsible for the observed signature.


Subject(s)
Hypothalamus , Leptin , Proteome , Proteomics , Rats, Wistar , Receptors, Leptin , Signal Transduction , Animals , Male , Leptin/metabolism , Receptors, Leptin/metabolism , Receptors, Leptin/genetics , Receptors, Leptin/deficiency , Hypothalamus/metabolism , Hypothalamus/drug effects , Rats , Signal Transduction/drug effects , Proteomics/methods , Proteome/metabolism , Obesity/metabolism , Energy Metabolism/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...