Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.795
1.
Cell Mol Biol (Noisy-le-grand) ; 70(4): 53-60, 2024 Apr 28.
Article En | MEDLINE | ID: mdl-38678627

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.


Antioxidants , Hypothalamus , Motor Cortex , Protoporphyrins , Rats, Wistar , Superoxide Dismutase , Animals , Female , Hypothalamus/metabolism , Hypothalamus/drug effects , Hypothalamus/enzymology , Antioxidants/metabolism , Protoporphyrins/pharmacology , Motor Cortex/drug effects , Motor Cortex/metabolism , Motor Cortex/enzymology , Superoxide Dismutase/metabolism , Catalase/metabolism , Rats , Oxidative Stress/drug effects , Glutathione Peroxidase/metabolism , Eating/drug effects , Glutathione Transferase/metabolism , Body Weight/drug effects , Glutathione Reductase/metabolism , Malondialdehyde/metabolism
2.
Arthritis Rheumatol ; 74(2): 212-222, 2022 02.
Article En | MEDLINE | ID: mdl-34398520

OBJECTIVE: To investigate whether thermogenesis and the hypothalamus may be involved in the physiopathology of experimental arthritis (EA). METHODS: EA was induced in male Lewis rats by intradermal injection of Freund's complete adjuvant (CFA). Food intake, body weight, plasma cytokines, thermographic analysis, gene and protein expression of thermogenic markers in brown adipose tissue (BAT) and white adipose tissue (WAT), and hypothalamic AMP-activated protein kinase (AMPK) were analyzed. Virogenetic activation of hypothalamic AMPK was performed. RESULTS: We first demonstrated that EA was associated with increased BAT thermogenesis and browning of subcutaneous WAT leading to elevated energy expenditure. Moreover, rats experiencing EA showed inhibition of hypothalamic AMPK, a canonical energy sensor modulating energy homeostasis at the central level. Notably, specific genetic activation of AMPK in the ventromedial nucleus of the hypothalamus (a key site modulating energy metabolism) reversed the effect of EA on energy balance, brown fat, and browning, as well as promoting amelioration of synovial inflammation in experimental arthritis. CONCLUSION: Overall, these data indicate that EA promotes a central catabolic state that can be targeted and reversed by the activation of hypothalamic AMPK. This might provide new therapeutic alternatives to treat rheumatoid arthritis (RA)-associated metabolic comorbidities, improving the overall prognosis in patients with RA.


AMP-Activated Protein Kinases/physiology , Arthritis/metabolism , Arthritis/physiopathology , Hypothalamus/enzymology , Thermogenesis , Animals , Arthritis/complications , Male , Rats , Rats, Inbred Lew
3.
J Clin Invest ; 131(24)2021 12 15.
Article En | MEDLINE | ID: mdl-34730112

The positive regulatory (PR) domain containing 13 (PRDM13) putative chromatin modifier and transcriptional regulator functions downstream of the transcription factor PTF1A, which controls GABAergic fate in the spinal cord and neurogenesis in the hypothalamus. Here, we report a recessive syndrome associated with PRDM13 mutation. Patients exhibited intellectual disability, ataxia with cerebellar hypoplasia, scoliosis, and delayed puberty with congenital hypogonadotropic hypogonadism (CHH). Expression studies revealed Prdm13/PRDM13 transcripts in the developing hypothalamus and cerebellum in mouse and human. An analysis of hypothalamus and cerebellum development in mice homozygous for a Prdm13 mutant allele revealed a significant reduction in the number of Kisspeptin (Kiss1) neurons in the hypothalamus and PAX2+ progenitors emerging from the cerebellar ventricular zone. The latter was accompanied by ectopic expression of the glutamatergic lineage marker TLX3. Prdm13-deficient mice displayed cerebellar hypoplasia and normal gonadal structure, but delayed pubertal onset. Together, these findings identify PRDM13 as a critical regulator of GABAergic cell fate in the cerebellum and of hypothalamic kisspeptin neuron development, providing a mechanistic explanation for the cooccurrence of CHH and cerebellar hypoplasia in this syndrome. To our knowledge, this is the first evidence linking disrupted PRDM13-mediated regulation of Kiss1 neurons to CHH in humans.


Cerebellum/abnormalities , Histone-Lysine N-Methyltransferase , Hypogonadism , Hypothalamus/enzymology , Mutation , Nervous System Malformations , Transcription Factors , Animals , Cerebellum/enzymology , Developmental Disabilities/enzymology , Developmental Disabilities/genetics , Disease Models, Animal , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Humans , Hypogonadism/enzymology , Hypogonadism/genetics , Mice , Mice, Mutant Strains , Nervous System Malformations/enzymology , Nervous System Malformations/genetics , Neurons/enzymology , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Nat Metab ; 3(10): 1415-1431, 2021 10.
Article En | MEDLINE | ID: mdl-34675439

Current pharmacological therapies for treating obesity are of limited efficacy. Genetic ablation or loss of function of AMP-activated protein kinase alpha 1 (AMPKα1) in steroidogenic factor 1 (SF1) neurons of the ventromedial nucleus of the hypothalamus (VMH) induces feeding-independent resistance to obesity due to sympathetic activation of brown adipose tissue (BAT) thermogenesis. Here, we show that body weight of obese mice can be reduced by intravenous injection of small extracellular vesicles (sEVs) delivering a plasmid encoding an AMPKα1 dominant negative mutant (AMPKα1-DN) targeted to VMH-SF1 neurons. The beneficial effect of SF1-AMPKα1-DN-loaded sEVs is feeding-independent and involves sympathetic nerve activation and increased UCP1-dependent thermogenesis in BAT. Our results underscore the potential of sEVs to specifically target AMPK in hypothalamic neurons and introduce a broader strategy to manipulate body weight and reduce obesity.


AMP-Activated Protein Kinases/metabolism , Adipose Tissue, Brown/enzymology , Extracellular Vesicles/metabolism , Hypothalamus/enzymology , Obesity/metabolism , Animals , Energy Metabolism , Mice , Thermogenesis , Weight Loss
5.
Pflugers Arch ; 473(8): 1199-1211, 2021 08.
Article En | MEDLINE | ID: mdl-34075446

Obesogenic diets (ODs) can affect AMPK activation in several sites as the colon, liver, and hypothalamus. OD intake can impair the hypothalamic AMPK regulation of energy homeostasis. Despite consuming ODs, not all subjects have the propensity to develop or progress to obesity. The obesity propensity is more associated with energy intake than expenditure dysregulations and may have a link with AMPK activity. While the effects of ODs are studied widely, few evaluate the short-term effects of terminating OD intake. Withdrawing from OD (WTD) is thought to improve or reverse the damages caused by the intake. Therefore, here we applied an OD intake and WTD protocol aiming to evaluate AMPK protein content and phosphorylation in the colon, liver, and hypothalamus and their relationship with obesity propensity. To this end, male Wistar rats (60 days) received control or high-sugar/high-fat (HSHF) OD for 30 days. Half of the animals were OD-withdrawn and fed the control diet for 48 h. After intake, we found a reduction in AMPK phosphorylation in the hypothalamus and colon, and after WTD, we found an increase in its hepatic and hypothalamic phosphorylation. The decrease in colon pAMPK/AMPK could be linked with hypothalamic pAMPK/AMPK after HSHF intake, while the increase in hepatic pAMPK/AMPK could have prevented the increase in hypothalamic pAMPK/AMPK. In the obesity-prone rats, we found higher levels of hypothalamic and colon pAMPK/AMPK despite the higher body mass gain. Our results highlight the relevance in multi-organ investigations and animal phenotype evaluation when studying the energy metabolism regulations.


AMP-Activated Protein Kinases/metabolism , Brain-Gut Axis , Colon/enzymology , Hypothalamus/enzymology , Liver/enzymology , Obesity/enzymology , Animals , Diet, High-Fat/adverse effects , Dietary Sugars/adverse effects , Male , Obesity/etiology , Rats, Wistar
6.
Gen Comp Endocrinol ; 311: 113838, 2021 09 15.
Article En | MEDLINE | ID: mdl-34181935

To investigate whether there are important interactions in play in broilers between thyroid hormones and the central regulation of energy homeostasis through AMP-activated protein kinase (AMPK), we induced a functional hyperthyroid and hypothyroid state in broiler chicks, and quantified systemic and hypothalamic AMPK related gene expression and related protein. Thyroid state was manipulated through dietary supplementation of triiodothyronine (T3) or methimazole (MMI) for 7 days. A hypothalamic AMPK suppressor, 0.1% α-lipoic acid (α-LA) was used to assess the effects of the T3 and MMI feed formulations on the AMPK pathways. Feed intake and body weight were reduced in both hypothyroid and hyperthyroid conditions. In hyperthyroid conditions (T3 supplementation) expression of the AMPKα1 subunit increased, while in hypothyroid conditions (MMI supplementation) active phosphorylated AMPK levels in the hypothalamus dropped, but gene expression of the AMPKα1 and α2 subunit increased. For FAS and ACC (involved in fatty acid metabolism), and CRH, TRH and CNR1 (anorexigenic neuropeptides stimulating energy expenditure) there were indications that their regulation in response to thyroid state might be modulated through AMPK pathways. Our results indicate that the expression of hypothalamic AMPK as well as that of several other genes from AMPK pathways are involved in thyroid-hormone-induced changes in appetite, albeit differently according to thyroid state.


AMP-Activated Protein Kinases , Chickens , Hypothalamus/enzymology , Thyroid Gland/physiopathology , AMP-Activated Protein Kinases/metabolism , Animals , Chickens/metabolism , Eating , Energy Metabolism , Homeostasis , Thyroid Gland/metabolism
7.
Sci Rep ; 11(1): 4757, 2021 02 26.
Article En | MEDLINE | ID: mdl-33637828

Ethanolamine plasmalogens (EPls), unique alkenylacyl-glycerophospholipids, are the only known ligands of G-protein-coupled receptor 61-a novel receptor co-localised with gonadotropin-releasing hormone receptors on anterior pituitary gonadotrophs. Brain EPl decreases with age. Commercial EPl-extracted from the cattle brain (unidentified age)-can independently stimulate FSH secretion from gonadotrophs. We hypothesised that there exists an age-related difference in the quality, quantity, and ability of bovine brain EPls to stimulate bovine gonadotrophs. We compared the brains of young (about 26 month old heifers) and old (about 90 month old cows) Japanese Black bovines, including EPls obtained from both groups. Additionally, mRNA expressions of the EPl biosynthesis enzymes, glyceronephosphate O-acyltransferase, alkylglycerone phosphate synthase, and fatty acyl-CoA reductase 1 (FAR1) were evaluated in young and old hypothalami. The old-brain EPl did not stimulate FSH secretion from gonadotrophs, unlike the young-brain EPl. Molecular species of EPl were compared using two-dimensional liquid chromatography-mass spectrometry. We identified 20 EPl molecular species of which three and three exhibited lower (P < 0.05) and higher (P < 0.05) ratios, respectively, in old compared to young brains. In addition, quantitative reverse transcription-polymerase chain reaction detected higher FAR1 levels in the POA, but not in the ARC&ME tissues, of old cows than that of fertile young heifers. Therefore, old-brain EPl may be associated with age-related infertility.


Age Factors , Gonadotrophs/drug effects , Plasmalogens/metabolism , Plasmalogens/pharmacology , Animals , Brain/metabolism , Cattle , Female , Follicle Stimulating Hormone/metabolism , Gene Expression Regulation , Hypothalamus/chemistry , Hypothalamus/enzymology , Plasmalogens/chemistry
8.
Gen Comp Endocrinol ; 300: 113617, 2021 01 01.
Article En | MEDLINE | ID: mdl-32950578

The mitosis-associated protein aurora kinase A (AURKA) regulates the maturation of germ cells. We have previously reported using transcriptome analysis that AURKA is expressed in yak testes. Although Tibetan sheep possess an immense economic value, their reproductive rate is low. Herein, the expression and functions of AURKA in the hypothalamus-pituitary-testicular (HPT) axis in Tibetan sheep from Tianzhu were investigated. The cDNA sequence of sheep AURKA was cloned and bioinformatics techniques were used to predict its structure. Tissue expression of AURKA was determined by qPCR, immunoblotting, immunostaining, and immunohistochemistry. The AURKA coding sequence was found to be 1218 bp in length, encoding a 405-amino acid polypeptide chain. Furthermore, the highest sequence similarity of AURKA with the corresponding sequence in other species was seen in goat and cattle; the least degree of similarity was seen in the domestic cat. In addition, AURKA expression was elevated in the testes compared to that in the hypothalamus and pituitary (p < 0.01). Moreover, AURKA was mainly localized in the hypothalamic paraventricular nucleus (magnocellular), chromophobe cells of the pituitary, and spermatogenic cells of the testis. These results indicated that AURKA might participate in sheep reproductive regulation, thus providing a reference for the study of AURKA function in the reproductive process of Tibetan sheep from Tianzhu.


Aurora Kinase A/metabolism , Hypothalamus/enzymology , Pituitary Gland/enzymology , Sheep/metabolism , Testis/enzymology , Amino Acid Sequence , Animals , Aurora Kinase A/chemistry , Aurora Kinase A/genetics , Gene Expression Profiling , Gene Expression Regulation, Enzymologic , Male , Phylogeny , Tibet
9.
Gen Comp Endocrinol ; 299: 113604, 2020 12 01.
Article En | MEDLINE | ID: mdl-32866475

Many temperate zone animals engage in seasonal reproductive physiology and behavior as a strategy to maximise the propagation of the species. The hypothalamus integrates environmental cues and hormonal signalling to optimize the timing of reproduction. Recent work has revealed that epigenetic modifications, such as DNA methylation, vary across seasonal reproductive states. Multiple hormones act in the hypothalamus to permit or inhibit reproductive physiology, and the increase in thyroid hormone triiodothyronine (T3) has been implicated in the initiation of breeding in many species. The objective of this study was to examine the effect of T3 on the photoperiod-dependent regulation of reproductive physiology and hypothalamic DNA methyltransferase enzyme expression in female Siberian hamsters (Phodopus sungorus). We tested the hypothesis that T3 in short days (SD) would stimulate hypothalamic Rfrp3 and de novo DNA methyltransferase (Dnmt) expression in female Siberian hamsters. 10 weeks of SD lengths induced a decrease in body and uterine mass. Hamsters maintained in SD were found to express lower levels of GnRH, Rfrp3, Dnmt3a and Dnmt3b. Two weeks of daily T3 injections did not affect body mass, uterine mass, Gnrh, Rfrp3, Dnmt3a or Dnmt3b expression in neuroendocrine tissues. SD significantly lowered Tshß mRNA expression and T3 reduced Tshß in LD hamsters. Our data indicate sex-dependent effects of T3 for the neuroendocrine regulation of seasonal reproduction in hamsters.


Gene Expression Regulation, Enzymologic/drug effects , Hypothalamus/enzymology , Methyltransferases/metabolism , Phodopus/physiology , Photoperiod , Reproduction , Triiodothyronine/pharmacology , Animals , Female , Hypothalamus/drug effects , Male , Methyltransferases/genetics , Seasons , Sex Factors , Siberia
10.
J Clin Invest ; 130(11): 6093-6108, 2020 11 02.
Article En | MEDLINE | ID: mdl-32780722

Recent genome-wide association studies (GWAS) identified DUSP8, encoding a dual-specificity phosphatase targeting mitogen-activated protein kinases, as a type 2 diabetes (T2D) risk gene. Here, we reveal that Dusp8 is a gatekeeper in the hypothalamic control of glucose homeostasis in mice and humans. Male, but not female, Dusp8 loss-of-function mice, either with global or corticotropin-releasing hormone neuron-specific deletion, had impaired systemic glucose tolerance and insulin sensitivity when exposed to high-fat diet (HFD). Mechanistically, we found impaired hypothalamic-pituitary-adrenal axis feedback, blunted sympathetic responsiveness, and chronically elevated corticosterone levels driven by hypothalamic hyperactivation of Jnk signaling. Accordingly, global Jnk1 ablation, AAV-mediated Dusp8 overexpression in the mediobasal hypothalamus, or metyrapone-induced chemical adrenalectomy rescued the impaired glucose homeostasis of obese male Dusp8-KO mice, respectively. The sex-specific role of murine Dusp8 in governing hypothalamic Jnk signaling, insulin sensitivity, and systemic glucose tolerance was consistent with functional MRI data in human volunteers that revealed an association of the DUSP8 rs2334499 risk variant with hypothalamic insulin resistance in men. Further, expression of DUSP8 was increased in the infundibular nucleus of T2D humans. In summary, our findings suggest the GWAS-identified gene Dusp8 as a novel hypothalamic factor that plays a functional role in the etiology of T2D.


Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Type 2/enzymology , Dual-Specificity Phosphatases/metabolism , Hypothalamus/enzymology , Insulin Resistance , MAP Kinase Kinase 4/metabolism , Signal Transduction , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 2/genetics , Dual-Specificity Phosphatases/genetics , MAP Kinase Kinase 4/genetics , Mice , Mice, Knockout
11.
Int J Neuropsychopharmacol ; 23(8): 481-490, 2020 11 26.
Article En | MEDLINE | ID: mdl-32710540

BACKGROUND: Accompanied with profound efficacy, atypical antipsychotics (AAPs) contribute to metabolic adverse effects with few effective strategies to attenuate. Serotonin 5-HT2C receptor (HTR2C) plays a critical role in hyperphagia and weight gain induced by AAPs, and expression of phosphatase tensin homolog (PTEN) in the hypothalamus also affects feeding behavior and weight change. Moreover, PTEN has a physical crosstalk between PTEN and a region in the third intracellular loop (3L4F) of the HTR2C. Tat-3L4F has the property to disrupt crosstalk between PTEN and HTR2C. This is the first study to our knowledge to investigate the effect of Tat-3L4F on olanzapine-induced metabolic abnormalities and PTEN/ phosphatidylinositol 3-kinase/protein kinase B expression in the hypothalamus in rats. METHODS: The effects of Tat-3L4F were investigated through measuring body weight, food intake, and blood glucose. In addition, PTEN/phosphatidylinositol 3-kinase/protein kinase B level in the hypothalamus was detected by immunofluorescence assay and western blot. Metabolites in the liver tissue were detected by liquid chromatography-mass spectrometry and analyzed by multivariate analyses and pairwise comparison. RESULTS: Our results showed that hyperphagia and weight gain were evident in the olanzapine alone-fed rats but was attenuated after Tat-3L4F treatment. In addition, oral glucose tolerance test indicated blood glucose at 120 minutes was higher in the olanzapine alone-treated group than in groups treated with vehicle and olanzapine + Tat-3L4F (10 µmol kg-1 per day). Furthermore, compared with olanzapine alone treatment, treatment with Tat-3L4F (10 µmol kg-1 per day) significantly inhibited PTEN expression in the hypothalamus. The olanzapine alone-treated group had the highest bile acid level, followed by the olanzapine with Tat-3L4F (1 µmol kg-1) group, olanzapine with Tat-3L4F (10 µmol kg-1) group, and vehicle group. CONCLUSIONS: Our present results reveal that Tat-3L4F is a potential pharmacological strategy for suppressing hyperphagia and weight gain induced by olanzapine, which acts through disrupting crosstalk between HTR2C and PTEN as a result of PTEN downregulation in the hypothalamus.


Antipsychotic Agents/toxicity , Appetite Depressants/pharmacology , Feeding Behavior/drug effects , Hypothalamus/drug effects , Olanzapine/toxicity , PTEN Phosphohydrolase/metabolism , Receptor, Serotonin, 5-HT2C/drug effects , Recombinant Fusion Proteins/pharmacology , Weight Gain/drug effects , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Female , Hypothalamus/enzymology , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats, Sprague-Dawley , Receptor, Serotonin, 5-HT2C/metabolism , Signal Transduction
12.
J Ethnopharmacol ; 261: 113061, 2020 Oct 28.
Article En | MEDLINE | ID: mdl-32525065

ETHNOPHARMACOLOGICAL RELEVANCE: TG-decoction (Tiao Geng decoction) is the extract of a Chinese herb mixture that has been used for treating menopausal symptoms for over 30 years. We have previously reported anti-aging and anti-oxidative effects of the TG-decoction on hypothalamic neurons in ovariectomized (OVX) rats. AIM OF THE STUDY: The present study further investigates the effects of TG-decoction on the prevention of aging-related ultrastructural changes in menopausal hypothalamic neurons and the likely molecular mechanism. MATERIALS AND METHODS: A total of 120 four-month-old female SPF Sprague Dawley rats were divided into six groups. Five groups were ovariectomized (OVX) and one group served as a sham control. Three OVX groups received TG-decoction at three different doses. The remaining two OVX groups served as positive and negative controls by receiving estradiol valerate and saline solution. The sham group received saline. After one month, aging-related ultrastructural alterations in hypothalamic neurons were evaluated using transmission electron microscopy. Nissl staining was used to assess the pathomorphological changes of the hypothalamic neurons. Cell apoptosis was evaluated by TUNEL. Expression of Bcl-2 family genes was studied using qRT-PCR. Expression of the apoptosis-related proteins ASK1, MKK7, JNK, c-Jun, Bax, Casp3 and Bcl-2 was studied using western blotting. RESULTS: Ovariectomy of female rats led to visible damage and aging-like alterations in the mitochondria and endoplasmic reticulum as well as large deposits of lipofuscin in hypothalamic tissue. TG-decoction treatment prevented this visible damage and lipofuscin deposition, increased the number of nerve cells and normally-shaped Nissl bodies, and reduced the number of TUNEL-positive cells. Expression of Bcl-2 gene was increased, while Bax gene reduced. Expression of the proteins ASK1, MKK7, JNK, c-Jun, Bax and Casp3 was reduced, while that of Bcl-2 was increased. CONCLUSION: TG-decoction reduces aging-related ultrastructural changes in hypothalamic neurons, likely by suppressing ASK1/MKK7/JNK-mediated apoptosis in neuronal mitochondria or nuclei.


Apoptosis/drug effects , Drugs, Chinese Herbal/pharmacology , Hypothalamus/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Menopause/drug effects , Mitogen-Activated Protein Kinases/metabolism , Neurons/drug effects , Age Factors , Animals , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/enzymology , Endoplasmic Reticulum/ultrastructure , Female , Hypothalamus/enzymology , Hypothalamus/pathology , Menopause/metabolism , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondria/ultrastructure , Neurons/enzymology , Neurons/ultrastructure , Ovariectomy , Rats, Sprague-Dawley , Signal Transduction , Syndrome
13.
J Neuroendocrinol ; 32(3): e12836, 2020 03.
Article En | MEDLINE | ID: mdl-32062869

The present study aimed to determine whether an i.c.v. administration of allopregnanolone (ALLO) rapidly modifies the hypothalamic and ovarian 3ß-hydroxysteroid dehydrogenase (3ß-HSD) enzymatic activity and gene expression in in vivo and ex vivo systems in pro-oestrus (PE) and dioestrus I (DI) rats. Animals were injected with vehicle, ALLO, bicuculline or bicuculline plus ALLO and were then killed. In the in vivo experiment, the hypothalamus, ovaries and serum were extracted and analysed. In the ex vivo experiment, the superior mesenteric ganglion - ovarian nerve plexus - ovary system was extracted and incubated during 120 minutes at 37 ºC. The serum and ovarian compartment fluids were used to determine progesterone by radioimmunoanalysis. In the in vivo experiments, ALLO caused a decrease in hypothalamic and ovarian 3ß-HSD enzymatic activity during PE. During DI, ALLO increased hypothalamic and ovarian 3ß-HSD activity and gene expression. The ovarian 3ß-HSD activity increased in both stages in the ex vivo system; gene expression increased only during DI. ALLO induced an increase in serum progesterone only in D1 and in the ovarian incubation liquids in both stages. All findings were reversed by an injection of bicuculline before ALLO. Ovarian steroidogenic changes could be attributed to signals coming from ganglion neurones, which are affected by the acute central neurosteroid stimulation. The i.c.v. administration of ALLO via the GABAergic system altered 3ß-HSD activity and gene expression, modulating the neuroendocrine axis. The present study reveals the action that ALLO exerts on the GABAA receptor in both the central and peripheral nervous system and its relationship with hormonal variations. ALLO is involved in the "fine tuning" of neurosecretory functions as a potent modulator of reproductive processes in female rats.


3-Hydroxysteroid Dehydrogenases/metabolism , Hypothalamus/drug effects , Neurosteroids/administration & dosage , Ovary/drug effects , Pregnanolone/administration & dosage , Animals , Diestrus/drug effects , Diestrus/metabolism , Female , Gene Expression/drug effects , Hypothalamus/enzymology , Injections, Intraventricular , Ovary/metabolism , Proestrus/drug effects , Proestrus/metabolism , Progesterone/blood , Rats
14.
Front Endocrinol (Lausanne) ; 11: 622581, 2020.
Article En | MEDLINE | ID: mdl-33633690

Rho-kinase 1 (ROCK1) has been implicated in diverse metabolic functions throughout the body, with promising evidence identifying ROCK1 as a therapeutic target in diabetes and obesity. Considering these metabolic roles, several pharmacological inhibitors have been developed to elucidate the mechanisms underlying ROCK1 function. Y27632 and fasudil are two common ROCK1 inhibitors; however, they have varying non-specific selectivity to inhibit other AGC kinase subfamily members and whole-body pharmacological approaches lack tissue-specific insight. As a result, interpretation of studies with these inhibitors is difficult, and alternative approaches are needed to elucidate ROCK1's tissue specific metabolic functions. Fortunately, recent technological advances utilizing molecular carriers or genetic manipulation have facilitated discovery of ROCK1's tissue-specific mechanisms of action. In this article, we review the tissue-specific roles of ROCK1 in the regulation of energy balance and substrate utilization. We highlight prominent metabolic roles in liver, adipose, and skeletal muscle, in which ROCK1 regulates energy expenditure, glucose uptake, and lipid metabolism via inhibition of AMPK2α and paradoxical modulation of insulin signaling. Compared to ROCK1's roles in peripheral tissues, we also describe contradictory functions of ROCK1 in the hypothalamus to increase energy expenditure and decrease food intake via leptin signaling. Furthermore, dysregulated ROCK1 activity in either of these tissues results in metabolic disease phenotypes. Overall, tissue-specific approaches have made great strides in deciphering the many critical metabolic functions of ROCK1 and, ultimately, may facilitate the development of novel treatments for metabolic disorders.


Adipose Tissue/enzymology , Hypothalamus/enzymology , Liver/enzymology , Metabolic Diseases/enzymology , Muscle, Skeletal/enzymology , rho-Associated Kinases/metabolism , Adipose Tissue/pathology , Animals , Energy Metabolism/physiology , Humans , Hypothalamus/pathology , Insulin Resistance/physiology , Lipid Metabolism/physiology , Liver/pathology , Metabolic Diseases/pathology , Muscle, Skeletal/pathology , Obesity/enzymology , Obesity/pathology
15.
Am J Physiol Regul Integr Comp Physiol ; 318(1): R122-R134, 2020 01 01.
Article En | MEDLINE | ID: mdl-31692367

Hypothalamic AMPK plays a major role in the regulation of whole body metabolism and energy balance. Present evidence has demonstrated that this canonical mechanism is evolutionarily conserved. Thus, recent data demonstrated that inhibition of AMPKα2 in fish hypothalamus led to decreased food intake and liver capacity to use and synthesize glucose, lipids, and amino acids. We hypothesize that a signal of abundance of nutrients from the hypothalamus controls hepatic metabolism. The vagus nerve is the most important link between the brain and the liver. We therefore examined in the present study whether surgical transection of the vagus nerve in rainbow trout is sufficient to alter the effect in liver of central inhibition of AMPKα2. Thus, we vagotomized (VGX) or not (Sham) rainbow trout and then intracerebroventricularly administered adenoviral vectors tagged with green fluorescent protein alone or linked to a dominant negative isoform of AMPKα2. The inhibition of AMPKα2 led to reduced food intake in parallel with changes in the mRNA abundance of hypothalamic neuropeptides [neuropeptide Y (npy), agouti-related protein 1 (agrp1), and cocaine- and amphetamine-related transcript (cartpt)] involved in food intake regulation. Central inhibition of AMPKα2 resulted in the liver having decreased capacity to use and synthesize glucose, lipids, and amino acids. Notably, these effects mostly disappeared in VGX fish. These results support the idea that autonomic nervous system actions mediate the actions of hypothalamic AMPKα2 on liver metabolism. Importantly, this evidence indicates that the well-established role of hypothalamic AMPK in energy balance is a canonical evolutionarily preserved mechanism that is also present in the fish lineage.


AMP-Activated Protein Kinases/metabolism , Energy Metabolism/physiology , Hypothalamus/enzymology , Liver/metabolism , Oncorhynchus mykiss/physiology , Vagus Nerve/physiology , AMP-Activated Protein Kinases/genetics , Adenoviridae , Animals , Feeding Behavior/physiology , Gene Expression Regulation, Enzymologic/physiology , Liver/innervation , Vagotomy
16.
Aging (Albany NY) ; 11(21): 9500-9529, 2019 11 10.
Article En | MEDLINE | ID: mdl-31708494

Researchers have long assumed that systematic estrogen fading might contribute to the sustained progression of menopausal degenerate syndromes, although definitive evidence has not been presented. Whether such findings represent a causal contribution or are the result of opportunistic messengers sent from the reproductive system to the brain is also a vital question. We constructed a multiscale network of the ovariectomy (OVX) induced estrogen receptors depletion (ER-depletion) model and integrated targeted proteomic, targeted lipidomic, cytochemical, and histopathological data across three tissues from the ovariectomy rodent model. We found that compared to control rats, OVX rats showed increased renal and uterine prostaglandin D2 synthase (Ptgds) expression and decreased hypothalamic Ptgds expression, abnormal Ptgds metabolites, the degenerate renal function profiles and decreased cognitive ability (learning and memory) in Morris water maze test. Importantly, we observed a regulatory relationship among ER (particularly ERß), the degree of the pathological phenotype, learning behavior test and the 'hypothalamus-uterus-kidney (HUK) axis functions. Collectively, this study elucidates that ER depletion promoted HUK aging is mostly attributed to a renal ERß/Ptgds signalling imbalance.


Intramolecular Oxidoreductases/metabolism , Kidney/metabolism , Lipid Metabolism/genetics , Lipocalins/metabolism , Menopause/metabolism , Receptors, Estrogen/deficiency , Animals , Eicosanoids/blood , Eicosanoids/urine , Female , Hypothalamus/enzymology , Maze Learning , Menopause/genetics , Ovariectomy , Proteome , Rats, Sprague-Dawley , Signal Transduction , Uterus/enzymology
17.
Neurochem Int ; 131: 104563, 2019 12.
Article En | MEDLINE | ID: mdl-31589911

Peripheral lipopolysaccharide (LPS) injection induces systemic inflammation through the activation of the inhibitor of nuclear factor kappa B (NF-κB) kinase (IKK)/NF-κB signaling pathway, which promotes brain dysfunction resulting in conditions including anorexia. LPS-mediated reduction of food intake is associated with activation of NF-κB signaling and phosphorylation of the transcription factor signal transducer and activator of transcription 3 (STAT3) in the hypothalamus. We recently reported phospholipase C-related catalytically inactive protein (PRIP) as a new negative regulator of phosphatidylinositol 3-kinase/AKT signaling. AKT regulates the IKK/NF-κB signaling pathway; therefore, this study aimed to investigate the role of PRIP/AKT signaling in LPS-mediated neuroinflammation-induced anorexia. PRIP gene (Prip1 and Prip2) knockout (Prip-KO) mice intraperitoneally (ip) administered with LPS exhibited increased anorexia responses compared with wild-type (WT) controls. Although few differences were observed between WT and Prip-KO mice in LPS-elicited plasma pro-inflammatory cytokine elevation, hypothalamic pro-inflammatory cytokines were significantly upregulated in Prip-KO rather than WT mice. Hypothalamic AKT and IKK phosphorylation and IκB degradation were significantly increased in Prip-KO rather than WT mice, indicating further promotion of AKT-mediated NF-κB signaling. Consistently, hypothalamic STAT3 was further phosphorylated in Prip-KO rather than WT mice. Furthermore, suppressor of cytokine signaling 3 (Socs3), a negative feedback regulator for STAT3 signaling, and cyclooxogenase-2 (Cox2), a candidate molecule in LPS-induced anorexigenic responses, were upregulated in the hypothalamus in Prip-KO rather than WT mice. Pro-inflammatory cytokines were upregulated in hypothalamic microglia isolated from Prip-KO rather than WT mice. Together, these findings indicate that PRIP negatively regulates LPS-induced anorexia caused by pro-inflammatory cytokine expression in the hypothalamus, which is mediated by AKT-activated NF-κB signaling. Importantly, hypothalamic microglia participate in this PRIP-mediated process. Elucidation of PRIP-mediated neuroinflammatory responses may provide novel insights into the pathophysiology of many brain dysfunctions.


Anorexia/enzymology , Encephalitis/enzymology , Hypothalamus/enzymology , Intracellular Signaling Peptides and Proteins/metabolism , Animals , Anorexia/chemically induced , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Eating , Encephalitis/chemically induced , Intracellular Signaling Peptides and Proteins/genetics , Lipopolysaccharides , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , NF-kappa B/metabolism , Oncogene Protein v-akt/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/genetics
18.
Physiol Behav ; 209: 112617, 2019 10 01.
Article En | MEDLINE | ID: mdl-31319109

To assess the hypothesis that Na+/K+-ATPase (NKA) is involved in the central regulation of food intake in fish, we observed in a first experiment with rainbow trout (Oncorhynchus mykiss) that intracerebroventricular (ICV) treatment with ouabain decreased food intake. We hypothesized that this effect relates to modulation of glucosensing mechanisms in brain areas (hypothalamus, hindbrain, and telencephalon) involved in food intake control. Therefore, we evaluated in a second experiment, the effect of ICV administration of ouabain, in the absence or in the presence of glucose, on NKA activity, mRNA abundance of different NKA subunits, parameters related to glucosensing, transcription factors, and appetite-related neuropeptides in brain areas involved in the control of food intake. NKA activity and mRNA abundance of nkaα1a and nkaα1c in brain were inhibited by ouabain treatment and partially by glucose. The anorectic effect of ouabain is opposed to the orexigenic effect reported in mammals. The difference might relate to the activity of glucosensing as well as downstream mechanisms involved in food intake regulation. Ouabain inhibited glucosensing mechanisms, which were activated by glucose in hypothalamus and telencephalon. Transcription factors and neuropeptides displayed responses comparable to those elicited by glucose when ouabain was administered alone, but not when glucose and ouabain were administered simultaneously. Ouabain might therefore affect other processes, besides glucosensing mechanisms, generating changes in membrane potential and/or intracellular pathways finally modulating transcription factors and neuropeptide mRNA abundance leading to modified food intake.


Brain Chemistry/physiology , Eating/physiology , Glucose/metabolism , Oncorhynchus mykiss/physiology , Sodium-Potassium-Exchanging ATPase/metabolism , Animals , Brain/drug effects , Brain/enzymology , Brain Chemistry/drug effects , Eating/drug effects , Enzyme Inhibitors/pharmacology , Hypothalamus/drug effects , Hypothalamus/enzymology , Hypothalamus/metabolism , Infusions, Intraventricular , Neuropeptides/metabolism , Ouabain/pharmacology , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Telencephalon/drug effects , Telencephalon/enzymology , Telencephalon/metabolism
19.
Thyroid ; 29(9): 1336-1343, 2019 09.
Article En | MEDLINE | ID: mdl-31303139

Background: Inflammation is associated with marked changes in cellular thyroid hormone (TH) metabolism in triiodothyronine (T3) target organs. In the hypothalamus, type 2 deiodinase (D2), the main T3 producing enzyme, increases upon inflammation, leading to an increase in local T3 availability, which in turn decreases thyrotropin releasing hormone expression in the paraventricular nucleus. Type 3 deiodinase (D3), the T3 inactivating enzyme, decreases during inflammation, which might also contribute to the increased T3 availability in the hypothalamus. While it is known that D2 is regulated by nuclear factor κB (NF-κB) during inflammation, the underlying mechanisms of D3 regulation are unknown. Therefore, the aim of the present study was to investigate inflammation-induced D3 regulation using in vivo and in vitro models. Methods: Mice were injected with a sublethal dose of bacterial endotoxin (lipopolysaccharide [LPS]) to induce a systemic acute-phase response. A human neuroblastoma (SK-N-AS) cell line was used to test the involvement of the thyroid hormone receptor alpha 1 (TRα1) as well as the activator protein-1 (AP-1) and NF-κB inflammatory pathways in the inflammation-induced decrease of D3. Results: D3 expression in the hypothalamus was decreased 24 hours after LPS injection in mice. This decrease was similar in mice lacking the TRα. Incubation of SK-N-AS cells with LPS robustly decreased both D3 mRNA expression and activity. This led to increased intracellular T3 concentrations. The D3 decrease was prevented when NF-κB or AP-1 was inhibited. TRα1 mRNA expression decreased in SK-N-AS cells incubated with LPS, but knockdown of the TRα in SK-N-AS cells did not prevent the LPS-induced D3 decrease. Conclusions: We conclude that the inflammation-induced D3 decrease in the hypothalamus is mediated by the inflammatory pathways NF-κB and AP-1, but not TRα1. Furthermore, the observed decrease modulates intracellular T3 concentrations. Our results suggest a concerted action of inflammatory modulators to regulate both hypothalamic D2 and D3 activities to increase the local TH concentrations.


Hypothalamus/enzymology , Inflammation/metabolism , Iodide Peroxidase/genetics , Animals , Cell Line, Tumor , Down-Regulation , Female , Humans , Iodide Peroxidase/physiology , Lipopolysaccharides , Male , Mice , NF-kappa B/physiology , RNA, Messenger/analysis , Signal Transduction , Thyroid Hormone Receptors alpha/physiology , Transcription Factor AP-1/physiology , Iodothyronine Deiodinase Type II
20.
Nutrients ; 11(7)2019 Jul 23.
Article En | MEDLINE | ID: mdl-31340540

Selenium, an essential trace element known mainly for its antioxidant properties, is critical for proper brain function and regulation of energy metabolism. Whole-body knockout of the selenium recycling enzyme, selenocysteine lyase (Scly), increases susceptibility to metabolic syndrome and diet-induced obesity in mice. Scly knockout mice also have decreased selenoprotein expression levels in the hypothalamus, a key regulator of energy homeostasis. This study investigated the role of selenium in whole-body metabolism regulation using a mouse model with hypothalamic knockout of Scly. Agouti-related peptide (Agrp) promoter-driven Scly knockout resulted in reduced weight gain and adiposity while on a high-fat diet (HFD). Scly-Agrp knockout mice had reduced Agrp expression in the hypothalamus, as measured by Western blot and immunohistochemistry (IHC). IHC also revealed that while control mice developed HFD-induced leptin resistance in the arcuate nucleus, Scly-Agrp knockout mice maintained leptin sensitivity. Brown adipose tissue from Scly-Agrp knockout mice had reduced lipid deposition and increased expression of the thermogenic marker uncoupled protein-1. This study sheds light on the important role of selenium utilization in energy homeostasis, provides new information on the interplay between the central nervous system and whole-body metabolism, and may help identify key targets of interest for therapeutic treatment of metabolic disorders.


Agouti-Related Protein/metabolism , Diet, High-Fat , Hypothalamus/enzymology , Leptin/metabolism , Lyases/deficiency , Neurons/metabolism , Obesity/prevention & control , Adipose Tissue, Brown/enzymology , Adipose Tissue, Brown/physiopathology , Adiposity , Animals , Disease Models, Animal , Female , Gene Knockout Techniques , Hypothalamus/physiopathology , Lyases/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Obesity/enzymology , Obesity/genetics , Obesity/physiopathology , Signal Transduction , Uncoupling Protein 1/metabolism , Weight Gain
...