Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49.796
Filter
1.
J Agric Food Chem ; 72(28): 15765-15777, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-38970495

ABSTRACT

Konjac glucomannan (KGM), high-viscosity dietary fiber, is utilized in weight management. Previous investigations on the appetite-suppressing effects of KGM have centered on intestinal responses to nutrients and gastric emptying rates, with less focus on downstream hypothalamic neurons of satiety hormones. In our studies, the molecular mechanisms through which KGM and its degradation products influence energy homeostasis via the adipocyte-hypothalamic axis have been examined. It was found that high-viscosity KGM more effectively stimulates enteroendocrine cells to release glucagon-like peptide-1 (GLP-1) and reduces ghrelin production, thereby activating hypothalamic neurons and moderating short-term satiety. Conversely, low-viscosity DKGM has been shown to exhibit stronger anti-inflammatory properties in the hypothalamus, enhancing hormone sensitivity and lowering the satiety threshold. Notably, both KGM and DKGM significantly reduced leptin signaling and fatty acid signaling in adipose tissue and activated brown adipose tissue thermogenesis to suppress pro-opiomelanocortin (POMC) expression and activate agouti-related protein (AgRP) expression, thereby reducing food intake and increasing energy expenditure. Additionally, high-viscosity KGM has been found to activate the adipocyte-hypothalamus axis more effectively than DKGM, thereby promoting greater daily energy expenditure. These findings provide novel insights into the adipocyte-hypothalamic axis for KGM to suppress appetite and reduce weight.


Subject(s)
Adipocytes , Appetite Regulation , Diet, High-Fat , Energy Metabolism , Hypothalamus , Mice, Inbred C57BL , Animals , Mice , Energy Metabolism/drug effects , Hypothalamus/metabolism , Hypothalamus/drug effects , Diet, High-Fat/adverse effects , Male , Appetite Regulation/drug effects , Adipocytes/metabolism , Adipocytes/drug effects , Humans , Glucagon-Like Peptide 1/metabolism , Ghrelin/metabolism , Leptin/metabolism , Agouti-Related Protein/metabolism , Agouti-Related Protein/genetics , Thermogenesis/drug effects , Pro-Opiomelanocortin/metabolism , Pro-Opiomelanocortin/genetics , Obesity/metabolism , Obesity/physiopathology , Obesity/diet therapy , Mannans
2.
J Clin Invest ; 134(14)2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39007271

ABSTRACT

The melanocortin-3 receptor (MC3R) regulates GABA release from agouti-related protein (AgRP) nerve terminals and thus tonically suppresses multiple circuits involved in feeding behavior and energy homeostasis. Here, we examined the role of the MC3R and the melanocortin system in regulating the response to various anorexigenic agents. The genetic deletion or pharmacological inhibition of the MC3R, or subthreshold doses of an MC4R agonist, improved the dose responsiveness to glucagon-like peptide 1 (GLP1) agonists, as assayed by inhibition of food intake and weight loss. An enhanced anorectic response to the acute satiety factors peptide YY (PYY3-36) and cholecystokinin (CCK) and the long-term adipostatic factor leptin demonstrated that increased sensitivity to anorectic agents was a generalized result of MC3R antagonism. We observed enhanced neuronal activation in multiple hypothalamic nuclei using Fos IHC following low-dose liraglutide in MC3R-KO mice (Mc3r-/-), supporting the hypothesis that the MC3R is a negative regulator of circuits that control multiple aspects of feeding behavior. The enhanced anorectic response in Mc3r-/- mice after administration of GLP1 analogs was also independent of the incretin effects and malaise induced by GLP1 receptor (GLP1R) analogs, suggesting that MC3R antagonists or MC4R agonists may have value in enhancing the dose-response range of obesity therapeutics.


Subject(s)
Liraglutide , Mice, Knockout , Receptor, Melanocortin, Type 3 , Receptor, Melanocortin, Type 4 , Animals , Mice , Receptor, Melanocortin, Type 4/metabolism , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/agonists , Liraglutide/pharmacology , Receptor, Melanocortin, Type 3/genetics , Receptor, Melanocortin, Type 3/metabolism , Receptor, Melanocortin, Type 3/agonists , Male , Appetite Depressants/pharmacology , Glucagon-Like Peptide 1/metabolism , Cholecystokinin/metabolism , Mice, Inbred C57BL , Eating/drug effects , Leptin/metabolism , Peptide YY/metabolism , Peptide YY/genetics , Hypothalamus/metabolism
3.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article in English | MEDLINE | ID: mdl-39000495

ABSTRACT

Patients with asthma experience elevated rates of mental illness. However, the molecular links underlying such lung-brain crosstalk remain ambiguous. Hypothalamic dysfunction is observed in many psychiatric disorders, particularly those with an inflammatory component due to many hypothalamic regions being unprotected by the blood-brain barrier. To gain a better insight into such neuropsychiatric sequelae, this study investigated gene expression differences in the hypothalamus following lung inflammation (asthma) induction in mice, using RNA transcriptome profiling. BALB/c mice were challenged with either bacterial lipopolysaccharide (LPS, E. coli) or ovalbumin (OVA) allergens or saline control (n = 7 per group), and lung inflammation was confirmed via histological examination of postmortem lung tissue. The majority of the hypothalamus was micro-dissected, and total RNA was extracted for sequencing. Differential expression analysis identified 31 statistically significant single genes (false discovery rate FDR5%) altered in expression following LPS exposure compared to controls; however, none were significantly changed following OVA treatment, suggesting a milder hypothalamic response. When gene sets were examined, 48 were upregulated and 8 were downregulated in both asthma groups relative to controls. REACTOME enrichment analysis suggests these gene sets are involved in signal transduction metabolism, immune response and neuroplasticity. Interestingly, we identified five altered gene sets directly associated with neurotransmitter signaling. Intriguingly, many of these altered gene sets can influence mental health and or/neuroinflammation in humans. These findings help characterize the links between asthma-induced lung inflammation and the brain and may assist in identifying relevant pathways and therapeutic targets for future intervention.


Subject(s)
Asthma , Disease Models, Animal , Hypothalamus , Lipopolysaccharides , Lung , Mice, Inbred BALB C , Transcriptome , Animals , Asthma/genetics , Asthma/metabolism , Asthma/pathology , Hypothalamus/metabolism , Mice , Lung/metabolism , Lung/pathology , Ovalbumin , Gene Expression Profiling , Female , Gene Expression Regulation
4.
Int J Mol Sci ; 25(13)2024 Jul 07.
Article in English | MEDLINE | ID: mdl-39000563

ABSTRACT

Circadian rhythms regulate physiological processes in approximately 24 h cycles, and their disruption is associated with various diseases. Inflammation may perturb circadian rhythms, though these interactions remain unclear. This study examined whether systemic inflammation induced by an intraperitoneal injection of lipopolysaccharide (LPS) could alter central and peripheral circadian rhythms and diurnal neuroimmune dynamics. Mice were randomly assigned to two groups: the saline control group and the LPS group. The diurnal expression of circadian clock genes and inflammatory cytokines were measured in the hypothalamus, hippocampus, and liver. Diurnal dynamic behaviors of microglia were also assessed. Our results revealed that the LPS perturbed circadian gene oscillations in the hypothalamus, hippocampus, and liver. Furthermore, systemic inflammation induced by the LPS could trigger neuroinflammation and perturb the diurnal dynamic behavior of microglia in the hippocampus. These findings shed light on the intricate link between inflammation and circadian disruption, underscoring their significance in relation to neurodegenerative diseases.


Subject(s)
Circadian Rhythm , Inflammation , Lipopolysaccharides , Animals , Mice , Male , Microglia/metabolism , Microglia/immunology , Hypothalamus/metabolism , Hypothalamus/immunology , Hippocampus/metabolism , Cytokines/metabolism , Liver/metabolism , Liver/pathology , Liver/immunology , Mice, Inbred C57BL , Circadian Clocks/genetics , Neuroimmunomodulation
5.
Int J Mol Sci ; 25(13)2024 Jun 23.
Article in English | MEDLINE | ID: mdl-39000000

ABSTRACT

Somatostatin (SS) plays crucial regulatory roles in animal growth and reproduction by affecting the synthesis and secretion of growth hormone (GH). However, the mechanism by which SS regulates growth and development in goats is still unclear. In order to investigate the regulatory networks of the hypothalamus and pituitary in goats affected by SS DNA vaccines, in this study, we used a previously established oral attenuated Salmonella typhimurium SS DNA vaccine, X9241 (ptCS/2SS-asd), to treat wethers. We analyzed the protein changes in hypothalamic and pituitary tissues using a TMT-based proteomics approach. Additionally, we examined the metabolic profiles of the serum of control and immunized wethers through untargeted metabolomics using liquid chromatography-mass spectrometry (LC-MS). Key signaling pathways were identified based on differentially expressed metabolites (DEMs) and differentially expressed proteins (DEPs). Furthermore, the effect of critical DEPs on signaling pathways was confirmed through Western blotting (WB) experiments, which elucidated the mechanism of active SS immunization in wethers. A proteomics analysis revealed that the expression of 58 proteins in the hypothalamus and 124 in the pituitary gland was significantly altered following SS vaccine treatment (fold change > 1.2 or < 0.83, p < 0.05). In the hypothalamus, many DEPs were associated with gene ontology (GO) terms related to neuronal signaling. In contrast, most DEPs were associated with metabolic pathways. In the pituitary gland, the DEPs were largely related to immune and nutrient metabolism functions, with significant enrichment in KEGG pathways, particularly those involving the metabolic pathway, sphingolipid signaling, and the cGMP-PKG signaling pathway. A metabolomic analysis further showed that active SS immunization in wethers led to significant alterations in seven serum metabolites. Notably, the sphingolipid signaling pathway, secondary bile acid synthesis, sphingolipid metabolism, and lysine synthesis were significantly disrupted. SS vaccines induced marked changes in hypothalamic-pituitary proteins in wethers, facilitating alterations in their growth processes. This study not only provides insights into the mechanism of the SS gene in regulating GH secretion in wethers but also establishes a basis for hormone immunoregulation technology to enhance livestock production performance.


Subject(s)
Goats , Hypothalamus , Pituitary Gland , Proteomics , Somatostatin , Vaccines, DNA , Animals , Somatostatin/metabolism , Proteomics/methods , Hypothalamus/metabolism , Vaccines, DNA/immunology , Pituitary Gland/metabolism , Metabolomics/methods , Signal Transduction , Metabolome
6.
Int J Mol Sci ; 25(13)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-39000096

ABSTRACT

The arginine vasopressin (AVP)-magnocellular neurosecretory system (AVPMNS) in the hypothalamus plays a critical role in homeostatic regulation as well as in allostatic motivational behaviors. However, it remains unclear whether adult neurogenesis exists in the AVPMNS. By using immunoreaction against AVP, neurophysin II, glial fibrillar acidic protein (GFAP), cell division marker (Ki67), migrating neuroblast markers (doublecortin, DCX), microglial marker (Ionized calcium binding adaptor molecule 1, Iba1), and 5'-bromo-2'-deoxyuridine (BrdU), we report morphological evidence that low-rate neurogenesis and migration occur in adult AVPMNS in the rat hypothalamus. Tangential AVP/GFAP migration routes and AVP/DCX neuronal chains as well as ascending AVP axonal scaffolds were observed. Chronic water deprivation significantly increased the BrdU+ nuclei within both the supraaoptic (SON) and paraventricular (PVN) nuclei. These findings raise new questions about AVPMNS's potential hormonal role for brain physiological adaptation across the lifespan, with possible involvement in coping with homeostatic adversities.


Subject(s)
Cell Movement , Doublecortin Protein , Neurogenesis , Neurons , Animals , Rats , Neurons/metabolism , Neurons/cytology , Male , Paraventricular Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Hypothalamus/metabolism , Hypothalamus/cytology , Arginine Vasopressin/metabolism
7.
Sci Rep ; 14(1): 15506, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38969725

ABSTRACT

Relatively low levels of antioxidant enzymes coupled with high oxygen metabolism result in the formation of numerous oxidative DNA damages in the tissues of the central nervous system. Recently, kynurenic acid (KYNA), knowns for its neuroprotective properties, has gained increasing attention in this context. Therefore, our hypothesis assumed that increased KYNA levels in the brain would positively influence mRNA expression of selected enzymes of the base excision repair pathway as well as enhance their efficiency in excising damaged nucleobases in specific areas of the sheep brain. The study was conducted on adult anestrous sheep (n = 18), in which two different doses of KYNA (20 and 100 µg/day) were infused into the third brain ventricle for three days. Molecular and biochemical analysis included the hypothalamus (preoptic and mediol-basal areas), hippocampus (CA3 field) and amygdala (central amygdaloid nucleus), dissected from the brain of sheep euthanized immediately after the last infusion. The results revealed a significant increase P < 0.001) in the relative mRNA abundance of N-methylpurine DNA glycosylase (MPG) following administration of both dose of KYNA across all examined tissues. The transcription of thymine-DNA glycosylase (TDG) increased significantly (P < 0.001) in all tissues in response to the lower KYNA dose compared to the control group. Moreover, 8-oxoguanine (8-oxoG) DNA glycosylase (OGG1) mRNA levels were also higher in both animal groups (P < 0.001). In addition, in the hypothalamus, hippocampus and amygdala, AP endonuclease 1 (APE1) mRNA expression increased under both doses of KYNA. Moreover, the both dose of KYNA significantly stimulated the efficiency of 8-oxoG excision in hypothalamus and amygdala (P < 0.05-0.001). The lower and higher doses of KYNA significantly influenced the effectiveness of εA and εC in all structures (P < 0.01-0.001). In conclusion, the favorable effect of KYNA in the brain may include the protection of genetic material in nerve and glial cells by stimulating the expression and efficiency of BER pathway enzymes.


Subject(s)
Brain , DNA Glycosylases , DNA Repair , Kynurenic Acid , Animals , DNA Repair/drug effects , Sheep , Kynurenic Acid/metabolism , DNA Glycosylases/metabolism , DNA Glycosylases/genetics , Brain/metabolism , Brain/drug effects , Hypothalamus/metabolism , Hypothalamus/drug effects , RNA, Messenger/metabolism , RNA, Messenger/genetics , DNA Damage/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Female , Hippocampus/metabolism , Hippocampus/drug effects , Excision Repair
8.
Nat Commun ; 15(1): 5803, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987241

ABSTRACT

Mammalian hibernators survive prolonged periods of cold and resource scarcity by temporarily modulating normal physiological functions, but the mechanisms underlying these adaptations are poorly understood. The hibernation cycle of thirteen-lined ground squirrels (Ictidomys tridecemlineatus) lasts for 5-7 months and comprises weeks of hypometabolic, hypothermic torpor interspersed with 24-48-h periods of an active-like interbout arousal (IBA) state. We show that ground squirrels, who endure the entire hibernation season without food, have negligible hunger during IBAs. These squirrels exhibit reversible inhibition of the hypothalamic feeding center, such that hypothalamic arcuate nucleus neurons exhibit reduced sensitivity to the orexigenic and anorexigenic effects of ghrelin and leptin, respectively. However, hypothalamic infusion of thyroid hormone during an IBA is sufficient to rescue hibernation anorexia. Our results reveal that thyroid hormone deficiency underlies hibernation anorexia and demonstrate the functional flexibility of the hypothalamic feeding center.


Subject(s)
Anorexia , Ghrelin , Hibernation , Hypothalamus , Sciuridae , Animals , Hibernation/physiology , Sciuridae/physiology , Anorexia/physiopathology , Anorexia/metabolism , Hypothalamus/metabolism , Ghrelin/metabolism , Ghrelin/deficiency , Leptin/deficiency , Leptin/metabolism , Arcuate Nucleus of Hypothalamus/metabolism , Neurons/metabolism , Neurons/physiology , Male , Thyroid Hormones/metabolism , Arousal/physiology , Female , Seasons , Feeding Behavior/physiology
9.
Sci Rep ; 14(1): 15996, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987609

ABSTRACT

Alzheimer's disease (AD) is a neurological condition that is connected with a decline in a person's memory as well as their cognitive ability. One of the key topics of AD research has been the exploration of metabolic causes. We investigated the effects of treadmill exercise and intranasal insulin on learning and memory impairment and the expression of IGF1, BDNF, and GLUT4 in hypothalamus. The animals were put into 9 groups at random. In this study, we examined the impact of insulin on spatial memory in male Wistar rats and analyzed the effects of a 4-week pretreatment of moderate treadmill exercise and insulin on the mechanisms of improved hypothalamic glucose metabolism through changes in gene and protein expression of IGF1, BDNF, and GLUT4. We discovered that rat given Aß25-35 had impaired spatial learning and memory, which was accompanied by higher levels of Aß plaque burden in the hippocampus and lower levels of IGF1, BDNF, and GLUT4 mRNA and protein expression in the hypothalamus. Additionally, the administration of exercise training and intranasal insulin results in the enhancement of spatial learning and memory impairments, the reduction of plaque burden in the hippocampus, and the enhancement of the expression of IGF1, BDNF, and GLUT4 in the hypothalamus of rats that were treated with Aß25-35. Our results show that the improvement of learning and spatial memory due to the improvement of metabolism and upregulation of the IGF1, BDNF, and GLUT4 pathways can be affected by pretreatment exercise and intranasal insulin.


Subject(s)
Alzheimer Disease , Disease Models, Animal , Glucose Transporter Type 4 , Hypothalamus , Insulin-Like Growth Factor I , Insulin , Physical Conditioning, Animal , Rats, Wistar , Signal Transduction , Animals , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/therapy , Insulin-Like Growth Factor I/metabolism , Male , Insulin/metabolism , Rats , Hypothalamus/metabolism , Signal Transduction/drug effects , Glucose Transporter Type 4/metabolism , Glucose Transporter Type 4/genetics , Amyloid beta-Peptides/metabolism , Brain-Derived Neurotrophic Factor/metabolism , Brain-Derived Neurotrophic Factor/genetics , Hippocampus/metabolism , Hippocampus/drug effects , Administration, Intranasal , Peptide Fragments , Spatial Memory/drug effects , Spatial Learning/drug effects
10.
Sci Rep ; 14(1): 16082, 2024 Jul 12.
Article in English | MEDLINE | ID: mdl-38992091

ABSTRACT

Regulation of physiological homeostasis, including energy balance, is thought to be modified by low levels of adult neurogenesis in the hypothalamus. Hormones such as oestradiol can influence both embryonic and adult hypothalamic neurogenic programs, demonstrating a sensitivity of hypothalamic neural progenitor cells to endogenous hormones. Previously we showed that gestational exposure to environmental levels of the xenoestrogen bisphenol A (BPA) changed neural progenitor cell behaviors in the embryo; however, we did not examine if these changes were permanent to affect adult neurogenesis. Here we investigated whether adult neuro- and/or gliogenesis were altered in mice prenatally exposed to BPA and placed on a high-fat diet challenge. Gestationally exposed adult female mice on a standard diet gained less weight than non-BPA controls, whereas gestationally exposed BPA females on a high-fat diet gained more weight than controls. Males exposed to gestational BPA showed no differences in weight gain relative to control males. Concomitantly, adult neurogenesis was increased in the VMH, DMH, and PVN of adult female mice exposed to BPA on standard diet, suggesting that disrupted adult neurogenesis might perturb normal energy balance regulation in females. These results add to growing evidence that low-dose BPA exposure in utero causes changes to adult hypothalamic function.


Subject(s)
Benzhydryl Compounds , Energy Metabolism , Homeostasis , Hypothalamus , Neurogenesis , Phenols , Prenatal Exposure Delayed Effects , Animals , Benzhydryl Compounds/toxicity , Female , Phenols/toxicity , Neurogenesis/drug effects , Pregnancy , Mice , Hypothalamus/drug effects , Hypothalamus/metabolism , Prenatal Exposure Delayed Effects/chemically induced , Homeostasis/drug effects , Energy Metabolism/drug effects , Male , Diet, High-Fat/adverse effects
11.
FASEB J ; 38(14): e23770, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-38995817

ABSTRACT

Repeated bouts of high-intensity interval training (HIIT) induce an improvement in metabolism via plasticity of melanocortin circuits and attenuated hypothalamic inflammation. HIF-1α, which plays a vital role in hypothalamus-mediated regulation of peripheral metabolism, is enhanced in the hypothalamus by HIIT. This study aimed to investigate the effects of HIIT on hypothalamic HIF-1α expression and peripheral metabolism in obese mice and the underlying molecular mechanisms. By using a high-fat diet (HFD)-induced obesity mouse model, we determined the effect of HIIT on energy balance and the expression of the hypothalamic appetite-regulating neuropeptides, POMC and NPY. Moreover, hypothalamic HIF-1α signaling and its downstream glycolytic enzymes were explored after HIIT intervention. The state of microglia and microglial NF-κB signaling in the hypothalamus were also examined in vivo. In vitro by using an adenovirus carrying shRNA-HIF1ß, we explored the impact of HIF-1 signaling on glycolysis and NF-κB inflammatory signaling in BV2 cells. Food intake was suppressed and whole-body metabolism was improved in exercised DIO mice, accompanied by changes in the expression of POMC and NPY. Moreover, total and microglial HIF-1α signaling were obviously attenuated in the hypothalamus, consistent with the decreased levels of glycolytic enzymes. Both HFD-induced microglial activation and hypothalamic NF-κB signaling were significantly suppressed following HIIT in vivo. In BV2 cells, after HIF-1 complex knockdown, glycolysis and NF-κB inflammatory signaling were significantly attenuated. The data indicate that HIIT improves peripheral metabolism probably via attenuated HFD-induced microglial activation and microglial NF-κB signaling in the hypothalamus, which could be mediated by suppressed microglial HIF-1α signaling.


Subject(s)
Hypothalamus , Hypoxia-Inducible Factor 1, alpha Subunit , Inflammation , Mice, Inbred C57BL , Microglia , Signal Transduction , Animals , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Microglia/metabolism , Male , Mice , Hypothalamus/metabolism , Inflammation/metabolism , High-Intensity Interval Training , Obesity/metabolism , Diet, High-Fat/adverse effects , Physical Conditioning, Animal/physiology , NF-kappa B/metabolism , Pro-Opiomelanocortin/metabolism , Pro-Opiomelanocortin/genetics , Neuropeptide Y/metabolism
12.
Sci Rep ; 14(1): 15384, 2024 07 04.
Article in English | MEDLINE | ID: mdl-38965316

ABSTRACT

Disruptions in energy homeostasis can lead to diseases like obesity and diabetes, affecting millions of people each year. Tanycytes, the adult stem cells in the hypothalamus, play crucial roles in assisting hypothalamic neurons in maintaining energy balance. Although tanycytes have been extensively studied in rodents, our understanding of human tanycytes remains limited. In this study, we utilized single-cell transcriptomics data to explore the heterogeneity of human embryonic tanycytes, investigate their gene regulatory networks, analyze their intercellular communication, and examine their developmental trajectory. Our analysis revealed the presence of two clusters of ß tanycytes and three clusters of α tanycytes in our dataset. Surprisingly, human embryonic tanycytes displayed significant similarities to mouse tanycytes in terms of marker gene expression and transcription factor activities. Trajectory analysis indicated that α tanycytes were the first to be generated, giving rise to ß tanycytes in a dorsal-ventral direction along the third ventricle. Furthermore, our CellChat analyses demonstrated that tanycytes generated earlier along the developmental lineages exhibited increased intercellular communication compared to those generated later. In summary, we have thoroughly characterized the heterogeneity of human embryonic tanycytes from various angles. We are confident that our findings will serve as a foundation for future research on human tanycytes.


Subject(s)
Ependymoglial Cells , Single-Cell Analysis , Transcriptome , Humans , Ependymoglial Cells/metabolism , Ependymoglial Cells/cytology , Gene Regulatory Networks , Mice , Animals , Gene Expression Profiling , Cell Communication/genetics , Hypothalamus/metabolism , Hypothalamus/cytology
13.
Nat Commun ; 15(1): 5979, 2024 Jul 16.
Article in English | MEDLINE | ID: mdl-39013907

ABSTRACT

Neuronal activity undergoes significant changes during vigilance states, accompanied by an accommodation of energy demands. While the astrocyte-neuron lactate shuttle has shown that lactate is the primary energy substrate for sustaining neuronal activity in multiple brain regions, its role in regulating sleep/wake architecture is not fully understood. Here we investigated the involvement of astrocytic lactate supply in maintaining consolidated wakefulness by downregulating, in a cell-specific manner, the expression of monocarboxylate transporters (MCTs) in the lateral hypothalamus of transgenic mice. Our results demonstrate that reduced expression of MCT4 in astrocytes disrupts lactate supply to wake-promoting orexin neurons, impairing wakefulness stability. Additionally, we show that MCT2-mediated lactate uptake is necessary for maintaining tonic firing of orexin neurons and stabilizing wakefulness. Our findings provide both in vivo and in vitro evidence supporting the role of astrocyte-to-orexinergic neuron lactate shuttle in regulating proper sleep/wake stability.


Subject(s)
Astrocytes , Hypothalamic Area, Lateral , Lactic Acid , Mice, Transgenic , Monocarboxylic Acid Transporters , Neurons , Orexins , Sleep , Wakefulness , Animals , Astrocytes/metabolism , Wakefulness/physiology , Orexins/metabolism , Sleep/physiology , Monocarboxylic Acid Transporters/metabolism , Monocarboxylic Acid Transporters/genetics , Neurons/metabolism , Lactic Acid/metabolism , Mice , Hypothalamic Area, Lateral/metabolism , Male , Hypothalamus/metabolism , Mice, Inbred C57BL , Muscle Proteins
14.
CNS Neurosci Ther ; 30(6): e14801, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38887187

ABSTRACT

BACKGROUND AND OBJECTIVE: Amyotrophic lateral sclerosis (ALS) causes motor neuron loss and progressive paralysis. While traditionally viewed as motor neuron disease (MND), ALS also affects non-motor regions, such as the hypothalamus. This study aimed to quantify the hypothalamic subregion volumes in patients with ALS versus healthy controls (HCs) and examine their associations with demographic and clinical features. METHODS: Forty-eight participants (24 ALS patients and 24 HCs) underwent structural MRI. A deep convolutional neural network was used for the automated segmentation of the hypothalamic subunits, including the anterior-superior (a-sHyp), anterior-inferior (a-iHyp), superior tuberal (supTub), inferior tuberal (infTub), and posterior (posHyp). The neural network was validated using FreeSurfer v7.4.1, with individual head size variations normalized using total intracranial volume (TIV) normalization. Statistical analyses were performed for comparisons using independent sample t-tests. Correlations were calculated using Pearson's and Spearman's tests (p < 0.05). The standard mean difference (SMD) was used to compare the mean differences between parametric variables. RESULTS: The volume of the left a-sHyp hypothalamic subunit was significantly lower in ALS patients than in HCs (p = 0.023, SMD = -0.681). No significant correlation was found between the volume of the hypothalamic subunits, body mass index (BMI), and ALSFRS-R in patients with ALS. However, right a-sHyp (r = 0.420, p = 0.041) was correlated with disease duration, whereas right supTub (r = -0.471, p = 0.020) and left postHyp (r = -0.406, p = 0.049) were negatively correlated with age. There was no significant difference in the volume of hypothalamic subunits between males and females, and no significant difference was found between patients with revised ALS Functional Rating Scale (ALSFRS-R) scores ≤41 and >41 and those with a disease duration of 9 months or less. DISCUSSION AND CONCLUSION: The main finding suggests atrophy of the left a-sHyp hypothalamic subunit in patients with ALS, which is supported by previous research as an extra-motor neuroimaging finding for ALS.


Subject(s)
Amyotrophic Lateral Sclerosis , Hypothalamus , Magnetic Resonance Imaging , Humans , Amyotrophic Lateral Sclerosis/diagnostic imaging , Amyotrophic Lateral Sclerosis/pathology , Male , Female , Middle Aged , Hypothalamus/diagnostic imaging , Hypothalamus/pathology , Aged , Adult
15.
Sci Adv ; 10(25): eadn8350, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38905332

ABSTRACT

The suprachiasmatic nucleus (SCN) sets the phase of oscillation throughout the brain and body. Anatomical evidence reveals a portal system linking the SCN and the organum vasculosum of the lamina terminalis (OVLT), begging the question of the direction of blood flow and the nature of diffusible signals that flow in this specialized vasculature. Using a combination of anatomical and in vivo two-photon imaging approaches, we unequivocally show that blood flows unidirectionally from the SCN to the OVLT, that blood flow rate displays daily oscillations with a higher rate at night than in the day, and that circulating vasopressin can access portal vessels. These findings highlight a previously unknown central nervous system communication pathway, which, like that of the pituitary portal system, could allow neurosecretions to reach nearby target sites in OVLT, avoiding dilution in the systemic blood. In both of these brain portal pathways, the target sites relay signals broadly to both the brain and the rest of the body.


Subject(s)
Suprachiasmatic Nucleus , Suprachiasmatic Nucleus/physiology , Animals , Mice , Hypothalamus/metabolism , Hypothalamus/blood supply , Brain/blood supply , Brain/physiology , Brain/metabolism , Portal System , Male , Vasopressins/metabolism , Vasopressins/blood , Cerebrovascular Circulation/physiology , Circadian Rhythm/physiology
16.
PLoS Biol ; 22(6): e3002641, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38833481

ABSTRACT

In utero exposure to maternal obesity programs increased obesity risk. Animal models show that programmed offspring obesity is preceded by hyperphagia, but the mechanisms that mediate these changes are unknown. Using a mouse model of maternal obesity, we observed increased intake of a high-fat diet (HFD) in offspring of obese mothers that precedes the development of obesity. Through small RNA sequencing, we identified programmed overexpression of hypothalamic miR-505-5p that is established in the fetus, lasts to adulthood and is maintained in hypothalamic neural progenitor cells cultured in vitro. Metabolic hormones and long-chain fatty acids associated with obesity increase miR-505-5p expression in hypothalamic neurons in vitro. We demonstrate that targets of miR-505-5p are enriched in fatty acid metabolism pathways and overexpression of miR-505-5p decreased neuronal fatty acid metabolism in vitro. miR-505-5p targets are associated with increased BMI in human genetic studies. Intra-cerebroventricular injection of miR-505-5p in wild-type mice increased HFD intake, mimicking the phenotype observed in offspring exposed to maternal obesity. Conversely, maternal exercise intervention in an obese mouse pregnancy rescued the programmed increase of hypothalamic miR-505-5p in offspring of obese dams and reduced HFD intake to control offspring levels. This study identifies a novel mechanism by which maternal obesity programs obesity in offspring via increased intake of high-fat foods.


Subject(s)
Diet, High-Fat , Fatty Acids , Hypothalamus , MicroRNAs , Obesity, Maternal , Animals , Female , Humans , Male , Mice , Pregnancy , Diet, High-Fat/adverse effects , Fatty Acids/metabolism , Hypothalamus/metabolism , Mice, Inbred C57BL , MicroRNAs/metabolism , MicroRNAs/genetics , Neurons/metabolism , Obesity/metabolism , Obesity/genetics , Obesity, Maternal/metabolism , Prenatal Exposure Delayed Effects/metabolism , Prenatal Exposure Delayed Effects/genetics
17.
Nutrients ; 16(11)2024 May 31.
Article in English | MEDLINE | ID: mdl-38892653

ABSTRACT

The onset of puberty, which is under the control of the hypothalamic-pituitary-gonadal (HPG) axis, is influenced by various factors, including obesity, which has been associated with the earlier onset of puberty. Obesity-induced hypothalamic inflammation may cause premature activation of gonadotropin-releasing hormone (GnRH) neurons, resulting in the development of precocious or early puberty. Mechanisms involving phoenixin action and hypothalamic microglial cells are implicated. Furthermore, obesity induces structural and cellular brain alterations, disrupting metabolic regulation. Imaging studies reveal neuroinflammatory changes in obese individuals, impacting pubertal timing. Magnetic resonance spectroscopy enables the assessment of the brain's neurochemical composition by measuring key metabolites, highlighting potential pathways involved in neurological changes associated with obesity. In this article, we present evidence indicating a potential association among obesity, hypothalamic inflammation, and precocious puberty.


Subject(s)
Hypothalamus , Pediatric Obesity , Puberty, Precocious , Humans , Pediatric Obesity/complications , Hypothalamus/metabolism , Child , Puberty, Precocious/etiology , Puberty/physiology , Inflammation , Female , Gonadotropin-Releasing Hormone/metabolism , Male , Hypothalamo-Hypophyseal System/metabolism
18.
Metab Brain Dis ; 39(5): 679-690, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38842661

ABSTRACT

Subarachnoid hemorrhage (SAH) is a serious hemorrhagic event with high mortality and morbidity. Multiple injurious events produced by SAH can lead to a series of pathophysiologic processes in the hypothalamus that can severely impact patients' life. These pathophysiologic processes usually result in physiologic derangements and dysfunction of the brain and multiple organs. This dysfunction involved multiple dimensions of the genome and metabolome. In our study, we induced the SAH model in rats to obtain hypothalamic tissue and serum. The samples were subsequently analyzed by transcriptomics and metabolomics. Next, the functional enrichment analysis of the differentially expressed genes and metabolites were performed by GO and KEGG pathway analysis. Through transcriptomic analysis of hypothalamus samples, 263 up-regulated differential genes, and 207 down-regulated differential genes were identified in SAH groups compared to Sham groups. In the KEGG pathway analysis, a large number of differential genes were found to be enriched in IL-17 signaling pathway, PI3K-Akt signaling pathway, and bile secretion. Liquid chromatography-mass spectrometry metabolomics technology was conducted on the serum of SAH rats and identified 11 up-regulated and 26 down-regulated metabolites in positive ion model, and 1 up-regulated and 10 down-regulated metabolites in negative ion model. KEGG pathways analysis showed that differentially expressed metabolites were mainly enriched in pathways of bile secretion and primary bile acid biosynthesis. We systematically depicted the neuro- and metabolism-related biomolecular changes occurring in the hypothalamus after SAH by performing transcriptomics and metabolomics studies. These biomolecular changes may provide new insights into hypothalamus-induced metabolic changes and gene expression after SAH.


Subject(s)
Hypothalamus , Metabolomics , Rats, Sprague-Dawley , Subarachnoid Hemorrhage , Transcriptome , Animals , Subarachnoid Hemorrhage/metabolism , Subarachnoid Hemorrhage/genetics , Rats , Hypothalamus/metabolism , Male , Gene Expression Profiling , Metabolome
19.
Physiol Behav ; 283: 114601, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-38838800

ABSTRACT

AIM: The hypothesis of this study is to determine the effects of intracerebroventricular (icv) prokineticin 2 infusion on food consumption and body weight and to elucidate whether it has effects on energy expenditure via the hypothalamus-pituitary-thyroid (HPT) axis in adipose tissue. MATERIAL AND METHODS: A total of 40 rats were used in the study and 4 groups were established: Control, Sham, Prokineticin 1.5 and Prokineticin 4.5 (n=10). Except for the Control group, rats were treated intracerebroventricularly via osmotic minipumps, the Sham group was infused with aCSF (vehicle), and the Prokineticin 1.5 and Prokineticin 4.5 groups were infused with 1.5 nMol and 4.5 nMol prokineticin 2, respectively. Food and water consumption and body weight were monitored during 7-day infusion in all groups. At the end of the infusion, the rats were decapitated and serum TSH, fT4 and fT3 levels were determined by ELISA. In addition, PGC-1α and UCP1 gene expression levels in white adipose tissue (WAT) and brown adipose tissue (BAT), TRH from rat hypothalamic tissue were determined by real-time PCR. RESULTS: Icv prokineticin 2 (4.5 nMol) infusion had no effect on water consumption but reduced daily food consumption and body weight (p<0.05). Icv prokineticin 2 (4.5 nMol) infusion significantly increased serum TSH, fT4 and fT3 levels when compared to Control and Sham groups (p<0.05). Also, icv prokineticin 2 (4.5 nMol) infusion increased the expression of TRH in the hypothalamus tissue and expression of PGC-1α UCP1 in the WAT and BAT (p<0.05). CONCLUSION: Icv prokineticin 2 (4.5 nMol) infusion may suppress food consumption via its receptors in the hypothalamus and reduce body weight by stimulating energy expenditure and thermogenesis in adipose tissue through the HPT axis.


Subject(s)
Body Weight , Eating , Energy Metabolism , Gastrointestinal Hormones , Infusions, Intraventricular , Thyroid Gland , Animals , Energy Metabolism/drug effects , Energy Metabolism/physiology , Male , Body Weight/drug effects , Eating/drug effects , Eating/physiology , Thyroid Gland/drug effects , Thyroid Gland/metabolism , Rats , Gastrointestinal Hormones/metabolism , Gastrointestinal Hormones/administration & dosage , Uncoupling Protein 1/metabolism , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Neuropeptides/metabolism , Neuropeptides/administration & dosage , Thyrotropin/blood , Thyrotropin/metabolism , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/drug effects , Thyroxine/blood , Thyroxine/administration & dosage , Drinking/drug effects , Triiodothyronine/administration & dosage , Triiodothyronine/blood , Triiodothyronine/pharmacology , Rats, Wistar , Hypothalamus/metabolism , Hypothalamus/drug effects , Adipose Tissue, White/metabolism , Adipose Tissue, White/drug effects
20.
Neurobiol Aging ; 141: 102-112, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38850591

ABSTRACT

The hypothalamus regulates homeostasis across the lifespan and is emerging as a regulator of aging. In murine models, aging-related changes in the hypothalamus, including microinflammation and gliosis, promote accelerated neurocognitive decline. We investigated relationships between hypothalamic microstructure and features of neurocognitive aging, including cortical thickness and cognition, in a cohort of community-dwelling older adults (age range 65-97 years, n=124). Hypothalamic microstructure was evaluated with two magnetic resonance imaging diffusion metrics: mean diffusivity (MD) and fractional anisotropy (FA), using a novel image processing pipeline. Hypothalamic MD was cross-sectionally positively associated with age and it was negatively associated with cortical thickness. Hypothalamic FA, independent of cortical thickness, was cross-sectionally positively associated with neurocognitive scores. An exploratory analysis of longitudinal neurocognitive performance suggested that lower hypothalamic FA may predict cognitive decline. No associations between hypothalamic MD, age, and cortical thickness were identified in a younger control cohort (age range 18-63 years, n=99). To our knowledge, this is the first study to demonstrate that hypothalamic microstructure is associated with features of neurocognitive aging in humans.


Subject(s)
Aging , Cognition , Cognitive Aging , Hypothalamus , Humans , Aged , Male , Female , Aged, 80 and over , Hypothalamus/diagnostic imaging , Hypothalamus/pathology , Middle Aged , Adult , Cognitive Aging/physiology , Aging/pathology , Aging/psychology , Young Adult , Magnetic Resonance Imaging , Adolescent , Cohort Studies , Cross-Sectional Studies , Cerebral Cortex/diagnostic imaging , Cerebral Cortex/pathology , Anisotropy
SELECTION OF CITATIONS
SEARCH DETAIL
...