5,479 results on '"Arcuate Nucleus"'
Search Results
2. Effect of galanin-like peptide on hypothalamic kisspeptin expression in female Zucker fatty rats
- Author
-
Sakata, Akiko, Iwata, Kinuyo, Nakao, Kimihiko, Kunimura, Yuyu, Suzuki, Shunji, Ozawa, Hitoshi, and Ishii, Hirotaka
- Published
- 2025
- Full Text
- View/download PDF
3. Altered GnRH neuron-glia networks close to interface of polycystic ovary syndrome: Molecular mechanism and clinical perspectives
- Author
-
Dai, Ruoxi and Sun, Yan
- Published
- 2025
- Full Text
- View/download PDF
4. Opposing effects of nicotine on hypothalamic arcuate nucleus POMC and NPY neurons
- Author
-
Ramírez-Sánchez, E., Mondragón-García, A., Garduño, J., Hernández-Vázquez, F., Ortega-Tinoco, S., and Hernández-López, S.
- Published
- 2024
- Full Text
- View/download PDF
5. Selective Colocalization of GHSR and GLP-1R in a Subset of Hypothalamic Neurons and Their Functional Interaction.
- Author
-
Aguggia, Julieta, Fernandez, Gimena, Cassano, Daniela, Mustafá, Emilio R, Rodríguez, Silvia S, Cantel, Sonia, Fehrentz, Jean-Alain, Raingo, Jesica, Schiöth, Helgi B, Habib, Abdella M, Francesco, Pablo N De, and Perello, Mario
- Subjects
GLUCAGON-like peptide-1 receptor ,G protein coupled receptors ,CALCIUM channels ,GHRELIN ,NEURONS - Abstract
The GH secretagogue receptor (GHSR) and the glucagon-like peptide-1 receptor (GLP-1R) are G protein-coupled receptors with critical, yet opposite, roles in regulating energy balance. Interestingly, these receptors are expressed in overlapping brain regions. However, the extent to which they target the same neurons and engage in molecular crosstalk remains unclear. To explore the potential colocalization of GHSR and GLP-1R in specific neurons, we performed detailed mapping of cells positive for both receptors using GHSR-eGFP reporter mice or wild-type mice infused with fluorescent ghrelin, alongside an anti-GLP-1R antibody. We found that GHSR+ and GLP-1R+ cells are largely segregated in the mouse brain. The highest overlap was observed in the hypothalamic arcuate nucleus, where 15% to 20% of GHSR+ cells were also GLP-1R+ cells. Additionally, we examined RNA-sequencing datasets from mouse and human brains to assess the fraction and distribution of neurons expressing both receptors, finding that double-positive Ghsr+/Glp1r+ cells are highly segregated, with a small subset of double-positive Ghsr+/Glp1r+ cells representing <10% of all Ghsr+ or Glp1r+ cells, primarily enriched in the hypothalamus. Furthermore, we conducted functional studies using patch-clamp recordings in a heterologous expression system to assess potential crosstalk in regulating presynaptic calcium channels. We provide the first evidence that liraglutide-evoked GLP-1R activity inhibits presynaptic channels, and that the presence of one GPCR attenuates the inhibitory effects of ligand-evoked activity mediated by the other on presynaptic calcium channels. In conclusion, while GHSR and GLP-1R can engage in molecular crosstalk, they are largely segregated across most neuronal types within the brain. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
6. Central δ/κ opioid receptor signaling pathways mediate chronic and/or acute suckling-induced LH suppression in rats during late lactation
- Author
-
Yoshihisa UENOYAMA, Miku NONOGAKI, Hitomi TSUCHIDA, Marina TAKIZAWA, Sena MATSUZAKI, Naoko INOUE, and Hiroko TSUKAMURA
- Subjects
arcuate nucleus ,δ opioid receptor ,enkephalin ,κ opioid receptor ,luteinizing hormone ,Reproduction ,QH471-489 ,Internal medicine ,RC31-1245 - Abstract
In mammals, secretion of tonic (pulsatile) gonadotropin-releasing hormone (GnRH)/luteinizing hormone (LH) is often suppressed during lactation. Suppression of GnRH/LH pulses in lactating dams is assumed to be caused by suckling stimuli and a chronic negative energy balance due to milk production. The present study aimed to investigate whether the central enkephalin-δ opioid receptor (DOR) signaling mediated the suppression of LH secretion by acute suckling stimuli and/or chronic negative energy balance due to milk production in rats during late lactation when dams were under a heavy energy demand. On postpartum day 16, the number of Penk (enkephalin mRNA)-expressing cells in the arcuate nucleus was significantly higher in lactating rats than in non-lactating control rats. Pulsatile LH secretion was suppressed in rats with chronic suckling or acute 1-h suckling stimuli 6 h after pup removal on day 16 of lactation. Central DOR antagonism significantly increased the mean LH concentrations and the baseline of LH pulses in rats with chronic suckling but not with acute suckling stimuli on day 16 of lactation. Besides, central κ opioid receptor (KOR) antagonism increased the amplitude of LH pulses in rats with the acute suckling stimuli on day 16 of lactation. These results suggest that central DOR signaling mediates the suppression of LH secretion caused by a negative energy balance in rats receiving chronic suckling during late lactation. On the other hand, central KOR signaling likely mediates acute suckling stimuli-induced suppression of LH secretion in rats during late lactation.
- Published
- 2024
- Full Text
- View/download PDF
7. Negative feedback control of hypothalamic feeding circuits by the taste of food.
- Author
-
Aitken, Tara J., Liu, Zhengya, Ly, Truong, Shehata, Sarah, Sivakumar, Nilla, La Santa Medina, Naymalis, Gray, Lindsay A., Zhang, Jingkun, Dundar, Naz, Barnes, Chris, and Knight, Zachary A.
- Subjects
- *
LEPTIN receptors , *TASTE testing of food , *FOOD consumption , *MIRROR images , *PEPTIDES , *SWEETNESS (Taste) , *HUNGER - Abstract
The rewarding taste of food is critical for motivating animals to eat, but whether taste has a parallel function in promoting meal termination is not well understood. Here, we show that hunger-promoting agouti-related peptide (AgRP) neurons are rapidly inhibited during each bout of ingestion by a signal linked to the taste of food. Blocking these transient dips in activity via closed-loop optogenetic stimulation increases food intake by selectively delaying the onset of satiety. We show that upstream leptin-receptor-expressing neurons in the dorsomedial hypothalamus (DMHLepR) are tuned to respond to sweet or fatty tastes and exhibit time-locked activation during feeding that is the mirror image of downstream AgRP cells. These findings reveal an unexpected role for taste in the negative feedback control of ingestion. They also reveal a mechanism by which AgRP neurons, which are the primary cells that drive hunger, are able to influence the moment-by-moment dynamics of food consumption. • Hunger-promoting AgRP neurons are transiently inhibited by the taste of food • Optogenetic reversal of this gustatory inhibition extends meal duration • Upstream neurons in the DMH are tuned to respond to sweet or fatty tastes • DMH neurons integrate taste cues with slower, post-ingestive nutrient signals Aitken et al. show that the taste of food inhibits hunger-promoting neurons in the hypothalamus and that this gustatory feedback controls the duration of a meal. This reveals how appetitive taste cues, which are traditionally thought to promote food intake, can simultaneously initiate the process of meal termination. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. Alpha-synuclein expression in GnRH neurons of young and old bovine hypothalami.
- Author
-
Niyonzima, Yvan Bienvenu, Asato, Yuuki, Murakami, Tomoaki, and Kadokawa, Hiroya
- Subjects
- *
PREOPTIC area , *CENTRAL nervous system , *CONGO red (Staining dye) , *GONADOTROPIN releasing hormone , *AMYLOID plaque , *HYPOTHALAMUS - Abstract
Context: Understanding of central nervous system mechanisms related to age-related infertility remains limited. Fibril α-synuclein, distinct from its monomer form, is implicated in age-related diseases and propagates among neurons akin to prions. Aims: We compared α-synuclein expression in gonadotropin-releasing hormone-expressing neurons (GnRH neurons) in the pre-optic area, arcuate nucleus, and median eminence of healthy heifers and aged cows to determine its role in age-related infertility. Methods: We analysed mRNA and protein expression, along with fluorescent immunohistochemistry for GnRH and α-synuclein, followed by Congo red staining to detect amyloid deposits, and confocal microscopy. Key results: Both mRNA and protein expressions of α-synuclein were confirmed by reverse transcription-polymerase chain reaction (RT-PCR) and western blots in bovine cortex, hippocampus, and anterior and posterior hypothalamus tissues. Significant differences in α-synuclein mRNA expression were observed in the cortex and hippocampus between young and old cows. Western blots showed five bands of α-synuclein, probably reflecting monomer, dimer, and oligomers, in the cortex, hippocampus, hypothalamus tissues, and there were significant differences in some bands between young and old cows. Bright-field and polarised light microscopy did not detect obvious amyloid deposition in aged hypothalami; however, higher-sensitive confocal microscopy unveiled strong positive signal of Congo red and α-synuclein in GnRH neurons in aged hypothalami. Additionally, α-synuclein expression was detected in immortalised GnRH neurons, GT1-7 cells. Conclusion: Alpha-synuclein was expressed in GnRH neurons, and some differences were observed between young and old hypothalami. Implications: Alpha-synuclein may play an important role in aging-related infertility. In our investigation into age-related infertility, we examined the presence of α-synuclein, a protein associated with brain aging, particularly in gonadotropin-releasing hormone (GnRH) neurons in hypothalami. Contrasting samples from young and old cows, we observed significant differences in α-synuclein levels across the specific hypothalamus regions linked to reproductive function. These findings suggest that α-synuclein may play a pivotal role in the mechanisms underlying age-related infertility, paving the way for further inquiries into potential therapeutic avenues in reproductive health. Image by Hiroya Kadokawa. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
9. Deletion of Nuclear Progesterone Receptors From Kisspeptin Cells Does Not Impair Negative Feedback in Female Mice.
- Author
-
Dillon, Kendra M, Lohr, Dayanara B, Novak, Alyssa G, Petriv, Anna-Maria V, Neifert, Nicole T, and Moore, Aleisha M
- Subjects
PROGESTERONE receptors ,KISSPEPTINS ,PULSE generators ,MESSENGER RNA ,GONADOTROPIN releasing hormone - Abstract
Reproductive function in mammals depends on the ability of progesterone (P4) to suppress pulsatile gonadotrophin-releasing hormone (GnRH) and luteinizing hormone (LH) secretion in a homeostatic-negative feedback loop. Previous research identified that cells upstream from GnRH neurons expressing the nuclear progesterone receptor (PGR) are required for P4-negative feedback. However, the identity of these cells and the mechanism by which they reduce GnRH/LH pulsatile secretion is unknown. We aimed to address the hypothesis that PGR expressed by a neural population in the arcuate nucleus recently identified as the GnRH pulse generator, cells expressing kisspeptin, neurokinin B, and dynorphin (KNDy cells), mediate P4-negative feedback. To achieve this, we used female mice with the PGR gene conditionally deleted from kisspeptin cells (KPRKO mice) and observed a substantial decrease in the percentage of KNDy neurons coexpressing PGR messenger RNA (mRNA) (11% in KPRKO mice vs 86% in wild-type [WT] mice). However, KPRKO mice did not display changes in the frequency or amplitude of LH pulses in diestrus or estrus, nor in the ability of exogenous P4 to blunt a postcastration increase in LH. Further, mRNA expression of arcuate kisspeptin and dynorphin, which are excitatory and inhibitory to GnRH secretion, respectively, remained unaltered in KPRKO mice compared to WT controls. Together, these findings show that the near-complete loss of PGR signaling from KNDy cells does not affect negative feedback regulation of GnRH pulse generation in mice, suggesting that feedback through this receptor can occur via a small number of KNDy cells or a yet unidentified cell population. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Microglia in physiological conditions and the importance of understanding their homeostatic functions in the arcuate nucleus.
- Author
-
Guzmán-Ruíz, Mara A., Guerrero Vargas, Natalí N., Jair Ramírez-Carreto, Ricardo, Carlos González-Orozco, Juan, Adair Torres-Hernández, Bryan, Valle-Rodríguez, Michelle, Guevara-Guzmán, Rosalinda, and Chavarría, Anahí
- Subjects
MICROGLIA ,CONDITIONED response ,NEURONS ,HOMEOSTASIS ,HYPOTHALAMUS - Abstract
Microglia are highly dynamic cells that have been mainly studied under pathological conditions. The present review discusses the possible implication of microglia as modulators of neuronal electrical responses in physiological conditions and hypothesizes how these cells might modulate hypothalamic circuits in health and during obesity. Microglial cells studied under physiological conditions are highly diverse, depending on the developmental stage and brain region. The evidence also suggests that neuronal electrical activity modulates microglial motility to control neuronal excitability. Additionally, we show that the expression of genes associated with neuronmicroglia interaction is down-regulated in obese mice compared to control-fed mice, suggesting an alteration in the contact-dependent mechanisms that sustain hypothalamic arcuate-median eminence neuronal function. We also discuss the possible implication of microglial-derived signals for the excitability of hypothalamic neurons during homeostasis and obesity. This review emphasizes the importance of studying the physiological interplay between microglia and neurons to maintain proper neuronal circuit function. It aims to elucidate how disruptions in the normal activities of microglia can adversely affect neuronal health. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. Arcuate dopaminergic/GABAergic neurons project within the hypothalamus and to the median eminence.
- Author
-
Mittal, Somya, Arenkiel, Benjamin R., and Lyons-Warren, Ariel M.
- Subjects
- *
GABAERGIC neurons , *GABA transporters , *DOPAMINERGIC neurons , *NEURAL circuitry , *TYROSINE hydroxylase - Abstract
Cotransmission, meaning the release of multiple neurotransmitters from one synapse, allows for increased diversity of signaling in the brain. Dopamine (DA) and γ-aminobutyric acid (GABA) are known to coexpress in many regions such as the olfactory bulb and the ventral tegmental area. Tuberoinfundibular dopaminergic neurons (TIDA) in the arcuate nucleus of the hypothalamus (Arc) project to the median eminence (ME) and regulate prolactin release from the pituitary, and prior work suggests dopaminergic Arc neurons also cotransmit GABA. However, the extent of cotransmission, and the projection patterns of these neurons have not been fully revealed. Here, we used a genetic intersectional reporter expression approach to selectively label cells that express both tyrosine hydroxylase (TH) and vesicular GABA transporter (VGAT). Through this approach, we identified cells capable of both DA and GABA cotransmission in the Arc, periventricular (Pe), paraventricular (Pa), ventromedial, and the dorsolateral hypothalamic nuclei, in addition to a novel population in the caudate putamen. The highest density of labeled cells was in the Arc, 6.68% of DAPI-labeled cells at Bregma −2.06 mm, and in the Pe, 2.83% of DAPI-labeled cells at Bregma −1.94 mm. Next, we evaluated the projections of these DA/GABA cells by injecting an mCherry virus that fluoresces in DA/GABA cells. We observed a cotransmitting DA/GABA population, with projections within the Arc, and to the Pa and ME. These data suggest DA/GABA Arc neurons are involved in prolactin release as a subset of TIDA neurons. Further investigation will elucidate the interactions of dopamine and GABA in the hypothalamus. NEW & NOTEWORTHY: Cotransmitting dopaminergic (DA) and γ-aminobutyric acid (GABA)ergic (DA/GABA) neurons contribute to the complexity of neural circuits. Using a new genetic technique, we characterized the locations, density, and projections of hypothalamic DA/GABA neurons. DA/GABA cells are mostly in the arcuate nucleus (Arc), from which they project locally within the arcuate, to the median eminence (ME), and to the paraventricular (Pa) nucleus. There is also a small and previously unreported group of DA/GABA cells in the caudate putamen. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Post‐weaning social isolation modifies neonatal anoxia‐induced changes in energy metabolism and growth of rats.
- Author
-
Cruz‐Ochoa, Natalia Andrea, Motta‐Teixeira, Lívia Clemente, Cruz‐Ochoa, Pablo Felipe, Lopez‐Paredes, Santiago, Ochoa‐Amaya, Julieta Esperanza, Takada, Silvia Honda, Xavier, Gilberto Fernando, and Nogueira, Maria Inês
- Subjects
- *
SOCIAL isolation , *ENERGY metabolism , *WEIGHT gain , *WEIGHT loss , *PREMATURE infants - Abstract
Neonatal oxygen deficiency in rats may disturb growth and long‐term metabolic homeostasis. In order to facilitate metabolic evaluation, the subjects are usually housed individually. However, social isolation associated with individually housed conditions alters animal behavior, which may influence the experimental results. This study investigated the effects of social isolation on neonatal anoxia‐induced changes in growth and energy metabolism. Male and female Wistar rats were exposed, on postnatal day 2 (P2), to either 25‐min of anoxia or control treatment. From P27 onward, part of the subjects of each group was isolated in standard cages, and the remaining subjects were housed in groups. At P34 or P95, the subjects were fasted for 18 h, refeed for 1 h, and then perfused 30 min later. Glycemia, leptin, insulin, and morphology of the pancreas were evaluated at both ages. For subjects perfused at P95, body weight and food intake were recorded up to P90, and the brain was collected for Fos and NeuN immunohistochemistry. Results showed that male rats exposed to neonatal anoxia and social isolation exhibited increased body weight gain despite the lack of changes in food intake. In addition, social isolation (1) decreased post‐fasting weight loss and post‐fasting food intake and (2) increased glycemia, insulin, and leptin levels of male and female rats exposed to anoxia and control treatments, both at P35 and P95. Furthermore, although at P35, anoxia increased insulin levels of males, it decreased the area of the β‐positive cells in the pancreas of females. At P95, anoxia increased post‐prandial weight loss of males, post‐fasting food intake, insulin, and leptin, and decreased Fos expression in the arcuate nucleus (ARC) of males and females. Hyperphagia was associated with possible resistance to leptin and insulin, suspected by the high circulating levels of these hormones and poor neuronal activation of ARC. This study demonstrated that continuous social isolation from weaning modifies, in a differentiated way, the long‐term energy metabolism and growth of male and female Wistar rats exposed to neonatal anoxia or even control treatments. Therefore, social isolation should be considered as a factor that negatively influences experimental results and the outcomes of the neonatal injury. These results should also be taken into account in clinical procedures, since the used model simulates the preterm babies' conditions and some therapeutic approaches require isolation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. Thyrotropin-Releasing Hormone and Food Intake in Mammals: An Update.
- Author
-
Vargas, Yamili, Castro Tron, Ana Elena, Rodríguez Rodríguez, Adair, Uribe, Rosa María, Joseph-Bravo, Patricia, and Charli, Jean-Louis
- Subjects
THYROTROPIN releasing factor ,FOOD consumption ,RAPHE nuclei ,BRAIN stem ,NUCLEUS accumbens ,HYPOTHALAMUS ,INGESTION - Abstract
Thyrotropin-releasing hormone (TRH; pGlu-His-Pro-NH2) is an intercellular signal produced mainly by neurons. Among the multiple pharmacological effects of TRH, that on food intake is not well understood. We review studies demonstrating that peripheral injection of TRH generally produces a transient anorexic effect, discuss the pathways that might initiate this effect, and explain its short half-life. In addition, central administration of TRH can produce anorexic or orexigenic effects, depending on the site of injection, that are likely due to interaction with TRH receptor 1. Anorexic effects are most notable when TRH is injected into the hypothalamus and the nucleus accumbens, while the orexigenic effect has only been detected by injection into the brain stem. Functional evidence points to TRH neurons that are prime candidate vectors for TRH action on food intake. These include the caudal raphe nuclei projecting to the dorsal motor nucleus of the vagus, and possibly TRH neurons from the tuberal lateral hypothalamus projecting to the tuberomammillary nuclei. For other TRH neurons, the anatomical or physiological context and impact of TRH in each synaptic domain are still poorly understood. The manipulation of TRH expression in well-defined neuron types will facilitate the discovery of its role in food intake control in each anatomical scene. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
14. Microglia in physiological conditions and the importance of understanding their homeostatic functions in the arcuate nucleus
- Author
-
Mara A. Guzmán-Ruíz, Natalí N. Guerrero Vargas, Ricardo Jair Ramírez-Carreto, Juan Carlos González-Orozco, Bryan Adair Torres-Hernández, Michelle Valle-Rodríguez, Rosalinda Guevara-Guzmán, and Anahí Chavarría
- Subjects
arcuate nucleus ,hypothalamus ,microglia ,microglial-neuronal interaction ,physiological function ,obesity ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Microglia are highly dynamic cells that have been mainly studied under pathological conditions. The present review discusses the possible implication of microglia as modulators of neuronal electrical responses in physiological conditions and hypothesizes how these cells might modulate hypothalamic circuits in health and during obesity. Microglial cells studied under physiological conditions are highly diverse, depending on the developmental stage and brain region. The evidence also suggests that neuronal electrical activity modulates microglial motility to control neuronal excitability. Additionally, we show that the expression of genes associated with neuron-microglia interaction is down-regulated in obese mice compared to control-fed mice, suggesting an alteration in the contact-dependent mechanisms that sustain hypothalamic arcuate-median eminence neuronal function. We also discuss the possible implication of microglial-derived signals for the excitability of hypothalamic neurons during homeostasis and obesity. This review emphasizes the importance of studying the physiological interplay between microglia and neurons to maintain proper neuronal circuit function. It aims to elucidate how disruptions in the normal activities of microglia can adversely affect neuronal health.
- Published
- 2024
- Full Text
- View/download PDF
15. Hypothalamic POMC neuron-specific knockout of MC4R affects insulin sensitivity by regulating Kir2.1
- Author
-
Hengru Guo, Ying Xin, Saifei Wang, Xiaoning Zhang, Yanqi Ren, Bo Qiao, Hongjiang Li, Jing Wu, Xiao Hao, Lijun Xu, Yushan Yan, and Haohao Zhang
- Subjects
MC4R ,Arcuate nucleus ,Kir2.1 ,Insulin resistance ,Energy expenditure ,Therapeutics. Pharmacology ,RM1-950 ,Biochemistry ,QD415-436 - Abstract
Abstract Background Imbalance in energy regulation is a major cause of insulin resistance and diabetes. Melanocortin-4 receptor (MC4R) signaling at specific sites in the central nervous system has synergistic but non-overlapping functions. However, the mechanism by which MC4R in the arcuate nucleus (ARC) region regulates energy balance and insulin resistance remains unclear. Methods The MC4Rflox/flox mice with proopiomelanocortin (POMC) -Cre mice were crossed to generate the POMC-MC4Rflox/+ mice. Then POMC-MC4Rflox/+ mice were further mated with MC4Rflox/flox mice to generate the POMC-MC4Rflox/flox mice in which MC4R is selectively deleted in POMC neurons. Bilateral injections of 200 nl of AAV-sh-Kir2.1 (AAV-sh-NC was used as control) were made into the ARC of the hypothalamus. Oxygen consumption, carbon dioxide production, respiratory exchange ratio and energy expenditure were measured by using the CLAMS; Total, visceral and subcutaneous fat was analyzed using micro-CT. Co-immunoprecipitation assays (Co-IP) were used to analyze the interaction between MC4R and Kir2.1 in GT1-7 cells. Results POMC neuron-specific ablation of MC4R in the ARC region promoted food intake, impaired energy expenditure, leading to increased weight gain and impaired systemic glucose homeostasis. Additionally, MC4R ablation reduced the activation of POMC neuron, and is not tissue-specific for peripheral regulation, suggesting the importance of its central regulation. Mechanistically, sequencing analysis and Co-IP assay demonstrated a direct interaction of MC4R with Kir2.1. Knockdown of Kir2.1 in POMC neuron-specific ablation of MC4R restored the effect of MC4R ablation on energy expenditure and systemic glucose homeostasis, indicating by reduced body weight and ameliorated insulin resistance. Conclusion Hypothalamic POMC neuron-specific knockout of MC4R affects energy balance and insulin sensitivity by regulating Kir2.1. Kir2.1 represents a new target and pathway that could be targeted in obesity.
- Published
- 2024
- Full Text
- View/download PDF
16. Hypothalamic POMC neuron-specific knockout of MC4R affects insulin sensitivity by regulating Kir2.1
- Author
-
Guo, Hengru, Xin, Ying, Wang, Saifei, Zhang, Xiaoning, Ren, Yanqi, Qiao, Bo, Li, Hongjiang, Wu, Jing, Hao, Xiao, Xu, Lijun, Yan, Yushan, and Zhang, Haohao
- Published
- 2024
- Full Text
- View/download PDF
17. Conditional Deletion of β-Catenin in the Mediobasal Hypothalamus Impairs Adaptive Energy Expenditure in Response to High-Fat Diet and Exacerbates Diet-Induced Obesity.
- Author
-
Rizwan, Mohammed Z., Kamstra, Kaj, Pretz, Dominik, Shepherd, Peter R., Tups, Alexander, and Grattan, David R.
- Subjects
- *
HIGH-fat diet , *REGULATION of body weight , *GENETIC regulation , *SYNAPTIC vesicles , *HYPOTHALAMUS , *CATENINS , *METABOLIC disorders , *INSULIN - Abstract
β-Catenin is a bifunctional molecule that is an effector of the wingless-related integration site (Wnt) signaling to control gene expression and contributes to the regulation of cytoskeleton and neurotransmitter vesicle trafficking. In its former role, β-catenin binds transcription factor 7-like 2 (TCF7L2), which shows strong genetic associations with the pathogenesis of obesity and type-2 diabetes. Here, we sought to determine whether β-catenin plays a role in the neuroendocrine regulation of body weight and glucose homeostasis. Bilateral injections of adeno-associated virus type-2 (AAV2)-mCherry-Cre were placed into the arcuate nucleus of adult male and female β-cateninflox mice, to specifically delete β-catenin expression in the mediobasal hypothalamus (MBH-β-cat KO). Metabolic parameters were then monitored under conditions of low-fat (LFD) and high-fat diet (HFD). On LFD, MBH-β-cat KO mice showed minimal metabolic disturbances, but on HFD, despite having only a small difference in weekly caloric intake, the MBH-β-cat KO mice were significantly heavier than the control mice in both sexes (p < 0.05). This deficit seemed to be due to a failure to show an adaptive increase in energy expenditure seen in controls, which served to offset the increased calories by HFD. Bothmale and female MBH-β-cat KO mice were highly glucose intolerant when on HFD and displayed a significant reduction in both leptin and insulin sensitivity compared with controls. This study highlights a critical role for β-catenin in the hypothalamic circuits regulating body weight and glucose homeostasis and reveals potential mechanisms by which genetic variation in this pathway could impact on development of metabolic disease. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. Long-term Recordings of Arcuate Nucleus Kisspeptin Neurons Across the Mouse Estrous Cycle.
- Author
-
Vas, Szilvia, Wall, Ellen, Zhou, Ziyue, Kalmar, Lajos, Han, Su Young, and Herbison, Allan E
- Subjects
KISSPEPTIN neurons ,GONADOTROPIN ,ESTRUS - Abstract
The arcuate nucleus kisspeptin (ARN
KISS ) neurons represent the GnRH pulse generator that likely drives pulsatile gonadotropin secretion in all mammals. Using an improved GCaMP fiber photometry system enabling long-term continuous recordings, we aimed to establish a definitive profile of ARNKISS neuronal activity across the murine estrous cycle. As noted previously, a substantial reduction in the frequency of ARNKISS neuron synchronization events (SEs) occurs on late proestrus and extends into estrus. The SE amplitude remains constant throughout the cycle. During metestrus, we unexpectedly detected many multipeak SEs where many SEs occurred rapidly, within 160 seconds of each other. By applying a machine learning-based, k-means clustering analysis, we were further able to detect substantial within-stage variability in the patterns of pulse generator activity. Estrous cycle-dependent changes in SE activity occurred around the time of lights on and off. We also find that a mild stressor such as vaginal lavage reduces ARNKISS neuron SE frequency for up to 3 hours. These observations provide a comprehensive account of ARNKISS neuron activity across the estrous cycle, highlight a new pattern of multipeak SE activity, and introduce a new k-means clustering approach for analyzing ARNKISS neuron population behavior. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
19. The temporal and spatial pattern of leptin receptor‐expressing cells in the developing mouse hypothalamus.
- Author
-
Higgins, Matt B. A., Glendining, Kelly A., and Jasoni, Christine L.
- Subjects
- *
LEPTIN , *HYPOTHALAMUS , *PARAVENTRICULAR nucleus , *LEPTIN receptors , *ANIMAL behavior - Abstract
The arcuate nucleus is a crucial hypothalamic brain region involved in regulating body weight homeostasis. Neurons within the arcuate nucleus respond to peripheral metabolic signals, such as leptin, and relay these signals via neuronal projections to brain regions both within and outside the hypothalamus, ultimately causing changes in an animal's behaviour and physiology. There is a substantial amount of evidence to indicate that leptin is intimately involved with the postnatal development of arcuate nucleus melanocortin circuitry. Further, it is clear that leptin signalling directly in the arcuate nucleus is required for circuitry development. However, as leptin receptor long isoform (Leprb) mRNA is expressed in multiple nuclei within the developing hypothalamus, including the postsynaptic target regions of arcuate melanocortin projections, this raises the possibility that leptin also signals in these nuclei to promote circuitry development. Here, we used RT‐qPCR and RNAscope® to reveal the spatio‐temporal pattern of Leprb mRNA in the early postnatal mouse hypothalamus. We found that Leprb mRNA expression increased significantly in the arcuate nucleus, ventromedial nucleus and paraventricular nucleus of the hypothalamus from P8, in concert with the leptin surge. In the dorsomedial nucleus of the hypothalamus, increases in Leprb mRNA were slightly later, increasing significantly from P12. Using duplex RNAscope®, we found Leprb co‐expressed with Sim1, Pou3f2, Mc4r and Bdnf in the paraventricular nucleus at P8. Together, these data suggest that leptin may signal in a subset of neurons postsynaptic to arcuate melanocortin neurons, as well as within the arcuate nucleus itself, to promote the formation of arcuate melanocortin circuitry during the early postnatal period. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
20. Running the Female Power Grid Across Lifespan Through Brain Estrogen Signaling
- Author
-
Ingraham, Holly A, Herber, Candice B, and Krause, William C
- Subjects
Medical Physiology ,Biomedical and Clinical Sciences ,Aging ,Estrogen ,Women's Health ,Neurosciences ,1.1 Normal biological development and functioning ,Neurological ,Brain ,Estrogens ,Female ,Humans ,Hypothalamus ,Longevity ,Neurons ,Signal Transduction ,female physiology ,central estrogen signaling ,hypothalamus ,ventromedial hypothalamus ,arcuate nucleus ,sex-dependent neurocircuits ,reproduction ,menopause ,estrogen receptor alpha ,brain-bone connection ,women's health ,Biological Sciences ,Medical and Health Sciences ,Physiology ,Zoology ,Medical physiology - Abstract
The role of central estrogen in cognitive, metabolic, and reproductive health has long fascinated the lay public and scientists alike. In the last two decades, insight into estrogen signaling in the brain and its impact on female physiology is beginning to catch up with the vast information already established for its actions on peripheral tissues. Using newer methods to manipulate estrogen signaling in hormone-sensitive brain regions, neuroscientists are now identifying the molecular pathways and neuronal subtypes required for controlling sex-dependent energy allocation. However, the immense cellular complexity of these hormone-sensitive brain regions makes it clear that more research is needed to fully appreciate how estrogen modulates neural circuits to regulate physiological and behavioral end points. Such insight is essential for understanding how natural or drug-induced hormone fluctuations across lifespan affect women's health.
- Published
- 2022
21. Mechanism of Arcuate Kisspeptin Neuron Synchronization in Acute Brain Slices From Female Mice.
- Author
-
Morris, Paul G and Herbison, Allan E
- Subjects
KISSPEPTINS ,GLUTAMIC acid ,DYNORPHINS - Abstract
The mechanism by which arcuate kisspeptin (ARN
KISS ) neurons co-expressing glutamate, neurokinin B, and dynorphin intermittently synchronize their activity to drive pulsatile hormone secretion remains unclear in females. In order to study spontaneous synchronization within the ARNKISS neuron network, acute brain slices were prepared from adult female Kiss1-GCaMP6 mice. Analysis of both spontaneous synchronizations and those driven by high frequency stimulation of individual ARNKISS neurons revealed that the network exhibits semi-random emergent excitation dependent upon glutamate signaling through AMPA receptors. No role for NMDA receptors was identified. In contrast to male mice, ongoing tachykinin receptor tone within the slice operated to promote spontaneous synchronizations in females. As previously observed in males, we found that ongoing dynorphin transmission in the slice did not contribute to synchronization events. These observations indicate that a very similar AMPA receptor–dependent mechanism underlies ARNKISS neuron synchronizations in the female mouse supporting the "glutamate two-transition" model for kisspeptin neuron synchronization. However, a potentially important sex difference appears to exist with a more prominent facilitatory role for tachykinin transmission in the female. [ABSTRACT FROM AUTHOR]- Published
- 2023
- Full Text
- View/download PDF
22. Brain Regulation of Feeding and Energy Homeostasis
- Author
-
Affinati, Alison H., Elias, Carol F., Olson, David P., Myers, Martin G., Jr, and Ahima, Rexford S., editor
- Published
- 2023
- Full Text
- View/download PDF
23. Enkephalin-δ opioid receptor signaling partly mediates suppression of LH release during early lactation in rats
- Author
-
Hitomi TSUCHIDA, Marina TAKIZAWA, Miku NONOGAKI, Naoko INOUE, Yoshihisa UENOYAMA, and Hiroko TSUKAMURA
- Subjects
arcuate nucleus ,gnrh neuron ,kisspeptin neuron ,suckling stimulus ,Reproduction ,QH471-489 ,Internal medicine ,RC31-1245 - Abstract
Gonadal function is often suppressed during lactation in mammals including rodents, ruminants, and primates. This suppression is thought to be mostly due to the inhibition of the tonic (pulsatile) release of gonadotropin-releasing hormone (GnRH) and consequent gonadotropin. Accumulating evidence suggests that kisspeptin neurons in the arcuate nucleus (ARC) play a critical role in the regulation of pulsatile GnRH/gonadotropin release, and kisspeptin mRNA (Kiss1) and/or kisspeptin expression in the ARC are strongly suppressed by the suckling stimuli in lactating rats. This study aimed to examine whether the central enkephalin-δ-opioid receptor (DOR) signaling mediates the suckling-induced suppression of luteinizing hormone (LH) release in lactating rats. Central administration of a selective DOR antagonist increased the mean plasma LH levels and baseline of LH pulses in ovariectomized lactating mother rats compared to vehicle-injected control dams on day 8 of lactation without affecting the number of Kiss1-expressing cells and the intensity of Kiss1 mRNA signals in the ARC. Furthermore, the suckling stimuli significantly increased the number of enkephalin mRNA (Penk)-expressing cells and the intensity of Penk mRNA signals in the ARC compared to non-lactating control rats. Collectively, these results suggest that central DOR signaling, at least in part, mediates the suppression of LH release induced by suckling stimuli in lactating rats via indirect and/or direct inhibition of ARC kisspeptin neurons.
- Published
- 2023
- Full Text
- View/download PDF
24. Thyrotropin-Releasing Hormone and Food Intake in Mammals: An Update
- Author
-
Yamili Vargas, Ana Elena Castro Tron, Adair Rodríguez Rodríguez, Rosa María Uribe, Patricia Joseph-Bravo, and Jean-Louis Charli
- Subjects
TRH ,hypothalamus ,arcuate nucleus ,lateral hypothalamus ,nucleus accumbens ,raphe nuclei ,Microbiology ,QR1-502 - Abstract
Thyrotropin-releasing hormone (TRH; pGlu-His-Pro-NH2) is an intercellular signal produced mainly by neurons. Among the multiple pharmacological effects of TRH, that on food intake is not well understood. We review studies demonstrating that peripheral injection of TRH generally produces a transient anorexic effect, discuss the pathways that might initiate this effect, and explain its short half-life. In addition, central administration of TRH can produce anorexic or orexigenic effects, depending on the site of injection, that are likely due to interaction with TRH receptor 1. Anorexic effects are most notable when TRH is injected into the hypothalamus and the nucleus accumbens, while the orexigenic effect has only been detected by injection into the brain stem. Functional evidence points to TRH neurons that are prime candidate vectors for TRH action on food intake. These include the caudal raphe nuclei projecting to the dorsal motor nucleus of the vagus, and possibly TRH neurons from the tuberal lateral hypothalamus projecting to the tuberomammillary nuclei. For other TRH neurons, the anatomical or physiological context and impact of TRH in each synaptic domain are still poorly understood. The manipulation of TRH expression in well-defined neuron types will facilitate the discovery of its role in food intake control in each anatomical scene.
- Published
- 2024
- Full Text
- View/download PDF
25. Electrophysiological Comparison of Definitive Pro-opiomelanocortin Neurons in the Arcuate Nucleus and the Retrochiasmatic Area of Male and Female Mice.
- Author
-
Srour, Nader, Lavoie, Olivier, Khouma, Axelle, Minbashi Moeini, Moein, Plamondon, Julie, Kinkead, Richard, Michael, Natalie J., and Caron, Alexandre
- Subjects
- *
HYPOTHALAMUS , *LEPTIN receptors , *PATCH-clamp techniques (Electrophysiology) , *PROOPIOMELANOCORTIN , *NEURONS , *ELECTROPHYSIOLOGY , *LEPTIN - Abstract
[Display omitted] • 80% of POMC neurons of the retrochiasmatic area (RCA) express the leptin receptor. • 42% of POMC neurons of the arcuate nucleus (ARC) express the leptin receptor. • Leptin excites 14% of definitive POMCRCA and 25% of definitive POMCARC neurons. • Leptin inhibits 10% of definitive POMCRCA and 13% of definitive POMCARC neurons. • POMC neurons from females are less responsive to 100 nM of leptin than males. Pro-opiomelanocortin (POMC)-expressing neurons in the arcuate nucleus of the hypothalamus (ARC) are considered a major site of leptin action. Due to increasing evidence that POMC neurons are highly heterogeneous and indications that the conventional molecular tools to study their functions have important limitations, a reassessment of leptin's effects on definitive POMC neurons is needed. POMC neurons are also expressed in the retrochiasmatic area (RCA), where their function is poorly understood. Furthermore, the response of POMC neurons to leptin in females is largely unknown. Therefore, the present study aimed to determine the differences in leptin responsiveness of POMC neurons in the ARC and the RCA using a mouse model allowing adult-inducible fluorescent labeling. We performed whole-cell patch clamp electrophysiology on 154 POMC neurons from male and female mice. We confirmed and extended the model by which leptin depolarizes POMC neurons, in both the ARC and the RCA. Furthermore, we characterized the electrophysiological properties of an underappreciated subpopulation representing ∼10% of hypothalamic POMC neurons that are inhibited by leptin. We also provide evidence that sex does not appear to be a major determinant of basal properties and leptin responsiveness of POMC neurons, but that females are overall less responsive to leptin compared to males. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
26. Dietary wheat gluten induces astro‐ and microgliosis in the hypothalamus of male mice.
- Author
-
Rizwan, Mohammed Z., Kerbus, Romy, Kamstra, Kaj, Keerthisinghe, Pramuk, and Tups, Alexander
- Subjects
- *
GLIAL fibrillary acidic protein , *GLUTEN , *BARLEY , *HIGH-fat diet , *OATS , *LOW-fat diet , *HYPOTHALAMUS - Abstract
Gluten, which is found in cereals such as wheat, rye and barley, makes up a major dietary component in most western nations, and has been shown to promote body mass gain and peripheral inflammation in mice. In the current study, we investigated the impact of gluten on central inflammation that is typically associated with diet‐induced obesity. While we found no effect of gluten when added to a low‐fat diet (LFD), male mice fed high fat diet (HFD) enriched with gluten increased body mass and adiposity compared with mice fed HFD without gluten. We furthermore found that gluten, when added to the LFD, increases circulating C‐reactive protein levels. Gluten regardless of whether it was added to LFD or HFD led to a profound increase in the number of microglia and astrocytes in the arcuate nucleus of the hypothalamus, as detected by immunohistochemistry for ionised calcium binding adaptor molecule 1 (Iba‐1) and glial fibrillary acidic protein (GFAP), respectively. In mice fed LFD, gluten mimicked the immunogenic effects of HFD exposure and when added to HFD led to a further increase in the number of immunoreactive cells. Taken together, our results confirm a moderate obesogenic effect of gluten when fed to mice exposed to HFD and for the first‐time report gluten‐induced astro‐ and microgliosis suggesting the development of hypothalamic injury in rodents. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
27. Differential voluntary feed intake and whole transcriptome profiling in the hypothalamus of young sheep offered CP and phosphorus-deficient diets
- Author
-
D.J. Innes, N.J. Hudson, S.T. Anderson, D.P. Poppi, and S.P. Quigley
- Subjects
Arcuate nucleus ,Gene expression ,Lateral hypothalamus ,Rumen environment ,Ventromedial hypothalamus ,Animal culture ,SF1-1100 - Abstract
A reduction in voluntary feed intake is observed in ruminants consuming nutrient-deficient diets, such as those with a low CP or P content, and has been attributed to active metabolic regulation, rather than a physical constraint. The hypothalamus is the key integrator of feed intake regulation in mammals. The objectives of this experiment were to (1) establish a model of metabolic feed intake regulation in ruminants consuming diets of variable CP and P content, and (2) determine key biochemical pathways and influential points of regulation within the hypothalamus. Merino wethers [n = 40; 23.7 ± 1.4 kg liveweight (mean ± SD)] were fed one of five dietary treatments (n = 8/treatment) for 63 days in individual pens. The treatments included targeted combinations of high (H) and low (L) CP (110 and 55 g/kg DM) and high and low P (2.5 and 0.7 g/kg DM) with 9 MJ metabolisable energy (ME) per kg DM which were fed ad libitum (UMEI; unrestricted ME intake) resulting in four experimental diets (HCP-HP-UMEI, LCP-HP-UMEI, HCP-LP-UMEI and LCP-LP-UMEI). An additional nutritional treatment (HCP-HP-RMEI) restricted intake of the HCP-HP diet to an equivalent ME intake of wethers consuming the LCP-LP-UMEI treatment. Wethers offered the LCP-HP-UMEI, HCP-LP-UMEI and LCP-LP-UMEI treatments consumed 42, 32 and 49% less total DM (P ≤ 0.05), respectively than the HCP-HP-UMEI treatment, and this was not attributable to any physical limitation of the rumen. Plasma concentrations of urea nitrogen and inorganic phosphate indicated that these nutrient deficiencies were successfully established. To assess potential mechanisms, RNA-seq was conducted on samples from the arcuate nucleus (ARC), ventromedial hypothalamus and lateral hypothalamus of the wethers, yielding a total of 301, 8 and 148 differentially expressed genes across all pairwise comparisons, respectively. The expression of NPY, AGRP and CARTPT, known for their regulatory role in mammalian feed intake regulation, had a similar transcriptional response in the ARC of wethers consuming nutrient-deficient treatments and those consuming a ME-restricted treatment, despite these wethers expressing behaviours indicative of satiated and hungry states, respectively. In addition, genes involved with glycolysis (TPI1), the citric acid cycle (CS, OGDH, GLUD1, GOT1) and oxidative phosphorylation (COX5A, ATP5MC1, ATP5F1B, ATP5MC3) were downregulated in the ARC of wethers fed a nutrient deficient (LCP-LP-UMEI) relative to the non-deficient (HCP-HP-UMEI) treatment. In summary, a model for voluntary feed intake restriction was established to determine genome-wide molecular changes in the hypothalamus of young ruminants.
- Published
- 2023
- Full Text
- View/download PDF
28. Cell-specific transcriptome changes in the hypothalamic arcuate nucleus in a mouse deoxycorticosterone acetate-salt model of hypertension.
- Author
-
Wagner, Valerie A., Deng, Guorui, Claflin, Kristin E., Ritter, McKenzie L., Huxing Cui, Nakagawa, Pablo, Sigmund, Curt D., Morselli, Lisa L., Grobe, Justin L., and Kwitek, Anne E.
- Subjects
NEUROGLIA ,ANGIOTENSIN II ,G protein coupled receptors ,PEPTIDES ,TRANSCRIPTOMES ,G proteins ,BLOOD pressure - Abstract
A common preclinical model of hypertension characterized by low circulating renin is the "deoxycorticosterone acetate (DOCA)-salt" model, which influences blood pressure and metabolism through mechanisms involving the angiotensin II type 1 receptor (AT1R) in the brain. More specifically, AT1R within Agouti-related peptide (AgRP) neurons of the arcuate nucleus of the hypothalamus (ARC) has been implicated in selected effects of DOCA-salt. In addition, microglia have been implicated in the cerebrovascular effects of DOCA-salt and angiotensin II. To characterize DOCA-salt effects upon the transcriptomes of individual cell types within the ARC, we used single-nucleus RNA sequencing (snRNAseq) to examine this region from male C57BL/6J mice that underwent sham or DOCAsalt treatment. Thirty-two unique primary cell type clusters were identified. Sub-clustering of neuropeptide-related clusters resulted in identification of three distinct AgRP subclusters. DOCA-salt treatment caused subtype-specific changes in gene expression patterns associated with AT1R and G protein signaling, neurotransmitter uptake, synapse functions, and hormone secretion. In addition, two primary cell type clusters were identified as resting versus activated microglia, and multiple distinct subtypes of activated microglia were suggested by subcluster analysis. While DOCA-salt had no overall effect on total microglial density within the ARC, DOCA-salt appeared to cause a redistribution of the relative abundance of activated microglia subtypes. These data provide novel insights into cell-specific molecular changes occurring within the ARC during DOCA-salt treatment, and prompt increased investigation of the physiological and pathophysiological significance of distinct subtypes of neuronal and glial cell types. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
29. Glucose‐dependent insulinotropic polypeptide counteracts diet‐induced obesity along with reduced feeding, elevated plasma leptin and activation of leptin‐responsive and proopiomelanocortin neurons in the arcuate nucleus.
- Author
-
Han, Wanxin, Wang, Lei, Ohbayashi, Kento, Takeuchi, Masakazu, O'Farrell, Libbey, Coskun, Tamer, Rakhat, Yermek, Yabe, Daisuke, Iwasaki, Yusaku, Seino, Yutaka, and Yada, Toshihiko
- Subjects
- *
INGESTION , *LEPTIN receptors , *LEPTIN , *GLUCOSE tolerance tests , *WEIGHT gain , *PROOPIOMELANOCORTIN , *BODY weight - Abstract
Aim: To clarify the effects of glucose‐dependent insulinotropic polypeptide (GIP) receptor agonists (GIPRAs) on feeding and body weight. Materials and Methods: Acute and subchronic effects of subcutaneous GIPFA‐085, a long‐acting GIPRA, on blood glucose, food intake, body weight, respiratory exchange ratio and plasma leptin levels were measured in diet‐induced obese (DIO) mice and/or functional leptin‐deficient ob/ob mice. The effects of GIPFA‐085 on the hypothalamic arcuate nucleus (ARC) neurons from lean and DIO mice were studied by measuring cytosolic Ca2+ concentration ([Ca2+]i). Results: Single bolus GIPFA‐085 (30, 300 nmol/kg) dose‐dependently reduced blood glucose in glucose tolerance tests, elevated plasma leptin levels at 0.5‐6 hours and inhibited food intake at 2‐24 hours after injection in DIO mice. Daily GIPFA‐085 (300 nmol/kg) inhibited food intake and increased fat utilization on day 1, and reduced body weight gain on days 3‐12 of treatment in DIO, but not ob/ob, mice. GIPFA‐085 increased [Ca2+]i in the ARC leptin‐responsive and proopiomelanocortin (POMC) neurons. GIPFA‐085 and leptin cooperated to increase [Ca2+]i in ARC neurons and inhibit food intake. Conclusions: GIPFA‐085 acutely inhibits feeding and increases lipid utilization, and sustainedly lowers body weight in DIO mice via mechanisms involving rises in leptin and activation of ARC leptin‐responsive and POMC neurons. This study highlights the therapeutic potential of GIPRAs for treating obesity and diabetes. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
30. Hypothalamus
- Author
-
Kalsbeek, Andries, Fliers, Eric, Pfaff, Donald W., editor, Volkow, Nora D., editor, and Rubenstein, John L., editor
- Published
- 2022
- Full Text
- View/download PDF
31. Gut Microbiome Regulation of Appetite and Role in Neurological Disorders
- Author
-
Singh, Ankita, Verma, Om Prakash, Tripathi, Rajavashisth, Essa, Mohamed, Series Editor, Tripathi, Amit Kumar, editor, and Kotak, Malini, editor
- Published
- 2022
- Full Text
- View/download PDF
32. Insulin‐induced hypoglycaemia suppresses pulsatile luteinising hormone secretion and arcuate Kiss1 cell activation in female mice
- Author
-
McCosh, Richard B, Kreisman, Michael J, Tian, Katherine, Ho, Bryan S, Thackray, Varykina G, and Breen, Kellie M
- Subjects
Neurosciences ,Contraception/Reproduction ,Aetiology ,2.1 Biological and endogenous factors ,Reproductive health and childbirth ,Metabolic and endocrine ,Animals ,Arcuate Nucleus of Hypothalamus ,Fasting ,Female ,Hypoglycemia ,Insulins ,Kisspeptins ,Luteinizing Hormone ,Mice ,Mice ,Transgenic ,Neurons ,Ovariectomy ,Proto-Oncogene Proteins c-fos ,arcuate nucleus ,GnRH ,hypoglycaemia ,LH pulses ,metabolic stress ,Clinical Sciences ,Endocrinology & Metabolism - Abstract
Stress suppresses pulsatile luteinising hormone (LH) secretion in a variety of species, although the mechanism underlying this inhibition of reproductive function remains unclear. Metabolic stress, particularly hypoglycaemia, is a clinically-relevant stress type that is modelled with bolus insulin injection (insulin-induced hypoglycaemia). The present study utilised ovariectomised C57BL/6 mice to test the hypothesis that acute hypoglycaemia suppresses pulsatile LH secretion via central mechanisms. Pulsatile LH secretion was measured in 90-minute sampling periods immediately prior to and following i.p. injection of saline or insulin. The secretion of LH was not altered over time in fed animals or acutely fasted (5 hours) animals following an i.p. saline injection. By contrast, insulin elicited a robust suppression of pulsatile LH secretion in fasted animals, preventing LH pulses in five of six mice. To identify the neuroendocrine site of impairment, a kisspeptin challenge was performed in saline or insulin pre-treated animals in a cross-over design. LH secretion in response to exogenous kisspeptin was not different between animals pre-treated with saline or insulin, indicating normal gonadotrophin-releasing hormone cell and pituitary responses during acute hypoglycaemia. Based on this finding, the effect of insulin-induced hypoglycaemia on arcuate kisspeptin (Kiss1) cell function was determined using c-Fos as a marker of neuronal activation. Insulin caused a significant suppression in the percentage of Kiss1 cells in the arcuate nucleus that contained c-Fos compared to saline-injected controls. Taken together, these data support the hypothesis that insulin-induced hypoglycaemia suppresses pulsatile LH secretion in the female mouse via predominantly central mechanisms, which culminates in the suppression of the arcuate Kiss1 population.
- Published
- 2019
33. Should We Make More Bone or Not, As Told by Kisspeptin Neurons in the Arcuate Nucleus
- Author
-
Herber, Candice B and Ingraham, Holly A
- Subjects
Biomedical and Clinical Sciences ,Clinical Sciences ,Estrogen ,Neurosciences ,Contraception/Reproduction ,Underpinning research ,1.1 Normal biological development and functioning ,Animals ,Arcuate Nucleus of Hypothalamus ,Bone Density ,Bone Development ,Estradiol ,Gene Expression Regulation ,Humans ,Kisspeptins ,Neurons ,Receptors ,Estrogen ,Sex Characteristics ,Signal Transduction ,kisspeptin ,estrogen ,arcuate nucleus ,bone ,Paediatrics and Reproductive Medicine ,Obstetrics & Reproductive Medicine ,Reproductive medicine - Abstract
Since its initial discovery in 2002, the neuropeptide Kisspeptin (Kiss1) has been anointed as the master regulator controlling the onset of puberty in males and females. Over the last several years, multiple groups found that Kiss1 signaling is mediated by the 7TM surface receptor GPCR54. Kiss1 mRNA is highly enriched in the basal medial and lateral subregions of the arcuate nucleus (ARC) in the medial basal hypothalamus. Thus, Kiss1ARC neurons reside in a unique anatomical location ideal for sensing and responding to circulating steroid hormones as well as nutrients. Kiss1 expression is highly responsive to fluctuations of the gonadal hormone, estrogen, with nearly 90% of Kiss1ARC neurons expressing the nuclear hormone estrogen receptor alpha (ERa). Here we review recent research that extends the function of Kiss1ARC neurons beyond the regulation of puberty and highlight their emerging, novel roles in controlling energy allocation, behavioral outputs, and sex-dependent bone remodeling in females. Indeed, some of these previously unknown functions for Kiss1 neurons are quite striking as exemplified by the remarkable increase in bone mass after manipulating estrogen signaling in Kiss1ARC neurons. Taken together, we suggest that Kiss1ARC neurons are highly sensitive to nutritional and hormonal cues that dictate energy utilization and reproduction.
- Published
- 2019
34. Exploring the neurobiology of the premonitory phase of migraine preclinically – a role for hypothalamic kappa opioid receptors?
- Author
-
Caroline M. Kopruszinski, Robson Vizin, Moe Watanabe, Ashley L. Martinez, Luiz Henrique Moreira de Souza, David W. Dodick, Frank Porreca, and Edita Navratilova
- Subjects
Kappa opioid receptors (KOR) ,Hypothalamus ,Arcuate nucleus ,Premonitory phase ,Premonitory symptoms ,Migraine prevention ,Medicine - Abstract
Abstract Background The migraine premonitory phase is characterized in part by increased thirst, urination and yawning. Imaging studies show that the hypothalamus is activated in the premonitory phase. Stress is a well know migraine initiation factor which was demonstrated to engage dynorphin/kappa opioid receptors (KOR) signaling in several brain regions, including the hypothalamus. This study proposes the exploration of the possible link between hypothalamic KOR and migraine premonitory symptoms in rodent models. Methods Rats were treated systemically with the KOR agonist U-69,593 followed by yawning and urination monitoring. Apomorphine, a dopamine D1/2 agonist, was used as a positive control for yawning behaviors. Urination and water consumption following systemic administration of U-69,593 was also assessed. To examine if KOR activation specifically in the hypothalamus can promote premonitory symptoms, AAV8-hSyn-DIO-hM4Di (Gi-DREADD)-mCherry viral vector was microinjected into the right arcuate nucleus (ARC) of female and male KORCRE or KORWT mice. Four weeks after the injection, clozapine N-oxide (CNO) was administered systemically followed by the assessment of urination, water consumption and tactile sensory response. Results Systemic administration of U-69,593 increased urination but did not produce yawning in rats. Systemic KOR agonist also increased urination in mice as well as water consumption. Cell specific Gi-DREADD activation (i.e., inhibition through Gi-coupled signaling) of KORCRE neurons in the ARC also increased water consumption and the total volume of urine in mice but did not affect tactile sensory responses. Conclusion Our studies in rodents identified the KOR in a hypothalamic region as a mechanism that promotes behaviors consistent with clinically-observed premonitory symptoms of migraine, including increased thirst and urination but not yawning. Importantly, these behaviors occurred in the absence of pain responses, consistent with the emergence of the premonitory phase before the headache phase. Early intervention for preventive treatment even before the headache phase may be achievable by targeting the hypothalamic KOR.
- Published
- 2022
- Full Text
- View/download PDF
35. Silencing of hypothalamic FGF11 prevents diet-induced obesity
- Author
-
Jae Hyun Cho, Kyungchan Kim, Han Chae Cho, Jaemeun Lee, and Eun-Kyoung Kim
- Subjects
Fibroblast growth factor 11 ,Neuropeptide Y ,Hypothalamus ,Arcuate nucleus ,Brown adipose tissue ,Thermogenesis ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Fibroblast growth factor 11 (FGF11) is a member of the intracellular fibroblast growth factor family. Here, we report the central role of FGF11 in the regulation of metabolism. Lentiviral injection of Fgf11 shRNA into the arcuate nucleus of the mouse hypothalamus decreased weight gain and fat mass, increased brown adipose tissue thermogenesis, and improved glucose and insulin intolerances under high-fat diet conditions. Fgf11 was expressed in the NPY–expressing neurons, and Fgf11 knockdown considerably decreased Npy expression and projection, leading to increased expression of tyrosine hydroxylase in the paraventricular nucleus. Mechanistically, FGF11 regulated Npy gene expression through the glycogen synthase kinase 3–cAMP response element-binding protein pathway. Our study defines the physiological significance of hypothalamic FGF11 in the regulation of metabolism in response to overnutrition such as high-fat diet.
- Published
- 2022
- Full Text
- View/download PDF
36. Cell-specific transcriptome changes in the hypothalamic arcuate nucleus in a mouse deoxycorticosterone acetate-salt model of hypertension
- Author
-
Valerie A. Wagner, Guorui Deng, Kristin E. Claflin, McKenzie L. Ritter, Huxing Cui, Pablo Nakagawa, Curt D. Sigmund, Lisa L. Morselli, Justin L. Grobe, and Anne E. Kwitek
- Subjects
snRNAseq ,DOCA-salt ,arcuate nucleus ,AgRP neurons ,microglia ,mouse ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
A common preclinical model of hypertension characterized by low circulating renin is the “deoxycorticosterone acetate (DOCA)-salt” model, which influences blood pressure and metabolism through mechanisms involving the angiotensin II type 1 receptor (AT1R) in the brain. More specifically, AT1R within Agouti-related peptide (AgRP) neurons of the arcuate nucleus of the hypothalamus (ARC) has been implicated in selected effects of DOCA-salt. In addition, microglia have been implicated in the cerebrovascular effects of DOCA-salt and angiotensin II. To characterize DOCA-salt effects upon the transcriptomes of individual cell types within the ARC, we used single-nucleus RNA sequencing (snRNAseq) to examine this region from male C57BL/6J mice that underwent sham or DOCA-salt treatment. Thirty-two unique primary cell type clusters were identified. Sub-clustering of neuropeptide-related clusters resulted in identification of three distinct AgRP subclusters. DOCA-salt treatment caused subtype-specific changes in gene expression patterns associated with AT1R and G protein signaling, neurotransmitter uptake, synapse functions, and hormone secretion. In addition, two primary cell type clusters were identified as resting versus activated microglia, and multiple distinct subtypes of activated microglia were suggested by sub-cluster analysis. While DOCA-salt had no overall effect on total microglial density within the ARC, DOCA-salt appeared to cause a redistribution of the relative abundance of activated microglia subtypes. These data provide novel insights into cell-specific molecular changes occurring within the ARC during DOCA-salt treatment, and prompt increased investigation of the physiological and pathophysiological significance of distinct subtypes of neuronal and glial cell types.
- Published
- 2023
- Full Text
- View/download PDF
37. Thermoneutrality effects on developmental programming of obesity.
- Author
-
Desai, Mina, Torsoni, Adrianna S., Torsoni, Marcio A., Eisaghalian, Agnlia, Ferrini, Monica G., and Ross, Michael G.
- Subjects
BODY composition ,LOW birth weight ,GLUCOSE tolerance tests ,FAT ,ADIPOSE tissues - Abstract
Developmental programming studies using mouse models have housed the animals at human thermoneutral temperatures (22°C) which imposes constant cold stress. As this impacts energy homeostasis, we investigated the effects of two housing temperatures (22°C and 30°C) on obesity development in male and female offspring of Control and FR dams. Pregnant mice were housed at 22°C (cold-exposed, CE) or 30°C (thermoneutrality, TN) room temperature. At gestational age e10, mice were fed either an ad libitum diet (Control) or were 30% food-restricted (FR) to produce low birth weight newborns. Following delivery, all dams were fed an ad libitum diet and maternal mice continued to nurse their own pups. At 3 weeks of age, offspring were weaned to an ad libitum diet and housed at similar temperatures as their mothers. Body weights and food intake were monitored. At 6 months of age, body composition and glucose tolerance test were determined, after which, brain and adipose tissue were collected for analysis. FR/CE and FR/TN offspring exhibited hyperphagia and were significantly heavier with increased adiposity as compared to their respective Controls. There was sex-specific effects of temperature in both groups. Male offspring at TN were heavier with increased body fat, though the food intake was decreased as compared to CE males. This was reflected by hypertrophic adipocytes and increased arcuate nucleus satiety/appetite ratio. In contrast, female offspring were not impacted by housing temperature. Thus, unlike female offspring, there was a significant interaction of diet and temperature evident in the male offspring with accentuated adverse effects evident in FR/TN males. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
38. Enkephalin-δ Opioid Receptor Signaling Mediates Glucoprivic Suppression of LH Pulse and Gluconeogenesis in Female Rats.
- Author
-
Tsuchida, Hitomi, Nonogaki, Miku, Takizawa, Marina, Inoue, Naoko, Uenoyama, Yoshihisa, and Tsukamura, Hiroko
- Subjects
ENKEPHALINS ,GLUCONEOGENESIS ,LABORATORY rats - Abstract
Energy availability is an important regulator of reproductive function at various reproductive phases in mammals. Glucoprivation induced by 2-deoxy-D-glucose (2DG), an inhibitor of glucose utilization, as an experimental model of malnutrition suppresses the pulsatile release of GnRH/LH and induces gluconeogenesis. The present study was performed with the aim of examining whether enkephalin-δ-opioid receptor (DOR) signaling mediates the suppression of pulsatile GnRH/LH release and gluconeogenesis during malnutrition. The administration of naltrindole hydrochloride (NTI), a selective DOR antagonist, into the third ventricle blocked the suppression of LH pulses and part of gluconeogenesis induced by IV 2DG administration in ovariectomized rats treated with a negative feedback level of estradiol-17 β (OVX + low E2). The IV 2DG administration significantly increased the number of Penk (enkephalin gene)-positive cells coexpressing fos (neuronal activation marker gene) in the paraventricular nucleus (PVN), but not in the arcuate nucleus (ARC) in OVX + low E2 rats. Furthermore, double in situ hybridization for Penk / Pdyn (dynorphin gene) in the PVN revealed that approximately 35% of the PVN Penk -expressing cells coexpressed Pdyn. Double in situ hybridization for Penk / Crh (corticotropin-releasing hormone gene) in the PVN and Penk / Kiss1 (kisspeptin gene) in the ARC revealed that few Penk -expressing cells coexpressed Crh and Kiss1. Taken together, these results suggest that central enkephalin-DOR signaling mediates the suppression of pulsatile LH release during malnutrition. Moreover, the current study suggests that central enkephalin-DOR signaling is also involved in gluconeogenesis during malnutrition in female rats. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
39. Interleukin-11Rα2 in the hypothalamic arcuate nucleus affects depression-related behaviors and the AKT-BDNF pathway.
- Author
-
Yang, Mengyu, Tian, Shulei, Han, Xiaofeng, Xu, Lihong, You, Jingjing, Wu, Min, Cao, Yifan, Jiang, Yuting, Zheng, Ziteng, Liu, Jing, Meng, Fantao, Li, Chen, and Wang, Xuezhen
- Subjects
- *
PROTEIN kinase B , *BRAIN-derived neurotrophic factor , *HYPOTHALAMUS , *ADENO-associated virus , *PSYCHOLOGICAL stress , *CELLULAR signal transduction - Abstract
• IL-11Rα2 upregulation linked to CUS induced depression. • IL-11Rα2 or IL-11 overexpression produced depression-related behaviours. • Reduced BDNF and AKT signalling due to IL-11Rα2 or IL-11 overexpression. • Potential IL-11Rα2 targeting for therapy. Depression is a widespread emotional disorder with complex pathogenesis. An essential function of the hypothalamus is to regulate emotional disorders. However, further investigation is required to identify the pathogenic genes and molecular mechanisms that contribute to the onset of depression within the hypothalamus. Through RNA-sequencing analysis, this study identified the upregulated expression of interleukin-11 receptor alpha 2 (IL-11Rα2) in the hypothalamus of mice with chronic unpredictable stress (CUS)-induced depression. This substantial increase in IL-11Rα2, not IL-11Rα1 expression levels in the hypothalamus under the influence of CUS was found to be associated with depression-related behaviors. We further showed that IL-11Rα2 is expressed in the arcuate nucleus (ARC) proopiomelanocortin (POMC) neurons of the hypothalamus. Male and female mice exhibited behaviors association with depression, when IL-11Rα2 or its ligand IL-11 was overexpressed in the ARC POMC neurons through the action of an adeno-associated virus. In addition, reductions in the expression levels of proteins involved in the protein kinase B signaling pathways and brain-derived neurotrophic factor were observed upon overexpression of IL-11Rα2 in the hypothalamic ARC. This study emphasizes the importance of IL-11Rα2 in the hypothalamus ARC in the development of depression, and presents it as a potential novel target for depression treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
40. Voluntary exercise suppresses inflammation and improves insulin resistance in the arcuate nucleus and ventral tegmental area in mice on a high-fat diet.
- Author
-
Sasaki, Tomoyuki, Sugiyama, Mariko, Kuno, Mitsuhiro, Miyata, Takashi, Kobayashi, Tomoko, Yasuda, Yoshinori, Onoue, Takeshi, Takagi, Hiroshi, Hagiwara, Daisuke, Iwama, Shintaro, Suga, Hidetaka, Banno, Ryoichi, and Arima, Hiroshi
- Subjects
- *
EXERCISE physiology , *TUMOR necrosis factors , *INSULIN resistance , *HIGH-fat diet , *WEIGHT gain - Abstract
• We investigated the effects of exercise on HFD-induced inflammation in the CNS. • HFD induced inflammation with microglial activation in the ARC and VTA. • The voluntary exercise suppressed HFD-induced inflammation in the ARC and VTA. • Insulin resistance in the ARC and VTA was improved with reduction of inflammation. • The voluntary exercise may have a direct effect on reducing inflammation in the ARC. A high-fat diet (HFD) causes inflammation with an increase in microglial activity in the hypothalamic arcuate nucleus (ARC) and ventral tegmental area (VTA), resulting in insulin resistance in both regions. This leads to a deterioration in glucose and energy metabolism. The effect of voluntary exercise on HFD-induced inflammation in the central nervous system (CNS) remains unclear. To clarify the effects of voluntary exercise on the CNS, 8-week-old male C57BL6 mice were fed a chow diet (CHD) or HFD for 4 weeks; each group was further divided into running exercise (EX+) on a wheel and no exercise (EX-) groups. The expression of the inflammatory cytokine, tumor necrosis factor alpha (TNFα), in the ARC and VTA was significantly increased in the HFD/EX- group, with an increase of microglial activity noted, compared to the CHD/EX- group. The expression of TNFα was significantly suppressed, with a decrease of microglial activity, in the HFD/EX+ compared to HFD/EX- group. Insulin resistance in the ARC and VTA was improved with the suppression of TNFα expression. The HFD/EX- group showed significant weight gain and impaired glucose metabolism compared to the CHD/EX- group. The HFD/EX+ group showed an improvement in glucose and energy metabolism compared to the HFD/EX- group. In addition, voluntary wheel running suppressed HFD-induced inflammation in the ARC, with a decrease in microglial activity observed independently of weight changes. Our data suggest that voluntary exercise prevents obesity and improves glucose metabolism by suppressing inflammation in the ARC and VTA under HFD conditions. [Display omitted] [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
41. Super-Obese Patient-Derived iPSC Hypothalamic Neurons Exhibit Obesogenic Signatures and Hormone Responses
- Author
-
Rajamani, Uthra, Gross, Andrew R, Hjelm, Brooke E, Sequeira, Adolfo, Vawter, Marquis P, Tang, Jie, Gangalapudi, Vineela, Wang, Yizhou, Andres, Allen M, Gottlieb, Roberta A, and Sareen, Dhruv
- Subjects
Pharmacology and Pharmaceutical Sciences ,Biomedical and Clinical Sciences ,Nutrition ,Human Genome ,Stem Cell Research - Induced Pluripotent Stem Cell ,Genetics ,Obesity ,Stem Cell Research - Induced Pluripotent Stem Cell - Human ,Stem Cell Research ,Neurosciences ,2.1 Biological and endogenous factors ,1.1 Normal biological development and functioning ,Metabolic and endocrine ,Cancer ,Body Mass Index ,Brain ,Cell Differentiation ,Female ,Ghrelin ,Humans ,Induced Pluripotent Stem Cells ,Leptin ,Male ,Neurons ,Obesity ,Morbid ,Signal Transduction ,arcuate nucleus ,hypothalamic neurons ,iPSC ,obesity ,Biological Sciences ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences - Abstract
The hypothalamus contains neurons that integrate hunger and satiety endocrine signals from the periphery and are implicated in the pathophysiology of obesity. The limited availability of human hypothalamic neurons hampers our understanding of obesity disease mechanisms. To address this, we generated human induced pluripotent stem cells (hiPSCs) from multiple normal body mass index (BMI; BMI ≤ 25) subjects and super-obese (OBS) donors (BMI ≥ 50) with polygenic coding variants in obesity-associated genes. We developed a method to reliably differentiate hiPSCs into hypothalamic-like neurons (iHTNs) capable of secreting orexigenic and anorexigenic neuropeptides. Transcriptomic profiling revealed that, although iHTNs maintain a fetal identity, they respond appropriately to metabolic hormones ghrelin and leptin. Notably, OBS iHTNs retained disease signatures and phenotypes of high BMI, exhibiting dysregulated respiratory function, ghrelin-leptin signaling, axonal guidance, glutamate receptors, and endoplasmic reticulum (ER) stress pathways. Thus, human iHTNs provide a powerful platform to study obesity and gene-environment interactions.
- Published
- 2018
42. Actin related protein 2/3 complex subunit 1 up‐regulation in the hypothalamus prevents high‐fat diet induced obesity.
- Author
-
Ni, Weimin, Zhang, Jie, Wang, Bing, Liang, Feng, Bao, Long, Li, Pengfei, and Fang, Yan
- Subjects
- *
LEPTIN , *HIGH-fat diet , *ACTIN , *HYPOTHALAMUS , *TRANSCRIPTION factors , *MICROFILAMENT proteins - Abstract
Obesity is a major health crisis in the modern society. Studies have shown that the consumption of a high‐fat diet (HFD) induces hypothalamic inflammation and leptin resistance, which consequently favours body mass gain. Actin related protein 2/3 complex subunit 1 (ARPC1B), an actin‐binding protein, is highly expressed in immune cells. Recent studies have shown that ARPC1B has a certain anti‐inflammatory effect. While ARPC1B expression is decreased in the hypothalamus of mice fed a HFD, the role of ARPC1B in HFD‐induced obesity remains unclear. Thus, we investigated whether ARPC1B up‐regulation in the hypothalamic arcuate nucleus (ARC) could inhibit the development of obesity. Herein, ARPC1B overexpression lentiviral particles were stereotaxically injected into the ARC of male C57BL/6J mice (7 weeks old) fed with HFD. Overexpression of ARPC1B in the hypothalamic ARC attenuated HFD‐induced ARC inflammation, reduced body‐weight gain and feed efficiency. Furthermore, up‐regulation of ARC ARPC1B improved the glucose tolerance and reduced subcutaneous/epididymal fat mass accumulation, which decreased the serum total cholesterol, serum triglyceride and leptin levels. In addition, upon ARPC1B overexpression in the hypothalamic ARC, intraperitoneal injection of leptin increased the phosphorylation level of signal transducer and activator of transcription 3 (STAT3), an important transcription factor for leptin's action, in the ARC of obese mice. Accordingly, we suggest that up‐regulation of ARPC1B in the hypothalamic ARC may improve the HFD‐induced hypothalamic inflammation and leptin resistance. Our findings demonstrate that ARPC1B is a promising target for the treatment of diet‐induced obesity. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
43. The Effects of Insulin on Spike Activity of the Suprachiasmatic Nucleus Neurones and Functional State of Afferent Inputs from the Arcuate Nucleus in Rats.
- Author
-
Inyushkin, A. N., Mistryugov, K. A., Ledyaeva, O. V., Romanova, I. D., Isakova, T. S., and Inyushkin, A. A.
- Subjects
- *
NEURONS , *SUPRACHIASMATIC nucleus , *HYPOTHALAMUS , *AFFERENT pathways , *INSULIN , *ACTION potentials , *RATS , *CLOCK genes - Abstract
In experiments on the sagittal hypothalamic slices of male Wistar rats, the effects of 15 nM insulin on the level of spike activity, parameters of spike information coding by the suprachiasmatic nucleus neurones, and functional state of afferent inputs to the neurones from the arcuate nucleus were studied. Application of insulin induced a decrease in the frequency of action potential generation and an increase in the entropy of interspike interval distribution in 33.3% of neurones recorded; in 12% of cells, the responses of opposite direction were found; in the remaining 54.7% neurones, spike activity did not change. The responses of the entropy of interspike interval distribution suggest the related changes in a degree of interspike interval irregularity induced by insulin. To characterise afferent inputs to the suprachiasmatic nucleus neurones from the arcuate nucleus, electrophysiological technique of the construction and analysis of the peristimulus time histogram (PSTH) was used. Statistically significant responses to the stimulation of the arcuate nucleus were recorded in 24 of 38 neurones of the suprachiasmatic nucleus. In 6 of the neurones, the responses were in the form of a short-latency (< 20 ms) excitation, in 1 neurone in the form of a long-latency excitation, in 6 neurones in the form of a short-latency inhibition; in 11 neurones complex two- or three-phase responses in the form of different compositions of excitation and inhibition were observed. Application of 15 nM insulin induced a qualitative transformation of the responses (disappearing of the initial responses or emergence of new responses) in 5 neurones initially responded to stimulation, and in 1 neurone initially not responded to stimulation of the arcuate nucleus. Statistically significant changes in the latency or duration of the responses in the presence of insulin were not found. The results of the study suggest the ability of insulin to influence the activity level and the spike code of a respectively numerous population of neurones in the suprachiasmatic nucleus circadian oscillator as well as modulate the functional state of afferent inputs to the circadian oscillator from hypothalamic arcuate nucleus playing an important role in the control of appetite and metabolism. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
44. A morphometric analysis of the circumolivary fiber bundle of the human brainstem.
- Author
-
Brendel, Victor A., Schmeisser, Michael J., and Schumann, Sven
- Subjects
MEDULLA oblongata ,RETICULAR formation ,BRAIN stem ,FIBERS ,ANATOMICAL variation ,KNEE - Abstract
The circumolivary fiber bundle (CFB) is considered to be an anatomical variation, which can be found on the surface of the human medulla oblongata. The macroscopical fiber bundle runs downwards from either the anterior median fissure, the pyramid, or both, around the inferior pole of the olive and turns upwards to reach the restiform body of the inferior cerebellar peduncle. Multiple fiber systems feed the constitution of the CFB (collateral corticospinal fibers, fibers connecting to the reticular formation, anterior external arcuate fibers). With this examination we provide a systematic analysis of the frequency of occurrence (6.14%), size, and laterality of the CFB. Including all three fiber bundle parts (descending part, genu, and ascending part), the left-sided sizes were increased. Likewise, the appearance of an unilateral left-sided CFB could be detected in more than 60% of our cases. Our morphometrical analysis currently covers the largest sample of investigated brainstem sides (n = 489) so far. This investigation should widen the perspective on how anatomists, neuroradiologists, and neurosurgeons expect the anterolateral surface of the human medulla oblongata. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
45. The Neuroanatomical Organization of Hypothalamic Feeding Circuits
- Author
-
Gruber, Tim, Woods, Stephen C., Tschöp, Matthias H., García-Cáceres, Cristina, Ludwig, Mike, Series Editor, Campbell, Rebecca, Series Editor, Grinevich, Valery, editor, and Dobolyi, Árpád, editor
- Published
- 2021
- Full Text
- View/download PDF
46. The molecular mechanism of Xiaoyaosan in treating major depressive disorder: Integrated analysis of DNA methylation and RNA sequencing of the arcuate nucleus in rats.
- Author
-
Wang R, Zou T, Wang Y, Liu Y, Mo X, Chen Y, Li X, and Chen J
- Abstract
Ethnopharmacological Relevance: Xiaoyaosan, a classic Chinese herbal formula, exhibits promising antidepressant effects. However, its specific antidepressant mechanisms remain incompletely understood. Previous studies have highlighted the significant role of DNA methylation in the pathogenesis of major depressive disorder (MDD). Yet, whether the effects of Xiaoyaosan are linked to DNA methylation and its regulation remains unclear., Aim of the Study: This study aims to explore and verify the molecular mechanism of Xiaoyaosan in treating MDD via integrated analysis of DNA methylation and RNA sequencing., Materials and Methods: In this study, a chronic unpredictable mild stress (CUMS) model was established to induce MDD in rats, which were subsequently orally treated with Xiaoyaosan, with fluoxetine as a positive control. Antidepressant effects were assessed by the open field test, sucrose preference test, and forced swimming test. Whole-genome bisulfite sequencing (WGBS) and bulk RNA sequencing were performed in the arcuate nucleus of hypothalamus to assess methylation changes and identify differentially expressed genes. Bioinformatics analyses were conducted to explore methylation alterations, RNA sequencing profiles, and their shared epigenetic as well as gene expression changes, to identify candidate genes. Finally, RT-PCR was used to validate the key differential genes., Results: Xiaoyaosan effectively reversed depressive-like behaviors. Further, Xiaoyaosan treatment involved multiple epigenetic modifications. The results of differentially methylated genes showed that there were 1353 overlapped genes between M-vs-C-hypo gene and X-vs-M-hyper gene, 5326 overlapped genes between M-vs-C-hyper gene and X-vs-M-hypo gene. GO and KEGG enrichment analyses indicated these intersecting genes were involved in biological regulation, transcription factors, appetite and endocrine control systems, etc. The analysis of differentially expressed genes from RNA sequencing revealed that there were 25 overlapping genes between the M vs C hypomethylated group and the X vs M hypermethylated group, while 81 overlapping genes were identified between the M vs C hypermethylated group and the X vs M hypomethylated group. Those differential genes regulated by methylation enriched in processes related to brain and neuronal growth, neuropeptide and hormone activation, as well as biological processes and molecular functions associated with protein translation, synthesis, transport, and localization. The integrated analysis of DNA methylation and RNA sequencing screened 14 potentially differential genes, which were associated with appetite regulation, energy metabolism, and neuroreceptor ligands. PCR verification found that Lmx1b, Abcc5, Gpc3 and Cfb showed statistical differences., Conclusions: The antidepressant mechanism of Xiaoyaosan involves the biological regulation in the arcuate nucleus of hypothalamus, including transcription factors, neurotransmitter regulation, neural development, appetite regulation peptides, and endocrine control systems. The methylation level and regulation at the gene locus of Lmx1b, Abcc5, Gpc3, and Cfb may play a key role in the treatment of Xiaoyaosan. These findings provide new insights into the therapeutic mechanisms of Xiaoyaosan., Competing Interests: Declaration of competing interest The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024. Published by Elsevier B.V.)
- Published
- 2024
- Full Text
- View/download PDF
47. Selective Colocalization of GHSR and GLP-1R in a Subset of Hypothalamic Neurons and Their Functional Interaction.
- Author
-
Aguggia J, Fernandez G, Cassano D, Mustafá ER, Rodríguez SS, Cantel S, Fehrentz JA, Raingo J, Schiöth HB, Habib AM, De Francesco PN, and Perello M
- Subjects
- Animals, Mice, Humans, Male, Ghrelin metabolism, Mice, Inbred C57BL, Mice, Transgenic, Receptors, Ghrelin metabolism, Receptors, Ghrelin genetics, Neurons metabolism, Neurons drug effects, Glucagon-Like Peptide-1 Receptor metabolism, Glucagon-Like Peptide-1 Receptor genetics, Hypothalamus metabolism
- Abstract
The GH secretagogue receptor (GHSR) and the glucagon-like peptide-1 receptor (GLP-1R) are G protein-coupled receptors with critical, yet opposite, roles in regulating energy balance. Interestingly, these receptors are expressed in overlapping brain regions. However, the extent to which they target the same neurons and engage in molecular crosstalk remains unclear. To explore the potential colocalization of GHSR and GLP-1R in specific neurons, we performed detailed mapping of cells positive for both receptors using GHSR-eGFP reporter mice or wild-type mice infused with fluorescent ghrelin, alongside an anti-GLP-1R antibody. We found that GHSR+ and GLP-1R+ cells are largely segregated in the mouse brain. The highest overlap was observed in the hypothalamic arcuate nucleus, where 15% to 20% of GHSR+ cells were also GLP-1R+ cells. Additionally, we examined RNA-sequencing datasets from mouse and human brains to assess the fraction and distribution of neurons expressing both receptors, finding that double-positive Ghsr+/Glp1r+ cells are highly segregated, with a small subset of double-positive Ghsr+/Glp1r+ cells representing <10% of all Ghsr+ or Glp1r+ cells, primarily enriched in the hypothalamus. Furthermore, we conducted functional studies using patch-clamp recordings in a heterologous expression system to assess potential crosstalk in regulating presynaptic calcium channels. We provide the first evidence that liraglutide-evoked GLP-1R activity inhibits presynaptic channels, and that the presence of one GPCR attenuates the inhibitory effects of ligand-evoked activity mediated by the other on presynaptic calcium channels. In conclusion, while GHSR and GLP-1R can engage in molecular crosstalk, they are largely segregated across most neuronal types within the brain., (© The Author(s) 2024. Published by Oxford University Press on behalf of the Endocrine Society. All rights reserved. For commercial re-use, please contact reprints@oup.com for reprints and translation rights for reprints. All other permissions can be obtained through our RightsLink service via the Permissions link on the article page on our site—for further information please contact journals.permissions@oup.com. See the journal About page for additional terms.)
- Published
- 2024
- Full Text
- View/download PDF
48. The roles of DNA methylation and hydroxymethylation at short interspersed nuclear elements in the hypothalamic arcuate nucleus during puberty
- Author
-
Yihang Shen, Hongchao Zhao, Lei Zhang, Yanping Hu, Li Cai, Jun Li, and Shasha Zhou
- Subjects
arcuate nucleus ,DNA methylation ,DNA hydroxymethylation ,puberty ,retroelement ,short interspersed nuclear elements ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Puberty is the gateway to adult reproductive competence, encompassing a suite of complex, integrative, and coordinated changes in neuroendocrine functions. However, the regulatory mechanisms of transcriptional reprogramming in the arcuate nucleus (ARC) during onset of puberty are still not fully understood. To understand the role of epigenetics in regulating gene expression, mouse hypothalamic ARCs were isolated at 4 and 8 weeks, and the transcriptome, DNA hydroxymethylation, DNA methylation, and chromatin accessibility were assessed via RNA sequencing (RNA-seq), reduced representation bisulfite sequencing (RRBS-seq), reduced representation hydroxymethylation profiling (RRHP)-seq, and assay for transposase-accessible chromatin (ATAC-seq), respectively. The overall DNA hydroxymethylation and DNA methylation changes in retroelements (REs) were associated with gene expression modeling for puberty in the ARC. We focused on analyzing DNA hydroxymethylation and DNA methylation at two short interspersed nuclear elements (SINEs) located on the promoter of the 5-hydroxytryptamine receptor 6 (Htr6) gene and the enhancer of the KISS-1 metastasis suppressor (Kiss1) gene and investigated their regulatory roles in gene expression. Our data uncovered a novel epigenetic mechanism by which SINEs regulate gene expression during puberty.
- Published
- 2021
- Full Text
- View/download PDF
49. Kisspeptin-neuron control of LH pulsatility and ovulation
- Author
-
Harvey Stevenson, Samuel Bartram, Mikaela Maria Charalambides, Sruthi Murthy, Theo Petitt, Anjali Pradeep, Owen Vineall, Ikenna Abaraonye, Amelia Lancaster, Kanyada Koysombat, Bijal Patel, and Ali Abbara
- Subjects
kisspeptin ,ovulation ,oestrogen receptor ,KNDy neurons ,arcuate nucleus ,anteroventral periventricular nucleus ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 - Abstract
Feedback from oestradiol (E2) plays a critical role in the regulation of major events in the physiological menstrual cycle including the release of gonadotrophins to stimulate follicular growth, and the mid-cycle luteinising hormone (LH) surge that leads to ovulation. E2 predominantly exerts its action via oestrogen receptor-alpha (ERα), however, as gonadotrophin releasing hormone (GnRH) neurons lack ERα, E2-feedback is posited to be indirectly mediated via upstream neurons. Kisspeptin (KP) is a neuropeptide expressed in hypothalamic KP-neurons that control GnRH secretion and plays a key role in the central mechanism regulating the hypothalamic-pituitary-gonadal (HPG) axis. In the rodent arcuate (ARC) nucleus, KP is co-expressed with Neurokinin B and Dynorphin; and thus, these neurons are termed ‘Kisspeptin-Neurokinin B-Dynorphin’ (KNDy) neurons. ARC KP-neurons function as the ‘GnRH pulse generator’ to regulate GnRH pulsatility, as well as mediating negative feedback from E2. A second KP neuronal population is present in the rostral periventricular area of the third ventricle (RP3V), which includes anteroventral periventricular (AVPV) nucleus and preoptic area neurons. These RP3V KP-neurons mediate positive feedback to induce the mid-cycle luteinising hormone (LH) surge and subsequent ovulation. Here, we describe the role of KP-neurons in these two regions in mediating this differential feedback from oestrogens. We conclude by considering reproductive diseases for which exploitation of these mechanisms could yield future therapies.
- Published
- 2022
- Full Text
- View/download PDF
50. Tamoxifen-mediated changes in estrogen-dependent gene expression in hypothalamic neurons responsible for hot flashes
- Author
-
Misquez, Amanda Marie
- Subjects
Physiology ,Endocrinology ,Neurosciences ,Arcuate nucleus ,estrogen-receptor-alpha ,gene expression ,hot flash ,Medial Preoptic Area ,Tamoxifen - Abstract
Tamoxifen (Tmx) is a selective estrogen receptor modulator widely used as a chemotherapeutic drug in the treatment regimen for estrogen-sensitive breast cancer. Tmx treatment leads to many side effects; the most prominently reported side effect is hot flashes. Hot flashes are episodes of thermodysregulation and have been shown to be caused by estrogen-receptor α (ERα) signaling in the hypothalamus. In this thesis, we delivered a custom, bi-functional fluorescent reporter to three estrogen-sensitive nuclei in the hypothalamus that regulate body temperature: the medial preoptic area (MPA), the arcuate nucleus (ARC), and the ventrolateral region of the ventromedial hypothalamus (VMHvl). The reporter labels estrogen-sensitive neurons that express ERα and reports whether neurons alter estrogen-dependent gene transcription and expression in response to Tmx treatment. Upon delivering the fluorescent reporter to the three thermoregulatory and estrogen-sensitive regions of interest (the MPA, ARC, and VMHvl), we observed that Tmx treatment in Esr1 Cre and Kiss1 Cre mice showed no significant differences in ERE-dependent gene expression in the MPA, ARC, and VMHvl compared to control, Oil-treated mice. Therefore, we are unable to detect Tmx-induced changes in gene expression in estrogen-sensitive neurons of the MPA, ARC, or VMHvl. We conclude that Tmx does not alter estrogen-dependent gene expression in the hypothalamus under the conditions we used. As our Tamoxifen treatment paradigm is sufficient to induce thermodysregulation, this implies the intriguing possibility that Tamoxifen induces hot flashes without widespread changes in hypothalamic gene expression.
- Published
- 2023
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.