Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 155
1.
ACS Chem Neurosci ; 15(11): 2350-2358, 2024 Jun 05.
Article En | MEDLINE | ID: mdl-38757688

Growth hormone-releasing hormone (Ghrh) neurons in the dorsomedial ventromedial hypothalamic nucleus (VMNdm) express the metabolic transcription factor steroidogenic factor-1 and hypoglycemia-sensitive neurochemicals of diverse chemical structures, transmission modes, and temporal signaling profiles. Ghrh imposes neuromodulatory control of coexpressed transmitters. Multiple metabolic sensory mechanisms are employed in the brain, including screening of the critical nutrient glucose or the energy currency ATP. Here, combinatory laser-catapult-microdissection/single-cell multiplex qPCR tools were used to investigate whether these neurons possess molecular machinery for monitoring cellular metabolic status and if these biomarkers exhibit sex-specific sensitivity to insulin-induced hypoglycemia. Data show that hypoglycemia up- (male) or downregulated (female) Ghrh neuron glucokinase (Gck) mRNA; Ghrh gene silencing decreased baseline and hypoglycemic patterns of Gck gene expression in each sex. Ghrh neuron glucokinase regulatory protein (Gckr) transcript levels were respectively diminished or augmented in hypoglycemic male vs female rats; this mRNA profile was decreased by Ghrh siRNA in both sexes. Gene transcripts encoding catalytic alpha subunits of the energy monitor 5-AMP-activated protein kinase (AMPK), i.e., Prkaa1 and 2, were increased by hypoglycemia in males, yet only the former mRNA was hypoglycemia-sensitive in females. Ghrh siRNA downregulated baseline and hypoglycemia-associated Prkaa subunit mRNAs in males but elicited divergent changes in Prkaa2 transcripts in eu- vs hypoglycemic females. Results provide unique evidence that VMNdm Ghrh neurons express the characterized metabolic sensor biomarkers glucokinase and AMPK and that the corresponding gene profiles exhibit distinctive sex-dimorphic transcriptional responses to hypoglycemia. Data further document Ghrh neuromodulation of baseline and hypoglycemic transcription patterns of these metabolic gene profiles.


Growth Hormone-Releasing Hormone , Hypoglycemia , Neurons , RNA, Messenger , Rats, Sprague-Dawley , Sex Characteristics , Ventromedial Hypothalamic Nucleus , Animals , Growth Hormone-Releasing Hormone/metabolism , Growth Hormone-Releasing Hormone/genetics , Hypoglycemia/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Male , Female , Neurons/metabolism , Rats , RNA, Messenger/metabolism , Glucokinase/metabolism , Glucokinase/genetics , Dorsomedial Hypothalamic Nucleus/metabolism
2.
Cell Metab ; 36(2): 377-392.e11, 2024 02 06.
Article En | MEDLINE | ID: mdl-38194970

Recent studies have shown that the hypothalamus functions as a control center of aging in mammals that counteracts age-associated physiological decline through inter-tissue communications. We have identified a key neuronal subpopulation in the dorsomedial hypothalamus (DMH), marked by Ppp1r17 expression (DMHPpp1r17 neurons), that regulates aging and longevity in mice. DMHPpp1r17 neurons regulate physical activity and WAT function, including the secretion of extracellular nicotinamide phosphoribosyltransferase (eNAMPT), through sympathetic nervous stimulation. Within DMHPpp1r17 neurons, the phosphorylation and subsequent nuclear-cytoplasmic translocation of Ppp1r17, regulated by cGMP-dependent protein kinase G (PKG; Prkg1), affect gene expression regulating synaptic function, causing synaptic transmission dysfunction and impaired WAT function. Both DMH-specific Prkg1 knockdown, which suppresses age-associated Ppp1r17 translocation, and the chemogenetic activation of DMHPpp1r17 neurons significantly ameliorate age-associated dysfunction in WAT, increase physical activity, and extend lifespan. Thus, these findings clearly demonstrate the importance of the inter-tissue communication between the hypothalamus and WAT in mammalian aging and longevity control.


Aging , Longevity , Mice , Animals , Neurons/metabolism , Synaptic Transmission , Adipose Tissue/metabolism , Hypothalamus/metabolism , Dorsomedial Hypothalamic Nucleus/metabolism , Mammals/metabolism , Cyclic GMP-Dependent Protein Kinase Type I/metabolism
3.
J Neurosci ; 42(21): 4267-4277, 2022 05 25.
Article En | MEDLINE | ID: mdl-35440490

Torpor is a naturally occurring, hypometabolic, hypothermic state engaged by a wide range of animals in response to imbalance between the supply and demand for nutrients. Recent work has identified some of the key neuronal populations involved in daily torpor induction in mice, in particular, projections from the preoptic area of the hypothalamus to the dorsomedial hypothalamus (DMH). The DMH plays a role in thermoregulation, control of energy expenditure, and circadian rhythms, making it well positioned to contribute to the expression of torpor. We used activity-dependent genetic TRAPing techniques to target DMH neurons that were active during natural torpor bouts in female mice. Chemogenetic reactivation of torpor-TRAPed DMH neurons in calorie-restricted mice promoted torpor, resulting in longer and deeper torpor bouts. Chemogenetic inhibition of torpor-TRAPed DMH neurons did not block torpor entry, suggesting a modulatory role for the DMH in the control of torpor. This work adds to the evidence that the preoptic area of the hypothalamus and the DMH form part of a circuit within the mouse hypothalamus that controls entry into daily torpor.SIGNIFICANCE STATEMENT Daily heterotherms, such as mice, use torpor to cope with environments in which the supply of metabolic fuel is not sufficient for the maintenance of normothermia. Daily torpor involves reductions in body temperature, as well as active suppression of heart rate and metabolism. How the CNS controls this profound deviation from normal homeostasis is not known, but a projection from the preoptic area to the dorsomedial hypothalamus has recently been implicated. We demonstrate that the dorsomedial hypothalamus contains neurons that are active during torpor. Activity in these neurons promotes torpor entry and maintenance, but their activation alone does not appear to be sufficient for torpor entry.


Dorsomedial Hypothalamic Nucleus , Torpor , Animals , Female , Mice , Dorsomedial Hypothalamic Nucleus/metabolism , Hypothalamus/metabolism , Neurons/physiology , Preoptic Area , Torpor/physiology
4.
Mol Metab ; 55: 101401, 2022 01.
Article En | MEDLINE | ID: mdl-34823066

OBJECTIVE: The paraventricular nucleus of hypothalamus (PVN), an integrative center in the brain, orchestrates a wide range of physiological and behavioral responses. While the PVN melanocortin 4 receptor (MC4R) signaling (PVNMC4R+) is involved in feeding regulation, the neuroanatomical organization of PVNMC4R+ connectivity and its role in other physiological regulations are incompletely understood. Here we aimed to better characterize the input-output organization of PVNMC4R+ neurons and test their physiological functions beyond feeding. METHODS: Using a combination of viral tools, we mapped PVNMC4R+ circuits and tested the effects of chemogenetic activation of PVNMC4R+ neurons on thermoregulation, cardiovascular control, and other behavioral responses beyond feeding. RESULTS: We found that PVNMC4R+ neurons innervate many different brain regions that are known to be important not only for feeding but also for neuroendocrine and autonomic control of thermoregulation and cardiovascular function, including but not limited to the preoptic area, median eminence, parabrachial nucleus, pre-locus coeruleus, nucleus of solitary tract, ventrolateral medulla, and thoracic spinal cord. Contrary to these broad efferent projections, PVNMC4R+ neurons receive monosynaptic inputs mainly from other hypothalamic nuclei (preoptic area, arcuate and dorsomedial hypothalamic nuclei, supraoptic nucleus, and premammillary nucleus), the circumventricular organs (subfornical organ and vascular organ of lamina terminalis), the bed nucleus of stria terminalis, and the parabrachial nucleus. Consistent with their broad efferent projections, chemogenetic activation of PVNMC4R+ neurons not only suppressed feeding but also led to an apparent increase in heart rate, blood pressure, and brown adipose tissue temperature. These physiological changes accompanied acute transient hyperactivity followed by hypoactivity and resting-like behavior. CONCLUSIONS: Our results elucidate the neuroanatomical organization of PVNMC4R+ circuits and shed new light on the roles of PVNMC4R+ pathways in autonomic control of thermoregulation, cardiovascular function, and biphasic behavioral activation.


Paraventricular Hypothalamic Nucleus/metabolism , Receptor, Melanocortin, Type 4/metabolism , Animals , Body Temperature Regulation/physiology , Brain/metabolism , Dorsomedial Hypothalamic Nucleus/metabolism , Gene Knock-In Techniques/methods , Hypothalamus/metabolism , Mice, 129 Strain , Mice, Inbred C57BL , Neurons/metabolism , Receptor, Melanocortin, Type 4/physiology , Spinal Cord/metabolism
5.
Mol Metab ; 54: 101366, 2021 12.
Article En | MEDLINE | ID: mdl-34728342

OBJECTIVE: The regulation of food intake is a major research area in the study of obesity, which plays a key role in the development of metabolic syndrome. Gene targeting studies have clarified the roles of hypothalamic neurons in feeding behavior, but the deletion of a gene has a long-term effect on neurophysiology. Our understanding of short-term changes such as appetite under physiological conditions is therefore still limited. METHODS: Targeted recombination in active populations (TRAP) is a newly developed method for labeling active neurons by using tamoxifen-inducible Cre recombination controlled by the promoter of activity-regulated cytoskeleton-associated protein (Arc/Arg3.1), a member of immediate early genes. Transgenic mice for TRAP were fasted overnight, re-fed with normal diet, and injected with 4-hydroxytamoxifen 1 h after the refeeding to label the active neurons. The role of labeled neurons was examined by expressing excitatory or inhibitory designer receptors exclusively activated by designer drugs (DREADDs). The labeled neurons were extracted and RNA sequencing was performed to identify genes that are specifically expressed in these neurons. RESULTS: Fasting-refeeding activated and labeled neurons in the compact part of the dorsomedial hypothalamus (DMH) that project to the paraventricular hypothalamic nucleus. Chemogenetic activation of the labeled DMH neurons decreased food intake and developed place preference, an indicator of positive valence. Chemogenetic activation or inhibition of these neurons had no influence on the whole-body glucose metabolism. The labeled DMH neurons expressed prodynorphin (pdyn), gastrin-releasing peptide (GRP), cholecystokinin (CCK), and thyrotropin-releasing hormone receptor (Trhr) genes. CONCLUSIONS: We identified a novel cell type of DMH neurons that can inhibit food intake and promote feeding-induced positive valence. Our study provides insight into the role of DMH and its molecular mechanism in the regulation of appetite and emotion.


Dorsomedial Hypothalamic Nucleus/metabolism , Eating , Neurons/metabolism , Animals , Cholecystokinin/genetics , Enkephalins/genetics , Feeding Behavior , Male , Mice , Mice, Transgenic , Protein Precursors/genetics
6.
Neurosci Lett ; 764: 136295, 2021 11 01.
Article En | MEDLINE | ID: mdl-34655712

Cholecystokinin (CCK) is an appetite-suppressing hormone that acts in the dorsomedial hypothalamus (DMH) in adult rats to suppress food intake. It remains unknown, however, whether CCK has the same affect in young animals, despite the rising prevalence of childhood obesity and drastic need for research in this area. At the synaptic level, CCK has been shown to inhibit putative orexigenic DMH neurons in young male rats by increasing GABA release onto these neurons via a CCK2 receptor and nitric oxide-dependent pathway. Whether this pathway leads to appetite suppression in young rats is not known. We therefore investigated whether intra-DMH administration of CCK, with or without inhibitors of the CCK2 receptor and nitric oxide signaling pathways, affects food intake in young, male, fasted Sprague-Dawley rats. We implanted bilateral guide cannulas into the DMH and allowed animals to recover from the surgery. Animals were then fasted for 24 h, following which they received intra-DMH microinjections of vehicle, CCK-8S, or CCK-8S combined with either LY-225910 (CCK2 receptor antagonist), L-NAME (a nitric oxide synthase inhibitor), or ODQ (a soluble guanylate cyclase inhibitor) and were given access to regular chow. Following a two hour refeeding period during which food intake, latency to feed, and body weight were measured, brains were subsequently removed to confirm cannula placement in the DMH. The effect of CCK on these parameters in rats given a high fat diet were also measured. Here we show that intra-DMH administration of CCK suppresses food intake and body weight in young rats. This effect requires activation of CCK2 receptors and nitric oxide signaling. Finally, CCK has no effect on consumption of a high fat diet when administered into the DMH. Overall, these findings demonstrate a potential pathway through which CCK might suppress appetite in young rats.


Appetite Regulation/physiology , Cholecystokinin/metabolism , Dorsomedial Hypothalamic Nucleus/metabolism , Nitric Oxide/metabolism , Pediatric Obesity/physiopathology , Animals , Cholecystokinin/administration & dosage , Humans , Male , Microinjections , Models, Animal , Pediatric Obesity/prevention & control , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin B/metabolism
7.
J Neuroendocrinol ; 33(8): e13006, 2021 08.
Article En | MEDLINE | ID: mdl-34235800

In hungry animals, neuropeptide Y (NPY) neurones in the arcuate nucleus (ArcN) are activated to suppress energy expenditure, in part by decreasing brown adipose tissue sympathetic nerve activity (BAT SNA); however, the NPY receptor subtype and brain neurocircuitry are unclear. In the present study, we investigated the inhibition of BAT SNA by exogenous and endogenous NPY via binding to Y1 receptors (NPY1R) in the hypothalamic paraventricular nucleus (PVN) and dorsomedial hypothalamus (DMH), in anaesthetised male rats. Downstream projections of PVN/DMH NPY1R-expressing neurones were identified using male Npy1r-cre mice and localised unilateral DMH or PVN injections of an adeno-associated virus, which allows for the cre-dependent expression of a fluorescent protein (mCherry) in the cell bodies, axon fibres and nerve terminals of NPY1R-containing neurones. Nanoinjections of NPY into the DMH of cooled rats decreased BAT SNA, as well as mean arterial pressure (MAP) and heart rate (HR), and these responses were reversed by subsequent injection of the selective NPY1R antagonist, BIBO3304. In warmed rats, with little to no BAT SNA, bilateral nanoinjections of BIBO3304 into the DMH or PVN increased BAT SNA, MAP and HR. DMH NPY1R-expressing neurones projected heavily to the raphe pallidus (RPa), which houses BAT presympathetic neurones, as well as the PVN. In anaesthetised mice, DMH BIBO3304 increased splanchnic SNA, MAP and HR, all of which were reversed by nonselective blockade of the PVN with muscimol, suggesting that DMH-to-PVN connections are involved in this DMH BIBO3304 disinhibition. PVN Y1R expressing neurones also projected to the RPa, as well as to the nucleus tractus solitarius. We conclude that NPY tonically released in the DMH and PVN suppresses BAT SNA, MAP and HR via Y1R. Downstream neuropathways for BAT SNA may utilise direct projections to the RPa. Release of tonic NPY inhibition of BAT SNA may contribute to feeding- and diet-induced thermogenesis.


Cardiovascular System/drug effects , Dorsomedial Hypothalamic Nucleus/drug effects , Neuropeptide Y/pharmacology , Paraventricular Hypothalamic Nucleus/drug effects , Thermogenesis/drug effects , Animals , Blood Pressure/drug effects , Cardiovascular System/innervation , Dorsomedial Hypothalamic Nucleus/metabolism , Heart Rate/drug effects , Male , Mice , Mice, Transgenic , Paraventricular Hypothalamic Nucleus/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Neuropeptide Y/metabolism , Receptors, Neuropeptide Y/physiology , Sympathetic Nervous System/drug effects , Sympathetic Nervous System/metabolism
8.
Proc Natl Acad Sci U S A ; 118(13)2021 03 30.
Article En | MEDLINE | ID: mdl-33753517

Leptin-deficient ob/ob mice eat voraciously, and their food intake is markedly reduced by leptin treatment. In order to identify potentially novel sites of leptin action, we used PhosphoTRAP to molecularly profile leptin-responsive neurons in the hypothalamus and brainstem. In addition to identifying several known leptin responsive populations, we found that neurons in the dorsomedial hypothalamus (DMH) of ob/ob mice expressing protein phosphatase 1 regulatory subunit 17 (PPP1R17) constitutively express cFos and that this is suppressed by leptin treatment. Because ob mice are hyperphagic, we hypothesized that activating PPP1R17 neurons would increase food intake. However, chemogenetic activation of PPP1R17 neurons decreased food intake and body weight of ob/ob mice while inhibition of PPP1R17 neurons increased them. Similarly, in a scheduled feeding protocol that elicits increased consumption, mice also ate more when PPP1R17 neurons were inhibited and ate less when they were activated. Finally, we found that pair-feeding of ob mice reduced cFos expression to a similar extent as leptin and that reducing the amount of food available during scheduled feeding in DMHPpp1r17 neurons also decreased cFos in DMHPpp1r17 neurons. Finally, these neurons do not express the leptin receptor, suggesting that the effect of leptin on these neurons is indirect and secondary to reduced food intake. In aggregate, these results show that PPP1R17 neurons in the DMH are activated by increased food intake and in turn restrict intake to limit overconsumption, suggesting that they function to constrain binges of eating.


Bulimia/physiopathology , Dorsomedial Hypothalamic Nucleus/physiopathology , Eating/physiology , Leptin/physiology , Neural Inhibition , Neurons/physiology , Proteins/metabolism , Animals , Bulimia/genetics , Dorsomedial Hypothalamic Nucleus/drug effects , Dorsomedial Hypothalamic Nucleus/metabolism , Eating/genetics , Leptin/genetics , Leptin/pharmacology , Mice , Mice, Obese , Neurons/drug effects , Neurons/metabolism , Proteins/genetics , Proto-Oncogene Proteins c-fos/metabolism , Satiety Response
9.
Eur J Pharmacol ; 899: 174033, 2021 May 15.
Article En | MEDLINE | ID: mdl-33727058

The dorsomedial hypothalamus (DMH) receives dense orexinergic innervation. Intra-DMH application of orexins increases arterial pressure and heart rate in rats. We studied the effects of orexin-A on DMH neurons, including those innervating the medullary cardiovascular center, the rostral ventrolateral medulla (RVLM), by using whole-cell recordings in brain slices. In the presence of tetrodotoxin, orexin-A (30-1000 nM) depolarized 56% of DMH neurons (EC50 82.4 ± 4.4 nM). Under voltage-clamp recording, orexin-A (300 nM) induced three types of responses characterized by different current-voltage relationships, namely unchanged, increased, and decreased slope conductance in 68%, 14%, and 18% of orexin-A-responsive neurons, respectively. The reversal potential of the decreased-conductance response was near the equilibrium potential of K+ and became more positive in a high-K+ solution, suggesting that K+ conductance blockade is the underlying mechanism. In a low-Na+ solution, unchanged-, increased-, and decreased-conductance responses were observed in 56%, 11%, and 33% of orexin-A-responsive neurons, respectively, implying that a non-selective cation current (NSCC) underlies orexin-A-induced responses in a small population of DMH neurons. KBR-7943 (70 µM), an inhibitor of Na+-Ca2+ exchanger (NCX), suppressed orexin-A-induced depolarization in 7 of 10 neurons. In the presence of KBR-7943, the majority of orexin-A-responsive neurons exhibited decreased-conductance responses. These findings suggest that NCX activation may underlie orexin-A-induced depolarization in the majority of orexin-responsive DMH neurons. Of 19 RVLM-projecting DMH neurons identified by retrograde labeling, 17 (90%) were orexin-A responsive. In conclusion, orexin-A directly excited over half of DMH neurons, including those innervating the RVLM, through decreasing K+ conductance, activating NCX, and/or increasing NSCC.


Dorsomedial Hypothalamic Nucleus/drug effects , Medulla Oblongata/drug effects , Neurons/drug effects , Orexins/pharmacology , Animals , Dorsomedial Hypothalamic Nucleus/cytology , Dorsomedial Hypothalamic Nucleus/metabolism , Female , In Vitro Techniques , Male , Medulla Oblongata/cytology , Medulla Oblongata/metabolism , Membrane Potentials , Neural Pathways/drug effects , Neural Pathways/metabolism , Neuroanatomical Tract-Tracing Techniques , Neurons/metabolism , Potassium/metabolism , Rats, Sprague-Dawley , Sodium-Calcium Exchanger/metabolism
10.
Physiol Behav ; 229: 113253, 2021 02 01.
Article En | MEDLINE | ID: mdl-33220330

Glucagon-like peptide 2 (GLP-2), a member of Glucagon peptide family involved in regulating energy metabolism, can be produced and secreted by preproglucagonergic (PPG) neurons in the brain. GLP-2 reduces food intake but at which brain sites GLP-2 exerts its feeding-suppress effects are still unclear. In this study, we used the stereological microinjection technique and behavioral test to examine the functions of locally delivered GLP-2 into DMH on feeding behavior. We compared effects of different concentration of GLP-2 on the food intake behavior in free-feeding rats and fasted-refeeding rats. We found that GLP-2 inhibited food intake in fasted rats after a short-term intervention in a dose-dependent manner. Importantly, the effects of locally delivered GLP-2 can be blocked by specific GLP-1 receptor antagonist Exendin(9-39), but not the melanocortin-4 receptor antagonist SHU9119, indicating the involvement of specificity of GLP-2 signaling in regulating the feeding behavior. Taken together, our data revealed that GLP-2 peptide pharmacologically inhibited food intake in DMH and this effect could be blocked functionally by Exendin(9-39).


Dorsomedial Hypothalamic Nucleus , Eating , Glucagon-Like Peptide 2 , Peptide Fragments , Animals , Dorsomedial Hypothalamic Nucleus/metabolism , Glucagon-Like Peptide 1 , Glucagon-Like Peptide-1 Receptor/metabolism , Male , Rats , Rats, Sprague-Dawley
11.
Anat Rec (Hoboken) ; 304(5): 1094-1104, 2021 05.
Article En | MEDLINE | ID: mdl-33040447

The hypothalamus is involved in the regulation of rhythms, autonomic, endocrine, and behavioral functions and may also participate in aging development and control. The aim of this work was to study the expression of calbindin (CB) and calretinin (CR) in the ventromedial (VMH) and dorsomedial (DMH) hypothalamic nuclei in young and old rats of both sexes by immunohistochemistry and western blotting. In young animals, the largest number of CB-immunoreactive (IR) neurons was detected in the ventral part of DMH (DMHv) and smaller percentage was found in its dorsal part (DMHd), in the dorsomedial part of the VMH (VMHdm) and in the ventrolateral part of the VMH (VMHvl). In aged animals, the percentage of CB-IR neurons significantly decreased in all studied nuclei, including DMHv, DMHd, VMHdm and VMHvl. CR-IR neurons were found in moderate number in the DMHv, DMHd, VMHdm and VMHvl of young rats. In aged rats, the percentage of CR-IR neurons significantly increased in the DMHv, DMHd, VMHdm and VMHvl. Less than one third of IR neurons colocalized CB and CR in young and aged rats. The expression of CB significantly decreased, and the expression of CR significantly increased in the DMH and VMH during aging by western blot analysis. Thus, there are opposite changes of the calcium-binding proteins expression in the hypothalamic nuclei involved in the metabolic and sexual regulation during aging.


Aging/metabolism , Calbindin 2/metabolism , Calbindins/metabolism , Dorsomedial Hypothalamic Nucleus/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Female , Male , Neurons/metabolism , Rats
12.
Bull Exp Biol Med ; 169(5): 698-700, 2020 Sep.
Article En | MEDLINE | ID: mdl-32989637

We studied the expression of sirtuin 1 (Sirt1) in the dorsomedial and ventromedial nuclei of the hypothalamus in young (2 months) and old (2 years) rats by immunohistochemical methods and Western blotting. In aged males and females, a decrease in Sirt1 expression in dorsomedial nucleus was observed. In ventromedial nucleus, the expression of Sirt1 did not change with age. The results confirm the hypothesis that aging is associated with a decrease in the content of sirtuins in the hypothalamic nuclei.


Dorsomedial Hypothalamic Nucleus/metabolism , Sirtuin 1/metabolism , Aging/physiology , Animals , Blotting, Western , Female , Hypothalamus/metabolism , Immunohistochemistry , Male , Rats , Sirtuin 1/genetics
13.
Mol Metab ; 37: 100985, 2020 07.
Article En | MEDLINE | ID: mdl-32311511

OBJECTIVE: Hypothalamic arcuate proopiomelanocortin (Arc-POMC) neurons are involved in different physiological processes such as the regulation of energy balance, glucose homeostasis, and stress-induced analgesia. Since these neurons heterogeneously express different biological markers and project to many hypothalamic and extrahypothalamic areas, it is proposed that Arc-POMC neurons could be classified into different subpopulations having diverse physiological roles. The aim of the present study was to characterize the contribution of the subpopulation of Arc-POMC neurons cosecreting gamma-aminobutyric acid (GABA) neurotransmitter in the control of energy balance. METHODS: Arc-Pomc expression restricted to GABAergic-POMC neurons was achieved by crossing a reversible Pomc-deficient mouse line (arcPomc-) with a tamoxifen-inducible Gad2-CreER transgenic line. Pomc expression was rescued in the compound arcPomc-/-:Gad2-CreER female and male mice by tamoxifen treatment at postnatal days 25 (P25) or 60 (P60), and body weight, daily food intake, fasting glycemia, and fasting-induced hyperphagia were measured. POMC recovery was quantified by immunohistochemistry and semiquantitative RT-PCR. Neuropeptide Y (NPY) and GABAergic neurons were identified by in situ hybridization. Arc-POMC neurons projecting to the dorsomedial hypothalamic nucleus (DMH) were studied by stereotactic intracerebral injection of fluorescent retrobeads into the DMH. RESULTS: Tamoxifen treatment of arcPomc-/-:Gad2-CreER mice at P60 resulted in Pomc expression in ∼23-25% of Arc-POMC neurons and ∼15-23% of Pomc mRNA levels, compared to Gad2-CreER control mice. Pomc rescue in GABAergic-POMC neurons at P60 normalized food intake, glycemia, and fasting-induced hyperphagia, while significantly reducing body weight. Energy balance was also improved in arcPomc-/-:Gad2-CreER mice treated with tamoxifen at P25. Distribution analysis of rescued POMC immunoreactive fibers revealed that the DMH is a major target site of GABAergic-POMC neurons. Further, the expression of the orexigenic neuropeptide Y (NPY) in the DMH was increased in arcPomc-/- obese mice but was completely restored after Pomc rescue in arcPomc-/-:Gad2-CreER mice. Finally, we found that ∼75% of Arc-POMC neurons projecting to the DMH are GABAergic. CONCLUSIONS: In the present study, we show that the expression of Pomc in the subpopulation of Arc-GABAergic-POMC neurons is sufficient to maintain normal food intake. In addition, we found that DMH-NPY expression is negatively correlated with Pomc expression in GABAergic-POMC neurons, suggesting that food intake may be regulated by an Arc-GABAergic-POMC → DMH-NPY pathway.


Eating/genetics , GABAergic Neurons/classification , Pro-Opiomelanocortin/metabolism , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Body Weight , Dorsomedial Hypothalamic Nucleus/metabolism , Eating/physiology , Energy Metabolism/physiology , Female , Gene Expression/genetics , Gene Expression Regulation/genetics , Hypothalamus/metabolism , Leptin/metabolism , Male , Mice , Mice, Obese , Neuropeptide Y/metabolism , Pro-Opiomelanocortin/genetics
14.
Nitric Oxide ; 100-101: 1-6, 2020 08 01.
Article En | MEDLINE | ID: mdl-32283261

The hypothalamus is the most important integrator of autonomic and endocrine regulation in the body and it also has a fundamental role in ageing development and lifespan control. In order to better understand the role of NO-ergic system in the hypothalamic regulation of ageing, the purpose of this study was to investigate the expression of neuronal nitric oxide synthase (nNOS) in the arcuate (ARC), ventromedial (VMH) and dorsomedial (DMH) hypothalamic nuclei in young (2-3-month-old) and old (24-month-old) male and female rats using immunohistochemistry and western blot analysis. In young animals, only single nNOS-immunoreactive (IR) neurons were detected in ARC, and nNOS-IR neurons were found in the VMH (19 ± 3.2% in females and 14.5 ± 2.6% in males) and DMH (17 ± 4.0% in females and 21 ± 2.8% in males). In aged animals, the number of nNOS-IR neurons increased in all studied nuclei, including ARC (36 ± 3.1% in females and 33.5 ± 3.7% in males), VMH (83 ± 4.3% in females and 58 ± 2.1% in males) and DMH (57 ± 1.9% in females and 54 ± 1.8% in males). The expression of nNOS also significantly increased in the ARC, VMH and DMH during ageing by western blot analysis. In conclusion, ageing is accompanied by increasing of nNOS expression in the hypothalamus and this process is related to regions involved in the control of feeding behavior.


Aging/metabolism , Arcuate Nucleus of Hypothalamus/metabolism , Dorsomedial Hypothalamic Nucleus/metabolism , Nitric Oxide Synthase Type I/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Female , Immunohistochemistry , Male , Neurons/metabolism , Rats
15.
Science ; 367(6482): 1105-1112, 2020 03 06.
Article En | MEDLINE | ID: mdl-32139538

The mechanism by which psychological stress elicits various physiological responses is unknown. We discovered a central master neural pathway in rats that drives autonomic and behavioral stress responses by connecting the corticolimbic stress circuits to the hypothalamus. Psychosocial stress signals from emotion-related forebrain regions activated a VGLUT1-positive glutamatergic pathway from the dorsal peduncular cortex and dorsal tenia tecta (DP/DTT), an unexplored prefrontal cortical area, to the dorsomedial hypothalamus (DMH), a hypothalamic autonomic center. Genetic ablation and optogenetics revealed that the DP/DTT→DMH pathway drives thermogenic, hyperthermic, and cardiovascular sympathetic responses to psychosocial stress without contributing to basal homeostasis. This pathway also mediates avoidance behavior from psychosocial stressors. Given the variety of stress responses driven by the DP/DTT→DMH pathway, the DP/DTT can be a potential target for treating psychosomatic disorders.


Dorsomedial Hypothalamic Nucleus/metabolism , Social Behavior , Stress, Psychological/metabolism , Vesicular Glutamate Transport Protein 1/metabolism , Animals , Emotions/physiology , Female , Glutamic Acid/metabolism , Homeostasis , Male , Neurons/metabolism , Prefrontal Cortex/metabolism , Prosencephalon/metabolism , Psychophysiologic Disorders/therapy , Rats , Rats, Inbred LEC , Rats, Wistar , Signal Transduction
16.
Neurosci Lett ; 714: 134550, 2020 01 01.
Article En | MEDLINE | ID: mdl-31634502

BACKGROUND: Cannabinoid receptor 1 (CB1R) is a GPCR expressed widely in the brain as well as in peripheral metabolic organs. Although pharmacological blockade of CB1R has been effective for the treatment of obesity and tobacco addiction, precise distribution of CB1R within the brain and potential changes by obesity or nicotine exposure have not been thoroughly addressed. METHODS: To examine CB1R distribution within the central energy center, we performed immunostaining and qPCR analysis of micro-dissected hypothalamic nuclei from male C57BL/6 mice. To address the effect of nicotine on food intake and body weight, and on potential changes of CB1R levels in the hypothalamus, mice kept on a high fat diet (HFD) for four weeks were challenged with nicotine intraperitoneally. RESULTS: Validity of the micro-dissected samples was confirmed by the expression of established nucleus-enriched genes. The expression levels of CB1R in the arcuate and lateral nuclei of the hypothalamus were higher than paraventricular and ventral-dorsal medial nuclei. Nicotine administration led to a significant suppression of food intake and body weight either under standard or high fat diet. Neither HFD nor nicotine alone altered CB1R levels in any nucleus tested. By contrast, treatment of HFD-fed mice with nicotine led to a significant increase in CB1R levels in the arcuate, paraventricular and lateral nuclei. CONCLUSIONS: CB1R was widely distributed in multiple hypothalamic nuclei. The expression of CB1R was augmented only when mice were treated with HFD and nicotine in combination. These data suggest that the exposure to nicotine may provoke an enhanced endocannabinoid response in diet-induced obesity.


Arcuate Nucleus of Hypothalamus/metabolism , Diet, High-Fat , Dorsomedial Hypothalamic Nucleus/metabolism , Hypothalamic Area, Lateral/metabolism , Nicotine/pharmacology , Paraventricular Hypothalamic Nucleus/metabolism , Receptor, Cannabinoid, CB1/biosynthesis , Animals , Body Weight/drug effects , Eating/drug effects , Male , Mice , Microdissection/methods , Neuropeptide Y/metabolism , Receptors, Corticotropin-Releasing Hormone/metabolism
17.
Brain Res ; 1727: 146538, 2020 01 15.
Article En | MEDLINE | ID: mdl-31705862

The surging obesity epidemic calls for a deeper understanding of central nervous system (CNS) mechanisms underlying the biologically defended level of body weight. Here, we analyzed global gene expression in four hypothalamic and two brainstem nuclei involved in energy homeostatic control of body weight in diet-induced obese (DIO) and lean rats. Male Sprague-Dawley rats were offered ad libitum chow, or a two-choice diet consisting of a high palatable high sugar/fat diet and chow for 40 weeks. At termination, the hypothalamic arcuate nucleus (ARC), dorsomedial hypothalamus (DMH), paraventricular nucleus (PVN) and lateral hypothalamus area (LHA), as well as the brainstem area postrema (AP) and nucleus of the solitary tract (NTS), were isolated by laser capture microdissection (LCM) followed by mRNA sequencing. Global gene expression analyses revealed a total of 88 differentially expressed genes (DEGs) in DIO rats. Transcriptome changes were mainly observed in the DMH and NTS and associated with neuropeptide signaling and regulation of signaling transduction pathways, suggesting a key role of these brain regions in body weight regulation.


Dorsomedial Hypothalamic Nucleus/metabolism , Obesity/genetics , Solitary Nucleus/metabolism , Transcriptome , Animals , Disease Models, Animal , Gene Expression Profiling , Male , RNA, Messenger/metabolism , Rats, Sprague-Dawley
18.
J Neurosci ; 39(33): 6449-6459, 2019 08 14.
Article En | MEDLINE | ID: mdl-31235650

Neuronal circuits regulating hunger and satiety synthesize information encoding the energy state of the animal and translate those signals into motivated behaviors to meet homeostatic needs. Proopiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus are activated by energy surfeits and inhibited by energy deficits. When activated, these cells inhibit food intake and facilitate weight loss. Conversely, decreased activity in POMC cells is associated with increased food intake and obesity. Circulating nutrients and hormones modulate the activity of POMC neurons over protracted periods of time. However, recent work indicates that calcium activity in POMC cells changes in response to food cues on times scales consistent with the rapid actions of amino acid transmitters. Indeed, the frequency of spontaneous IPSCs (sIPSCs) onto POMC neurons increases during caloric deficits. However, the afferent brain regions responsible for this inhibitory modulation are currently unknown. Here, through the use of brain region-specific deletion of GABA release in both male and female mice we show that neurons in the dorsomedial hypothalamus (DMH) are responsible for the majority of sIPSCs in POMC neurons as well as the fasting-induced increase in sIPSC frequency. Further, the readily releasable pool of GABA vesicles and the release probability of GABA is increased at DMH-to-POMC synapses following an overnight fast. Collectively these data provide evidence that DMH-to-POMC GABA circuitry conveys inhibitory neuromodulation onto POMC cells that is sensitive to the animal's energy state.SIGNIFICANCE STATEMENT Activation of proopiomelanocortin (POMC) cells signals satiety, whereas GABAergic cells in the dorsomedial hypothalamus (DMH) can increase food consumption. However, communication between these cells, particularly in response to changes in metabolic state, is unknown. Here, through targeted inhibition of DMH GABA release, we show that DMH neurons contribute a significant portion of spontaneously released GABA onto POMC cells and are responsible for increased GABAergic inhibition of POMC cells during fasting, likely mediated through increased release probability of GABA at DMH terminals. These data provide important information about inhibitory modulation of metabolic circuitry and provide a mechanism through which POMC neurons could be inhibited, or disinhibited, rapidly in response to food availability.


Dorsomedial Hypothalamic Nucleus/metabolism , GABAergic Neurons/metabolism , Inhibitory Postsynaptic Potentials/physiology , Neurons/metabolism , Pro-Opiomelanocortin/metabolism , Animals , Female , Male , Mice , Mice, Transgenic
19.
Neurosci Lett ; 703: 79-85, 2019 06 11.
Article En | MEDLINE | ID: mdl-30902570

Thyrotropin-releasing hormone (TRH) plays an important role in the regulation of energy balance. While the regulation of TRH in the paraventricular nucleus (PVN) in response to changes of energy balance has been well studied, how TRH is regulated in the dorsomedial hypothalamus (DMH) in maintaining energy homeostasis remains unclear. Here, we assessed the effects of food restriction and exercise on hypothalamic Trh expression using Otsuka Long-Evens Tokushima Fatty (OLETF) rats. Sedentary ad lib fed OLETF rats (OLETF-SED) became hyperphagic and obese. These alterations were prevented in OLETF rats with running wheel access (OLETF-RW) or food restriction in which their food was pair-fed (OLETF-PF) to the intake of lean control rats (LETO-SED). Evaluation of hypothalamic gene expression revealed that Trh mRNA expression was increased in the PVN of OLETF-SED rats and normalized in OLETF-RW and OLETF-PF rats compared to LETO-SED rats. In contrast, the expression of Trh in the DMH was decreased in OLETF-SED rats relative to LETO-SED rats. This alteration was reversed in OLETF-RW rats as seen in LETO-SED rats, but food restriction resulted in a significant increase in DMH Trh expression in OLETF-PF rats compared to LETO-SED rats. Strikingly, while Trh mRNA expression was decreased in the PVN of intact rats in response to acute food deprivation, food deprivation resulted in increased expression of Trh in the DMH. Together, these results demonstrate the differential regulation of Trh expression in the PVN and DMH in OLETF rats and suggest that DMH TRH also contributes to hypothalamic regulation of energy balance.


Dorsomedial Hypothalamic Nucleus/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , RNA, Messenger/metabolism , Thyrotropin-Releasing Hormone/metabolism , Animals , Blood Glucose , Body Weight , Diet, Reducing , Eating , Leptin/blood , Male , Obesity/physiopathology , Obesity/psychology , Physical Conditioning, Animal , Rats , Thyrotropin-Releasing Hormone/genetics
20.
Psychopharmacology (Berl) ; 236(6): 1863-1874, 2019 Jun.
Article En | MEDLINE | ID: mdl-30694375

RATIONALE: The endocannabinoid system plays an important role in the organization of panic-like defensive behavior. Threatening situations stimulate brain areas, such as the dorsomedial hypothalamus (DMH). However, there is a lack of studies addressing the role of the DMH endocannabinoid system in panic-like responses. OBJECTIVES: We aimed to verify which mechanisms underlie anandamide-mediated responses in the DMH. METHODS: To test the hypothesis that the anandamide produces panicolytic-like effects, we treated mice with intra-DMH microinjections of vehicle or increasing doses of anandamide (0.5, 5, or 50 pmol) and then performed confrontation with the South American snake Epicrates cenchria assisi. RESULTS: Intra-DMH anandamide treatment yielded a U-shaped dose-response curve with no effect of the lowest (0.5 pmol) or the highest (50 pmol) dose and significant inhibition of panic-like responses at the intermediate (5 pmol) dose. In addition, this panicolytic-like effect was prevented by pretreatment of the DMH with the CB1 receptor antagonist AM251 (100 pmol). However, pretreatment of the DMH with the TRPV1 receptor antagonist 6-iodo-nordihydrocapsaicin (3 nmol) restored the panicolytic-like effect of the highest dose of anandamide. Immunohistochemistry revealed that CB1 receptors were present primarily on axonal fibers, while TRPV1 receptors were found almost exclusively surrounding the perikarya in DMH. CONCLUSIONS: The present results suggest that anandamide exerts a panicolytic-like effect in the DMH by activation of CB1 receptors and that TRPV1 receptors are related to the lack of effect of the highest dose of anandamide.


Arachidonic Acids/administration & dosage , Cannabinoid Receptor Agonists/administration & dosage , Dorsomedial Hypothalamic Nucleus/metabolism , Endocannabinoids/administration & dosage , Panic/physiology , Polyunsaturated Alkamides/administration & dosage , Receptor, Cannabinoid, CB1/biosynthesis , TRPV Cation Channels/biosynthesis , Animals , Boidae , Brazil , Dorsomedial Hypothalamic Nucleus/drug effects , Dose-Response Relationship, Drug , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Panic/drug effects , Piperidines/administration & dosage , Pyrazoles/administration & dosage , Rats , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/antagonists & inhibitors , TRPV Cation Channels/antagonists & inhibitors
...