Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 262
Filter
Add more filters











Publication year range
1.
Nature ; 632(8025): 585-593, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38987598

ABSTRACT

The most successful obesity therapeutics, glucagon-like peptide-1 receptor (GLP1R) agonists, cause aversive responses such as nausea and vomiting1,2, effects that may contribute to their efficacy. Here, we investigated the brain circuits that link satiety to aversion, and unexpectedly discovered that the neural circuits mediating these effects are functionally separable. Systematic investigation across drug-accessible GLP1R populations revealed that only hindbrain neurons are required for the efficacy of GLP1-based obesity drugs. In vivo two-photon imaging of hindbrain GLP1R neurons demonstrated that most neurons are tuned to either nutritive or aversive stimuli, but not both. Furthermore, simultaneous imaging of hindbrain subregions indicated that area postrema (AP) GLP1R neurons are broadly responsive, whereas nucleus of the solitary tract (NTS) GLP1R neurons are biased towards nutritive stimuli. Strikingly, separate manipulation of these populations demonstrated that activation of NTSGLP1R neurons triggers satiety in the absence of aversion, whereas activation of APGLP1R neurons triggers strong aversion with food intake reduction. Anatomical and behavioural analyses revealed that NTSGLP1R and APGLP1R neurons send projections to different downstream brain regions to drive satiety and aversion, respectively. Importantly, GLP1R agonists reduce food intake even when the aversion pathway is inhibited. Overall, these findings highlight NTSGLP1R neurons as a population that could be selectively targeted to promote weight loss while avoiding the adverse side effects that limit treatment adherence.


Subject(s)
Anti-Obesity Agents , Avoidance Learning , Glucagon-Like Peptide-1 Receptor , Neural Pathways , Rhombencephalon , Satiety Response , Animals , Female , Male , Mice , Anti-Obesity Agents/adverse effects , Anti-Obesity Agents/pharmacology , Area Postrema/metabolism , Area Postrema/drug effects , Avoidance Learning/drug effects , Avoidance Learning/physiology , Eating/drug effects , Eating/physiology , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucagon-Like Peptide-1 Receptor/metabolism , Mice, Inbred C57BL , Neural Pathways/drug effects , Neurons/metabolism , Neurons/physiology , Neurons/drug effects , Obesity/metabolism , Rhombencephalon/cytology , Rhombencephalon/drug effects , Rhombencephalon/metabolism , Rhombencephalon/physiology , Satiety Response/drug effects , Satiety Response/physiology , Solitary Nucleus/cytology , Solitary Nucleus/drug effects , Solitary Nucleus/metabolism , Solitary Nucleus/physiology , Food
2.
Molecules ; 27(2)2022 Jan 10.
Article in English | MEDLINE | ID: mdl-35056733

ABSTRACT

Flavonoids and polyunsaturated fatty acids due to low cytotoxicity in vitro studies are suggested as potential substances in the prevention of diseases associated with oxidative stress. We examined novel 6-hydroxy-flavanone and 7-hydroxy-flavone conjugates with selected fatty acids (FA) of different length and saturation and examined their cytotoxic and antioxidant potential. Our findings indicate that the conjugation with FA affects the biological activity of both the original flavonoids. The conjugation of 6-hydroxy-flavanone increased its cytotoxicity towards prostate cancer PC3 cells. The most noticeable effect was found for oleate conjugate. A similar trend was observed for 7-hydroxy-flavone conjugates with the most evident effect for oleate and stearate. The cytotoxic potential of all tested conjugates was not specific towards PC3 because the viability of human keratinocytes HaCaT cells decreased after exposure to all conjugates. Additionally, we showed that esterification of the two flavonoids decreased their antioxidant activity compared to that of the original compounds. Of all the tested compounds, only 6-sorbic flavanone showed a slight increase in antioxidant potential compared to that of the original compound. Our data show that conjugated flavonoids are better absorbed and enhance cytotoxic effects, but the presence of FA lowered the antioxidant potential.


Subject(s)
Antineoplastic Agents/pharmacology , Antioxidants/pharmacology , Fatty Acids/chemistry , Flavones/chemistry , Flavones/pharmacology , Animals , Antineoplastic Agents/chemistry , Antioxidants/chemistry , Drug Evaluation, Preclinical , Esterification , Humans , Keratinocytes/drug effects , Male , PC-3 Cells , Rats , Rhombencephalon/drug effects , Rhombencephalon/metabolism , Structure-Activity Relationship
3.
PLoS One ; 16(8): e0256207, 2021.
Article in English | MEDLINE | ID: mdl-34403440

ABSTRACT

Thyroid hormones are messengers that bind to specific nuclear receptors and regulate a wide range of physiological processes in the early stages of vertebrate embryonic development, including neurodevelopment and myelogenesis. We here tested the effects of reduced T3 availability upon the myelination process by treating zebrafish embryos with low concentrations of iopanoic acid (IOP) to block T4 to T3 conversion. Black Gold II staining showed that T3 deficiency reduced the myelin density in the forebrain, midbrain, hindbrain and the spinal cord at 3 and 7 dpf. These observations were confirmed in 3 dpf mbp:egfp transgenic zebrafish, showing that the administration of IOP reduced the fluorescent signal in the brain. T3 rescue treatment restored brain myelination and reversed the changes in myelin-related gene expression induced by IOP exposure. NG2 immunostaining revealed that T3 deficiency reduced the amount of oligodendrocyte precursor cells in 3 dpf IOP-treated larvae. Altogether, the present results show that inhibition of T4 to T3 conversion results in hypomyelination, suggesting that THs are part of the key signaling molecules that control the timing of oligodendrocyte differentiation and myelin synthesis from very early stages of brain development.


Subject(s)
Gene Expression Regulation, Developmental/drug effects , Larva/genetics , Myelin Sheath/genetics , Thyroxine/deficiency , Triiodothyronine/deficiency , Zebrafish/metabolism , Animals , Animals, Genetically Modified , Antigens/genetics , Antigens/metabolism , Embryo, Nonmammalian , Embryonic Development , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Iopanoic Acid/pharmacology , Larva/cytology , Larva/drug effects , Larva/growth & development , Mesencephalon/cytology , Mesencephalon/drug effects , Mesencephalon/growth & development , Mesencephalon/metabolism , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Myelin Sheath/drug effects , Myelin Sheath/metabolism , Neurogenesis/drug effects , Neurogenesis/genetics , Oligodendrocyte Transcription Factor 2/genetics , Oligodendrocyte Transcription Factor 2/metabolism , Oligodendroglia/cytology , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Prosencephalon/cytology , Prosencephalon/drug effects , Prosencephalon/growth & development , Prosencephalon/metabolism , Proteoglycans/genetics , Proteoglycans/metabolism , Rhombencephalon/cytology , Rhombencephalon/drug effects , Rhombencephalon/growth & development , Rhombencephalon/metabolism , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Spinal Cord/cytology , Spinal Cord/drug effects , Spinal Cord/growth & development , Spinal Cord/metabolism , Triiodothyronine/pharmacology , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
4.
Am J Physiol Regul Integr Comp Physiol ; 321(2): R250-R259, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34259025

ABSTRACT

The peptide hormone amylin reduces food intake and body weight and is an attractive candidate target for novel pharmacotherapies to treat obesity. However, the short half-life of native amylin and amylin analogs like pramlintide limits these compounds' potential utility in promoting sustained negative energy balance. Here, we evaluate the ability of the novel long-acting amylin/calcitonin receptor agonist ZP5461 to reduce feeding and body weight in rats, and also test the role of calcitonin receptors (CTRs) in the dorsal vagal complex (DVC) of the hindbrain in the energy balance effects of chronic ZP5461 administration. Acute dose-response studies indicate that systemic ZP5461 (0.5-3 nmol/kg) robustly suppresses energy intake and body weight gain in chow- and high-fat diet (HFD)-fed rats. When HFD-fed rats received chronic systemic administration of ZP5461 (1-2 nmol/kg), the compound initially produced reductions in energy intake and weight gain but failed to produce sustained suppression of intake and body weight. Using virally mediated knockdown of DVC CTRs, the ability of chronic systemic ZP5461 to promote early reductions in intake and body weight gain was determined to be mediated in part by activation of DVC CTRs, implicating the DVC as a central site of action for ZP5461. Future studies should address other dosing regimens of ZP5461 to determine whether an alternative dose/frequency of administration would produce more sustained body weight suppression.


Subject(s)
Amylin Receptor Agonists/pharmacology , Appetite Depressants/pharmacology , Eating/drug effects , Feeding Behavior/drug effects , Receptors, Calcitonin/agonists , Receptors, Islet Amyloid Polypeptide/drug effects , Rhombencephalon/drug effects , Vagus Nerve/drug effects , Weight Gain/drug effects , Animals , Dose-Response Relationship, Drug , Energy Intake/drug effects , Male , Rats, Sprague-Dawley , Receptors, Calcitonin/genetics , Receptors, Calcitonin/metabolism , Receptors, Islet Amyloid Polypeptide/genetics , Receptors, Islet Amyloid Polypeptide/metabolism , Rhombencephalon/metabolism , Signal Transduction , Time Factors , Vagus Nerve/metabolism
5.
Endocrinology ; 162(9)2021 09 01.
Article in English | MEDLINE | ID: mdl-34161572

ABSTRACT

Lowered glucose availability, sensed by the hindbrain, has been suggested to enhance gluconeogenesis and food intake as well as suppress reproductive function. In fact, our previous histological and in vitro studies suggest that hindbrain ependymal cells function as a glucose sensor. The present study aimed to clarify the hindbrain glucose sensor-hypothalamic neural pathway activated in response to hindbrain glucoprivation to mediate counterregulatory physiological responses. Administration of 2-deoxy-D-glucose (2DG), an inhibitor of glucose utilization, into the fourth ventricle (4V) of male rats for 0.5 hour induced messenger RNA (mRNA) expression of c-fos, a marker for cellular activation, in ependymal cells in the 4V, but not in the lateral ventricle, the third ventricle or the central canal without a significant change in blood glucose and testosterone levels. Administration of 2DG into the 4V for 1 hour significantly increased blood glucose levels, food intake, and decreased blood testosterone levels. Simultaneously, the expression of c-Fos protein was detected in the 4V ependymal cells; dopamine ß-hydroxylase-immunoreactive cells in the C1, C2, and A6 regions; neuropeptide Y (NPY) mRNA-positive cells in the C2; corticotropin-releasing hormone (CRH) mRNA-positive cells in the hypothalamic paraventricular nucleus (PVN); and NPY mRNA-positive cells in the arcuate nucleus (ARC). Taken together, these results suggest that lowered glucose availability, sensed by 4V ependymal cells, activates hindbrain catecholaminergic and/or NPY neurons followed by CRH neurons in the PVN and NPY neurons in the ARC, thereby leading to counterregulatory responses, such as an enhancement of gluconeogenesis, increased food intake, and suppression of sex steroid secretion.


Subject(s)
Glucose/metabolism , Neural Pathways/metabolism , Rhombencephalon/metabolism , Animals , Blood Glucose/metabolism , Eating/physiology , Energy Metabolism/drug effects , Energy Metabolism/physiology , Food Deprivation/physiology , Glucose/deficiency , Glucose/pharmacology , Hypothalamus/anatomy & histology , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothalamus/metabolism , Male , Neural Pathways/anatomy & histology , Neural Pathways/drug effects , Rats , Rats, Wistar , Rhombencephalon/anatomy & histology , Rhombencephalon/cytology , Rhombencephalon/drug effects
6.
Epilepsia ; 62(7): 1689-1700, 2021 07.
Article in English | MEDLINE | ID: mdl-33997963

ABSTRACT

OBJECTIVE: Fetal exposure to the anticonvulsant drug valproic acid (VPA), used to treat certain types of epilepsy, increases the risk for birth defects, including neural tube defects, as well as learning difficulties and behavioral problems. Here, we investigated neurotoxic effects of VPA exposure using zebrafish as a model organism. The capacity of folic acid (FA) supplementation to rescue the VPA-induced neuronal and behavioral perturbations was also examined. METHODS: Zebrafish embryos of different transgenic lines with neuronal green fluorescent protein expression were exposed to increasing concentrations of VPA with or without FA supplementation. Fluorescence microscopy was used to visualize alterations in brain structures and neural progenitor cells, as well as motor neurons and neurite sprouting. A twitching behavioral assay was used to examine the functional consequences of VPA and FA treatment. RESULTS: In zebrafish embryos, VPA exposure caused a decrease in the midbrain size, an increase in the midline gap of the hindbrain, and perturbed neurite sprouting of secondary motor neurons, in a concentration-dependent manner. VPA exposure also decreased the fluorescence intensity of neuronal progenitor cells in early developmental stages, indicating fewer cells. Furthermore, VPA exposure significantly altered embryonic twitching activity, causing hyperactivity in dark and hypoactivity in light. Supplementation of FA rescued the VPA-induced smaller midbrain size and hindbrain midline gap defects. FA treatment also increased the number of neuronal progenitor cells in VPA-treated embryos and salvaged neurite sprouting of the secondary motor neurons. FA rescued the VPA-induced alterations in twitching activity in light but not in dark. SIGNIFICANCE: We conclude that VPA exposure induces specific neurotoxic perturbations in developing zebrafish embryos, and that FA reversed most of the identified defects. The results demonstrate that zebrafish is a promising model to study VPA-induced teratogenesis and to screen for countermeasures.


Subject(s)
Anticonvulsants/toxicity , Behavior, Animal/drug effects , Folic Acid/therapeutic use , Neurotoxicity Syndromes/prevention & control , Neurotoxicity Syndromes/psychology , Valproic Acid/toxicity , Vitamins/therapeutic use , Zebrafish , Animals , Animals, Genetically Modified , Dietary Supplements , Embryonic Development/drug effects , Larva , Lighting , Mesencephalon/anatomy & histology , Mesencephalon/drug effects , Motor Neurons/drug effects , Neural Stem Cells/drug effects , Neural Tube Defects/chemically induced , Neurites/drug effects , Rhombencephalon/anatomy & histology , Rhombencephalon/drug effects , Valproic Acid/antagonists & inhibitors
7.
Nat Genet ; 53(3): 379-391, 2021 03.
Article in English | MEDLINE | ID: mdl-33603234

ABSTRACT

Rapid cellular responses to environmental stimuli are fundamental for development and maturation. Immediate early genes can be transcriptionally induced within minutes in response to a variety of signals. How their induction levels are regulated and their untimely activation by spurious signals prevented during development is poorly understood. We found that in developing sensory neurons, before perinatal sensory-activity-dependent induction, immediate early genes are embedded into a unique bipartite Polycomb chromatin signature, carrying active H3K27ac on promoters but repressive Ezh2-dependent H3K27me3 on gene bodies. This bipartite signature is widely present in developing cell types, including embryonic stem cells. Polycomb marking of gene bodies inhibits mRNA elongation, dampening productive transcription, while still allowing for fast stimulus-dependent mark removal and bipartite gene induction. We reveal a developmental epigenetic mechanism regulating the rapidity and amplitude of the transcriptional response to relevant stimuli, while preventing inappropriate activation of stimulus-response genes.


Subject(s)
Chromatin/genetics , Gene Expression Regulation, Developmental , Genes, Immediate-Early , Polycomb-Group Proteins/genetics , Animals , Chromatin/metabolism , Embryonic Stem Cells/physiology , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Epigenesis, Genetic , Histones/metabolism , Mice, Transgenic , Mutation , Polycomb-Group Proteins/metabolism , Promoter Regions, Genetic , RNA Polymerase II/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rhombencephalon/drug effects , Rhombencephalon/embryology , Sensory Receptor Cells/physiology
8.
Am J Physiol Regul Integr Comp Physiol ; 320(4): R471-R487, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33470901

ABSTRACT

Previous studies indicate that oxytocin (OT) administration reduces body weight in high-fat diet (HFD)-induced obese (DIO) rodents through both reductions in food intake and increases in energy expenditure. We recently demonstrated that chronic hindbrain [fourth ventricular (4V)] infusions of OT evoke weight loss in DIO rats. Based on these findings, we hypothesized that chronic 4V OT would elicit weight loss in DIO mice. We assessed the effects of 4V infusions of OT (16 nmol/day) or vehicle over 28 days on body weight, food intake, and body composition. OT reduced body weight by approximately 4.5% ± 1.4% in DIO mice relative to OT pretreatment body weight (P < 0.05). These effects were associated with reduced adiposity and adipocyte size [inguinal white adipose tissue (IWAT)] (P < 0.05) and attributed, in part, to reduced energy intake (P < 0.05) at a dose that did not increase kaolin intake (P = NS). OT tended to increase uncoupling protein-1 expression in IWAT (0.05 < P < 0.1) suggesting that OT stimulates browning of WAT. To assess OT-elicited changes in brown adipose tissue (BAT) thermogenesis, we examined the effects of 4V OT on interscapular BAT temperature (TIBAT). 4V OT (1 µg) elevated TIBAT at 0.75 (P = 0.08), 1, and 1.25 h (P < 0.05) postinjection; a higher dose (5 µg) elevated TIBAT at 0.75-, 1-, 1.25-, 1.5-, 1.75- (P < 0.05), and 2-h (0.05 < P < 0.1) postinjection. Together, these findings support the hypothesis that chronic hindbrain OT treatment evokes sustained weight loss in DIO mice by reducing energy intake and increasing BAT thermogenesis at a dose that is not associated with evidence of visceral illness.


Subject(s)
Anti-Obesity Agents/administration & dosage , Diet, High-Fat , Obesity/drug therapy , Oxytocin/administration & dosage , Rhombencephalon/drug effects , Weight Loss/drug effects , Adipocytes, Brown/drug effects , Adipocytes, Brown/metabolism , Adipocytes, Brown/pathology , Adipocytes, White/drug effects , Adipocytes, White/metabolism , Adipocytes, White/pathology , Adiposity/drug effects , Animals , Disease Models, Animal , Eating/drug effects , Energy Intake/drug effects , Infusions, Intraventricular , Leptin/blood , Male , Mice, Inbred C57BL , Obesity/metabolism , Obesity/pathology , Obesity/physiopathology , Rhombencephalon/physiopathology , Thermogenesis/drug effects , Uncoupling Protein 1/metabolism
9.
Int J Mol Sci ; 22(2)2021 Jan 13.
Article in English | MEDLINE | ID: mdl-33451134

ABSTRACT

The catecholamine norepinephrine (NE) links hindbrain metabolic-sensory neurons with key glucostatic control structures in the brain, including the ventromedial hypothalamic nucleus (VMN). In the brain, the glycogen reserve is maintained within the astrocyte cell compartment as an alternative energy source to blood-derived glucose. VMN astrocytes are direct targets for metabolic stimulus-driven noradrenergic signaling due to their adrenergic receptor expression (AR). The current review discusses recent affirmative evidence that neuro-metabolic stability in the VMN may be shaped by NE influence on astrocyte glycogen metabolism and glycogen-derived substrate fuel supply. Noradrenergic modulation of estrogen receptor (ER) control of VMN glycogen phosphorylase (GP) isoform expression supports the interaction of catecholamine and estradiol signals in shaping the physiological stimulus-specific control of astrocyte glycogen mobilization. Sex-dimorphic NE control of glycogen synthase and GP brain versus muscle type proteins may be due, in part, to the dissimilar noradrenergic governance of astrocyte AR and ER variant profiles in males versus females. Forthcoming advances in the understanding of the molecular mechanistic framework for catecholamine stimulus integration with other regulatory inputs to VMN astrocytes will undoubtedly reveal useful new molecular targets in each sex for glycogen mediated defense of neuronal metabolic equilibrium during neuro-glucopenia.


Subject(s)
Astrocytes/metabolism , Carbohydrate Metabolism/drug effects , Glycogen/metabolism , Norepinephrine/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Astrocytes/drug effects , Gene Expression Regulation , Glucose/metabolism , Humans , Neurons/metabolism , Norepinephrine/pharmacology , Receptors, Adrenergic/genetics , Receptors, Adrenergic/metabolism , Rhombencephalon/drug effects , Rhombencephalon/metabolism , Signal Transduction/drug effects , Ventromedial Hypothalamic Nucleus/drug effects
10.
Endocrinology ; 161(11)2020 11 01.
Article in English | MEDLINE | ID: mdl-32894768

ABSTRACT

Malnutrition suppresses reproductive functions in mammals, which is considered to be mostly due to the inhibition of pulsatile gonadotropin-releasing hormone (GnRH)/gonadotropin secretion. Accumulating evidence suggests that kisspeptin neurons in the arcuate nucleus (ARC) play a critical role in the regulation of pulsatile GnRH/gonadotropin release. The present study aimed to examine if the hypothalamic dynorphin A (Dyn) neurons mediate the suppression of GnRH/luteinizing hormone (LH) pulses during malnutrition. Ovariectomized rats treated with a negative feedback level of estradiol-17ß-treated (OVX+E2) were administered with intravenous (iv) or fourth cerebroventricle (4V) 2-deoxy-D-glucose (2DG), an inhibitor of glucose utilization, to serve as a malnutrition model. Central administration of a Dyn receptor antagonist blocked the iv- or 4V-2DG-induced suppression of LH pulses in OVX+E2 rats. The 4V 2DG administration significantly increased the number of Pdyn (Dyn gene)-positive cells co-expressing fos in the paraventricular nucleus (PVN), but not in the ARC and supraoptic nucleus (SON), and the iv 2DG treatment significantly increased the number of fos and Pdyn-co-expressing cells in the PVN and SON, but decreased it in the ARC. The E2 treatment significantly increased Pdyn expression in the PVN, but not in the ARC and SON. Double in situ hybridization for Kiss1 (kisspeptin gene) and Oprk1 (Dyn receptor gene) revealed that around 60% of ARC Kiss1-expressing cells co-expressed Oprk1. These results suggest that the PVN Dyn neurons, at least in part, mediate LH pulse suppression induced by the hindbrain or peripheral glucoprivation, and Dyn neurons may directly suppress the ARC kisspeptin neurons in female rats.


Subject(s)
Dynorphins/metabolism , Food Deprivation/physiology , Luteinizing Hormone/metabolism , Neurons/physiology , Paraventricular Hypothalamic Nucleus , Rhombencephalon/metabolism , Animals , Down-Regulation/drug effects , Female , Glucose/metabolism , Glucose/pharmacology , Kisspeptins/metabolism , Malnutrition/metabolism , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Rats , Rats, Wistar , Rhombencephalon/drug effects
11.
Peptides ; 133: 170351, 2020 11.
Article in English | MEDLINE | ID: mdl-32579900

ABSTRACT

Hypothalamic orexin neurons project to many brain areas, including hindbrain structures such as the nucleus of the solitary tract (NTS) and area postrema (AP), where orexin 1 receptors (OX1Rs) are expressed. Hindbrain administration of orexin-A increases feeding and meal size, and blockade of hindbrain OX1Rs with the selective antagonist SB334867 has the opposite effect. Here we asked whether hindbrain OX1R stimulation or blockade alter rats' sensitivity to gastrointestinal satiety signals. Rats received 4th intracerebroventricular (icv) injections of vehicle or orexin-A, at a dose with no effect on its own, prior to an intragastric (IG) infusion of saline or a satiating volume of Ensure. IG Ensure suppressed subsequent chow intake, but orexin-A pretreatment significantly attenuated this IG nutrient-induced satiety at 2 h into the dark phase. In a second experiment, rats received NTS injections of vehicle or orexin-A before intraperitoneal (IP) injection of vehicle or the satiation hormone cholecystokinin (CCK). NTS orexin-A pretreatment completely blocked the intake-suppressive effect of CCK on dark-phase chow intake. Finally, we investigated the role of endogenous hindbrain OX1R activation by pretreating rats with 4th-icv injection of vehicle or SB334867 followed by IG infusion of saline or Ensure just before a chocolate Ensure licking test session. IG nutrient infusion suppressed Ensure intake, and blockade of hindbrain OX1Rs significantly prolonged that intake-suppressive effect. We conclude that hindbrain OX1Rs are a mechanism though which hypothalamic orexin neurons can reduce animals' sensitivity to gastrointestinal nutrient load, allowing them to consume more food.


Subject(s)
Cholecystokinin/metabolism , Orexin Receptors/metabolism , Rhombencephalon/physiology , Animals , Benzoxazoles/pharmacology , Chocolate , Cholecystokinin/pharmacology , Eating , Feeding Behavior/drug effects , Feeding Behavior/physiology , Male , Naphthyridines/pharmacology , Neurons/metabolism , Nutrients/metabolism , Orexin Receptor Antagonists/pharmacology , Orexins/metabolism , Orexins/pharmacology , Rats, Wistar , Rhombencephalon/drug effects , Rhombencephalon/metabolism , Urea/analogs & derivatives , Urea/pharmacology
12.
Am J Physiol Regul Integr Comp Physiol ; 318(6): R1068-R1077, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32320636

ABSTRACT

Severe trauma can produce a postinjury "metabolic self-destruction" characterized by catabolic metabolism and hyperglycemia. The severity of the hyperglycemia is highly correlated with posttrauma morbidity and mortality. Although no mechanism has been posited to connect severe trauma with a loss of autonomic control over metabolism, traumatic injury causes other failures of autonomic function, notably, gastric stasis and ulceration ("Cushing's ulcer"), which has been connected with the generation of thrombin. Our previous studies established that proteinase-activated receptors (PAR1; "thrombin receptors") located on astrocytes in the autonomically critical nucleus of the solitary tract (NST) can modulate gastric control circuit neurons to cause gastric stasis. Hindbrain astrocytes have also been implicated as important detectors of low glucose or glucose utilization. When activated, these astrocytes communicate with hindbrain catecholamine neurons that, in turn, trigger counterregulatory responses (CRR). There may be a convergence between the effects of thrombin to derange hindbrain gastrointestinal control and the hindbrain circuitry that initiates CRR to increase glycemia in reaction to critical hypoglycemia. Our results suggest that thrombin acts within the NST to increase glycemia through an astrocyte-dependent mechanism. Blockade of purinergic gliotransmission pathways interrupted the effect of thrombin to increase glycemia. Our studies also revealed that thrombin, acting in the NST, produced a rapid, dramatic, and potentially lethal suppression of respiratory rhythm that was also a function of purinergic gliotransmission. These results suggest that the critical connection between traumatic injury and a general collapse of autonomic regulation involves thrombin action on astrocytes.


Subject(s)
Astrocytes/drug effects , Blood Glucose , Neurons/drug effects , Rhombencephalon/drug effects , Thrombin/pharmacology , Animals , Male , Phrenic Nerve/drug effects , Rats , Rats, Sprague-Dawley , Respiratory Rate/drug effects , Solitary Nucleus/drug effects
13.
Am J Physiol Regul Integr Comp Physiol ; 318(5): R1014-R1023, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32292065

ABSTRACT

In rats, overnight fasting reduces the ability of systemic cholecystokinin-8 (CCK) to suppress food intake and to activate cFos in the caudal nucleus of the solitary tract (cNTS), specifically within glucagon-like peptide-1 (GLP-1) and noradrenergic (NA) neurons of the A2 cell group. Systemic CCK increases vagal sensory signaling to the cNTS, an effect that is amplified by leptin and reduced by ghrelin. Since fasting reduces plasma leptin and increases plasma ghrelin levels, we hypothesized that peripheral leptin administration and/or antagonism of ghrelin receptors in fasted rats would rescue the ability of CCK to activate GLP-1 neurons and a caudal subset of A2 neurons that coexpress prolactin-releasing peptide (PrRP). To test this, cFos expression was examined in ad libitum-fed and overnight food-deprived (DEP) rats after intraperitoneal CCK, after coadministration of leptin and CCK, or after intraperitoneal injection of a ghrelin receptor antagonist (GRA) before CCK. In fed rats, CCK activated cFos in ~60% of GLP-1 and PrRP neurons. Few or no GLP-1 or PrRP neurons expressed cFos in DEP rats treated with CCK alone, CCK combined with leptin, or GRA alone. However, GRA pretreatment increased the ability of CCK to activate GLP-1 and PrRP neurons and also enhanced the hypophagic effect of CCK in DEP rats. Considered together, these new findings suggest that reduced behavioral sensitivity to CCK in fasted rats is at least partially due to ghrelin-mediated suppression of hindbrain GLP-1 and PrRP neural responsiveness to CCK.


Subject(s)
Appetite Regulation/drug effects , Cholecystokinin/administration & dosage , Eating/drug effects , Fasting/metabolism , Feeding Behavior/drug effects , Ghrelin/blood , Neurons/drug effects , Rhombencephalon/drug effects , Animals , Glucagon-Like Peptide 1/metabolism , Leptin/blood , Male , Neurons/metabolism , Prolactin-Releasing Hormone/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Rats, Sprague-Dawley , Receptors, Ghrelin/metabolism , Rhombencephalon/metabolism , Signal Transduction
14.
Acta Neurobiol Exp (Wars) ; 80(1): 57-65, 2020.
Article in English | MEDLINE | ID: mdl-32214275

ABSTRACT

The catecholamine norepinephrine (NE) links hindbrain metabolic­sensory neurons with downstream gluco­regulatory loci, including the ventromedial hypothalamic nucleus (VMN). Exogenous NE up­regulates VMN expression of glutamate decarboxylase (GAD), biomarker for the gluco­inhibitory transmitter γ­aminobutryic acid (GABA). Brain glycogen phosphorylase (GP)­muscle (GPmm) and ­brain (GPbb) variants are stimulated in vitro by NE or energy deficiency, respectively. Current research investigated whether lactoprivic­driven VMN NE signaling regulates GABA and if VMN GPmm and GPbb profiles react differently to that deficit cue. Male rats were pretreated by caudal fourth ventricle delivery of the selective catecholamine neurotoxin 6­hydroxydopamine (6OHDA) ahead of the monocarboxylate transporter inhibitor alpha­cyano­4­hydroxycinnamic acid (4CIN). Micropunch­dissected VMN tissue was analyzed by Western blot and ELISA to assess NE­dependent 4CIN regulation of GAD and GP variant protein expression and NE activity. 4CIN caused 6OHDA­reversible augmentation of VMN NE content and plasma glucose and counter­regulatory hormone levels. 6OHDA stimulated basal VMN GAD expression, but prevented 4CIN stimulation of this profile. Neurotoxin inhibited or increased baseline VMN GPmm and GPbb levels, respectively, in non­4CIN­injected rats. 6OHDA deterred 4CIN inhibition of GPmm, but did not prevent drug stimulation of GPbb. Results affirm hindbrain lactoprivic regulation of glucostasis. Hindbrain NE exerts opposite effects on VMN GABA transmission during hindbrain lactostasis vs. ­privation. VMN norepinephrine­ vs. energy­sensitive GP variants are subject to dissimilar NE regulation during energy homeostasis, and respond differently to hindbrain lactoprivation.


Subject(s)
Glucose/metabolism , Glycogen/metabolism , Rhombencephalon/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Coumaric Acids/pharmacology , Dopamine beta-Hydroxylase/biosynthesis , Dopamine beta-Hydroxylase/genetics , Enzyme Induction/drug effects , Glutamate Decarboxylase/biosynthesis , Glutamate Decarboxylase/genetics , Glycogen Phosphorylase/metabolism , Isoenzymes/biosynthesis , Isoenzymes/genetics , Male , Norepinephrine/pharmacology , Norepinephrine/physiology , Oxidopamine/toxicity , Rats , Rats, Sprague-Dawley , Rhombencephalon/drug effects
15.
Int J Biol Macromol ; 151: 855-860, 2020 May 15.
Article in English | MEDLINE | ID: mdl-32068062

ABSTRACT

The aim of this study was to evaluate the spatial structure and potential antifatigue activity of polysaccharide fractions which was extracted from Inonotus obliquus. The first polysaccharide fractions of Inonotus obliquus (PIO-1) were obtained after hot-water extraction and purification by DEAE cellulose-52 chromatography. Results of the forced swimming test showed that the doses (50 mg/kg) of PIO-1 could increase the climbing duration and swimming time as well as reduced the immobility time in the PIO treated mice. The fatigue related metabolic parameters showed that PIO-1 decreased the level of blood lactic acid (BLA), urea nitrogen (BUN) and lactic dehydrogenase (LDH). Additionally, PIO-1 significantly decreased the 5-HT concentrations in the mice brain. The results of monosaccharide analysis showed that the molar ratio of mannose, glucose, galactose, xylose and arabinose with the molar ratio of 1.0:1.9:3.5:18.5:5.7. The molecular morphology of the PIO-1 observed under atomic force microscopy (AFM). There were many spherical and heterogeneous clumps existed in the images. Therefore, current study indicated polysaccharide PIO-1 not only has great potential to postpone physical fatigue but also shown potential to improve mental fatigue.


Subject(s)
Fungal Polysaccharides/chemistry , Fungal Polysaccharides/pharmacology , Inonotus/chemistry , Animals , Biomarkers , Fatigue/drug therapy , Fungal Polysaccharides/isolation & purification , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Mice , Microscopy, Atomic Force , Molecular Structure , Monosaccharides/analysis , Monosaccharides/chemistry , Muscle, Skeletal/metabolism , Rhombencephalon/drug effects , Rhombencephalon/metabolism , Structure-Activity Relationship
16.
Sci Rep ; 9(1): 16347, 2019 11 08.
Article in English | MEDLINE | ID: mdl-31705012

ABSTRACT

Thirst has evolved for vertebrate terrestrial adaptation. We previously showed that buccal drying induced a series of drinking behaviours (migration to water-taking water into the mouth-swallowing) in the amphibious mudskipper goby, thereby discovering thirst in ray-finned fish. However, roles of dipsogenic/antidipsogenic hormones, which act on the thirst center in terrestrial tetrapods, have remained unclear in the mudskipper thirst. Here we examined the hormonal effects on the mudskipper drinking behaviours, particularly the antagonistic interaction between angiotensin II (AngII) and atrial natriuretic peptide (ANP) which is important for thirst regulation in mammalian 'forebrain'. Expectedly, intracerebroventricular injection of ANP in mudskippers reduced AngII-increased drinking rate. ANP also suppressed the neural activity at the 'hindbrain' region for the swallowing reflex, and the maintenance of buccopharyngeal water due to the swallowing inhibition may attenuate the motivation to move to water. Thus, the hormonal molecules involved in drinking regulation, as well as the influence of buccopharyngeal water, appear to be conserved in distantly related species to solve osmoregulatory problems, whereas hormonal control of thirst at the forebrain might have been acquired only in tetrapod lineage during evolution.


Subject(s)
Angiotensin II/administration & dosage , Atrial Natriuretic Factor/administration & dosage , Biological Evolution , Drinking Behavior/physiology , Ecosystem , Thirst/physiology , Water-Electrolyte Balance/physiology , Animals , Drinking Behavior/drug effects , Prosencephalon/drug effects , Prosencephalon/physiology , Rhombencephalon/drug effects , Rhombencephalon/physiology , Skates, Fish , Thirst/drug effects , Vasoconstrictor Agents/administration & dosage , Water-Electrolyte Balance/drug effects
17.
Neuroscience ; 411: 211-221, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31085279

ABSTRACT

Neural substrates for estrogen regulation of glucose homeostasis remain unclear. Female rat dorsal vagal complex (DVC) A2 noradrenergic neurons are estrogen- and metabolic-sensitive. The ventromedial hypothalamic nucleus (VMN) is a key component of the brain network that governs counter-regulatory responses to insulin-induced hypoglycemia (IIH). Here, the selective estrogen receptor-alpha (ERα) or -beta (ERß) antagonists MPP and PHTPP were administered separately to the caudal fourth ventricle to address the premise that these hindbrain ER variants exert distinctive control of VMN reactivity to IIH in the female sex. Data show that ERα governs hypoglycemic patterns of VMN astrocyte glycogen metabolic enzyme, e.g. glycogen synthase and phosphorylase protein expression, whereas ERß mediates local glycogen breakdown. DVC ERs also regulate VMN neurotransmitter signaling of energy sufficiency [γ-aminobutyric acid] or deficiency [nitric oxide, steroidogenic factor-1] during IIH. Neither hindbrain ER mediates IIH-associated diminution of VMN norepinephrine (NE) content. Both ERs oppose hypoglycemic hyperglucagonemia, while ERß contributes to reduced corticosterone output. Outcomes reveal that input from the female hindbrain to the VMN is critical for energy reserve mobilization, metabolic transmitter signaling, and counter-regulatory hormone secretion during hypoglycemia, and that ERs control those cues. Evidence that VMN NE content is not controlled by hindbrain ERα or -ß implies that these receptors may regulate VMN function via NE-independent mechanisms, or alternatively, that other neurotransmitter signals to the VMN may control local substrate receptivity to NE.


Subject(s)
Glycogen/metabolism , Hypoglycemia/metabolism , Receptors, Estrogen/metabolism , Rhombencephalon/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Estrogen Receptor Antagonists/pharmacology , Female , Nitric Oxide Synthase Type I/metabolism , Piperidines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Rats , Rhombencephalon/drug effects , Steroidogenic Factor 1/metabolism , Ventromedial Hypothalamic Nucleus/drug effects
18.
Neuroscience ; 409: 253-260, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30954669

ABSTRACT

Estrogen receptor-alpha (ERα) and -beta (ERß) occur in key elements of the brain gluco-homeostatic network in both sexes, including the hindbrain dorsal vagal complex (DVC), but the influence of distinct receptor populations on this critical function is unclear. The ventromedial hypothalamic nucleus (VMN) maintains glucose balance by integrating nutrient, endocrine, and neurochemical cues, including metabolic sensory information supplied by DVC A2 noradrenergic neurons. Current research utilized the selective ERα and ERß antagonists MPP and PHTPP to characterize effects of DVC ERs on VMN norepinephrine (NE) activity and metabolic neurotransmitter signaling in insulin-induced hypoglycemic (IIH) male rats. Data show that ERß inhibits VMN glycogen synthase and stimulates phosphorylase protein expression, while attenuating hypoglycemic augmentation of glycogen content. Furthermore, both ERs attenuate VMN glucose concentrations during IIH. Hypoglycemic up-regulation of nitric oxide (NO) and brain-derived neurotrophic factor (BDNF) signaling was correspondingly driven by ERα or -ß, whereas GABA and steroidogenic factor-1 were respectively suppressed independently of ER input or by ERß. IIH intensified VMN NE accumulation by ERß-dependent mechanisms, but did not alter NE levels in other gluco-regulatory loci. ERß amplified the magnitude of insulin-induced decline in blood glucose. Both ERs regulate corticosterone, but not glucagon secretion during IIH and oppose hypoglycemic diminution of circulating free fatty acids. These findings identify distinguishing versus common VMN functions targeted by DVC ERα and -ß. Sex differences in hypoglycemic VMN NE accumulation, glycogen metabolism, and transmitter signaling may involve, in part, discrepant regulatory involvement or differential magnitude of impact of these hindbrain ERs.


Subject(s)
Glucose/metabolism , Glycogen/metabolism , Hypoglycemia/metabolism , Receptors, Estrogen/metabolism , Rhombencephalon/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Animals , Brain-Derived Neurotrophic Factor/metabolism , Corticosterone/metabolism , Male , Nitric Oxide/metabolism , Norepinephrine/metabolism , Piperidines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Estrogen/antagonists & inhibitors , Rhombencephalon/drug effects , Ventromedial Hypothalamic Nucleus/drug effects
19.
Physiol Behav ; 206: 191-199, 2019 07 01.
Article in English | MEDLINE | ID: mdl-30980855

ABSTRACT

Glucagon-like peptide 1 receptors (GLP-1R) are expressed in the lateral septum (LS) of rats and mice, and we have published that endogenous LS GLP-1 affects feeding and motivation for food in rats. Here we asked if these effects are also observed in mice. In separate dose-response studies using male C57Bl6J mice, intra-LS GLP-1 or the GLP-1R antagonist Exendin 9 (Ex9) was delivered shortly before dark onset, at doses subthreshold for effect when injected intracerebroventricularly (icv). Intra-LS GLP-1 significantly suppressed chow intake early in the dark phase and tended to reduce overnight intake. However, blockade of LS GLP-1R with Ex9 had no effect on ad libitum dark onset chow intake. We then asked if LS GLP-1R blockade blunts nutrient preload-induced intake suppression. Mice were trained to consume Ensure immediately before dark onset, which suppressed subsequent chow intake, and intra-LS Ex9 attenuated that preload-induced intake suppression. We also found that restraint stress robustly activates hindbrain GLP-1-producing neurons, and that LS GLP-1R blockade attenuates 30-min restraint stress-induced hypophagia in mice. Furthermore, we have reported that in the rat, GLP-1R in the dorsal subregion of the LS (dLS) affect motivation for food. We examined this in food-restricted mice responding for sucrose pellets on a progressive ratio (PR) schedule. Intra-dLS GLP-1R stimulation significantly suppressed, and Ex9 significantly increased, operant responding, and the Ex9 effect remained after mice returned to ad libitum conditions. Similarly, we found that stimulation of dLS GLP-1 suppressed licking for sucrose and conversely, Ex9 increased licking under ad libitum feeding conditions. Together, our data suggest that endogenous activation of LS GLP-1R plays a role in feeding in mice under some but not all conditions, and that these receptors strongly influence motivation for food.


Subject(s)
Eating/drug effects , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Motivation/drug effects , Satiation/drug effects , Septal Nuclei/drug effects , Animals , Mice , Neurons/drug effects , Restraint, Physical , Rhombencephalon/drug effects , Stress, Psychological
20.
Neuropsychopharmacology ; 44(10): 1742-1751, 2019 09.
Article in English | MEDLINE | ID: mdl-30959513

ABSTRACT

The overlap in neurobiological circuitry mediating the physiological and behavioral response to satiation and noxious/stressful stimuli are not well understood. The interaction between serotonin (5-HT) and glucagon-like peptide-1 (GLP-1) could play a role as upstream effectors involved in mediating associations between anorectic and noxious/stressful stimuli. We hypothesize that 5-HT acts as an endogenous modulator of the central GLP-1 system to mediate satiation and malaise in rats. Here, we investigate whether interactions between central 5-HT and GLP-1 signaling are behaviorally and physiologically relevant for the control of food intake and pica (i.e., behavioral measure of malaise). Results show that the anorexia and body weight changes induced by administration of exogenous hindbrain 5-HT are dependent on central GLP-1 receptor signaling. Furthermore, anatomical evidence shows mRNA expression of 5-HT2C and 5-HT3 receptors on GLP-1-producing preproglucagon (PPG) neurons in the medial nucleus tractus solitarius by fluorescent in situ hybridization, suggesting that PPG neurons are likely to express both of these receptors. Behaviorally, the hypophagia induced by the pharmacological activation of both of these receptors is also dependent on GLP-1 signaling. Finally, 5-HT3, but not 5-HT2C receptors, are required for the anorectic effects of the interoceptive stressor LiCl, suggesting the hypophagia induced by these 5-HT receptors may be driven by different mechanisms. Our findings highlight 5-HT as a novel endogenous modulator of the central GLP-1 system and suggest that the central interaction between 5-HT and GLP-1 is involved in the control of food intake in rats.


Subject(s)
Feeding Behavior/drug effects , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/antagonists & inhibitors , Receptor, Serotonin, 5-HT2C/metabolism , Receptors, Serotonin, 5-HT3/metabolism , Serotonin/pharmacology , Stress, Psychological/metabolism , Weight Loss/drug effects , Animals , Anorexia/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , Lithium Chloride/pharmacology , Male , Neurons/drug effects , Neurons/metabolism , Ondansetron/pharmacology , Peptide Fragments/pharmacology , Pica/metabolism , Proglucagon , Rats , Rhombencephalon/drug effects , Rhombencephalon/metabolism , Serotonin/metabolism , Serotonin 5-HT2 Receptor Agonists/pharmacology , Serotonin 5-HT3 Receptor Agonists/pharmacology , Serotonin 5-HT3 Receptor Antagonists/pharmacology , Signal Transduction , Solitary Nucleus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL