78 results on '"Dunn-Meynell AA"'
Search Results
2. The F.DIO obesity-prone rat is insulin resistant prior to obesity onset
- Author
-
Levin, Be, Magnan, Christophe, Migrenne, S., Chua, Jr Sc, Dunn-Meynell, Aa, Douared, Laetitia, Laboratoire de physiopathologie de la nutrition (LPN), and Centre National de la Recherche Scientifique (CNRS)-Université Paris Diderot - Paris 7 (UPD7)
- Published
- 2005
3. Role of neuronal glucosensing in the regulation of energy homeostasis.
- Author
-
Levin BE, Kang L, Sanders NM, and Dunn-Meynell AA
- Abstract
Glucosensing is a property of specialized neurons in the brain that regulate their membrane potential and firing rate as a function of ambient glucose levels. These neurons have several similarities to beta- and alpha-cells in the pancreas, which are also responsive to ambient glucose levels. Many use glucokinase as a rate-limiting step in the production of ATP and its effects on membrane potential and ion channel function to sense glucose. Glucosensing neurons are organized in an interconnected distributed network throughout the brain that also receives afferent neural input from glucosensors in the liver, carotid body, and small intestines. In addition to glucose, glucosensing neurons can use other metabolic substrates, hormones, and peptides to regulate their firing rate. Consequently, the output of these 'metabolic sensing' neurons represents their integrated response to all of these simultaneous inputs. The efferents of these neurons regulate feeding, neuroendocrine and autonomic function, and thereby energy expenditure and storage. Thus, glucosensing neurons play a critical role in the regulation of energy homeostasis. Defects in the ability to sense glucose and regulatory hormones like leptin and insulin may underlie the predisposition of some individuals to develop diet-induced obesity. [ABSTRACT FROM AUTHOR]
- Published
- 2006
- Full Text
- View/download PDF
4. Erythropoietin-derived peptide treatment reduced neurological deficit and neuropathological changes in a mouse model of tauopathy.
- Author
-
Choi YB, Dunn-Meynell AA, Marchese M, Blumberg BM, Gaindh D, Dowling PC, and Lu W
- Subjects
- Animals, Brain metabolism, Disease Models, Animal, Female, Humans, Mice, Mice, Transgenic, tau Proteins genetics, tau Proteins metabolism, Erythropoietin, Tauopathies drug therapy
- Abstract
Background: Prominent activation of microglial immune/inflammatory processes is a characteristic feature of brains of patients with tauopathies including Alzheimer's disease (AD), suggesting that neuroinflammation may be a critical factor in their pathogenesis. Strategies aimed at developing new therapeutics for tauopathies based on anti-inflammation or immunomodulation are likely to be promising avenues of research. We previously developed JM4-a 19'mer cyclic peptide derived from the first loop of human erythropoietin. This peptide possesses beneficial immune modulatory and tissue protective effects while lacking the undesirable side effects of full-length erythropoietin. In this preclinical study, we investigated the effect of chronic JM4 treatment on the PS19 mouse that carries the P301S mutant human tau gene, linked to a form of frontotemporal dementia. This transgenic mouse has been widely used as a model of tauopathies including AD and related dementias., Methods: Daily subcutaneous treatment of female PS19 mice with JM4 was initiated before disease onset and continued on for the animals' lifespan. The progression of neurological deficit and the lifespan of these mice were assessed. To evaluate the effect of JM4 treatment on cognition of these animals, the PS19 mice underwent Barnes maze test and elevated plus maze test. In addition, neuronal loss, phosphorylated tau aggregation, and microglial activation were assessed using immunohistochemistry of PS19 mouse brain sections., Results: JM4 treatment of PS19 mice initiated before disease onset reduced neurological deficit, prolonged lifespan, and rescued memory impairment. The beneficial effects of JM4 were accompanied by reductions in neuronal loss, phosphorylated tau aggregation, and microglial activation in the PS19 mouse brain., Limitations: Use of a single dose of JM4 and female mice only., Conclusion: JM4 is a potential novel therapeutic agent for the treatment of tauopathies including AD and related dementias.
- Published
- 2021
- Full Text
- View/download PDF
5. In vivo Bioluminescence Imaging Used to Monitor Disease Activity and Therapeutic Response in a Mouse Model of Tauopathy.
- Author
-
Dunn-Meynell AA, Dowling P, Marchese M, Rodriguez E, Blumberg B, Choi YB, Gaindh D, and Lu W
- Abstract
Many studies of tauopathy use transgenic mice that overexpress the P301S mutant form of tau. Neuronal damage in these mice is associated with astrogliosis and induction of glial fibrillary acidic protein (GFAP) expression. GFAP-luc transgenic mice express firefly luciferase under the GFAP promoter, allowing bioluminescence to be measured non-invasively as a surrogate biomarker for astrogliosis. We bred double transgenic mice possessing both P301S and GFAP-luc cassettes and compared them to control mice bearing only the GFAP-luc transgene. We used serial bioluminescent images to define the onset and the time course of astrogliosis in these mice and this was correlated with the development of clinical deficit. Mice containing both GFAP-luc and P301S transgenes showed increased luminescence indicative of astroglial activation in the brain and spinal cord. Starting at 5 months old, the onset of clinical deterioration in these mice corresponded closely to the initial rise in the luminescent signal. Post mortem analysis showed the elevated luminescence was correlated with hyperphosphorylated tau deposition in the hippocampus of double transgenic mice. We used this method to determine the therapeutic effect of JM4 peptide [a small peptide immunomodulatory agent derived from human erythropoietin (EPO)] on double transgenic mice. JM4 treatment significantly decreased the intensity of luminescence, neurological deficit and hyperphosphorylated tau in mice with both the P301S and GFAP-luc transgenes. These findings indicate that bioluminescence imaging (BLI) is a powerful tool for quantifying GFAP expression in living P301S mice and can be used as a noninvasive biomarker of tau-induced neurodegeneration in preclinical therapeutic trials., (Copyright © 2019 Dunn-Meynell, Dowling, Marchese, Rodriguez, Blumberg, Choi, Gaindh and Lu.)
- Published
- 2019
- Full Text
- View/download PDF
6. Amylin Selectively Signals Onto POMC Neurons in the Arcuate Nucleus of the Hypothalamus.
- Author
-
Lutz TA, Coester B, Whiting L, Dunn-Meynell AA, Boyle CN, Bouret SG, Levin BE, and Le Foll C
- Subjects
- Agouti-Related Protein metabolism, Animals, Animals, Newborn, Feeding Behavior, Female, Islet Amyloid Polypeptide genetics, Male, Mice, Mice, Knockout, Mice, Obese, Neuropeptide Y metabolism, Rats, Sprague-Dawley, Receptor Activity-Modifying Protein 1 genetics, Receptor Activity-Modifying Protein 1 metabolism, Receptor Activity-Modifying Protein 3 genetics, Receptor Activity-Modifying Protein 3 metabolism, Ventromedial Hypothalamic Nucleus metabolism, alpha-MSH metabolism, Arcuate Nucleus of Hypothalamus metabolism, Islet Amyloid Polypeptide metabolism, Leptin metabolism, MAP Kinase Signaling System, Neurons metabolism, Paraventricular Hypothalamic Nucleus metabolism, Pro-Opiomelanocortin metabolism
- Abstract
Amylin phosphorylates ERK (p-ERK) in the area postrema to reduce eating and synergizes with leptin to phosphorylate STAT3 in the arcuate (ARC) and ventromedial (VMN) hypothalamic nuclei to reduce food intake and body weight. The current studies assessed potential amylin and amylin-leptin ARC/VMN interactions on ERK signaling and their roles in postnatal hypothalamic pathway development. In amylin knockout mice, the density of agouti-related protein (AgRP)-immunoreactive (IR) fibers in the hypothalamic paraventricular nucleus (PVN) was increased, while the density of α-melanocyte-stimulating hormone (αMSH) fibers was decreased. In mice deficient of the amylin receptor components RAMP1/3, both AgRP and αMSH-IR fiber densities were decreased, while only αMSH-IR fiber density was decreased in rats injected neonatally in the ARC/VMN with an adeno-associated virus short hairpin RNA against the amylin core receptor. Amylin induced p-ERK in ARC neurons, 60% of which was present in POMC-expressing neurons, with none in NPY neurons. An amylin-leptin interaction was shown by an additive effect on ARC ERK signaling in neonatal rats and a 44% decrease in amylin-induced p-ERK in the ARC of leptin receptor-deficient and of ob/ob mice. Together, these results suggest that amylin directly acts, through a p-ERK-mediated process, on POMC neurons to enhance ARC-PVN αMSH pathway development., (© 2018 by the American Diabetes Association.)
- Published
- 2018
- Full Text
- View/download PDF
7. Early postnatal amylin treatment enhances hypothalamic leptin signaling and neural development in the selectively bred diet-induced obese rat.
- Author
-
Johnson MD, Bouret SG, Dunn-Meynell AA, Boyle CN, Lutz TA, and Levin BE
- Subjects
- Animals, Arcuate Nucleus of Hypothalamus drug effects, Arcuate Nucleus of Hypothalamus pathology, Arcuate Nucleus of Hypothalamus physiopathology, Body Weight drug effects, Diet, High-Fat, Dietary Fats, Female, Hypothalamus drug effects, Hypothalamus pathology, Islet Amyloid Polypeptide pharmacology, Male, Neurogenesis drug effects, Paraventricular Hypothalamic Nucleus drug effects, Paraventricular Hypothalamic Nucleus pathology, Paraventricular Hypothalamic Nucleus physiopathology, Postnatal Care, Rats, Treatment Outcome, Hypothalamus physiopathology, Islet Amyloid Polypeptide administration & dosage, Leptin metabolism, Obesity drug therapy, Obesity physiopathology
- Abstract
Selectively bred diet-induced obese (DIO) rats become obese on a high-fat diet and are leptin resistant before becoming obese. Compared with diet-resistant (DR) neonates, DIO neonates have impaired leptin-dependent arcuate (ARC) neuropeptide Y/agouti-related peptide (NPY/AgRP) and α-melanocyte-stimulating hormone (α-MSH; from proopiomelanocortin (POMC) neurons) axon outgrowth to the paraventricular nucleus (PVN). Using phosphorylation of STAT3 (pSTAT3) as a surrogate, we show that reduced DIO ARC leptin signaling develops by postnatal day 7 (P7) and is reduced within POMC but not NPY/AgRP neurons. Since amylin increases leptin signaling in adult rats, we treated DIO neonates with amylin during postnatal hypothalamic development and assessed leptin signaling, leptin-dependent ARC-PVN pathway development, and metabolic changes. DIO neonates treated with amylin from P0-6 and from P0-16 increased ARC leptin signaling and both AgRP and α-MSH ARC-PVN pathway development, but increased only POMC neuron number. Despite ARC-PVN pathway correction, P0-16 amylin-induced reductions in body weight did not persist beyond treatment cessation. Since amylin enhances adult DIO ARC signaling via an IL-6-dependent mechanism, we assessed ARC-PVN pathway competency in IL-6 knockout mice and found that the AgRP, but not the α-MSH, ARC-PVN pathway was reduced. These results suggest that both leptin and amylin are important neurotrophic factors for the postnatal development of the ARC-PVN pathway. Amylin might act as a direct neurotrophic factor in DIO rats to enhance both the number of POMC neurons and their α-MSH ARC-PVN pathway development. This suggests important and selective roles for amylin during ARC hypothalamic development.
- Published
- 2016
- Full Text
- View/download PDF
8. IL-6 ameliorates defective leptin sensitivity in DIO ventromedial hypothalamic nucleus neurons.
- Author
-
Larsen L, Le Foll C, Dunn-Meynell AA, and Levin BE
- Subjects
- Animals, Cells, Cultured, Dietary Fats, Male, Obesity chemically induced, Rats, Rats, Sprague-Dawley, Interleukin-6 immunology, Leptin immunology, Neurons metabolism, Obesity immunology, Receptors, Leptin metabolism, Ventromedial Hypothalamic Nucleus immunology
- Abstract
Rats selectively bred to develop diet-induced obesity (DIO) have an early onset reduction in the sensitivity of their ventromedial hypothalamic nucleus (VMN) neurons to leptin compared with diet-resistant (DR) rats. This reduced sensitivity includes decreased leptin receptor (Lepr-b) mRNA expression, leptin receptor binding, leptin-induced phosphorylation of STAT3 (pSTAT3), and impaired leptin excitation (LepE) of VMN neurons. When administered exogenously, the pancreatic peptide, amylin, acts synergistically to reduce food intake and body weight in obese, leptin-resistant DIO rats by increasing VMN leptin signaling, likely by stimulation of microglia IL-6, which acts on its receptor to increase leptin-induced pSTAT3. Here, we demonstrate that incubation of cultured VMN neurons of outbred rats with IL-6 increases their leptin sensitivity. Control, dissociated DIO VMN neurons express 66% less Lepr-b and 75% less Bardet Biedl Syndrome-6 (BBS6) mRNA and have reduced leptin-induced activation of LepE neurons compared with DR neurons. Incubation for 4 days with IL-6 increased DIO neuron Lepr-b expression by 77% and BBS6 by 290% and corrected their defective leptin activation of LepE neurons to DR levels. Since BBS6 enhances trafficking of Lepr-b to the cell membrane, the increases in Lepr-b and BBS6 expression appear to account for correction of the reduced leptin excitation of DIO LepE neurons to that of control DR rats. These data support prior findings suggesting that IL-6 mediates the leptin-sensitizing effects of amylin on VMN neurons and that the inherent leptin resistance of DIO rats can be effectively reversed at a cellular level by IL-6.
- Published
- 2016
- Full Text
- View/download PDF
9. Endogenous VMH amylin signaling is required for full leptin signaling and protection from diet-induced obesity.
- Author
-
Dunn-Meynell AA, Le Foll C, Johnson MD, Lutz TA, Hayes MR, and Levin BE
- Subjects
- Animals, Arcuate Nucleus of Hypothalamus metabolism, Diet, High-Fat, Eating, Glucose Intolerance genetics, Insulin Resistance genetics, Iodine Radioisotopes, Islet Amyloid Polypeptide genetics, Leptin genetics, Male, Obesity genetics, RNA, Small Interfering genetics, Radionuclide Imaging, Rats, Receptors, Calcitonin genetics, Receptors, Calcitonin metabolism, STAT3 Transcription Factor genetics, Ventromedial Hypothalamic Nucleus diagnostic imaging, Weight Gain, Islet Amyloid Polypeptide metabolism, Leptin metabolism, Obesity physiopathology, Ventromedial Hypothalamic Nucleus metabolism
- Abstract
Amylin enhances arcuate (ARC) and ventromedial (VMN) hypothalamic nuclei leptin signaling and synergistically reduces food intake and body weight in selectively bred diet-induced obese (DIO) rats. Since DIO (125)I-amylin dorsomedial nucleus-dorsomedial VMN binding was reduced, we postulated that this contributed to DIO ventromedial hypothalamus (VMH) leptin resistance, and that impairing VMH (ARC + VMN) calcitonin receptor (CTR)-mediated signaling by injecting adeno-associated virus (AAV) expressing a short hairpin portion of the CTR mRNA would predispose diet-resistant (DR) rats to obesity on high-fat (45%) diet (HFD). Depleting VMH CTR by 80-90% in 4-wk-old male DR rats reduced their ARC and VMN (125)I-labeled leptin binding by 57 and 51%, respectively, and VMN leptin-induced phospho-signal transducer and activator of transcription 3-positive neurons by 59% vs. AAV control rats. After 6 wk on chow, VMH CTR-depleted DR rats ate and gained the equivalent amount of food and weight but had 18% heavier fat pads (relative to carcass weight), 144% higher leptin levels, and were insulin resistant compared with control AAV DR rats. After 6 wk more on HFD, VMH CTR-depleted DR rats ate the same amount but gained 28% more weight, had 60% more carcass fat, 254% higher leptin levels, and 132% higher insulin areas under the curve during an oral glucose tolerance test than control DR rats. Therefore, impairing endogenous VMH CTR-mediated signaling reduced leptin signaling and caused DR rats to become more obese and insulin resistant, both on chow and HFD. These results suggest that endogenous VMH amylin signaling is required for full leptin signaling and protection from HFD-induced obesity.
- Published
- 2016
- Full Text
- View/download PDF
10. Role of VMH ketone bodies in adjusting caloric intake to increased dietary fat content in DIO and DR rats.
- Author
-
Le Foll C, Dunn-Meynell AA, Miziorko HM, and Levin BE
- Subjects
- 3-Hydroxybutyric Acid pharmacology, Animals, Calcium metabolism, Diet, High-Fat, Glucose pharmacology, Male, Neurons drug effects, Obesity etiology, Oleic Acid pharmacology, Rats, Rats, Sprague-Dawley, Ventromedial Hypothalamic Nucleus drug effects, Energy Intake physiology, Ketone Bodies metabolism, Neurons metabolism, Obesity metabolism, Ventromedial Hypothalamic Nucleus metabolism
- Abstract
The objective of this study was to determine the potential role of astrocyte-derived ketone bodies in regulating the early changes in caloric intake of diet induced-obese (DIO) versus diet-resistant (DR) rats fed a 31.5% fat high-energy (HE) diet. After 3 days on chow or HE diet, DR and DIO rats were assessed for their ventromedial hypothalamic (VMH) ketone bodies levels and neuronal ventromedial hypothalamic nucleus (VMN) sensing using microdialysis coupled to continuous food intake monitoring and calcium imaging in dissociated neurons, respectively. DIO rats ate more than DR rats over 3 days of HE diet intake. On day 3 of HE diet intake, DR rats reduced their caloric intake while DIO rats remained hyperphagic. Local VMH astrocyte ketone bodies production was similar between DR and DIO rats during the first 6 h after dark onset feeding but inhibiting VMH ketone body production in DR rats on day 3 transiently returned their intake of HE diet to the level of DIO rats consuming HE diet. In addition, dissociated VMN neurons from DIO and DR rats were equally sensitive to the largely excitatory effects of β-hydroxybutyrate. Thus while DR rats respond to increased VMH ketone levels by decreasing their intake after 3 days of HE diet, this is not the case of DIO rats. These data suggest that DIO inherent leptin resistance prevents ketone bodies inhibitory action on food intake.
- Published
- 2015
- Full Text
- View/download PDF
11. Amylin-induced central IL-6 production enhances ventromedial hypothalamic leptin signaling.
- Author
-
Le Foll C, Johnson MD, Dunn-Meynell AA, Boyle CN, Lutz TA, and Levin BE
- Subjects
- Animals, Astrocytes metabolism, Gene Expression Regulation drug effects, Gene Expression Regulation physiology, Interleukin-6 genetics, Leptin genetics, Male, Mice, Mice, Knockout, Microglia metabolism, Neurons metabolism, RNA, Messenger, Rats, Rats, Sprague-Dawley, STAT3 Transcription Factor genetics, STAT3 Transcription Factor metabolism, Ventromedial Hypothalamic Nucleus cytology, Interleukin-6 metabolism, Islet Amyloid Polypeptide pharmacology, Leptin metabolism, Signal Transduction physiology, Ventromedial Hypothalamic Nucleus metabolism
- Abstract
Amylin acts acutely via the area postrema to reduce food intake and body weight, but it also interacts with leptin over longer periods of time, possibly via the ventromedial hypothalamus (VMH), to increase leptin signaling and phosphorylation of STAT3. We postulated that amylin enhances VMH leptin signaling by inducing interleukin (IL)-6, which then interacts with its gp130 receptor to activate STAT3 signaling and gene transcription downstream of the leptin receptor. We found that components of the amylin receptor (RAMPs1-3, CTR1a,b) are expressed in cultured VMH astrocytes, neurons, and microglia, as well as in micropunches of arcuate and ventromedial hypothalamic nuclei (VMN). Amylin exposure for 5 days increased IL-6 mRNA expression in VMH explants and microglia by two- to threefold, respectively, as well as protein abundance in culture supernatants by five- and twofold, respectively. Amylin had no similar effects on cultured astrocytes or neurons. In rats, 5 days of amylin treatment decreased body weight gain and/or food intake and increased IL-6 mRNA expression in the VMN. Similar 5-day amylin treatment increased VMN leptin-induced phosphorylation of STAT3 expression in wild-type mice and rats infused with lateral ventricular IgG but not in IL-6 knockout mice or rats infused with ventricular IL-6 antibody. Lateral ventricular infusion of IL-6 antibody also prevented the amylin-induced decrease of body weight gain. These results show that amylin-induced VMH microglial IL-6 production is the likely mechanism by which amylin treatment interacts with VMH leptin signaling to increase its effect on weight loss., (© 2015 by the American Diabetes Association. Readers may use this article as long as the work is properly cited, the use is educational and not for profit, and the work is not altered.)
- Published
- 2015
- Full Text
- View/download PDF
12. Role of FAT/CD36 in fatty acid sensing, energy, and glucose homeostasis regulation in DIO and DR rats.
- Author
-
Le Foll C, Dunn-Meynell AA, and Levin BE
- Subjects
- Adipose Tissue metabolism, Animal Feed, Animals, Body Weight physiology, Disease Models, Animal, Insulin metabolism, Leptin, Male, Rats, Blood Glucose metabolism, CD36 Antigens metabolism, Energy Intake physiology, Fatty Acids metabolism, Homeostasis physiology, Obesity metabolism
- Abstract
Hypothalamic fatty acid (FA) sensing neurons alter their activity utilizing the FA translocator/receptor, FAT/CD36. Depletion of ventromedial hypothalamus (VMH) CD36 with adeno-associated viral vector expressing CD36 shRNA (AAV CD36 shRNA) leads to redistribution of adipose stores and insulin resistance in outbred rats. This study assessed the requirement of VMH CD36-mediated FA sensing for the regulation of energy and glucose homeostasis in postnatal day 5 (P5) and P21 selectively bred diet-induced obese (DIO) and diet-resistant (DR) rats using VMH AAV CD36 shRNA injections. P5 CD36 depletion altered VMH neuronal FA sensing predominantly in DIO rats. After 10 wk on a 45% fat diet, DIO rats injected with VMH AAV CD36 shRNA at P21 ate more and gained more weight than DIO AAV controls, while DR AAV CD36 shRNA-injected rats gained less weight than DR AAV controls. VMH CD36 depletion increased inguinal fat pad weights and leptin levels in DIO and DR rats. Although DR AAV CD36 shRNA-injected rats became as obese as DIO AAV controls, only DIO control and CD36 depleted rats became insulin-resistant on a 45% fat diet. VMH CD36 depletion stunted linear growth in DIO and DR rats. DIO rats injected with AAV CD36 shRNA at P5 had increased fat mass, mostly due to a 45% increase in subcutaneous fat. They were also insulin-resistant with an associated 71% increase of liver triglycerides. These results demonstrate that VMH CD36-mediated FA sensing is a critical factor in the regulation of energy and glucose homeostasis and fat deposition in DIO and DR rats.
- Published
- 2015
- Full Text
- View/download PDF
13. Regulation of hypothalamic neuronal sensing and food intake by ketone bodies and fatty acids.
- Author
-
Le Foll C, Dunn-Meynell AA, Miziorko HM, and Levin BE
- Subjects
- 3-Hydroxybutyric Acid blood, Animals, Hypothalamus drug effects, Male, Neurons drug effects, Rats, Eating drug effects, Fatty Acids pharmacology, Hypothalamus physiology, Ketone Bodies pharmacology, Neurons physiology
- Abstract
Metabolic sensing neurons in the ventromedial hypothalamus (VMH) alter their activity when ambient levels of metabolic substrates, such as glucose and fatty acids (FA), change. To assess the relationship between a high-fat diet (HFD; 60%) intake on feeding and serum and VMH FA levels, rats were trained to eat a low-fat diet (LFD; 13.5%) or an HFD in 3 h/day and were monitored with VMH FA microdialysis. Despite having higher serum levels, HFD rats had lower VMH FA levels but ate less from 3 to 6 h of refeeding than did LFD rats. However, VMH β-hydroxybutyrate (β-OHB) and VMH-to-serum β-OHB ratio levels were higher in HFD rats during the first 1 h of refeeding, suggesting that VMH astrocyte ketone production mediated their reduced intake. In fact, using calcium imaging in dissociated VMH neurons showed that ketone bodies overrode normal FA sensing, primarily by exciting neurons that were activated or inhibited by oleic acid. Importantly, bilateral inhibition of VMH ketone production with a 3-hydroxy-3-methylglutaryl-CoA synthase inhibitor reversed the 3- to 6-h HFD-induced inhibition of intake but had no effect in LFD-fed rats. These data suggest that a restricted HFD intake regimen inhibits caloric intake as a consequence of FA-induced VMH ketone body production by astrocytes.
- Published
- 2014
- Full Text
- View/download PDF
14. Tanycytic VEGF-A boosts blood-hypothalamus barrier plasticity and access of metabolic signals to the arcuate nucleus in response to fasting.
- Author
-
Langlet F, Levin BE, Luquet S, Mazzone M, Messina A, Dunn-Meynell AA, Balland E, Lacombe A, Mazur D, Carmeliet P, Bouret SG, Prevot V, and Dehouck B
- Subjects
- Animals, Blood-Brain Barrier drug effects, Deoxyglucose pharmacology, Ependyma cytology, Fasting, Male, Mice, Mice, Inbred C57BL, Signal Transduction, Tight Junctions metabolism, Vascular Endothelial Growth Factor A genetics, Arcuate Nucleus of Hypothalamus metabolism, Blood-Brain Barrier metabolism, Ependyma metabolism, Vascular Endothelial Growth Factor A metabolism
- Abstract
The delivery of blood-borne molecules conveying metabolic information to neural networks that regulate energy homeostasis is restricted by brain barriers. The fenestrated endothelium of median eminence microvessels and tight junctions between tanycytes together compose one of these. Here, we show that the decrease in blood glucose levels during fasting alters the structural organization of this blood-hypothalamus barrier, resulting in the improved access of metabolic substrates to the arcuate nucleus. These changes are mimicked by 2-deoxyglucose-induced glucoprivation and reversed by raising blood glucose levels after fasting. Furthermore, we show that VEGF-A expression in tanycytes modulates these barrier properties. The neutralization of VEGF signaling blocks fasting-induced barrier remodeling and significantly impairs the physiological response to refeeding. These results implicate glucose in the control of blood-hypothalamus exchanges through a VEGF-dependent mechanism and demonstrate a hitherto unappreciated role for tanycytes and the permeable microvessels associated with them in the adaptive metabolic response to fasting., (Copyright © 2013 Elsevier Inc. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
15. Large litter rearing enhances leptin sensitivity and protects selectively bred diet-induced obese rats from becoming obese.
- Author
-
Patterson CM, Bouret SG, Park S, Irani BG, Dunn-Meynell AA, and Levin BE
- Subjects
- Adiposity physiology, Agouti-Related Protein metabolism, Animals, Animals, Newborn, Arcuate Nucleus of Hypothalamus metabolism, Body Weight physiology, Breeding, Eating physiology, Female, Gestational Age, Immunohistochemistry, Lactation physiology, Leptin metabolism, Male, Obesity blood, Obesity etiology, Paraventricular Hypothalamic Nucleus metabolism, Protein Binding, Rats, Receptors, Leptin metabolism, Time Factors, Weaning, Weight Gain physiology, alpha-MSH metabolism, Diet, Leptin blood, Litter Size physiology, Obesity physiopathology
- Abstract
Because rearing rats in large litters (LLs) protects them from becoming obese, we postulated that LL rearing would protect rats selectively bred to develop diet-induced obesity (DIO) from becoming obese by overcoming their inborn central leptin resistance. Male and female DIO rats were raised in normal litters (NLs; 10 pups/dam) or LLs (16 pups/dam) and assessed for anatomical, biochemical, and functional aspects of leptin sensitivity at various ages when fed low-fat chow or a 31% fat high-energy (HE) diet. LL rearing reduced plasma leptin levels by postnatal day 2 (P2) and body weight gain by P8. At P16, LL DIO neonates had increased arcuate nucleus (ARC) binding of leptin to its extracellular receptors and at P28 an associated increase of their agouti-related peptide and alpha-MSH axonal projections to the paraventricular nucleus. Reduced body weight persisted and was associated with increased ARC leptin receptor binding and sensitivity to the anorectic effects of leptin, reduced adiposity, and enhanced insulin sensitivity in LL DIO rats fed chow until 10 wk of age. The enhanced ARC leptin receptor binding and reduced adiposity of LL DIO rats persisted after an additional 5 wk on the HE diet. Female LL DIO rats had similar reductions in weight gain on both chow and HE diet vs. normal litter DIO rats. We postulate that LL rearing enhances DIO leptin sensitivity by lowering plasma leptin levels and thereby increasing leptin receptor availability and that this both enhances the ARC-paraventricular nucleus pathway development and protects them from becoming obese.
- Published
- 2010
- Full Text
- View/download PDF
16. Mechanisms of amylin/leptin synergy in rodent models.
- Author
-
Turek VF, Trevaskis JL, Levin BE, Dunn-Meynell AA, Irani B, Gu G, Wittmer C, Griffin PS, Vu C, Parkes DG, and Roth JD
- Subjects
- Amyloid genetics, Amyloid metabolism, Amyloid pharmacology, Animals, Arcuate Nucleus of Hypothalamus drug effects, Arcuate Nucleus of Hypothalamus metabolism, Area Postrema drug effects, Area Postrema metabolism, Female, Islet Amyloid Polypeptide, Leptin genetics, Leptin metabolism, Leptin pharmacology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Proto-Oncogene Proteins c-fos metabolism, Rats, Rats, Sprague-Dawley, STAT3 Transcription Factor genetics, STAT3 Transcription Factor metabolism, Signal Transduction drug effects, Signal Transduction genetics, Amyloid physiology, Drug Synergism, Leptin physiology, Models, Animal, Rodentia genetics, Rodentia metabolism
- Abstract
The present studies aimed to identify mechanisms contributing to amylin/leptin synergy in reducing body weight and adiposity. We reasoned that if amylin/leptin harnessed complementary neuronal pathways, then in the leptin-sensitive state, amylin should augment leptin signaling/binding and that in the absence of endogenous amylin, leptin signaling should be diminished. Amylin (50 microg/kg, ip) amplified low-dose leptin-stimulated (15 microg/kg, ip) phosphorylated signal transducer and activator of transcription-3 signaling within the arcuate nucleus (ARC) in lean rats. Amylin (50 microg/kg x d) or leptin (125 microg/kg x d) infusion to lean rats decreased 28-d food intake (14 and 10%, respectively), body weight (amylin by 4.3%, leptin by 4.9%), and epididymal fat (amylin by 19%, leptin by 37%). Amylin/leptin co-infusion additively decreased food intake (by 26%) and reduced body weight (by 15%) and epididymal fat (by 78%; all P < 0.05 vs. all groups) in a greater than mathematically additive manner, consistent with synergy. Amylin increased leptin binding within the ventromedial hypothalamus (VMN) by 35% and dorsomedial hypothalamus by 47% (both P < 0.05 vs. vehicle). Amylin/leptin similarly increased leptin binding in the VMN by 40% and ARC by 70% (P < 0.05 vs. vehicle). In amylin-deficient mice, hypothalamic leptin receptor mRNA expression was reduced by 50%, leptin-stimulated phosphorylated signal transducer and activator of transcription-3 within ARC and VMN was reduced by 40%, and responsiveness to leptin's (1 mg/kg x d for 28 d) weight-reducing effects was attenuated (all P < 0.05 vs. wild-type controls). We suggest that amylin/leptin's marked weight- and fat-reducing effects are due to activation of intrinsic synergistic neuronal signaling pathways and further point to the integrated neurohormonal therapeutic potential of amylin/leptin agonism in obesity.
- Published
- 2010
- Full Text
- View/download PDF
17. Genetic and dietary effects on dendrites in the rat hypothalamic ventromedial nucleus.
- Author
-
Labelle DR, Cox JM, Dunn-Meynell AA, Levin BE, and Flanagan-Cato LM
- Subjects
- Animals, Body Weight genetics, Disease Models, Animal, Eating genetics, Male, Obesity genetics, Rats, Silver Staining methods, Dendrites genetics, Dendrites metabolism, Diet, Neurons cytology, Obesity metabolism, Ventromedial Hypothalamic Nucleus cytology
- Abstract
Both genetic and environmental factors contribute to individual differences in body weight regulation. The present study examined a possible role for the dendritic arbor of hypothalamic ventromedial nucleus (VMH) neurons in a model of diet-induced obesity (DIO) in male rats. Rats were screened and selectively bred for being either susceptible, i.e., exhibiting DIO, or diet resistant (DR) when exposed to a 31% fat diet. A 2x2 experimental design was used, based on these two strains of rats and exposure to rat chow versus the 31% fat diet for seven weeks. Golgi-impregnated neurons were measured for soma size and dendrite parameters, including number, length, and direction. As previously observed, each VMH neuron had a single long primary dendrite. Genetic background and diet did not affect soma size or the number of dendrites of VMH neurons. However, genetic background exerted a main effect on the length of the long primary dendrites. In particular, the long primary dendrites were approximately 12.5% shorter on the VMH neurons in the DIO rats compared with DR rats regardless of diet. This effect was isolated to the long primary dendrites extending in the dorsolateral direction, with these long primary dendrites 19% shorter for the DIO group compared with the DR group. This finding implicates the connectivity of the long primary dendrites on VMH neurons in the control of energy balance. The functional significance of these shortened dendrites and their afferents warrants further study.
- Published
- 2009
- Full Text
- View/download PDF
18. Characteristics and mechanisms of hypothalamic neuronal fatty acid sensing.
- Author
-
Le Foll C, Irani BG, Magnan C, Dunn-Meynell AA, and Levin BE
- Subjects
- Animals, CD36 Antigens metabolism, Carnitine O-Palmitoyltransferase antagonists & inhibitors, Carnitine O-Palmitoyltransferase metabolism, Coenzyme A Ligases antagonists & inhibitors, Coenzyme A Ligases metabolism, Enzyme Inhibitors pharmacology, Free Radical Scavengers pharmacology, Homeostasis, Ion Channels metabolism, KATP Channels antagonists & inhibitors, KATP Channels metabolism, Male, Membrane Potentials, Microscopy, Fluorescence, Mitochondrial Proteins metabolism, Neural Inhibition, Neurons drug effects, Neurons enzymology, Potassium Channel Blockers pharmacology, Rats, Rats, Sprague-Dawley, Reactive Oxygen Species metabolism, Time Factors, Uncoupling Protein 2, Ventromedial Hypothalamic Nucleus cytology, Ventromedial Hypothalamic Nucleus drug effects, Ventromedial Hypothalamic Nucleus enzymology, Calcium Signaling, Energy Metabolism, Glucose metabolism, Neurons metabolism, Oleic Acid metabolism, Ventromedial Hypothalamic Nucleus metabolism
- Abstract
We assessed the mechanisms by which specialized hypothalamic ventromedial nucleus (VMN) neurons utilize both glucose and long-chain fatty acids as signaling molecules to alter their activity as a potential means of regulating energy homeostasis. Fura-2 calcium (Ca(2+)) and membrane potential dye imaging, together with pharmacological agents, were used to assess the mechanisms by which oleic acid (OA) alters the activity of dissociated VMN neurons from 3- to 4-wk-old rats. OA excited up to 43% and inhibited up to 29% of all VMN neurons independently of glucose concentrations. In those neurons excited by both 2.5 mM glucose and OA, OA had a concentration-dependent effective excitatory concentration (EC(50)) of 13.1 nM. Neurons inhibited by both 2.5 mM glucose and OA had an effective inhibitory concentration (IC(50)) of 93 nM. At 0.5 mM glucose, OA had markedly different effects on these same neurons. Inhibition of carnitine palmitoyltransferase, reactive oxygen species formation, long-chain acetyl-CoA synthetase and ATP-sensitive K(+) channel activity or activation of uncoupling protein 2 (UCP2) accounted for only approximately 20% of OA's excitatory effects and approximately 40% of its inhibitory effects. Inhibition of CD36, a fatty acid transporter that can alter cell function independently of intracellular fatty acid metabolism, reduced the effects of OA by up to 45%. Thus OA affects VMN neuronal activity through multiple pathways. In glucosensing neurons, its effects are glucose dependent. This glucose-OA interaction provides a potential mechanism whereby such "metabolic sensing" neurons can respond to differences in the metabolic states associated with fasting and feeding.
- Published
- 2009
- Full Text
- View/download PDF
19. Relationship among brain and blood glucose levels and spontaneous and glucoprivic feeding.
- Author
-
Dunn-Meynell AA, Sanders NM, Compton D, Becker TC, Eiki J, Zhang BB, and Levin BE
- Subjects
- Analysis of Variance, Animals, Arcuate Nucleus of Hypothalamus drug effects, Behavior, Animal, Body Weight drug effects, Body Weight physiology, Feeding Behavior drug effects, Gene Expression Regulation drug effects, Gene Expression Regulation physiology, Glucokinase genetics, Glucokinase metabolism, Glucose analogs & derivatives, Glucose pharmacology, Hypoglycemic Agents pharmacology, Insulin pharmacology, Male, Microdialysis methods, RNA, Messenger metabolism, Rats, Rats, Sprague-Dawley, Time Factors, Ventromedial Hypothalamic Nucleus drug effects, Arcuate Nucleus of Hypothalamus metabolism, Blood Glucose physiology, Feeding Behavior physiology, Glucose deficiency, Ventromedial Hypothalamic Nucleus metabolism
- Abstract
Although several studies implicate small declines in blood glucose levels as stimulus for spontaneous meal initiation, no mechanism is known for how these dips might initiate feeding. To assess the role of ventromedial hypothalamus (VMH) (arcuate plus ventromedial nucleus) glucosensing neurons as potential mediators of spontaneous and glucoprivic feeding, meal patterns were observed, and blood and VMH microdialysis fluid were sampled in 15 rats every 10 min for 3.5 h after dark onset and 2 h after insulin (5 U/kg, i.v.) infusion. Blood glucose levels declined by 11% beginning approximately 5 min before 65% of all spontaneous meals, with no fall in VMH levels. After insulin, blood and VMH glucose reached nadirs by 30-40 min, and the same rats ate 60% faster and spent 84% more time eating during the ensuing hypoglycemia. Although 83% of first hypoglycemic meals were preceded by 5 min dips in VMH (but not blood) glucose levels, neither blood nor VMH levels declined before second meals, suggesting that low glucose, rather than changing levels, was the stimulus for glucoprivic meals. Furthermore, altering VMH glucosensing by raising or lowering glucokinase (GK) activity failed to affect spontaneous feeding, body or adipose weights, or glucose tolerance. However, chronic depletion by 26-70% of VMH GK mRNA reduced glucoprivic feeding. Thus, although VMH glucosensing does not appear to be involved in either spontaneous feeding or long-term body-weight regulation, it does participate in glucoprivic feeding, similar to its role in the counter-regulatory neurohumoral responses to glucoprivation.
- Published
- 2009
- Full Text
- View/download PDF
20. Three weeks of postweaning exercise in DIO rats produces prolonged increases in central leptin sensitivity and signaling.
- Author
-
Patterson CM, Bouret SG, Dunn-Meynell AA, and Levin BE
- Subjects
- Adipose Tissue physiology, Animals, Arcuate Nucleus of Hypothalamus physiology, Blood Glucose metabolism, Body Temperature Regulation physiology, Caloric Restriction, Eating physiology, Extracellular Signal-Regulated MAP Kinases biosynthesis, Immunohistochemistry, Insulin blood, Leptin blood, Male, Paraventricular Hypothalamic Nucleus physiology, Rats, STAT3 Transcription Factor genetics, STAT3 Transcription Factor physiology, Telemetry, Weight Gain physiology, alpha-MSH metabolism, Diet, Leptin physiology, Obesity physiopathology, Physical Conditioning, Animal physiology, Signal Transduction physiology
- Abstract
In rats selectively bred to develop diet-induced obesity (DIO) 3 wk of postweaning exercise reduces weight and adipose regain for 10 wk after exercise cessation, despite intake of 31% fat high-energy (HE) diet. To test the hypothesis that this effect is due to increased central leptin sensitivity, 4-wk-old DIO rats were fed the HE diet and left sedentary (Sed), exercised for 3 wk, and then remained sedentary for 10 additional weeks (Ex/Sed) or continued exercise for a full 13 wk (Ex). After 3 wk, leptin (5 mg/kg ip) induced a 36% decrease in 24-h food intake in Ex rats, while Sed rats had no change in 24-h intake. Ex rats also had 23% more leptin-induced phospho-STAT3 (pSTAT3)-expressing neurons in the arcuate nucleus (ARC) and 95% and 68% higher (125)I-labeled leptin receptor binding in the ventromedial and dorsomedial nuclei than did Sed rats, respectively. At 7 wk after onset, leptin decreased 24-h intake by 20% in Ex and 24% in Ex/Sed rats without altering Sed intake. After a total of 13 wk, compared with Sed rats, Ex and Ex/Sed rats had 58% and 38% less fat, respectively, but leptin failed to decrease food intake in any group. Nevertheless, Ex, but not Ex/Sed rats, still had 32% more ARC leptin-induced pSTAT3-expressing neurons than Sed rats. These data suggest that brief postweaning exercise in DIO rats that are inherently leptin resistant causes a sustained resistance to obesity on HE diet, which is, in part, due to increased central leptin sensitivity.
- Published
- 2009
- Full Text
- View/download PDF
21. Ventromedial nucleus neurons are less sensitive to leptin excitation in rats bred to develop diet-induced obesity.
- Author
-
Irani BG, Le Foll C, Dunn-Meynell AA, and Levin BE
- Subjects
- Animals, Arcuate Nucleus of Hypothalamus cytology, Arcuate Nucleus of Hypothalamus drug effects, Blood Glucose metabolism, Calcium Signaling drug effects, Dose-Response Relationship, Drug, Energy Intake drug effects, Genotype, Insulin blood, Leptin blood, Male, RNA, Messenger biosynthesis, RNA, Messenger genetics, Rats, Ventromedial Hypothalamic Nucleus cytology, Weight Gain drug effects, Diet, Leptin pharmacology, Neurons drug effects, Obesity etiology, Obesity genetics, Ventromedial Hypothalamic Nucleus drug effects
- Abstract
Maternal obesity accentuates offspring obesity in dams bred to develop diet-induced obesity (DIO) on a 31% fat, high energy (HE) diet but has no effect on offspring of diet-resistant (DR) dams. Only DIO dams became obese on HE diet when they and DR dams were fed 5% fat chow or HE diets throughout gestation and lactation. Leptin sensitivity of dissociated arcuate (ARC) and ventromedial (VMN) hypothalamic nucleus neurons from the 3- to 4-wk-old offspring was assessed using fura-2 calcium imaging to monitor leptin-induced changes in intracellular calcium ([Ca(2+)](i)) as an index of neuronal activity. At 0.1, 1, 10 fmol/l leptin, approximately 4 times more VMN and ARC neurons were excited than inhibited by leptin. In the VMN, leptin excited up to 41% fewer neurons, and these excited neurons were less sensitive to increasing doses of leptin in DIO compared with DR offspring. Also, maternal HE diet intake decreased the percentage of leptin-excited VMN neurons in both DIO and DR offspring and decreased the percentage of leptin-inhibited VMN neurons by 36% only in DIO offspring. In the ARC, there were no genotype or maternal diet effects on the percentage of ARC neurons excited by leptin. However, those DR neurons that were leptin excited were more sensitive to leptin than were those from DIO offspring. These data suggest that reduced responsiveness of DIO VMN neurons to leptin's excitatory effects may be an important contributing factor to the reduced anorectic and thermogenic leptin responsiveness of DIO rats in vivo.
- Published
- 2009
- Full Text
- View/download PDF
22. Ventromedial hypothalamic glucokinase is an important mediator of the counterregulatory response to insulin-induced hypoglycemia.
- Author
-
Levin BE, Becker TC, Eiki J, Zhang BB, and Dunn-Meynell AA
- Subjects
- Animals, Diabetes Mellitus, Experimental enzymology, Enzyme Activation, Homeostasis, Hypoglycemia chemically induced, Kinetics, Male, Rats, Rats, Sprague-Dawley, Glucokinase metabolism, Hypoglycemia enzymology, Insulin pharmacology, Ventromedial Hypothalamic Nucleus enzymology
- Abstract
Objective: The counterregulatory response to insulin-induced hypoglycemia is mediated by the ventromedial hypothalamus (VMH), which contains specialized glucosensing neurons, many of which use glucokinase (GK) as the rate-limiting step in glucose's regulation of neuronal activity. Since conditions associated with increased VMH GK expression are associated with a blunted counterregulatory response, we tested the hypothesis that increasing VMH GK activity would similarly attenuate, while decreasing GK activity would enhance the counterregulatory response to insulin-induced hypoglycemia., Research Design and Methods: The counterregulatory response to insulin-induced hypoglycemia was evaluated in Sprague-Dawley rats after bilateral VMH injections of 1) a GK activator drug (compound A) to increase VMH GK activity, 2) low-dose alloxan (4 mug) to acutely inhibit GK activity, 3) high-dose alloxan (24 microg), or 4) an adenovirus expressing GK short hairpin RNA (shRNA) to chronically reduce GK expression and activity., Results: Compound A increased VMH GK activity sixfold in vitro and reduced the epinephrine, norepinephrine, and glucagon responses to insulin-induced hypoglycemia by 40-62% when injected into the VMH in vivo. On the other hand, acute and chronic reductions of VMH GK mRNA or activity had a lesser and more selective effect on increasing primarily the epinephrine response to insulin-induced hypoglycemia by 23-50%., Conclusions: These studies suggest that VMH GK activity is an important regulator of the counterregulatory response to insulin-induced hypoglycemia and that a drug that specifically inhibited the rise in hypothalamic GK activity after insulin-induced hypoglycemia might improve the dampened counterregulatory response seen in tightly controlled diabetic subjects.
- Published
- 2008
- Full Text
- View/download PDF
23. Prior hypoglycemia enhances glucose responsiveness in some ventromedial hypothalamic glucosensing neurons.
- Author
-
Kang L, Sanders NM, Dunn-Meynell AA, Gaspers LD, Routh VH, Thomas AP, and Levin BE
- Subjects
- Adrenal Medulla drug effects, Adrenal Medulla physiology, Animals, Calcium metabolism, Cyclic AMP-Dependent Protein Kinases physiology, Glucokinase biosynthesis, Glucokinase genetics, Hexokinase biosynthesis, Hexokinase genetics, Hypoglycemia chemically induced, Hypoglycemic Agents, Insulin, Male, RNA, Messenger biosynthesis, RNA, Messenger genetics, Rats, Rats, Sprague-Dawley, Ventromedial Hypothalamic Nucleus cytology, Ventromedial Hypothalamic Nucleus drug effects, Glucose metabolism, Glucose pharmacology, Hypoglycemia physiopathology, Neurons drug effects, Ventromedial Hypothalamic Nucleus physiopathology
- Abstract
Antecedent insulin-induced hypoglycemia (IIH) reduces adrenomedullary responses (AMR) to subsequent bouts of hypoglycemia. The ventromedial hypothalamus [VMH: arcuate (ARC) + ventromedial nuclei] contains glucosensing neurons, which are thought to be mediators of these AMR. Since type 1 diabetes mellitus often begins in childhood, we used juvenile (4- to 5-wk-old) rats to demonstrate that a single bout of IIH (5 U/kg sc) reduced plasma glucose by 24% and peak epinephrine by 59% 1 day later. This dampened AMR was associated with 46% higher mRNA for VMH glucokinase, a key mediator of neuronal glucosensing. Compared with neurons from saline-injected rats, ventromedial nucleus glucose-excited neurons from insulin-injected rats demonstrated a leftward shift in their glucose responsiveness (EC50 = 0.45 and 0.10 mmol/l for saline and insulin, respectively, P = 0.05) and a 31% higher maximal activation by glucose (P = 0.05), although this maximum occurred at a higher glucose concentration (saline, 0.7 vs. insulin, 1.5 mmol/l). Although EC50 values did not differ, ARC glucose-excited neurons had 19% higher maximal activation, which occurred at a lower glucose concentration in insulin- than saline-injected rats (saline, 2.5 vs. insulin, 1.5 mmol/l). In addition, ARC glucose-inhibited neurons from insulin-injected rats were maximally inhibited at a fivefold lower glucose concentration (saline, 2.5 vs. insulin, 0.5 mmol/l), although this inhibition declined at >0.5 mmol/l glucose. These data suggest that the increased VMH glucokinase after IIH may contribute to the increased responsiveness of VMH glucosensing neurons to glucose and the associated blunting of the AMR.
- Published
- 2008
- Full Text
- View/download PDF
24. Three weeks of early-onset exercise prolongs obesity resistance in DIO rats after exercise cessation.
- Author
-
Patterson CM, Dunn-Meynell AA, and Levin BE
- Subjects
- Adipose Tissue physiology, Animal Feed, Animals, Brain-Derived Neurotrophic Factor genetics, Caloric Restriction, Eating physiology, Insulin blood, Leptin blood, Male, Neuropeptide Y genetics, Pro-Opiomelanocortin genetics, RNA, Messenger metabolism, Rats, Rats, Inbred Strains, Receptors, Leptin genetics, Suppressor of Cytokine Signaling 3 Protein, Suppressor of Cytokine Signaling Proteins genetics, Body Weight physiology, Hypothalamus physiology, Obesity physiopathology, Obesity prevention & control, Physical Conditioning, Animal physiology
- Abstract
We assessed the effect of early-onset exercise as a means of preventing childhood obesity using juvenile male rats selectively bred to develop diet-induced obesity (DIO) or to be diet resistant (DR) when fed a 31% fat high-energy diet. Voluntary wheel running begun at 36 days of age selectively reduced adiposity in DIO vs. DR rats. Other 4-wk-old DIO rats fed a high-energy diet and exercised (Ex) for 13 wk increased their core temperature, gained 22% less body weight, and had 39% lighter fat pads compared with sedentary (Sed) rats. When wheels were removed after 6 wk (6 wk Ex/7 wk Sed), rats gained less body weight over the next 7 wk than Sed rats and still had comparable adipose pad weights to 13-wk-exercised rats. In fact, only 3 wk of exercise sufficed to prevent obesity for 10 wk after wheel removal. Terminally, the 6-wk-Ex/7-wk-Sed rats had a 55% increase in arcuate nucleus proopiomelanocortin mRNA expression vs. Sed rats, suggesting that this contributed to their sustained obesity resistance. Finally, when Sed rats were calorically restricted for 6 wk to weight match them to Ex rats (6 wk Rstr/7 wk Al), they increased their intake and body weight when fed ad libitum and, after 7 wk more, had higher leptin levels and adiposity than Sed rats. Thus, early-onset exercise may favorably alter, while early caloric restriction may unfavorably influence, the development of the hypothalamic pathways controlling energy homeostasis during brain development.
- Published
- 2008
- Full Text
- View/download PDF
25. Maternal obesity increases hypothalamic leptin receptor expression and sensitivity in juvenile obesity-prone rats.
- Author
-
Gorski JN, Dunn-Meynell AA, and Levin BE
- Subjects
- Adipose Tissue, Adipose Tissue, Brown metabolism, Animals, Diet, Leptin metabolism, Leptin pharmacology, Neuropeptides genetics, Neuropeptides metabolism, Obesity genetics, Rats, Receptors, Leptin, Time Factors, Weight Gain, Hypothalamus metabolism, Obesity physiopathology, Receptors, Cell Surface genetics, Receptors, Cell Surface metabolism, Up-Regulation
- Abstract
In rats selectively bred to develop diet-induced obesity (DIO) or to be diet-resistant (DR), DIO maternal obesity selectively enhances the development of obesity and insulin resistance in their adult offspring. We postulated that the interaction between genetic predisposition and factors in the maternal environment alter the development of hypothalamic peptide systems involved in energy homeostasis regulation. Maternal obesity in the current studies led to increased body and fat pad weights and higher leptin and insulin levels in postnatal day 16 offspring of both DIO and DR dams. However, by 6 wk of age, most of these intergroup differences disappeared and offspring of obese DIO dams had unexpected increases in arcuate nucleus leptin receptor mRNA, peripheral insulin sensitivity, diet- and leptin-induced brown adipose temperature increase and 24-h anorectic response compared with offspring of lean DIO, but not lean DR dams. On the other hand, while offspring of obese DIO dams did have the highest ventromedial nucleus melanocortin-4 receptor expression, their anorectic and brown adipose thermogenic responses to the melanocortin agonist, Melanotan II (MTII), did not differ from those of offspring of lean DR or DIO dams. Thus, during their rapid growth phase, juvenile offspring of obese DIO dams have alterations in their hypothalamic systems regulating energy homeostasis, which ameliorates their genetic and perinatally determined predisposition toward leptin resistance. Because they later go onto become more obese, it is possible that interventions during this time period might prevent the subsequent development of obesity.
- Published
- 2007
- Full Text
- View/download PDF
26. Altered hypothalamic leptin, insulin, and melanocortin binding associated with moderate-fat diet and predisposition to obesity.
- Author
-
Irani BG, Dunn-Meynell AA, and Levin BE
- Subjects
- Adipose Tissue metabolism, Animals, Arcuate Nucleus of Hypothalamus metabolism, Blood Glucose, Dietary Fats pharmacology, Dorsomedial Hypothalamic Nucleus metabolism, Insulin blood, Iodine Radioisotopes, Leptin blood, Male, Obesity genetics, Rats, Rats, Mutant Strains, Receptor, Melanocortin, Type 4 metabolism, Substantia Nigra metabolism, Ventral Tegmental Area metabolism, Ventromedial Hypothalamic Nucleus metabolism, Weight Gain physiology, Hypothalamus metabolism, Insulin pharmacokinetics, Leptin pharmacokinetics, Melanocortins metabolism, Obesity metabolism
- Abstract
Rats with a genetic predisposition to develop diet-induced obesity (DIO) have a preexisting reduction in central leptin and insulin sensitivity. High-fat diets also reduce sensitivity to leptin, insulin, and melanocortin agonists. We postulated that such reduced sensitivities would be associated with decreased binding to the hypothalamic leptin, insulin, and melanocortin receptors in selectively bred DIO rats and in rats fed a high-energy (HE; 31% fat) diet for 7 wk. On HE diet, DIO rats gained 15% more weight and had 121% heavier fat pads and 70% higher leptin levels than low fat chow-fed DIO rats. Diet-resistant (DR) rats gained no more weight on HE diet but had 48% heavier fat pads and 70% higher leptin levels than chow-fed DR rats. Compared with DR rats, DIO (125)I-leptin binding was 41, 36, and 40% lower in the hypothalamic dorsomedial, arcuate, and dorsomedial portion of the ventromedial nuclei, respectively, and arcuate (125)I-insulin binding was 31% lower independent of diet. In contrast, hypothalamic melanocortin binding did not differ between DIO and DR rats. However, HE diet intake lowered lateral hypothalamic melanocortin-3 and melanocortin-4 receptor and hippocampal insulin binding of both DIO and DR rats and hypothalamic paraventricular nucleus melanocortin-4 receptor binding only in DR rats. Neither genotype nor diet affected substantia nigra or ventral tegmental area binding. These results corroborate our previous findings demonstrating a preexisting decrease in DIO hypothalamic leptin and insulin signaling and demonstrate that HE diet intake reduces hypothalamic melanocortin and hippocampal insulin binding.
- Published
- 2007
- Full Text
- View/download PDF
27. Postnatal environment overrides genetic and prenatal factors influencing offspring obesity and insulin resistance.
- Author
-
Gorski JN, Dunn-Meynell AA, Hartman TG, and Levin BE
- Subjects
- Aging, Animal Feed, Animals, Energy Metabolism, Fatty Acids analysis, Female, Gene Expression Regulation, Hormones blood, Hypothalamus metabolism, Male, Milk chemistry, Neuropeptides metabolism, RNA, Messenger metabolism, Rats, Receptors, Neuropeptide metabolism, Weight Gain, Insulin Resistance genetics, Insulin Resistance physiology, Obesity genetics, Obesity metabolism
- Abstract
There is growing evidence that the postnatal environment can have a major impact on the development of obesity and insulin resistance in offspring. We postulated that cross-fostering obesity-prone offspring to lean, obesity-resistant dams would ameliorate their development of obesity and insulin resistance, while fostering lean offspring to genetically obese dams would lead them to develop obesity and insulin resistance as adults. We found that obesity-prone pups cross-fostered to obesity-resistant dams remained obese but did improve their insulin sensitivity as adults. In contrast, obesity-resistant pups cross-fostered to genetically obese dams showed a diet-induced increase in adiposity, reduced insulin sensitivity, and associated changes in hypothalamic neuropeptide, insulin, and leptin receptors, which might have contributed to their metabolic defects. There was a selective increase in insulin levels and differences in fatty acid composition of obese dam milk which might have contributed to the increased adiposity, insulin resistance, and hypothalamic changes in obesity-resistant cross-fostered offspring. These results demonstrate that postnatal factors can overcome both genetic predisposition and prenatal factors in determining the development of adiposity, insulin sensitivity, and the brain pathways that mediate these functions.
- Published
- 2006
- Full Text
- View/download PDF
28. Differential effects of exercise on body weight gain and adiposity in obesity-prone and -resistant rats.
- Author
-
Levin BE and Dunn-Meynell AA
- Subjects
- Animals, Energy Intake physiology, Insulin blood, Leptin blood, Male, Rats, Rats, Sprague-Dawley, Adiposity physiology, Obesity physiopathology, Physical Exertion physiology, Weight Gain physiology
- Abstract
Objective: To determine the effect of exercise on weight gain and adiposity in obesity-prone and -resistant rats., Design: Body weight gain, fat pad weights, food intake, plasma leptin and insulin levels were assessed in outbred male Sprague-Dawley rats, which remained sedentary or were given unrestricted access to running wheels either before or after they developed diet-induced obesity (DIO) or diet-resistance (DR) on a high energy (HE; 31% fat) diet., Results: When fed a low fat (4.5%) chow diet, rats which would later develop DIO (n=6) after 3 weeks on HE diet ran the same amount as DR rats (n=6). Other rats were first made DIO (n=12) or DR (n=12) after 10 weeks on HE diet and then either kept sedentary or given running wheels for 4 weeks on HE diet. DIO and DR rats ran comparable amounts but only the DIO rats reduced their body weight gain, fat pad relative to body weights and plasma leptin levels significantly, compared to their sedentary controls. Exercise had no effect on food intake in either DIO or DR rats but reduced feed efficiency (weight gain/caloric intake) in both., Conclusion: Although DIO and DR rats ran similar amounts, the greater reduction in body weight gain and adiposity of exercising DIO rats suggests that they are more sensitive to some metabolic or physiologic system that prevents them from increasing their intake sufficiently to compensate for their net reduction in energy stores.
- Published
- 2006
- Full Text
- View/download PDF
29. Glucokinase is a critical regulator of ventromedial hypothalamic neuronal glucosensing.
- Author
-
Kang L, Dunn-Meynell AA, Routh VH, Gaspers LD, Nagata Y, Nishimura T, Eiki J, Zhang BB, and Levin BE
- Subjects
- Animals, Calcium metabolism, Cells, Cultured, Gene Expression Regulation, Enzymologic, Glucokinase genetics, Hypoglycemic Agents pharmacology, Male, RNA, Messenger genetics, RNA, Messenger metabolism, RNA, Small Interfering genetics, RNA, Small Interfering metabolism, Rats, Rats, Sprague-Dawley, Tolbutamide pharmacology, Ventromedial Hypothalamic Nucleus cytology, Ventromedial Hypothalamic Nucleus drug effects, Glucokinase metabolism, Glucose metabolism, Neurons enzymology, Neurons physiology, Ventromedial Hypothalamic Nucleus enzymology, Ventromedial Hypothalamic Nucleus physiology
- Abstract
To test the hypothesis that glucokinase is a critical regulator of neuronal glucosensing, glucokinase activity was increased, using a glucokinase activator drug, or decreased, using RNA interference combined with calcium imaging in freshly dissociated ventromedial hypothalamic nucleus (VMN) neurons or primary ventromedial hypothalamus (VMH; VMN plus arcuate nucleus) cultures. To assess the validity of our approach, we first showed that glucose-induced (0.5-2.5 mmol/l) changes in intracellular Ca(2+) concentration ([Ca(2+)](i)) oscillations, using fura-2 and changes in membrane potential (using a membrane potential-sensitive dye), were highly correlated in both glucose-excited and -inhibited neurons. Also, glucose-excited neurons increased (half-maximal effective concentration [EC(50)] = 0.54 mmol/l) and glucose-inhibited neurons decreased (half-maximal inhibitory concentration [IC(50)] = 1.12 mmol/l) [Ca(2+)](i) oscillations to incremental changes in glucose from 0.3 to 5 mmol/l. In untreated primary VMH neuronal cultures, the expression of glucokinase mRNA and the number of demonstrable glucosensing neurons fell spontaneously by half over 12-96 h without loss of viable neurons. Transfection of neurons with small interfering glucokinase RNA did not affect survival but did reduce glucokinase mRNA by 90% in association with loss of all demonstrable glucose-excited neurons and a 99% reduction in glucose-inhibited neurons. A pharmacological glucokinase activator produced a dose-related increase in [Ca(2+)](i) oscillations in glucose-excited neurons (EC(50) = 0.98 mmol/l) and a decrease in glucose-inhibited neurons (IC(50) = 0.025 micromol/l) held at 0.5 mmol/l glucose. Together, these data support a critical role for glucokinase in neuronal glucosensing.
- Published
- 2006
- Full Text
- View/download PDF
30. Cortical Fluoro-Jade staining and blunted adrenomedullary response to hypoglycemia after noncoma hypoglycemia in rats.
- Author
-
Tkacs NC, Pan Y, Raghupathi R, Dunn-Meynell AA, and Levin BE
- Subjects
- Animals, Biomarkers, Blood Glucose, Consciousness, Fluoresceins, Hypoglycemia chemically induced, Hypoglycemia pathology, Hypoglycemic Agents blood, Hypoglycemic Agents pharmacology, Insulin blood, Insulin pharmacology, Lactic Acid blood, Male, Organic Chemicals, Rats, Rats, Sprague-Dawley, Adrenal Medulla physiopathology, Autonomic Nervous System physiopathology, Cerebral Cortex pathology, Cerebral Cortex physiopathology, Hypoglycemia physiopathology
- Abstract
Intensive insulin therapy in patients with type 1 diabetes mellitus reduces long-term complications; however, intensive therapy is also associated with a three-fold increase in hypoglycemic episodes. The present study in conscious rats characterizes the physiologic and neuropathologic consequences of a single episode of moderate hypoglycemia. In this model, intravenous insulin is used to reduce plasma glucose to 30 to 35 mg/dL for 75 mins. This single hypoglycemic insult acutely induces hypoglycemia-associated autonomic failure (HAAF), with epinephrine responses to hypoglycemia reduced more than 36% from control. Neuropathology after this insult includes the appearance of dying cells, assessed with the marker Fluoro-jade B (FJ). After hypoglycemic insult, FJ+ cells were consistently seen in subdivisions of the medial prefrontal cortex, the orbital cortex, and the piriform cortex. There was a significant correlation between depth of hypoglycemia and number of FJ+ cells, suggesting that there is a critical threshold below which vulnerable cells begin to die. These data suggest that there is a population of cells that are vulnerable to moderate levels of hypoglycemia commonly experienced by patients with insulin-treated diabetes. These cells, which may be neurons, are primarily found in cortical regions implicated in visceral perception and autonomic control, raising the possibility that their loss contributes to clinically reported deficits in autonomic and perceptual responses to hypoglycemia.
- Published
- 2005
- Full Text
- View/download PDF
31. F-DIO obesity-prone rat is insulin resistant before obesity onset.
- Author
-
Levin BE, Magnan C, Migrenne S, Chua SC Jr, and Dunn-Meynell AA
- Subjects
- Animals, Arcuate Nucleus of Hypothalamus metabolism, Blood Glucose metabolism, Disease Models, Animal, Glucose antagonists & inhibitors, Glucose pharmacology, Hypoglycemic Agents blood, Hypoglycemic Agents pharmacology, Hypothalamic Area, Lateral metabolism, Insulin blood, Insulin pharmacology, Intracellular Signaling Peptides and Proteins metabolism, Liver metabolism, Neuropeptides metabolism, Obesity genetics, Orexins, Phenotype, Pro-Opiomelanocortin metabolism, Rats, Rats, Inbred F344, Rats, Inbred Strains, Diet adverse effects, Genetic Predisposition to Disease, Insulin Resistance genetics, Obesity etiology, Obesity physiopathology
- Abstract
We previously created a novel F-DIO rat strain derived by crossing rats selectively bred for the diet-induced obesity (DIO) phenotype with obesity-resistant Fischer F344 rats. The offspring retained the DIO phenotype through 3 backcrosses with F344 rats but also had exaggerated insulin responses to oral glucose before they became obese on a 31% fat high-energy (HE) diet. Here, we demonstrate that chow-fed rats from the subsequent randomly bred progeny required 57% lower glucose infusions to maintain euglycemia during a hyperinsulinemic clamp in association with 45% less insulin-induced hepatic glucose output inhibition and 80% lower insulin-induced glucose uptake than F344 rats. The DIO phenotype and exaggerated insulin response to oral glucose in the nonobese, chow-fed state persisted in the F6 generation. Also, compared with F344 rats, chow-fed F-DIO rats had 68% higher arcuate nucleus proopiomelanocortin mRNA expression which, unlike the increase in F344 rats, was decreased by 26% on HE diet. Further, F-DIO lateral hypothalamic orexin expression was 18% lower than in F344 rats and was increased rather than decreased by HE diet intake. Finally, both maternal obesity and 30% caloric restriction during the third week of gestation produced F-DIO offspring which were heavier and had higher leptin and insulin levels than lean F-DIO dam offspring. Third-gestational week dexamethasone also produced offspring with higher leptin and insulin levels but with lower body weight. Thus F-DIO rats represent a novel and potentially useful model for the study of DIO, insulin resistance, and perinatal factors that influence the development and persistence of obesity.
- Published
- 2005
- Full Text
- View/download PDF
32. Neuronal glucosensing: what do we know after 50 years?
- Author
-
Levin BE, Routh VH, Kang L, Sanders NM, and Dunn-Meynell AA
- Subjects
- Action Potentials physiology, Animals, Glucokinase metabolism, Glycolysis, Humans, Potassium Channels physiology, Glucose analysis, Glucose physiology, Neurons physiology, Signal Transduction physiology
- Abstract
Glucosensing neurons are specialized cells that use glucose as a signaling molecule to alter their action potential frequency in response to variations in ambient glucose levels. Glucokinase (GK) appears to be the primary regulator of most neuronal glucosensing, but other regulators almost certainly exist. Glucose-excited neurons increase their activity when glucose levels rise, and most use GK and an ATP-sensitive K(+) channel as the ultimate effector of glucose-induced signaling. Glucose-inhibited (GI) neurons increase their activity at low glucose levels. Although many use GK, it is unclear what the final pathway of GI neuronal glucosensing is. Glucosensing neurons are located in brain sites and respond to and integrate a variety of hormonal, metabolic, transmitter, and peptide signals involved in the regulation of energy homeostasis and other biological functions. Although it is still uncertain whether daily fluctuations in blood glucose play a specific regulatory role in these physiological functions, it is clear that large decreases in glucose availability stimulate food intake and counterregulatory responses that restore glucose levels to sustain cerebral function. Finally, glucosensing is altered in obesity and after recurrent bouts of hypoglycemia, and this altered sensing may contribute to the adverse outcomes of these conditions. Thus, although much is known, much remains to be learned about the physiological function of brain glucosensing neurons.
- Published
- 2004
- Full Text
- View/download PDF
33. The regulation of glucose-excited neurons in the hypothalamic arcuate nucleus by glucose and feeding-relevant peptides.
- Author
-
Wang R, Liu X, Hentges ST, Dunn-Meynell AA, Levin BE, Wang W, and Routh VH
- Subjects
- Animals, Arcuate Nucleus of Hypothalamus drug effects, In Vitro Techniques, Male, Membrane Potentials drug effects, Neurons drug effects, Patch-Clamp Techniques, Rats, Rats, Sprague-Dawley, Tolbutamide pharmacology, Alloxan pharmacology, Arcuate Nucleus of Hypothalamus physiology, Glucose pharmacology, Neurons physiology
- Abstract
Glucosensing neurons in the hypothalamic arcuate nucleus (ARC) were studied using electrophysiological and immunocytochemical techniques in neonatal male Sprague-Dawley rats. We identified glucose-excited and -inhibited neurons, which increase and decrease, respectively, their action potential frequency (APF) as extracellular glucose levels increase throughout the physiological range. Glucose-inhibited neurons were found predominantly in the medial ARC, whereas glucose-excited neurons were found in the lateral ARC. ARC glucose-excited neurons in brain slices dose-dependently increased their APF and decreased their ATP-sensitive K+ channel (KATP channel) currents as extracellular glucose levels increased from 0.1 to 10 mmol/l. However, glucose sensitivity was greatest as extracellular glucose decreased to <2.5 mmol/l. The glucokinase inhibitor alloxan increases KATP single-channel currents in glucose-excited neurons in a manner similar to low glucose. Leptin did not alter the activity of ARC glucose-excited neurons. Although insulin did not affect ARC glucose-excited neurons in the presence of 2.5 mmol/l (steady-state) glucose, they were stimulated by insulin in the presence of 0.1 mmol/l glucose. Neuropeptide Y (NPY) inhibited and alpha-melanocyte-stimulating hormone stimulated ARC glucose-excited neurons. ARC glucose-excited neurons did not show pro-opiomelanocortin immunoreactivity. These data suggest that ARC glucose-excited neurons may serve an integrative role in the regulation of energy balance.
- Published
- 2004
- Full Text
- View/download PDF
34. Third ventricular alloxan reversibly impairs glucose counterregulatory responses.
- Author
-
Sanders NM, Dunn-Meynell AA, and Levin BE
- Subjects
- Animals, Blood Glucose metabolism, Eating drug effects, Enzyme Inhibitors pharmacology, Enzyme Inhibitors poisoning, Fourth Ventricle, Glucokinase genetics, Hyperglycemia chemically induced, Hypothalamus metabolism, Injections, Injections, Intraventricular, Male, Medulla Oblongata, Neuropeptide Y genetics, Neuropeptide Y physiology, Pro-Opiomelanocortin genetics, Pro-Opiomelanocortin physiology, RNA, Messenger metabolism, Rats, Rats, Sprague-Dawley, Alloxan administration & dosage, Enzyme Inhibitors administration & dosage, Glucokinase antagonists & inhibitors, Glucokinase physiology, Glucose analogs & derivatives, Glucose metabolism, Third Ventricle pathology
- Abstract
Glucokinase (GK) is hypothesized to be the critical glucosensor of pancreatic beta-cells and hypothalamic glucosensing neurons. To understand the role of GK in glucoprivic counterregulatory responses, we injected alloxan, a GK inhibitor and toxin, into the third ventricle (3v) to target nearby GK-expressing neurons. Four and 6 days after 3v, but not 4v, alloxan injection, alloxan-treated rats ate only 30% and their blood glucose area under the curve was only 28% of saline controls' after systemic 2-deoxy-D-glucose. In addition, their hyperglycemic response to hindbrain glucoprivation induced with 5-thio-glucose was impaired, whereas fasting blood glucose levels and food intake after an overnight fast were elevated. These impaired responses were associated with the destruction of 3v tanycytes, reduced glial fibrillary acidic protein-immunoreactivity surrounding the 3v, neuronal swelling, and decreased arcuate nucleus neuropeptide Y (NPY) mRNA. Nevertheless, hypothalamic GK mRNA was significantly elevated. Two weeks after alloxan injection, 3v tanycyte destruction was reversed along with restoration of feeding and hyperglycemic responses to both systemic and hindbrain glucoprivation. At this time there were significant decreases in GK, NPY, and proopiomelanocortin mRNA. Thus, neural substrates near and around the 3v affected by alloxan may be critically involved in the expression of these glucoprivic responses.
- Published
- 2004
- Full Text
- View/download PDF
35. Chronic exercise lowers the defended body weight gain and adiposity in diet-induced obese rats.
- Author
-
Levin BE and Dunn-Meynell AA
- Subjects
- Agouti-Related Protein, Animals, Autoradiography, Corticotropin-Releasing Hormone biosynthesis, Diet, Energy Intake physiology, Energy Metabolism physiology, Insulin blood, Leptin blood, Male, Motor Activity physiology, Neuropeptide Y biosynthesis, Peptide Fragments biosynthesis, Pro-Opiomelanocortin biosynthesis, RNA, Messenger biosynthesis, Rats, Rats, Sprague-Dawley, Receptors, Cell Surface biosynthesis, Receptors, Leptin, Adipose Tissue physiology, Obesity physiopathology, Physical Exertion physiology, Weight Gain physiology
- Abstract
The effects of running wheel exercise and caloric restriction on the regulation of body weight, adiposity, and hypothalamic neuropeptide expression were compared in diet-induced obese male rats over 6 wk. Compared with sedentary controls, exercising rats had reduced body weight gain (24%), visceral (4 fat pads; 36%) and carcass (leptin; 35%) adiposity but not insulin levels. Hypothalamic arcuate nucleus (ARC) proopiomelanocortin (POMC) mRNA expression was 25% lower, but ARC neuropeptide Y (NPY), agouti- related peptide, dorsomedial nucleus (DMN) NPY, and paraventricular nucleus (PVN) corticotropin- releasing hormone (CRH) expression was comparable to controls. Sedentary rats calorically restricted to 85% of control body weight reduced their visceral adiposity (24%), leptin (64%), and insulin (21%) levels. ARC NPY (23%) and DMN NPY (60%) were increased, while ARC POMC (40%) and PVN CRH (14%) were decreased. Calorically restricted exercising rats an half as much as ad libitum-fed exercising rats and had less visceral obesity than comparably restricted sedentary rats. When sedentary restricted rats were refed after 4 wk, they increased intake and regained the weight gain and adiposity of sedentary controls. While refed exercising rats and sedentary rats ate comparable amounts, refed exercising rats regained weight and adiposity only to the level of ad libitum-fed exercising rats. Thus exercise lowers the defended level of weight gain and adiposity without a compensatory increase in intake and with a very different profile of hypothalamic neuropeptide expression from calorically restricted rats. This may be due to exercise-related factors other than plasma insulin and leptin.
- Published
- 2004
- Full Text
- View/download PDF
36. Obesity-prone rats have normal blood-brain barrier transport but defective central leptin signaling before obesity onset.
- Author
-
Levin BE, Dunn-Meynell AA, and Banks WA
- Subjects
- Aging metabolism, Animals, Biological Transport, DNA-Binding Proteins metabolism, Diet adverse effects, Eating drug effects, Female, Genotype, Leptin pharmacokinetics, Leptin pharmacology, Male, Obesity etiology, Phosphorylation, RNA, Messenger metabolism, Rats, Rats, Sprague-Dawley, Receptors, Cell Surface genetics, Receptors, Leptin, STAT3 Transcription Factor, Sex Characteristics, Sympathetic Nervous System physiology, Trans-Activators metabolism, Blood-Brain Barrier, Brain metabolism, Genetic Predisposition to Disease, Leptin metabolism, Obesity genetics, Signal Transduction
- Abstract
Rats selectively bred to develop diet-induced obesity (DIO) were compared with those bred to be diet resistant (DR) on a 31% fat high-energy diet with regard to their central leptin signaling and blood-brain barrier (BBB) transport. Peripheral leptin injection (15 mg/kg ip) into lean 4- to 5-wk-old rats produced 54% less anorexia in DIO than DR rats. DIO rats also had 21, 63, and 64% less leptin-induced immunoreactive phosphorylated signal transducer and activator of transcription 3 (pSTAT3) expression in the hypothalamic arcuate, ventromedial, and dorsomedial nuclei, respectively. However, hindbrain leptin-induced nucleus tractus solitarius pSTAT3 and generalized sympathetic (24-h urine norepinephrine) activation were comparable. Reduced central leptin signaling was not due to defective BBB transport since transport did not differ between lean 4- to 5-wk-old DIO and DR rats. Conversely, DIO leptin BBB transport was reduced when they became obese at 23 wk of age on low-fat chow or after 6 wk on high-energy diet. In addition, leptin receptor mRNA expression was 23% lower in the arcuate nuclei of 4- to 5-wk-old DIO compared with DR rats. Thus a preexisting reduction in hypothalamic but not brain stem leptin signaling might contribute to the development of DIO when dietary fat and caloric density are increased. Defects in leptin transport appear to be an acquired defect associated with the development of obesity and possibly age.
- Published
- 2004
- Full Text
- View/download PDF
37. A new obesity-prone, glucose-intolerant rat strain (F.DIO).
- Author
-
Levin BE, Dunn-Meynell AA, McMinn JE, Alperovich M, Cunningham-Bussel A, and Chua SC Jr
- Subjects
- Animals, Energy Intake genetics, Energy Metabolism genetics, Female, Glucose Intolerance physiopathology, Male, Motor Activity genetics, Obesity physiopathology, Phenotype, Pregnancy, Rats, Rats, Inbred F344, Rats, Sprague-Dawley, Disease Models, Animal, Glucose Intolerance genetics, Obesity genetics, Rats, Mutant Strains
- Abstract
Previous breeding for the diet-induced obese (DIO) trait from outbred Sprague-Dawley rats produced a substrain with selection characteristics suggesting a polygenic mode of inheritance. To assess this issue further, selectively bred DIO male rats were crossed with obesity-resistant inbred Fischer F344 dams. Male offspring were crossed twice more against female F344 dams. The resultant N3 (F.DIO) rats were then inbred three more times. On low-fat chow, 10-wk-old male and female DIO rats weighed 86 and 59% more than respective F344 rats. By the N3 (F.DIO) generation, they were only 12 and 10% heavier, respectively. After three additional inbreeding cycles, chow-fed F.DIO males had an exaggerated insulin response to oral glucose compared with F344 rats. After 3 wk on a 31% fat (high-energy) diet, male N3 F.DIO rats gained 16-20% more carcass and adipose weight with 98% higher plasma leptin levels, whereas F.DIO females gained 36-54% more carcass and adipose weight with 130% higher leptin levels than comparable F344 rats. After three inbreeding cycles, F.DIO males still gained more weight on high-energy diet and developed a threefold greater insulin response to oral glucose than F344 males. Preservation of the DIO and glucose intolerance traits through successive backcrosses and inbreeding cycles to produce the F.DIO strain lends further support to the idea that they inherited in a polygenic fashion.
- Published
- 2003
- Full Text
- View/download PDF
38. Abnormalities of leptin and ghrelin regulation in obesity-prone juvenile rats.
- Author
-
Levin BE, Dunn-Meynell AA, Ricci MR, and Cummings DE
- Subjects
- Adipose Tissue anatomy & histology, Animals, Arcuate Nucleus of Hypothalamus chemistry, Body Weight, Diet, Dietary Fats administration & dosage, Dorsomedial Hypothalamic Nucleus chemistry, Eating, Energy Intake, Ghrelin, Insulin blood, Male, Obesity blood, Obesity genetics, Organ Size, RNA, Messenger analysis, Rats, Rats, Sprague-Dawley, Receptors, Cell Surface genetics, Receptors, G-Protein-Coupled genetics, Receptors, Ghrelin, Receptors, Leptin, Signal Transduction, Ventromedial Hypothalamic Nucleus chemistry, Leptin blood, Obesity etiology, Peptide Hormones blood
- Abstract
Rats selectively bred to develop diet-induced obesity (DIO) spontaneously gain more body weight between 5 and 7 wk of age than do those bred to be diet resistant (DR). Here, chow-fed DIO rats ate 9% more and gained 19% more body weight from 5 to 6 wk of age than did DR rats but had comparable leptin and insulin levels. However, 6-wk-old DIO rats had 29% lower plasma ghrelin levels at dark onset but equivalent levels 6 h later compared with DR rats. When subsequently fed a high-energy (HE; 31% fat) diet for 10 days, DIO rats ate 70% more, gained more body and adipose depot weight, had higher leptin and insulin levels, and had 22% lower feed efficiency than DR rats fed HE diet. In DIO rats on HE diet, leptin levels increased significantly at 3 days followed by increased insulin levels at 7 days. These altered DIO leptin and ghrelin responses were associated with 10% lower leptin receptor mRNA expression in the arcuate (ARC), dorsomedial (DMN), and ventromedial hypothalamic nuclei and 13 and 15% lower ghrelin receptor (GHS-R) mRNA expression in the ARC and DMN than in the DR rats. These data suggest that increased ghrelin signaling is not a proximate cause of DIO, whereas reduced leptin sensitivity might play a causal role.
- Published
- 2003
- Full Text
- View/download PDF
39. Stress facilitates body weight gain in genetically predisposed rats on medium-fat diet.
- Author
-
Michel C, Levin BE, and Dunn-Meynell AA
- Subjects
- Animal Feed, Animals, Corticosterone blood, Corticotropin-Releasing Hormone genetics, Eating genetics, Energy Metabolism genetics, Exploratory Behavior physiology, Gene Expression physiology, Hypothalamo-Hypophyseal System metabolism, Male, Obesity genetics, Obesity metabolism, Phenotype, RNA, Messenger analysis, Rats, Rats, Sprague-Dawley, Receptors, Cell Surface genetics, Receptors, Glucocorticoid genetics, Receptors, Leptin, Dietary Fats pharmacology, Stress, Physiological genetics, Stress, Physiological metabolism, Weight Gain genetics
- Abstract
To assess the interaction between stress and energy homeostasis, we immobilized male Sprague-Dawley rats prone to diet-induced obesity (DIO) or diet resistance (DR) once for 20 min and then fed them either low-fat (4.5%) chow or a medium-fat (31%), high-energy (HE) diet for 9 days. Stressed, chow-fed DIO rats gained less, while stressed DIO rats on HE diet gained more body weight and had higher feed efficiency and plasma leptin levels than unstressed controls. Neither stress nor diet affected DR body weight gain. While stress-induced plasma corticosterone levels did not differ between phenotypes, DIO rats were initially more active in an open field and had higher hippocampal dentate gyrus and CA1 glucocorticoid receptor (GR) mRNA than DR rats, regardless of prior stress or diet. HE diet intake was associated with raised dentate gyrus and CA1 GR and amygdalar central nucleus (CeA) corticotropin-releasing hormone (CRH) mRNA expression, while stress was associated with reduced hypothalamic dorsomedial nucleus Ob-R mRNA and CeA CRH specifically in DIO rats fed HE diet. Thus a single stress triggers a complex interaction among weight gain phenotype, diet, and stress responsivity, which determines the body weight and adiposity of a given individual.
- Published
- 2003
- Full Text
- View/download PDF
40. Maternal obesity alters adiposity and monoamine function in genetically predisposed offspring.
- Author
-
Levin BE and Dunn-Meynell AA
- Subjects
- Amygdala anatomy & histology, Amygdala metabolism, Animals, Brain Chemistry genetics, Brain Chemistry physiology, Carrier Proteins metabolism, Diet, Female, Genotype, Hypothalamus anatomy & histology, Hypothalamus metabolism, Male, Membrane Glycoproteins metabolism, Norepinephrine Plasma Membrane Transport Proteins, Phenotype, Pregnancy, Rats, Rats, Sprague-Dawley, Serotonin Plasma Membrane Transport Proteins, Symporters genetics, Symporters metabolism, Adipose Tissue physiopathology, Biogenic Monoamines physiology, Body Composition genetics, Body Composition physiology, Membrane Transport Proteins, Nerve Tissue Proteins, Obesity genetics, Obesity physiopathology
- Abstract
The impact of maternal obesity on brain monoamine function in adult offspring of dams selectively bred to express diet-induced obesity (DIO) or diet resistance (DR) was assessed by making dams obese or lean during gestation and lactation. After 12 wk on chow and 4 wk on a 31% fat diet, offspring hypothalamic nucleus size and [(3)H]nisoxetine binding to norepinephrine transporters (NET) and [(3)H]paroxetine binding to serotonin transporters (SET) were measured. Offspring of obese DIO dams became more obese than all other groups, but maternal obesity did not alter weight gain in DR offspring (25). Maternal obesity was associated with 10-17% enlargement of ventromedial nuclei (VMN) and dorsomedial nuclei in both DIO and DR offspring. Offspring of obese DIO dams had 25-88% lower NET binding in the paraventricular nuclei (PVN), arcuate nuclei, VMN, and the central amygdalar nuclei, while offspring of obese DR dams had 43-67% higher PVN and 90% lower VMN NET binding and a generalized increase in SET binding across all hypothalamic areas compared with other groups. Thus maternal obesity was associated with alterations in offspring brain monoamine metabolism, which varied as a function of genotype and the development of offspring obesity.
- Published
- 2002
- Full Text
- View/download PDF
41. Reduced central leptin sensitivity in rats with diet-induced obesity.
- Author
-
Levin BE and Dunn-Meynell AA
- Subjects
- Animals, Anorexia chemically induced, Anorexia etiology, Arcuate Nucleus of Hypothalamus metabolism, Body Weight, Energy Intake, Food Deprivation physiology, Injections, Intraventricular, Leptin administration & dosage, Leptin blood, Male, Neuropeptide Y genetics, Obesity pathology, RNA, Messenger metabolism, Rats, Rats, Sprague-Dawley, Brain physiopathology, Dietary Fats administration & dosage, Leptin physiology, Obesity etiology, Obesity physiopathology
- Abstract
On low-fat chow diet, rats prone to diet-induced obesity (DIO) have increased arcuate nucleus neuropeptide Y (NPY) expression but similar leptin levels compared with diet-resistant (DR) rats (19). Here, body weight and leptin levels rose in DIO rats, and they defended their higher body weight after only 1 wk on a 31% fat high-energy (HE) diet. However, DIO NPY expression did not fall to DR levels until 4 wk when plasma leptin was 168% of DR levels. When switched to chow, DIO rats lost carcass fat (18). By 10 wk, leptin levels fell to 148% and NPY expression again rose to 150% of DR levels. During 4 wk of food restriction, DIO leptin fell by approximately 50% while NPY increased by 30%. While both returned to control levels by 8 wk, DIO rats still regained all lost weight when fed ad libitum. Finally, the anorexic effect of intracerebroventricular leptin (10 microg) was inversely correlated with subsequent 3-wk weight gain on HE diet. Thus NPY expression and food intake are less sensitive to the leptin's suppressive effects in DIO rats. While this may predispose them to develop DIO, it does not fully explain their defense of a higher body weight on HE diet.
- Published
- 2002
- Full Text
- View/download PDF
42. Glucokinase is the likely mediator of glucosensing in both glucose-excited and glucose-inhibited central neurons.
- Author
-
Dunn-Meynell AA, Routh VH, Kang L, Gaspers L, and Levin BE
- Subjects
- Animals, Brain drug effects, Brain enzymology, Carotid Artery, Internal, Genes, fos drug effects, Glucokinase genetics, Glucose administration & dosage, In Situ Hybridization, Infusions, Intra-Arterial, Neurons drug effects, Neurons enzymology, Obesity metabolism, Rats, Rats, Sprague-Dawley, Transcription, Genetic drug effects, Weight Gain, Brain physiology, Gene Expression Regulation, Enzymologic drug effects, Glucokinase metabolism, Glucose pharmacology, Neurons physiology
- Abstract
Specialized neurons utilize glucose as a signaling molecule to alter their firing rate. Glucose-excited (GE) neurons increase and glucose-inhibited (GI) neurons reduce activity as ambient glucose levels rise. Glucose-induced changes in the ATP-to-ADP ratio in GE neurons modulate the activity of the ATP-sensitive K(+) channel, which determines the rate of cell firing. The GI glucosensing mechanism is unknown. We postulated that glucokinase (GK), a high-Michaelis constant (K(m)) hexokinase expressed in brain areas containing populations of GE and GI neurons, is the controlling step in glucosensing. Double-label in situ hybridization demonstrated neuron-specific GK mRNA expression in locus ceruleus norepinephrine and in hypothalamic neuropeptide Y, pro-opiomelanocortin, and gamma-aminobutyric acid neurons, but it did not demonstrate this expression in orexin neurons. GK mRNA was also found in the area postrema/nucleus tractus solitarius region by RT-PCR. Intracarotid glucose infusions stimulated c-fos expression in the same areas that expressed GK. At 2.5 mmol/l glucose, fura-2 Ca(2+) imaging of dissociated ventromedial hypothalamic nucleus neurons demonstrated GE neurons whose intracellular Ca(2+) oscillations were inhibited and GI neurons whose Ca(2+) oscillations were stimulated by four selective GK inhibitors. Finally, GK expression was increased in rats with impaired central glucosensing (posthypoglycemia and diet-induced obesity) but was unaffected by a 48-h fast. These data suggest a critical role for GK as a regulator of glucosensing in both GE and GI neurons in the brain.
- Published
- 2002
- Full Text
- View/download PDF
43. Defense of body weight depends on dietary composition and palatability in rats with diet-induced obesity.
- Author
-
Levin BE and Dunn-Meynell AA
- Subjects
- Adipose Tissue physiology, Animals, Arcuate Nucleus of Hypothalamus physiology, Body Weight physiology, Diet, Dietary Sucrose pharmacology, Dynorphins genetics, Eating physiology, Food, Formulated, Gene Expression physiology, Leptin blood, Male, Neuropeptide Y genetics, Pro-Opiomelanocortin genetics, RNA, Messenger analysis, Rats, Rats, Sprague-Dawley, Animal Feed, Obesity physiopathology
- Abstract
Sprague-Dawley rats selectively bred for diet-induced obesity (DIO) or diet resistance (DR) were characterized on diets of differing energy content and palatability. Over 10 wk, DR rats on a high-energy (HE) diet (31% fat) gained weight similarly to DR rats fed chow (4.5% fat), but they became obese on a palatable liquid diet (Ensure). DIO rats gained 22% more weight on an HE diet and 50% more on Ensure than chow-fed DIO rats. DIO body weight gains plateaued when switched from HE diet to chow. But, Ensure-fed DIO rats switched to chow spontaneously reduced their intake and weight to that of rats switched from HE diet to chow. They also reduced their hypothalamic proopiomelanocortin and dynorphin but not neuropeptide Y mRNA expression by 17-40%. When reexposed to Ensure after 7 wk, they again overate and matched their body weights to rats maintained on Ensure throughout. All Ensure-fed rats had a selective reduction in dynorphin mRNA in the ventromedial hypothalamic nucleus. Thus genetic background, diet composition, and palatability interact to produce disparate levels of defended body weight and central neuropeptide expression.
- Published
- 2002
- Full Text
- View/download PDF
44. CNS sensing and regulation of peripheral glucose levels.
- Author
-
Levin BE, Dunn-Meynell AA, and Routh VH
- Subjects
- Animals, Diabetes Mellitus metabolism, Humans, Hypoglycemia metabolism, Obesity metabolism, Brain physiology, Chemoreceptor Cells physiology, Glucose metabolism
- Abstract
It is clear that the brain has evolved a mechanism for sensing levels of ambient glucose. Teleologically, this is likely to be a function of its requirement for glucose as a primary metabolic substrate. There is no question that the brain can sense and mount a counterregulatory response to restore very low levels of plasma and brain glucose. But it is less clear that the changes in glucose associated with normal diurnal rhythms and feeding cycles are sufficient to influence either ingestive behavior or the physiologic responses involved in regulating plasma glucose levels. Glucosensing neurons are clearly a distinct class of metabolic sensors with the capacity to respond to a variety of intero- and exteroceptive stimuli. This makes it likely that these glucosensing neurons do participate in physiologically relevant homeostatic mechanisms involving energy balance and the regulation of peripheral glucose levels. It is our challenge to identify the mechanisms by which these neurons sense and respond to these metabolic cues.
- Published
- 2002
- Full Text
- View/download PDF
45. Brain glucosensing and the K(ATP) channel.
- Author
-
Levin BE, Dunn-Meynell AA, and Routh VH
- Subjects
- ATP-Binding Cassette Transporters, Animals, Brain Chemistry genetics, Glucose metabolism, KATP Channels, Mice, Mice, Knockout, Potassium Channels genetics, Potassium Channels, Inwardly Rectifying, Brain Chemistry physiology, Glucose physiology, Potassium Channels physiology
- Published
- 2001
- Full Text
- View/download PDF
46. Sibutramine alters the central mechanisms regulating the defended body weight in diet-induced obese rats.
- Author
-
Levin BE and Dunn-Meynell AA
- Subjects
- Adipose Tissue anatomy & histology, Adipose Tissue drug effects, Animals, Arcuate Nucleus of Hypothalamus drug effects, Body Weight drug effects, Energy Intake, Epinephrine urine, Gene Expression Regulation drug effects, Homeostasis drug effects, Homeostasis physiology, Leptin blood, Neuropeptide Y genetics, Norepinephrine urine, Organ Size drug effects, Pro-Opiomelanocortin genetics, RNA, Messenger genetics, Rats, Appetite Depressants pharmacology, Arcuate Nucleus of Hypothalamus metabolism, Body Weight physiology, Cyclobutanes pharmacology, Feeding Behavior, Obesity physiopathology
- Abstract
Chronic administration of sibutramine lowers body weight, presumably by altering brain monoamine metabolism. Here the effect of sibutramine on sympathoadrenal function (24-h urine norepinephrine and epinephrine levels) and arcuate nucleus (ARC) neuropeptide Y (NPY) and proopiomelanocortin (POMC) expression was assessed in diet-induced obese rats fed a low-fat diet. Chronic (10 wk) sibutramine [5 mg. kg(-1). day(-1) ip; rats fed ad libitum and injected with sibutramine (AS)] lowered body weight by 15% but only transiently (3-4 wk) reduced intake compared with vehicle-treated controls [rats fed chow ad libitum and injected with vehicle daily (AV)]. Other rats food restricted (RS) to 90% of the weight of AS rats and then given sibutramine restored their body weights to the level of AS rats when allowed libitum food intake. After reequilibration, RS rats were again energy restricted to reduce their weight to 90% of AS rats, and additional vehicle-treated rats (RV) were restricted to keep their body weights at the level of AS rats for 3 wk more. Terminally, total adipose depot weights and leptin levels paralleled body weights (AV > AS = RV > RS), although AS rats had heavier abdominal and lighter peripheral depots than RV rats of comparable body weights. Sibutramine treatment increased sympathetic activity, attenuated the increased ARC NPY, and decreased POMC mRNA levels induced by energy restriction in RV rats. Thus sibutramine lowered the defended body weight in association with compensatory changes in those central pathways involved in energy homeostasis.
- Published
- 2000
- Full Text
- View/download PDF
47. Presumed apoptosis and reduced arcuate nucleus neuropeptide Y and pro-opiomelanocortin mRNA in non-coma hypoglycemia.
- Author
-
Tkacs NC, Dunn-Meynell AA, and Levin BE
- Subjects
- Animals, Brain pathology, Brain physiopathology, Hypoglycemia chemically induced, Hypoglycemia metabolism, Hypoglycemia pathology, Hypoglycemic Agents, In Situ Hybridization, In Situ Nick-End Labeling, Insulin, Male, Rats, Rats, Sprague-Dawley, Apoptosis, Arcuate Nucleus of Hypothalamus metabolism, Hypoglycemia physiopathology, Neuropeptide Y genetics, Pro-Opiomelanocortin genetics, RNA, Messenger antagonists & inhibitors
- Abstract
Hypoglycemia reduces sympathoadrenal responses to subsequent hypoglycemic bouts by an unknown mechanism. To assess whether such hypoglycemia-associated autonomic failure is due to actual brain damage, male Sprague-Dawley rats underwent 1-h bouts of insulin-induced (5 U/kg i.v.) hypoglycemia (1.6-2.8 mmol/l) 1 or 3 times on alternate days. Rats remained alert and were rescued with intravenous glucose at 60-80 min. Plasma epinephrine and corticosterone responses were significantly reduced during the second and third bouts. Brains from these rats were processed by the terminal transferase-mediated deoxyuridine triphosphate-biotin nick end-labeling (TUNEL) procedure as an index of apoptotic cell death at 24, 48, or 96 h after their first bout. At 48 h, but not 24 h, TUNEL+ cells were consistently seen only in the arcuate nucleus (arcuate hypothalamic nucleus [ARC]). Hypoglycemic rats had 188% more apoptotic ARC cells (1 bout 39+/-5; 3 bouts 37+/-4) than euglycemic controls (13+/-3;P = 0.001). In situ hybridization for neuropeptide Y (NPY) and proopiomelanocortin (POMC) mRNA was performed in sections of ARC containing maximal numbers of apoptotic cells as well as in other fresh frozen brains. After 1 bout, NPY (0.041+/-0.003) and POMC (0.119+/-0.022) mRNA were decreased, respectively, by 52 and 55% vs. controls (NPY 0.076+/-0.007; POMC 0.222+/-0.020; P = 0.01). NPY (0.029+/-0.002) but not POMC (0.093+/-0.013) fell 29% further after a third bout. NPY (r = -0.721; P = 0.001) and POMC (r = -0.756; P = 0.001) mRNA levels correlated negatively with the number of apoptotic ARC cells in the same sections. Thus, non-coma hypoglycemia produces apparent apoptotic cell death with reduced NPY and POMC expression selectively in the ARC. This may contribute to the reduced counterregulatory response following repeated bouts of hypoglycemia.
- Published
- 2000
- Full Text
- View/download PDF
48. Localization of glucokinase gene expression in the rat brain.
- Author
-
Lynch RM, Tompkins LS, Brooks HL, Dunn-Meynell AA, and Levin BE
- Subjects
- Animals, Arcuate Nucleus of Hypothalamus metabolism, Base Sequence genetics, Brain metabolism, Hypothalamus metabolism, In Situ Hybridization, Male, Potassium Channels metabolism, Punctures, RNA, Messenger metabolism, Rats, Rats, Sprague-Dawley, Reverse Transcriptase Polymerase Chain Reaction, Tissue Distribution, Brain physiology, Gene Expression physiology, Glucokinase genetics, Potassium Channels, Inwardly Rectifying
- Abstract
The brain contains a subpopulation of glucosensing neurons that alter their firing rate in response to elevated glucose concentrations. In pancreatic beta-cells, glucokinase (GK), the rate-limiting enzyme in glycolysis, mediates glucose-induced insulin release by regulating intracellular ATP production. A similar role for GK is proposed to underlie neuronal glucosensing. Via in situ hybridization, GK mRNA was localized to hypothalamic areas that are thought to contain relatively large populations of glucosensing neurons (the arcuate, ventromedial, dorsomedial, and paraventricular nuclei and the lateral area). GK also was found in brain areas without known glucosensing neurons (the lateral habenula, the bed nucleus stria terminalis, the inferior olive, the retrochiasmatic and medial preoptic areas, and the thalamic posterior paraventricular, interpeduncular, oculomotor, and anterior olfactory nuclei). Conversely, GK message was not found in the nucleus tractus solitarius, which contains glucosensing neurons, or in ependymal cells lining the third ventricle, where others have described its presence. In the arcuate nucleus, >75% of neuropeptide Y-positive neurons also expressed GK, and most GK+ neurons also expressed KIR6.2 (the pore-forming subunit of the ATP-sensitive K+ channel). The anatomic distribution of GK mRNA was confirmed in micropunch samples of hypothalamus via reverse transcription-polymerase chain reaction (RT-PCR). Nucleotide sequencing of the recovered PCR product indicated identity with nucleotides 1092-1411 (within exon 9 and 10) of hepatic and beta-cell GK. The specific anatomic localization of GK mRNA in hypothalamic areas known to contain glucosensing neurons and the coexpression of KIR6.2 and NPY in GK+ neurons support a role for GK as a primary determinant of glucosensing in neuropeptide neurons that integrate multiple signals relating to peripheral energy metabolism.
- Published
- 2000
- Full Text
- View/download PDF
49. Defense of body weight against chronic caloric restriction in obesity-prone and -resistant rats.
- Author
-
Levin BE and Dunn-Meynell AA
- Subjects
- Adipose Tissue pathology, Animals, Circadian Rhythm, Diet adverse effects, Disease Susceptibility, Insulin blood, Leptin blood, Male, Norepinephrine urine, Obesity etiology, Obesity metabolism, Organ Size physiology, Rats, Rats, Sprague-Dawley, Time Factors, Body Weight, Diet, Fat-Restricted, Energy Intake, Obesity pathology
- Abstract
Half of Sprague-Dawley rats develop and defend diet-induced obesity (DIO) or diet resistance (DR) when fed a high-energy (HE) diet. Here, adult male rats were made DIO or DR after 10 wk on HE diet. Then half of each group was food restricted for 8 wk on chow to maintain their body weights at 90% of their respective baselines. Rate and magnitude of weight loss were comparable, but maintenance energy intake and the degree of sympathetic activity (24-h urine norepinephrine) inhibition were 17 and 29% lower, respectively, in restricted DR than DIO rats. Restricted DIO rats reduced adipose depot weights, plasma leptin, and insulin levels by 35%. Restricted DR rats reduced none of these. When fed ad libitum, both DR and DIO rats returned to the body weights of their respective chow-fed phenotype controls within 2 wk. This was associated with increased adipose mass and leptin and insulin levels only in DIO rats. Thus DR rats appear to alter primarily their lean body mass, whereas DIO rats primarily alter their adipose mass during chronic caloric restriction and refeeding.
- Published
- 2000
- Full Text
- View/download PDF
50. Brain glucose sensing and body energy homeostasis: role in obesity and diabetes.
- Author
-
Levin BE, Dunn-Meynell AA, and Routh VH
- Subjects
- Animals, Energy Metabolism physiology, Glucose analysis, Humans, Rats, Brain metabolism, Diabetes Mellitus, Type 1 metabolism, Diabetes Mellitus, Type 2 metabolism, Glucose metabolism, Homeostasis physiology, Obesity metabolism
- Abstract
The brain has evolved mechanisms for sensing and regulating glucose metabolism. It receives neural inputs from glucosensors in the periphery but also contains neurons that directly sense changes in glucose levels by using glucose as a signal to alter their firing rate. Glucose-responsive (GR) neurons increase and glucose-sensitive (GS) decrease their firing rate when brain glucose levels rise. GR neurons use an ATP-sensitive K+ channel to regulate their firing. The mechanism regulating GS firing is less certain. Both GR and GS neurons respond to, and participate in, the changes in food intake, sympathoadrenal activity, and energy expenditure produced by extremes of hyper- and hypoglycemia. It is less certain that they respond to the small swings in plasma glucose required for the more physiological regulation of energy homeostasis. Both obesity and diabetes are associated with several alterations in brain glucose sensing. In rats with diet-induced obesity and hyperinsulinemia, GR neurons are hyporesponsive to glucose. Insulin-dependent diabetic rats also have abnormalities of GR neurons and neurotransmitter systems potentially involved in glucose sensing. Thus the challenge for the future is to define the role of brain glucose sensing in the physiological regulation of energy balance and in the pathophysiology of obesity and diabetes.
- Published
- 1999
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.