Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
Add more filters











Publication year range
1.
Alzheimers Res Ther ; 13(1): 32, 2021 01 27.
Article in English | MEDLINE | ID: mdl-33504364

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.


Subject(s)
Erythropoietin , Tauopathies , Animals , Brain/metabolism , Disease Models, Animal , Female , Humans , Mice , Mice, Transgenic , Tauopathies/drug therapy , tau Proteins/genetics , tau Proteins/metabolism
2.
Front Aging Neurosci ; 11: 252, 2019.
Article in English | MEDLINE | ID: mdl-31572168

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.

3.
Diabetes ; 67(5): 805-817, 2018 05.
Article in English | MEDLINE | ID: mdl-29467172

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.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Islet Amyloid Polypeptide/metabolism , Leptin/metabolism , MAP Kinase Signaling System , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Pro-Opiomelanocortin/metabolism , 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
4.
Am J Physiol Regul Integr Comp Physiol ; 311(6): R1032-R1044, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27629888

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.


Subject(s)
Hypothalamus/physiopathology , Islet Amyloid Polypeptide/administration & dosage , Leptin/metabolism , Obesity/drug therapy , Obesity/physiopathology , 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
5.
Am J Physiol Regul Integr Comp Physiol ; 311(4): R764-R770, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27534878

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.


Subject(s)
Interleukin-6/immunology , Leptin/immunology , Neurons/metabolism , Obesity/immunology , Receptors, Leptin/metabolism , Ventromedial Hypothalamic Nucleus/immunology , Animals , Cells, Cultured , Dietary Fats , Male , Obesity/chemically induced , Rats , Rats, Sprague-Dawley
6.
Am J Physiol Regul Integr Comp Physiol ; 310(4): R355-65, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26676252

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.


Subject(s)
Islet Amyloid Polypeptide/metabolism , Leptin/metabolism , Obesity/physiopathology , Ventromedial Hypothalamic Nucleus/metabolism , 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
7.
Am J Physiol Regul Integr Comp Physiol ; 308(10): R872-8, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25786485

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 ß-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.


Subject(s)
Energy Intake/physiology , Ketone Bodies/metabolism , Neurons/metabolism , Obesity/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , 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
8.
Diabetes ; 64(5): 1621-31, 2015 May.
Article in English | MEDLINE | ID: mdl-25409701

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.


Subject(s)
Interleukin-6/metabolism , Islet Amyloid Polypeptide/pharmacology , Leptin/metabolism , Signal Transduction/physiology , Ventromedial Hypothalamic Nucleus/metabolism , 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
9.
Am J Physiol Regul Integr Comp Physiol ; 308(3): R188-98, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25477422

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.


Subject(s)
Blood Glucose , CD36 Antigens/metabolism , Energy Intake/physiology , Fatty Acids/metabolism , Homeostasis/physiology , Obesity/metabolism , Adipose Tissue/metabolism , Animal Feed , Animals , Blood Glucose/metabolism , Body Weight/physiology , Disease Models, Animal , Insulin/metabolism , Leptin , Male , Rats
10.
Diabetes ; 63(4): 1259-69, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24379353

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 ß-hydroxybutyrate (ß-OHB) and VMH-to-serum ß-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.


Subject(s)
Eating/drug effects , Fatty Acids/pharmacology , Hypothalamus/physiology , Ketone Bodies/pharmacology , Neurons/physiology , 3-Hydroxybutyric Acid/blood , Animals , Hypothalamus/drug effects , Male , Neurons/drug effects , Rats
11.
Cell Metab ; 17(4): 607-17, 2013 Apr 02.
Article in English | MEDLINE | ID: mdl-23562080

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.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Blood-Brain Barrier/metabolism , Ependyma/metabolism , Vascular Endothelial Growth Factor A/metabolism , 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
12.
Endocrinology ; 151(9): 4270-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20668022

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.


Subject(s)
Diet , Leptin/blood , Litter Size/physiology , Obesity/physiopathology , 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
13.
Endocrinology ; 151(1): 143-52, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19875640

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.


Subject(s)
Amyloid/physiology , Drug Synergism , Leptin/physiology , Models, Animal , Rodentia , 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 , Rodentia/genetics , Rodentia/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
14.
Physiol Behav ; 98(4): 511-6, 2009 Oct 19.
Article in English | MEDLINE | ID: mdl-19698729

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.


Subject(s)
Dendrites/genetics , Dendrites/metabolism , Diet , Neurons/cytology , Obesity/metabolism , Ventromedial Hypothalamic Nucleus/cytology , Animals , Body Weight/genetics , Disease Models, Animal , Eating/genetics , Male , Obesity/genetics , Rats , Silver Staining/methods
15.
Am J Physiol Regul Integr Comp Physiol ; 297(3): R655-64, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19535676

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.


Subject(s)
Calcium Signaling , Energy Metabolism , Glucose/metabolism , Neurons/metabolism , Oleic Acid/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , 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
16.
J Neurosci ; 29(21): 7015-22, 2009 May 27.
Article in English | MEDLINE | ID: mdl-19474328

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.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Blood Glucose/physiology , Feeding Behavior/physiology , Glucose/deficiency , Ventromedial Hypothalamic Nucleus/metabolism , 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
17.
Am J Physiol Regul Integr Comp Physiol ; 296(3): R537-48, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19158409

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.


Subject(s)
Diet , Leptin/physiology , Obesity/physiopathology , Physical Conditioning, Animal/physiology , Signal Transduction/physiology , 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
18.
Am J Physiol Regul Integr Comp Physiol ; 296(3): R521-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19118094

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.


Subject(s)
Diet , Leptin/pharmacology , Neurons/drug effects , Obesity/etiology , Obesity/genetics , Ventromedial Hypothalamic Nucleus/drug effects , 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
19.
Diabetes ; 57(5): 1371-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18292346

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.


Subject(s)
Glucokinase/metabolism , Hypoglycemia/enzymology , Insulin/pharmacology , Ventromedial Hypothalamic Nucleus/enzymology , Animals , Diabetes Mellitus, Experimental/enzymology , Enzyme Activation , Homeostasis , Hypoglycemia/chemically induced , Kinetics , Male , Rats , Rats, Sprague-Dawley
20.
Am J Physiol Regul Integr Comp Physiol ; 294(3): R784-92, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18094065

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.


Subject(s)
Glucose/metabolism , Glucose/pharmacology , Hypoglycemia/physiopathology , Neurons/drug effects , Ventromedial Hypothalamic Nucleus/physiopathology , 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
SELECTION OF CITATIONS
SEARCH DETAIL