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










Publication year range
1.
Age (Dordr) ; 35(3): 533-47, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22411258

ABSTRACT

Biological aging alters the metabolism and volume of adipose tissue depots. Recent evidence suggests that circadian mechanisms play a role in promoting adipogenesis, obesity, and lipodystrophy. The current study compared cohorts of younger (5-9 months) and older (24-28 months) C57BL/6 mice as a function of biological age and circadian time. Advanced age significantly reduced the weight of the brown, epididymal, inguinal, and retroperitoneal adipose depots but not total body weight. The older mice reduced their physical activity by >50% and delayed their activity initiation after light offset. The expressed transcriptome in brown and white adipose depots and liver of both cohorts displayed evidence of circadian rhythmicity; however, the oscillating mRNAs differed significantly between age groups and across tissues. The amplitude of Cry1, a component of the negative arm of the circadian apparatus, and downstream regulators such as Rev-erbα were elevated in the older relative to the younger cohorts as a function of circadian time. Overall, transcript levels differed significantly for 557 (inguinal adipose), 1,016 (liver), and 1,021 (brown adipose) expressed sequences between the cohorts as a function of age. These included transcripts encoding proteins within the canonical and non-canonical Wnt pathways. Since the Wnt pathway regulates adipose stem cell differentiation and shares a critical enzyme, glycogen synthase kinase 3ß, with the circadian mechanism, the intersection between these two fundamental regulatory mechanisms merits further investigation with respect to biological aging of adipose tissues.


Subject(s)
Adipogenesis/physiology , Adipose Tissue/physiology , Aging/physiology , Circadian Rhythm/physiology , Animals , Cell Differentiation , Disease Models, Animal , Follow-Up Studies , Male , Mice , Mice, Inbred C57BL
2.
Curr Opin Clin Nutr Metab Care ; 14(6): 554-61, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21986477

ABSTRACT

PURPOSE OF REVIEW: Over the past decade, evidence has accumulated from basic science, clinical and epidemiological studies linking circadian mechanisms to adipose tissue biology and its related comorbidities, diabetes, metabolic syndrome and obesity. This review highlights recent in-vitro and in-vivo findings from murine, human and model organism studies. RECENT FINDINGS: High-fat diets attenuate circadian mechanisms in murine adipose depots and these effects appear to be due to obesity rather than hyperglycemia. Deletion of circadian regulatory genes such as AMPK1 and nocturnin alter the circadian biology of adipose tissue. Unlike the mouse, circadian gene oscillation in human adipose tissue appears to be independent of BMI and diabetes status, suggesting that circadian mechanistic variation occurs across species. Clues for future directions in this emerging field come from studies of the hibernation and torpor state in mammals and infection models involving the Drosophila metabolic organ or 'fat body'. SUMMARY: There is a growing consensus that circadian rhythms and metabolism are tightly regulated in adipose tissue and peripheral metabolic organs. Although central mechanisms are critical, autonomous clocks exist within the adipocytes themselves. Future circadian advances are likely to result from the studies of adipose tissue-specific gene deletions.


Subject(s)
Adipose Tissue/metabolism , Adipose Tissue/physiopathology , Circadian Rhythm/physiology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Adipocytes , Animals , Diet, High-Fat , Drosophila/genetics , Drosophila/metabolism , Gene Deletion , Humans , Mice , Models, Animal , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptome
3.
Physiol Behav ; 104(4): 546-54, 2011 Sep 26.
Article in English | MEDLINE | ID: mdl-21497617

ABSTRACT

The central nervous melanocortin system is a neural network linking nutrient-sensing systems with hypothalamic, limbic and hindbrain neurons regulating behavior and metabolic homeostasis. Primary melanocortin neurons releasing melanocortin receptor ligands residing in the hypothalamic arcuate nucleus are regulated by nutrient-sensing and metabolic signals. A smaller group of primary neurons releasing melanocortin agonists in the nucleus tractus solitarius in the brainstem are also regulated by signals of metabolic state. Two melanocortin receptors regulate energy homeostasis. Melanocortin-4 receptors regulate satiety and autonomic outputs controlling peripheral metabolism. The functions of melanocortin-3 receptors (MC3R) expressed in hypothalamic and limbic structures are less clear. Here we discuss published data and preliminary observations from our laboratory suggesting that neural MC3R regulate inputs into systems governing the synchronization of rhythms in behavior and metabolism with nutrient intake. Mice subjected to a restricted feeding protocol, where a limited number of calories are presented at a 24h interval, rapidly exhibit bouts of increased wakefulness and activity which anticipate food presentation. The full expression of these responses is dependent on MC3R. Moreover, MC3R knockout mice are unique in exhibiting a dissociation of weight loss from improved glucose homeostasis when subject to a restricted feeding protocol. While mice lacking MC3R fed ad libitum exhibit normal to moderate hyperinsulinemia, when subjected to a restricted protocol they develop hyperglycemia, glucose intolerance, and dyslipidemia. Collectively, our data suggest that the central nervous melanocortin system is a point convergence in the control of energy balance and the expression of rhythms anticipating nutrient intake.


Subject(s)
Energy Metabolism/physiology , Homeostasis/physiology , Receptor, Melanocortin, Type 3/physiology , Animals , Anticipation, Psychological/physiology , Circadian Clocks/physiology , Circadian Rhythm/physiology , Hypothalamus/physiology , Melanocortins/physiology , Neural Pathways/physiology , Satiety Response/physiology , Signal Transduction/physiology
4.
Nat Rev Endocrinol ; 7(2): 98-107, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21178997

ABSTRACT

Circadian rhythms make a critical contribution to endocrine functions that involve adipose tissue. These contributions are made at the systemic, organ and stem cell levels. The transcription factors and enzymes responsible for the maintenance of circadian rhythms in adipose depots and other peripheral tissues that are metabolically active have now been identified. Furthermore, the circadian regulation of glucose and lipid metabolism is well-established. Animal and human models provide strong evidence that disturbances in circadian pathways are associated with an increased risk of type 2 diabetes mellitus, obesity and their comorbidities. Thus, circadian mechanisms represent a novel putative target for therapy in patients with metabolic diseases.


Subject(s)
Adipose Tissue/metabolism , Circadian Clocks/physiology , Adipose Tissue/physiology , Animals , Glucose/metabolism , Humans , Lipid Metabolism
5.
Endocrinology ; 151(4): 1570-80, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20160133

ABSTRACT

The mechanisms linking intrauterine growth retardation (IUGR) with adulthood obesity and diabetes are unclear. These studies investigated energy homeostasis in 8- and 20-wk-old male and female mice subjected to protein deficiency in utero. Pregnant C57BL/6J female mice were fed a protein-deficient diet (6% protein). Undernourished offspring (UO) and controls (CO) were cross-fostered to lactating dams fed a 20% control diet. The 24-h profiles of energy expenditure, feeding behavior, physical activity, and whole-body substrate preference was assessed using 8-wk UO and CO weaned onto control diet. Blood chemistries, glucose tolerance, and expression of genes involved in hepatic lipid and glucose metabolism were analyzed in 8- and 20-wk-old CO and UO fed control or a high-fat diet. UO exhibited IUGR with catch-up growth at 8 wk of age and increased severity of diet-induced obesity and insulin resistance by 20 wk of age. Therefore, fetal malnutrition in the C57BL/6J mouse increases sensitivity to diet-induced obesity. Abnormal daily rhythms in food intake and metabolism, increased lipogenesis, and inflammation preceded obesity in the UO group. Arrhythmic expression of circadian oscillator genes was evident in brain, liver, and muscle of UO at 8 and 20 wk of age. Expression of the clock-associated nuclear receptor and transcription repressor Rev-erbalpha was reduced in liver and muscle of UO. Altered circadian physiology may be symptomatic of the metabolic dysregulation associated with IUGR, and altered feeding behavior and substrate metabolism may contribute to the obese phenotype.


Subject(s)
Circadian Rhythm/physiology , Obesity/etiology , Prenatal Exposure Delayed Effects/metabolism , Prenatal Nutritional Physiological Phenomena/physiology , Protein Deficiency/metabolism , ARNTL Transcription Factors/genetics , ARNTL Transcription Factors/metabolism , Adipose Tissue, White/metabolism , Analysis of Variance , Animals , Blood Glucose/metabolism , Brain/metabolism , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Eating/physiology , Energy Metabolism/physiology , Feeding Behavior/physiology , Female , Gene Expression , Glucose Tolerance Test , Insulin Resistance/physiology , Lipid Metabolism/physiology , Liver/metabolism , Male , Mice , Motor Activity/physiology , Muscle, Skeletal/metabolism , Neuropeptide Y/genetics , Neuropeptide Y/metabolism , Obesity/metabolism , Period Circadian Proteins/genetics , Period Circadian Proteins/metabolism , Pregnancy , Pro-Opiomelanocortin/genetics , Pro-Opiomelanocortin/metabolism , Severity of Illness Index
6.
FASEB J ; 24(3): 862-72, 2010 Mar.
Article in English | MEDLINE | ID: mdl-19837866

ABSTRACT

Melanocortin-3 receptors (Mc3rs) in the central nervous system are involved in expression of anticipatory rhythms and synchronizing clocks maintaining circadian rhythms during restricted feeding (RF) [mice housed under a 12-h light-dark cycle with lights on between zeitgeber time (ZT) 0 to ZT12 fed 60% of normal calories between ZT7 and ZT11]. Because the systems governing circadian rhythms are important for adaptation to RF, we investigated whether Mc3rs are required for metabolic adaption to RF. Mc3r(-/-) mice subjected to RF exhibited normal weight loss; however, they developed hyperinsulinemia, glucose intolerance, increased expression of lipogenic genes, and increased ketogenesis relative to controls. Rhythmic expression of transcription factors regulating liver clock activity and energy metabolism (Bmal1, Rev-erbalpha, Pgc1, Foxo1, Hnf4alpha, and Pck1) was severely compromised in Mc3r(-/-) mice during RF. Inhibition of neural melanocortin receptors by agouti-related peptide also attenuated rhythmicity in the hepatic expression of these genes during RF. Collectively, these data suggest that neural Mc3rs are important for adapting metabolism and maintaining rhythms of liver metabolism during periods when feeding is restricted to the light cycle.-Sutton, G. M., Begriche, K., Kumar, K. G., Gimble, J. M., Perez-Tilve, D., Nogueiras, R., McMillan, R. P., Hulver, M. W., Tschöp, M. H., Butler, A. A. Central nervous system melanocortin-3 receptors are required for synchronizing metabolism during entrainment to restricted feeding during the light cycle.


Subject(s)
Caloric Restriction , Central Nervous System/metabolism , Photoperiod , Receptor, Melanocortin, Type 3/physiology , Agouti-Related Protein/pharmacology , Animals , Central Nervous System/radiation effects , Electrophoresis, Polyacrylamide Gel , Energy Metabolism/drug effects , Energy Metabolism/genetics , Fatty Acids/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Glucose Intolerance/genetics , Glucose Tolerance Test , Hyperinsulinism/genetics , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Mutant Strains , Receptor, Melanocortin, Type 3/antagonists & inhibitors , Receptor, Melanocortin, Type 3/genetics
7.
Peptides ; 30(10): 1892-900, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19646498

ABSTRACT

Melanocortin receptor agonists act in the brain to regulate food intake and body weight and, independently of these actions, affect insulin sensitivity. These experiments investigated the function of novel non-selective melanocortin receptor agonists (BIM-22493, BIM-22511) that cross the blood-brain barrier when administered peripherally. Treatment of diet induced obese C57BL/6J (B6) mice with melanocortin agonists administered peripherally improved obesity, hyperinsulinemia (approximately 50%) and fatty liver disease. Specificity of function was determined using B6 melanocortin-3 and melanocortin-4 receptor knockout mice (MC3RKO, MC4RKO). Chow fed MC4RKO but not MC3RKO used for these tests exhibited obesity, hyperinsulinemia and severe hepatosteatosis associated with increased expression of insulin-stimulated genes involved in lipogenesis. Reduced food intake associated with acute BIM-22493 treatment, and weight loss associated with 14 days of treatment with BIM-22511, required functional MC4R but not MC3R. However, while 14 days of treatment with BIM-22511 did not affect body weight and even increased cumulative food intake in MC4RKO, a significant reduction (approximately 50%) in fasting insulin was still observed. Despite lowering insulin, chronic treatment with BIM-22511 did not improve hepatosteatosis in MC4RKO, and did not affect hepatic lipogenic gene expression. Together, these results demonstrate that peripherally administered melanocortin receptor agonists regulate body weight, liver metabolism and glucose homeostasis through independent pathways. MC4R are necessary for melanocortin agonist-induced weight loss and improvements in liver metabolism, but are not required for improvements in hyperinsulinemia. Agonists with activity at MC4R improve glucose homeostasis at least partially by causing weight loss, however other melanocortin receptors may have potential for treating aberrations in glucose homeostasis associated with obesity.


Subject(s)
Receptor, Melanocortin, Type 3/agonists , Receptor, Melanocortin, Type 3/genetics , Receptor, Melanocortin, Type 4/agonists , Receptor, Melanocortin, Type 4/genetics , alpha-MSH/analogs & derivatives , Animals , Diet , Eating , Energy Metabolism , Female , Glucose/metabolism , Humans , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/metabolism , Receptor, Melanocortin, Type 3/metabolism , Receptor, Melanocortin, Type 4/metabolism , Weight Loss , alpha-MSH/pharmacology
8.
Am J Physiol Regul Integr Comp Physiol ; 296(4): R845-54, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19176891

ABSTRACT

The satiating potency of CCK has been well characterized, including its mediation by capsaicin-sensitive vagal primary afferents. We have previously shown that peripherally administered CCK activates the MAPK-signaling cascade in a population of nucleus of the solitary tract (NTS) neurons and that preventing ERK1/2 phosphorylation partly attenuates CCK's satiating potency. The aim of this study was to identify the neurochemical phenotypes of the NTS neurons that exhibit CCK-induced activation of ERK1/2. Using confocal microscopy, we demonstrate that intraperitoneal CCK administration increases the number of neurons that express phosphorylated ERK1/2 (pERK1/2) in the medial and commissural subnuclei of the NTS and that CCK-induced expression of ERK1/2 is increased in tyrosine hydroxylase-immunoreactive neurons. Using Western blot analysis, we show that the robust increase in tyrosine hydroxylase phosphorylation obtained with intraperitoneal CCK is significantly attenuated in rats pretreated with the ERK-pathway blocker U0126 injected into the 4th ventricle. In addition, CCK injections increased pERK1/2 expression in POMC neurons in the NTS. In contrast, only the rare GAD67, neuronal nitric oxide synthase, and leptin-responsive neuron exhibited CCK-induced pERK immunoreactivity. We conclude that activation of POMC-immunoreactive neurons and tyrosine hydroxylase activity via the ERK-signaling pathway in the NTS likely contributes to CCK's satiating effects.


Subject(s)
Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/drug effects , Peptide Fragments/administration & dosage , Signal Transduction/drug effects , Sincalide/analogs & derivatives , Solitary Nucleus/drug effects , Animals , Butadienes/administration & dosage , Catecholamines/metabolism , Enzyme Activation , Female , Glutamate Decarboxylase/genetics , Green Fluorescent Proteins/genetics , Injections, Intraperitoneal , Injections, Intraventricular , Leptin/metabolism , Male , Mice , Mice, Transgenic , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Neurons/enzymology , Nitric Oxide Synthase Type I/metabolism , Nitriles/administration & dosage , Phenotype , Phosphorylation , Pro-Opiomelanocortin/genetics , Promoter Regions, Genetic , Protein Kinase Inhibitors/administration & dosage , Rats , Rats, Sprague-Dawley , Receptors, Leptin/genetics , Satiation/drug effects , Sincalide/administration & dosage , Solitary Nucleus/cytology , Solitary Nucleus/enzymology , Tyrosine 3-Monooxygenase/metabolism
9.
Cell Metab ; 8(6): 468-81, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19041763

ABSTRACT

Obesity and nutrient homeostasis are linked by mechanisms that are not fully elucidated. Here we describe a secreted protein, adropin, encoded by a gene, Energy Homeostasis Associated (Enho), expressed in liver and brain. Liver Enho expression is regulated by nutrition: lean C57BL/6J mice fed high-fat diet (HFD) exhibited a rapid increase, while fasting reduced expression compared to controls. However, liver Enho expression declines with diet-induced obesity (DIO) associated with 3 months of HFD or with genetically induced obesity, suggesting an association with metabolic disorders in the obese state. In DIO mice, transgenic overexpression or systemic adropin treatment attenuated hepatosteatosis and insulin resistance independently of effects on adiposity or food intake. Adropin regulated expression of hepatic lipogenic genes and adipose tissue peroxisome proliferator-activated receptor gamma, a major regulator of lipogenesis. Adropin may therefore be a factor governing glucose and lipid homeostasis, which protects against hepatosteatosis and hyperinsulinemia associated with obesity.


Subject(s)
Blood Proteins/physiology , Energy Metabolism , Lipid Metabolism , Proteins/physiology , Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , Amino Acid Sequence , Animals , Base Sequence , Benzoates/chemistry , Benzoates/metabolism , Benzylamines/chemistry , Benzylamines/metabolism , Blood Proteins/genetics , Blood Proteins/metabolism , Cells, Cultured , DNA-Binding Proteins/agonists , DNA-Binding Proteins/metabolism , Fasting , Fatty Liver/metabolism , Female , Humans , Intercellular Signaling Peptides and Proteins , Leptin/metabolism , Liver X Receptors , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Obesity/genetics , Obesity/metabolism , Orphan Nuclear Receptors , Peptides , Proteins/genetics , Proteins/metabolism , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Cytoplasmic and Nuclear/agonists , Receptors, Cytoplasmic and Nuclear/metabolism
10.
J Neurosci ; 28(48): 12946-55, 2008 Nov 26.
Article in English | MEDLINE | ID: mdl-19036988

ABSTRACT

Entrainment of anticipatory activity and wakefulness to nutrient availability is a poorly understood component of energy homeostasis. Restricted feeding (RF) paradigms with a periodicity of 24 h rapidly induce entrainment of rhythms anticipating food presentation that are independent of master clocks in the suprachiasmatic nucleus (SCN) but do require other hypothalamic structures. Here, we report that the melanocortin system, which resides in hypothalamic structures required for food entrainment, is required for expression of food entrainable rhythms. Food anticipatory activity was assessed in wild-type (WT) and melanocortin-3 receptor-deficient (Mc3r-/-) C57BL/J mice by wheel running, spontaneous locomotory movement, and measurement of wakefulness. WT mice housed in wheel cages subject to RF exhibited increased wheel activity during the 2 h preceding meal presentation, which corresponded with an increase in wakefulness around meal time and reduced wakefulness during the dark. WT mice also exhibited increased x- and z-movements centered around food initiation. The activity-based responses to RF were significantly impaired in mice lacking Mc3r. RF also failed to increase wakefulness in the 2 h before food presentation in Mc3r-/- mice. Food entrainment requires expression of Neuronal PAS domain 2 (Npas2) and Period2 (Per2) genes, components of the transcriptional machinery maintaining a clock rhythm. Analysis of cortical gene expression revealed severe abnormalities in rhythmic expression of clock genes (Bmal1, Npas2, Per2) under ad libitum and RF conditions. In summary, Mc3r are required for expression of anticipatory patterns of activity and wakefulness during periods of limited nutrient availability and for normal regulation of cortical clock function.


Subject(s)
Appetite Regulation/genetics , Appetite/genetics , Brain/metabolism , Circadian Rhythm/genetics , Pro-Opiomelanocortin/metabolism , Receptor, Melanocortin, Type 3/metabolism , ARNTL Transcription Factors , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Brain/anatomy & histology , Cell Cycle Proteins/genetics , Cerebral Cortex/anatomy & histology , Cerebral Cortex/metabolism , Feeding Behavior/physiology , Gene Expression Regulation/genetics , Hypothalamus/anatomy & histology , Hypothalamus/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/genetics , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Period Circadian Proteins , Receptor, Melanocortin, Type 3/genetics , Transcription Factors/genetics , Wakefulness/genetics
11.
Peptides ; 29(1): 104-11, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18054119

ABSTRACT

Melanocortin receptors are considered promising candidates for the treatment of behavioral and metabolic disorders ranging from obesity to anorexia and cachexia. These experiments examined the response of mice to peripheral injections of two compounds. PG932 is a derivative of SHU9119 which is non-selective antagonist of melanocortin-3 and melanocortin-4 receptors (Mc3r and Mc4r). PG946 is a derivative of a hybrid of alpha- and beta-MSH, and is a moderately selective Mc3r antagonist. SHU9119 increases food intake when administered intracerebroventricularly but is without effect when injected into the periphery. In contrast, PG932 was found to be highly effective at stimulating food intake when administered peripherally by intraperitoneal injection. The orexigenic effect of PG932 required functional Mc4r, suggesting that inhibition of this receptor is involved in the stimulation of food intake. PG946 did not significantly affect on feeding behavior. PG932 is thus a useful new compound for studies examining the regulation of appetite and energy balance, and may also prove useful for the treatment of cachectic conditions.


Subject(s)
Eating/drug effects , Melanocyte-Stimulating Hormones/administration & dosage , Melanocyte-Stimulating Hormones/pharmacology , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/pharmacology , Receptor, Melanocortin, Type 3/antagonists & inhibitors , Receptor, Melanocortin, Type 4/antagonists & inhibitors , Animals , Anorexia/chemically induced , Anorexia/drug therapy , Dizziness/chemically induced , Dizziness/drug therapy , Dose-Response Relationship, Drug , Injections, Intraperitoneal , Injections, Intraventricular , Lipopolysaccharides/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Melanocortin, Type 4/deficiency , Receptor, Melanocortin, Type 4/physiology
12.
Obesity (Silver Spring) ; 15(11): 2664-72, 2007 Nov.
Article in English | MEDLINE | ID: mdl-18070757

ABSTRACT

OBJECTIVE: To investigate the involvement of hypoadiponectinemia and inflammation in coupling obesity to insulin resistance in melanocortin-3 receptor and melanocortin-4 receptor knockout (KO) mice (Mc3/4rKO). RESEARCH METHODS AND PROCEDURES: Sera and tissue were collected from 6-month-old Mc3rKO, Mc4rKO, and wild-type C57BL6J litter mates maintained on low-fat diet or exposed to high-fat diet (HFD) for 1 or 3 months. Inflammation was assessed by both real-time polymerase chain reaction analysis of macrophage-specific gene expression and immunohistochemistry. RESULTS: Mc4rKO exhibited hypoadiponectinemia, exacerbated by HFD and obesity, previously reported in murine models of obesity. Mc4r deficiency was also associated with high levels of macrophage infiltration of adipose tissue, again exacerbated by HFD. In contrast, Mc3rKO exhibited normal serum adiponectin levels, irrespective of diet or obesity, and a delayed inflammatory response to HFD relative to Mc4rKO. DISCUSSION: Our findings suggest that severe insulin resistance of Mc4rKO fed a HFD, as reported in other models of obesity such as leptin-deficient (Lep(ob)/Lep(ob)) and KK-A(y) mice, is linked to reduced serum adiponectin and high levels of inflammation in adipose tissue. Conversely, maintenance of normal serum adiponectin may be a factor in the relatively mild insulin-resistant phenotype of severely obese Mc3rKO. Mc3rKO are, thus, a unique mouse model where obesity is not associated with reduced serum adiponectin levels. A delay in macrophage infiltration of adipose tissue of Mc3rKO during exposure to HFD may also be a factor contributing to the mild insulin resistance in this model.


Subject(s)
Adiponectin/metabolism , Inflammation/metabolism , Insulin Resistance/physiology , Obesity/metabolism , Receptor, Melanocortin, Type 3/metabolism , Receptor, Melanocortin, Type 4/metabolism , Adipocytes/drug effects , Adipocytes/metabolism , Adipocytes/pathology , Animals , Body Composition/drug effects , Body Composition/physiology , Dietary Fats/pharmacology , Disease Models, Animal , Humans , Inflammation/pathology , Inflammation/physiopathology , Leptin/blood , Male , Mice , Mice, Knockout , Obesity/pathology , Obesity/physiopathology , Receptor, Melanocortin, Type 3/genetics , Receptor, Melanocortin, Type 4/genetics
13.
Cell Metab ; 6(5): 398-405, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17983585

ABSTRACT

The burden of type 2 diabetes and its associated premature morbidity and mortality is rapidly growing, and the need for novel efficacious treatments is pressing. We report here that serotonin 2C receptor (5-HT(2C)R) agonists, typically investigated for their anorectic properties, significantly improve glucose tolerance and reduce plasma insulin in murine models of obesity and type 2 diabetes. Importantly, 5-HT(2C)R agonist-induced improvements in glucose homeostasis occurred at concentrations of agonist that had no effect on ingestive behavior, energy expenditure, locomotor activity, body weight, or fat mass. We determined that this primary effect on glucose homeostasis requires downstream activation of melanocortin-4 receptors (MC4Rs), but not MC3Rs. These findings suggest that pharmacological targeting of 5-HT(2C)Rs may enhance glucose tolerance independently of alterations in body weight and that this may prove an effective and mechanistically novel strategy in the treatment of type 2 diabetes.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Receptor, Melanocortin, Type 4/physiology , Serotonin 5-HT2 Receptor Agonists , Serotonin Receptor Agonists/pharmacology , Signal Transduction/drug effects , Absorptiometry, Photon , Animals , Blotting, Western , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/physiopathology , Gene Expression/drug effects , Glucose/metabolism , Glucose Intolerance , Glucose Tolerance Test , Homeostasis/drug effects , Immunohistochemistry , Insulin/blood , Male , Mice , Mice, Knockout , Mice, Obese , Neurons/drug effects , Neurons/metabolism , Piperazines/pharmacology , Polymerase Chain Reaction , Pro-Opiomelanocortin/genetics , Receptor, Melanocortin, Type 4/chemistry , Receptor, Melanocortin, Type 4/metabolism
14.
J Clin Invest ; 117(11): 3475-88, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17885689

ABSTRACT

Disruptions of the melanocortin signaling system have been linked to obesity. We investigated a possible role of the central nervous melanocortin system (CNS-Mcr) in the control of adiposity through effects on nutrient partitioning and cellular lipid metabolism independent of nutrient intake. We report that pharmacological inhibition of melanocortin receptors (Mcr) in rats and genetic disruption of Mc4r in mice directly and potently promoted lipid uptake, triglyceride synthesis, and fat accumulation in white adipose tissue (WAT), while increased CNS-Mcr signaling triggered lipid mobilization. These effects were independent of food intake and preceded changes in adiposity. In addition, decreased CNS-Mcr signaling promoted increased insulin sensitivity and glucose uptake in WAT while decreasing glucose utilization in muscle and brown adipose tissue. Such CNS control of peripheral nutrient partitioning depended on sympathetic nervous system function and was enhanced by synergistic effects on liver triglyceride synthesis. Our findings offer an explanation for enhanced adiposity resulting from decreased melanocortin signaling, even in the absence of hyperphagia, and are consistent with feeding-independent changes in substrate utilization as reflected by respiratory quotient, which is increased with chronic Mcr blockade in rodents and in humans with loss-of-function mutations in MC4R. We also reveal molecular underpinnings for direct control of the CNS-Mcr over lipid metabolism. These results suggest ways to design more efficient pharmacological methods for controlling adiposity.


Subject(s)
Central Nervous System/metabolism , Lipid Metabolism , Melanocortins/metabolism , Signal Transduction/physiology , Adipocytes/cytology , Adipocytes/metabolism , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Behavior, Animal/physiology , Eating , Glucose/metabolism , Humans , Insulin/metabolism , Melanocyte-Stimulating Hormones/administration & dosage , Melanocyte-Stimulating Hormones/metabolism , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , Receptors, Melanocortin , alpha-MSH/administration & dosage , alpha-MSH/analogs & derivatives , alpha-MSH/metabolism
15.
Physiol Behav ; 89(4): 517-24, 2006 Nov 30.
Article in English | MEDLINE | ID: mdl-16996546

ABSTRACT

Ingestive behavior is controlled by a complex interplay between signals conveying availability of (1) potentially ingestible food in the environment, (2) digestible food in the alimentary canal, (3) circulating fuels and (4) stored fuels. Each of these four classes of signals interact with specific sensors and neural circuits whose integrated output determines when food intake is initiated and when it is stopped. Because the final common path responsible for oromotor control is contained within complex neural pattern generators within the brainstem and is intimately linked to sensory information from the alimentary canal, at least part of the integration between the four classes of signals is thought to take place at the level of the caudal brainstem. Here we show that CCK, representing a class 2, or direct signal, and MC4-melanocortin receptor activity, representing a second order class 3/4, or indirect signal, converge in the nucleus of the solitary tract where they modulate activity of the mitogen-activated, extracellular-signal regulated kinases 1 and 2 (ERK) pathway to determine the level of satiation. Blockade of this signaling pathway attenuates suppression of deprivation-induced food intake by intraperitoneal CCK and fourth ventricular MTII injection. Additional findings suggest that specific ERK-phosphorylation sites on ion channels and enzymes involved in catecholamine synthesis of NTS neurons may be involved in ERK-mediated satiation and meal termination. Longer-term downstream effects of ERK activation might involve CREB-mediated gene transcription known to produce plasticity changes in neurocircuitry that could determine inter-meal intervals and the size of future meals.


Subject(s)
Appetite Regulation/physiology , Eating/physiology , Solitary Nucleus/physiology , Animals , Brain Stem/cytology , Brain Stem/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Gastrointestinal Tract/innervation , Gastrointestinal Tract/physiology , Humans , Neural Pathways/physiology , Prosencephalon/cytology , Prosencephalon/physiology , Satiation/physiology , Signal Transduction/physiology , Solitary Nucleus/cytology
16.
Neuron ; 51(2): 239-49, 2006 Jul 20.
Article in English | MEDLINE | ID: mdl-16846858

ABSTRACT

The neural pathways through which central serotonergic systems regulate food intake and body weight remain to be fully elucidated. We report that serotonin, via action at serotonin1B receptors (5-HT1BRs), modulates the endogenous release of both agonists and antagonists of the melanocortin receptors, which are a core component of the central circuitry controlling body weight homeostasis. We also show that serotonin-induced hypophagia requires downstream activation of melanocortin 4, but not melanocortin 3, receptors. These results identify a primary mechanism underlying the serotonergic regulation of energy balance and provide an example of a centrally derived signal that reciprocally regulates melanocortin receptor agonists and antagonists in a similar manner to peripheral adiposity signals.


Subject(s)
Eating/physiology , Neurons/physiology , Receptor, Melanocortin, Type 3/physiology , Receptor, Serotonin, 5-HT1B/physiology , Receptors, Melanocortin/physiology , Serotonin/physiology , Animals , Eating/drug effects , Electric Stimulation , Male , Mice , Mice, Inbred A , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Mice, Transgenic , Nerve Net/drug effects , Nerve Net/physiology , Neurons/drug effects , Pyridines/pharmacology , Receptor, Melanocortin, Type 4/agonists , Receptor, Melanocortin, Type 4/antagonists & inhibitors , Receptor, Melanocortin, Type 4/physiology , Receptors, Melanocortin/agonists , Receptors, Melanocortin/antagonists & inhibitors , Serotonin/pharmacology , Serotonin 5-HT1 Receptor Agonists
17.
Endocrinology ; 147(5): 2183-96, 2006 May.
Article in English | MEDLINE | ID: mdl-16469808

ABSTRACT

Loss of brain melanocortin receptors (Mc3rKO and Mc4rKO) causes increased adiposity and exacerbates diet-induced obesity (DIO). Little is known about how Mc3r or Mc4r genotype, diet, and obesity affect insulin sensitivity. Insulin resistance, assessed by insulin and glucose tolerance tests, Ser(307) phosphorylation of insulin receptor substrate 1, and activation of protein kinase B, was examined in control and DIO wild-type (WT), Mc3rKO and Mc4rKO C57BL/6J mice. Mc4rKO mice were hyperphagic and had increased metabolic efficiency (weight gain per kilojoule consumed) relative to WT; both parameters increased further on high-fat diet. Obesity of Mc3rKO was more dependent on fat intake, involving increased metabolic efficiency. Fat mass of DIO Mc3rKO and Mc4rKO was similar, although Mc4rKO gained weight more rapidly. Mc4rKO develop hepatic insulin resistance and severe hepatic steatosis with obesity, independent of diet. DIO caused further deterioration of insulin action in Mc4rKO of either sex and, in male Mc3rKO, compared with controls, associated with increased fasting insulin, severe glucose intolerance, and reduced insulin signaling in muscle and adipose tissue. DIO female Mc3rKO exhibited very modest perturbations in glucose metabolism and insulin sensitivity. Consistent with previous data suggesting impaired fat oxidation, both Mc3rKO and Mc4rKO had reduced muscle oxidative metabolism, a risk factor for weight gain and insulin resistance. Energy expenditure was, however, increased in Mc4rKO compared with Mc3rKO and controls, perhaps due to hyperphagia and metabolic costs associated with rapid growth. In summary, DIO affects insulin sensitivity more severely in Mc4rKO compared with Mc3rKO, perhaps due to a more positive energy balance.


Subject(s)
Diet , Receptor, Melanocortin, Type 3/physiology , Receptor, Melanocortin, Type 4/physiology , Adipose Tissue/metabolism , Animal Feed , Animals , Calorimetry , Cell Proliferation , Energy Metabolism , Fatty Acids/metabolism , Female , Genotype , Glucose/metabolism , Insulin/metabolism , Insulin Resistance , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Muscle, Skeletal/metabolism , Obesity , Oxygen/metabolism , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , RNA/metabolism , Receptor, Insulin/metabolism , Receptor, Melanocortin, Type 3/genetics , Receptor, Melanocortin, Type 4/genetics , Regression Analysis , Risk Factors , Sensitivity and Specificity , Serine/chemistry , Sex Factors , Signal Transduction , Time Factors
18.
Endocrinology ; 146(9): 3739-47, 2005 Sep.
Article in English | MEDLINE | ID: mdl-15961554

ABSTRACT

Signals from the gut and hypothalamus converge in the caudal brainstem to control ingestive behavior. We have previously shown that phosphorylation of ERK1/2 in the solitary nucleus (NTS) is necessary for food intake suppression by exogenous cholecystokinin (CCK). Here we test whether this intracellular signaling cascade is also involved in the integration of melanocortin-receptor (MCR) mediated inputs to the caudal brainstem. Using fourth ventricular-cannulated rats and Western blotting of NTS tissue, we show that the MC4R agonist melanotan II (MTII) rapidly and dose-dependently increases phosphorylation of both ERK1/2 and cAMP response element-binding protein (CREB). Sequential administration of fourth ventricular MTII and peripheral CCK at doses that alone produced submaximal stimulation of pERK1/2 produced an additive increase. Prior fourth ventricular administration of the MC4R antagonist SHU9119 completely abolished the CCK-induced increases in pERK and pCREB and, in freely feeding rats, SHU9119 significantly increased meal size and satiety ratio. Prior administration of the MAPK kinase inhibitor U0126 abolished the capacity of MTII to suppress 2-h food intake and significantly decreased MTII-induced ERK phosphorylation in the NTS. Furthermore, pretreatment with the cAMP inhibitor, cAMP receptor protein-Rp isomer, significantly attenuated stimulation of pERK induced by either CCK or MTII. The results demonstrate that activation of the ERK pathway is necessary for peripheral CCK and central MTII to suppress food intake. The cAMP-->ERK-->CREB cascade may thus constitute a molecular integrator for converging satiety signals from the gut and adiposity signals from the hypothalamus in the control of meal size and food intake.


Subject(s)
Cholecystokinin/pharmacology , Eating/drug effects , MAP Kinase Signaling System/drug effects , Receptors, Melanocortin/agonists , Solitary Nucleus/drug effects , alpha-MSH/analogs & derivatives , Animals , Cyclic AMP Response Element-Binding Protein/metabolism , Eating/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Fourth Ventricle , Injections, Intraventricular , MAP Kinase Signaling System/physiology , Male , Melanocyte-Stimulating Hormones/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Corticotropin/antagonists & inhibitors , Solitary Nucleus/physiology , alpha-MSH/pharmacology
19.
Histochem Cell Biol ; 123(2): 147-56, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15742197

ABSTRACT

Orexin-expressing neurons in the lateral hypothalamus with their wide projections throughout the brain are important for the regulation of sleep and wakefulness, ingestive behavior, and the coordination of these behaviors in the environmental context. To further identify downstream effector targets of the orexin system, we examined in detail orexin-A innervation of the caudal raphe nuclei in the medulla, known to harbor sympathetic preganglionic motor neurons involved in thermal, cardiovascular, and gastrointestinal regulation. All three components of the caudal raphe nuclei, raphe pallidus, raphe obscurus, and parapyramidal nucleus, are innervated by orexin-A-immunoreactive fibers. Using confocal microscopy, we demonstrate close anatomical appositions between varicose orexin-A immunoreactive axon profiles and sympathetic premotor neurons identified with either a transneuronal retrograde pseudorabies virus tracer injected into the interscapular brown fat pads, or with in situ hybridization of pro-TRH mRNA. Furthermore, orexin-A injected into the fourth ventricle induced c-Fos expression in the raphe pallidus and parapyramidal nucleus. These findings suggest that orexin neurons in the hypothalamus can modulate brown fat thermogenesis, cardiovascular, and gastrointestinal functions by acting directly on neurons in the caudal raphe nuclei, and support the idea that orexin's simultaneous stimulation of food intake and sympathetic activity might have evolved as a mechanism to stay alert while foraging.


Subject(s)
Adipose Tissue, Brown/metabolism , Cardiovascular Physiological Phenomena , Gastrointestinal Tract/physiology , Intracellular Signaling Peptides and Proteins/physiology , Neuropeptides/physiology , Raphe Nuclei/metabolism , Animals , Gene Expression , Immunohistochemistry , In Situ Hybridization/methods , Intracellular Signaling Peptides and Proteins/analysis , Intracellular Signaling Peptides and Proteins/pharmacology , Microscopy, Confocal , Neurons/chemistry , Neurons/metabolism , Neurons/physiology , Neuropeptides/analysis , Neuropeptides/pharmacology , Orexins , Proto-Oncogene Proteins c-fos/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Raphe Nuclei/drug effects , Rats , Rats, Sprague-Dawley , Sympathetic Nervous System/cytology , Sympathetic Nervous System/physiology , Thermogenesis , Thyrotropin-Releasing Hormone/genetics
20.
J Neurosci ; 24(45): 10240-7, 2004 Nov 10.
Article in English | MEDLINE | ID: mdl-15537896

ABSTRACT

Increased food intake is a major factor in the development of obesity, and the control of meal size is a valid approach to reduce food intake in humans. Meal termination, or satiety, is thought to be organized within the caudal brainstem where direct signals from the food handling alimentary canal and long-term signals from the forebrain converge in the solitary nucleus. Cholecystokinin (CCK) released from the gut after ingestion of food has been strongly implicated in nucleus tractus solitarius (NTS)-mediated satiation, but the exact cellular and intracellular signaling events are not understood. Using Western blotting and immunohistochemistry with phosphospecific antibodies, we demonstrate here that peripheral administration of CCK in rats leads to rapid activation of the extracellular signal-regulated kinase (ERK) signaling cascade in NTS neurons and that blockade of ERK signaling with microinfusion of a selective mitogen-activated ERK kinase inhibitor into the fourth ventricle attenuates the capacity of CCK to suppress food intake. In addition, we show that CCK-induced activation of ERK results in phosphorylation of the voltage-dependent potassium channel Kv4.2 and the nuclear transcription factor CREB (cAMP response element-binding protein). The results demonstrate that ERK signaling is necessary for exogenous CCK to suppress food intake in deprived rats and suggest that this pathway may also be involved in natural satiation and the period of satiety between meals through coupling of ERK activation to both cytosolic and nuclear effector mechanisms that have the potential to confer acute and long-term changes in neuronal functioning.


Subject(s)
Eating/drug effects , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Satiation/physiology , Signal Transduction/physiology , Sincalide/analogs & derivatives , Sincalide/pharmacology , Solitary Nucleus/physiology , Animals , Brain Stem/physiology , Butadienes/pharmacology , Cyclic AMP Response Element-Binding Protein/metabolism , Enzyme Activation/drug effects , Male , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Nitriles/pharmacology , Phosphorylation/drug effects , Potassium Channels, Voltage-Gated/metabolism , Protein Processing, Post-Translational/drug effects , Rats , Shal Potassium Channels , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...