Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Front Pharmacol ; 13: 852080, 2022.
Article in English | MEDLINE | ID: mdl-35308230

ABSTRACT

Praliciguat is a soluble guanylate cyclase stimulator that elicits hemodynamic, anti-inflammatory, and antifibrotic effects in preclinical models of metabolic dysfunction. We assessed the metabolic effects of praliciguat in a mouse diet-induced obesity (DIO) model housed at thermoneutrality. At 6 weeks old, male C57BL/6N mice were either maintained on low-fat diet (LFD, lean mice) or placed on 60% high-fat diet (HFD, DIO mice). At 14 weeks old, the DIO mice were either maintained on HFD or switched to HFD with praliciguat (6-mg/kg). Day 28 samples were collected for biomarker analysis. In a second study under the same paradigm, indirect calorimetry was performed on days 8, 9, 20, 21, 32, and 33 and an oral lipid tolerance test (LTT) on day 38. Mice treated 28 days with praliciguat had lower levels of fasting plasma insulin, C-peptide, triglycerides, and HOMA-IR (homeostatic model assessment for insulin resistance) than DIO controls. In addition, energy expenditure was higher in praliciguat-treated than in DIO control mice on days 9, 20, 32, and 33; and day-38 triglycerides were lower. HFD-induced increases in gene expression of liver TNF-ɑ, lipoprotein lipase (Lpl), and patatin-like phospholipase domain-containing protein 3 (Pnpla3) in control DIO mice were attenuated in praliciguat-treated DIO mice. The positive metabolic effects observed in praliciguat-treated mice were associated with the restoration of liver PI3K (pAKT-Thr308) signaling, but not MAPK (pERK). In conclusion, praliciguat-treated DIO mice had increased energy utilization, improved insulin sensitivity, and lower plasma triglycerides. These results illustrate metabolic effects associated with praliciguat treatment in DIO mice.

2.
J Neuroinflammation ; 18(1): 213, 2021 Sep 18.
Article in English | MEDLINE | ID: mdl-34537066

ABSTRACT

BACKGROUND: Inflammation in the central nervous system (CNS) is observed in many neurological disorders. Nitric oxide-soluble guanylate cyclase-cyclic guanosine monophosphate (NO-sGC-cGMP) signaling plays an essential role in modulating neuroinflammation. CYR119 is a CNS-penetrant sGC stimulator that amplifies endogenous NO-sGC-cGMP signaling. We evaluated target engagement and the effects of CYR119 on markers of neuroinflammation in vitro in mouse microglial cells and in vivo in quinolinic acid (QA)-induced and high-fat diet-induced rodent neuroinflammation models. METHODS: Target engagement was verified in human embryonic kidney (HEK) cells, rat primary neurons, mouse SIM-A9 cells, and in rats by measuring changes in cGMP and downstream targets of sGC signaling [phosphorylated vasodilator-stimulated phosphoprotein (pVASP), phosphorylated cAMP-response element binding (pCREB)]. In SIM-A9 cells stimulated with lipopolysaccharides (LPS), markers of inflammation were measured when cells were treated with or without CYR119. In rats, microinjections of QA and vehicle were administered into the right and left hemispheres of striatum, respectively, and then rats were dosed daily with either CYR119 (10 mg/kg) or vehicle for 7 days. The activation of microglia [ionized calcium binding adaptor molecule 1 (Iba1)] and astrocytes [glial fibrillary acidic protein (GFAP)] was measured by immunohistochemistry. Diet-induced obese (DIO) mice were treated daily with CYR119 (10 mg/kg) for 6 weeks, after which inflammatory genetic markers were analyzed in the prefrontal cortex. RESULTS: In vitro, CYR119 synergized with exogenous NO to increase the production of cGMP in HEK cells and in primary rat neuronal cell cultures. In primary neurons, CYR119 stimulated sGC, resulting in accumulation of cGMP and phosphorylation of CREB, likely through the activation of protein kinase G (PKG). CYR119 attenuated LPS-induced elevation of interleukin 6 (IL-6) and tumor necrosis factor (TNF) in mouse microglial cells. Following oral dosing in rats, CYR119 crossed the blood-brain barrier (BBB) and stimulated an increase in cGMP levels in the cerebral spinal fluid (CSF). In addition, levels of proinflammatory markers associated with QA administration or high-fat diet feeding were lower in rodents treated with CYR119 than in those treated with vehicle. CONCLUSIONS: These data suggest that sGC stimulation could provide neuroprotective effects by attenuating inflammatory responses in nonclinical models of neuroinflammation.


Subject(s)
Anti-Inflammatory Agents/metabolism , Central Nervous System/metabolism , Cyclic GMP/metabolism , Inflammation Mediators/metabolism , Neurons/metabolism , Soluble Guanylyl Cyclase/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Biomarkers/metabolism , Cells, Cultured , Central Nervous System/drug effects , Dose-Response Relationship, Drug , HEK293 Cells , Humans , Inflammation Mediators/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Rats , Rats, Sprague-Dawley
3.
Front Pharmacol ; 12: 656561, 2021.
Article in English | MEDLINE | ID: mdl-34108877

ABSTRACT

Effective treatments for neurodegenerative diseases remain elusive and are critically needed since the burden of these diseases increases across an aging global population. Nitric oxide (NO) is a gasotransmitter that binds to soluble guanylate cyclase (sGC) to produce cyclic guanosine monophosphate (cGMP). Impairment of this pathway has been demonstrated in neurodegenerative diseases. Normalizing deficient NO-cGMP signaling could address multiple pathophysiological features of neurodegenerative diseases. sGC stimulators are small molecules that synergize with NO, activate sGC, and increase cGMP production. Many systemic sGC stimulators have been characterized and advanced into clinical development for a variety of non-central nervous system (CNS) pathologies. Here, we disclose the discovery of CY6463, the first brain-penetrant sGC stimulator in clinical development for the treatment of neurodegenerative diseases, and demonstrate its ability to improve neuronal activity, mediate neuroprotection, and increase cognitive performance in preclinical models. In several cellular assays, CY6463 was demonstrated to be a potent stimulator of sGC. In agreement with the known effects of sGC stimulation in the vasculature, CY6463 elicits decreases in blood pressure in both rats and mice. Relative to a non-CNS penetrant sGC stimulator, rodents treated with CY6463 had higher cGMP levels in cerebrospinal fluid (CSF), functional-magnetic-resonance-imaging-blood-oxygen-level-dependent (fMRI-BOLD) signals, and cortical electroencephalographic (EEG) gamma-band oscillatory power. Additionally, CY6463 improved cognitive performance in a model of cognitive disruption induced by the administration of a noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist. In models of neurodegeneration, CY6463 treatment increased long-term potentiation (LTP) in hippocampal slices from a Huntington's disease mouse model and decreased the loss of dendritic spines in aged and Alzheimer's disease mouse models. In a model of diet-induced obesity, CY6463 reduced markers of inflammation in the plasma. Furthermore, CY6463 elicited an additive increase in cortical gamma-band oscillatory power when co-administered with donepezil: the standard of care in Alzheimer's disease. Together, these data support the clinical development of CY6463 as a novel treatment for neurodegenerative disorders.

4.
Br J Pharmacol ; 178(17): 3463-3475, 2021 09.
Article in English | MEDLINE | ID: mdl-33864386

ABSTRACT

BACKGROUND AND PURPOSE: Reduced bioavailability of NO, a hallmark of sickle cell disease (SCD), contributes to intravascular inflammation, vasoconstriction, vaso-occlusion and organ damage observed in SCD patients. Soluble guanylyl cyclase (sGC) catalyses synthesis of cGMP in response to NO. cGMP-amplifying agents, including NO donors and phosphodiesterase 9 inhibitors, alleviate TNFα-induced inflammation in wild-type C57BL/6 mice and in 'humanised' mouse models of SCD. EXPERIMENTAL APPROACH: Effects of the sGC stimulator olinciguat on intravascular inflammation and renal injury were studied in acute (C57BL6 and Berkeley mice) and chronic (Townes mice) mouse models of TNFα-induced and systemic inflammation associated with SCD. KEY RESULTS: Acute treatment with olinciguat attenuated increases in plasma biomarkers of endothelial cell activation and leukocyte-endothelial cell interactions in TNFα-challenged mice. Co-treatment with hydroxyurea, an FDA-approved SCD therapeutic agent, further augmented the anti-inflammatory effect of olinciguat. In the Berkeley mouse model of TNFα-induced vaso-occlusive crisis, a single dose of olinciguat attenuated leukocyte-endothelial cell interactions, improved blood flow and prolonged survival time compared to vehicle-treated mice. In Townes SCD mice, plasma biomarkers of inflammation and endothelial cell activation were lower in olinciguat- than in vehicle-treated mice. In addition, kidney mass, water consumption, 24-h urine excretion, plasma levels of cystatin C and urinary excretion of N-acetyl-ß-d-glucosaminidase and neutrophil gelatinase-associated lipocalin were lower in Townes mice treated with olinciguat than in vehicle-treated mice. CONCLUSION AND IMPLICATIONS: Our results suggest that the sGC stimulator olinciguat attenuates inflammation, vaso-occlusion and kidney injury in mouse models of SCD and systemic inflammation.


Subject(s)
Anemia, Sickle Cell , Vascular Diseases , Anemia, Sickle Cell/complications , Anemia, Sickle Cell/drug therapy , Animals , Humans , Inflammation , Mice , Mice, Inbred C57BL , Soluble Guanylyl Cyclase
5.
Am J Physiol Renal Physiol ; 319(4): F697-F711, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32865013

ABSTRACT

Praliciguat, a clinical-stage soluble guanylate cyclase (sGC) stimulator, increases cGMP via the nitric oxide-sGC pathway. Praliciguat has been shown to be renoprotective in rodent models of hypertensive nephropathy and renal fibrosis. In the present study, praliciguat alone and in combination with enalapril attenuated proteinuria in the obese ZSF1 rat model of diabetic nephropathy. Praliciguat monotherapy did not affect hemodynamics. In contrast, enalapril monotherapy lowered blood pressure but did not attenuate proteinuria. Renal expression of genes in pathways involved in inflammation, fibrosis, oxidative stress, and kidney injury was lower in praliciguat-treated obese ZSF1 rats than in obese control rats; fasting glucose and cholesterol were also lower with praliciguat treatment. To gain insight into how tubular mechanisms might contribute to its pharmacological effects on the kidneys, we studied the effects of praliciguat on pathological processes and signaling pathways in cultured human primary renal proximal tubular epithelial cells (RPTCs). Praliciguat inhibited the expression of proinflammatory cytokines and secretion of monocyte chemoattractant protein-1 in tumor necrosis factor-α-challenged RPTCs. Praliciguat treatment also attenuated transforming growth factor-ß-mediated apoptosis, changes to a mesenchyme-like cellular phenotype, and phosphorylation of SMAD3 in RPTCs. In conclusion, praliciguat improved proteinuria in the ZSF1 rat model of diabetic nephropathy, and its actions in human RPTCs suggest that tubular effects may contribute to its renal benefits, building upon strong evidence for the role of cGMP signaling in renal health.


Subject(s)
Apoptosis/drug effects , Diabetic Nephropathies/drug therapy , Guanylyl Cyclase C Agonists/pharmacology , Kidney Tubules, Proximal/drug effects , Nephritis/drug therapy , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Animals , Cell Line , Cytokines/metabolism , Diabetic Nephropathies/metabolism , Diabetic Nephropathies/pathology , Disease Models, Animal , Disease Progression , Enalapril/pharmacology , Humans , Inflammation Mediators/metabolism , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/pathology , Male , Nephritis/metabolism , Nephritis/pathology , Phosphorylation , Rats, Zucker , Signal Transduction , Smad3 Protein/metabolism
6.
Front Pharmacol ; 11: 419, 2020.
Article in English | MEDLINE | ID: mdl-32322204

ABSTRACT

Nitric oxide (NO)-soluble guanylate cyclase (sGC)-cyclic 3',5' GMP (cGMP) signaling plays a central role in regulation of diverse processes including smooth muscle relaxation, inflammation, and fibrosis. sGC is activated by the short-lived physiologic mediator NO. sGC stimulators are small-molecule compounds that directly bind to sGC to enhance NO-mediated cGMP signaling. Olinciguat, (R)-3,3,3-trifluoro-2-(((5-fluoro-2-(1-(2-fluorobenzyl)-5-(isoxazol-3-yl)-1H-pyrazol-3-yl)pyrimidin-4-yl)amino)methyl)-2-hydroxypropanamide, is a new sGC stimulator currently in Phase 2 clinical development. To understand the potential clinical utility of olinciguat, we studied its pharmacokinetics, tissue distribution, and pharmacologic effects in preclinical models. Olinciguat relaxed human vascular smooth muscle and was a potent inhibitor of vascular smooth muscle proliferation in vitro. These antiproliferative effects were potentiated by the phosphodiesterase 5 inhibitor tadalafil, which did not inhibit vascular smooth muscle proliferation on its own. Olinciguat was orally bioavailable and predominantly cleared by the liver in rats. In a rat whole body autoradiography study, olinciguat-derived radioactivity in most tissues was comparable to plasma levels, indicating a balanced distribution between vascular and extravascular compartments. Olinciguat was explored in rodent models to study its effects on the vasculature, the heart, the kidneys, metabolism, and inflammation. Olinciguat reduced blood pressure in normotensive and hypertensive rats. Olinciguat was cardioprotective in the Dahl rat salt-sensitive hypertensive heart failure model. In the rat ZSF1 model of diabetic nephropathy and metabolic syndrome, olinciguat was renoprotective and associated with lower circulating glucose, cholesterol, and triglycerides. In a mouse TNFα-induced inflammation model, olinciguat treatment was associated with lower levels of endothelial and leukocyte-derived soluble adhesion molecules. The pharmacological features of olinciguat suggest that it may have broad therapeutic potential and that it may be suited for diseases that have both vascular and extravascular pathologies.

8.
Cell Rep ; 22(6): 1522-1530, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29425507

ABSTRACT

The age-related effects of GDF11 have been a subject of controversy. Here, we find that elevated GDF11 causes signs of cachexia in mice: reduced food intake, body weight, and muscle mass. GDF11 also elicited a significant elevation in plasma Activin A, previously shown to contribute to the loss of skeletal muscle. The effects of GDF11 on skeletal muscle could be reversed by administration of antibodies to the Activin type II receptors. In addition to the effects on muscle, GDF11 increased plasma GDF15, an anorectic agent. The anorexia, but not the muscle loss, could be reversed with a GDF15-neutralizing antibody. GDF15 upregulation is due to GDF11-induced recruitment of SMAD2/3 to the GDF15 promoter. Inhibition of GDF15 can restore appetite but cannot restore the GDF11-induced loss of muscle mass, which requires blockade of ActRII signaling. These findings are relevant for treatment of cachexia.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cachexia , Growth Differentiation Factor 15/biosynthesis , Growth Differentiation Factors/metabolism , Activins/metabolism , Animals , Bone Morphogenetic Proteins/pharmacology , Growth Differentiation Factors/pharmacology , Male , Mice , Mice, Inbred C57BL , Up-Regulation
9.
Neuron ; 71(3): 488-97, 2011 Aug 11.
Article in English | MEDLINE | ID: mdl-21835345

ABSTRACT

VIDEO ABSTRACT: Serotonin 2C receptors (5-HT(2C)Rs) expressed by pro-opiomelanocortin (POMC) neurons of hypothalamic arcuate nucleus regulate food intake, energy homeostasis and glucose metabolism. However, the cellular mechanisms underlying the effects of 5-HT to regulate POMC neuronal activity via 5-HT(2C)Rs have not yet been identified. In the present study, we found the putative transient receptor potential C (TRPC) channels mediate the activation of a subpopulation of POMC neurons by mCPP (a 5-HT(2C)R agonist). Interestingly, mCPP-activated POMC neurons were found to be a distinct population from those activated by leptin. Together, our data suggest that 5-HT(2C)R and leptin receptors are expressed by distinct subpopulations of arcuate POMC neurons and that both 5-HT and leptin exert their actions in POMC neurons via TRPC channels.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiology , Neurons/physiology , Pro-Opiomelanocortin/physiology , Receptor, Serotonin, 5-HT2C/physiology , TRPC Cation Channels/physiology , Animals , Arcuate Nucleus of Hypothalamus/drug effects , G Protein-Coupled Inwardly-Rectifying Potassium Channels/physiology , Leptin/pharmacology , Leptin/physiology , Male , Membrane Potentials/physiology , Mice , Mice, Transgenic , Patch-Clamp Techniques/methods , Piperazines/pharmacology , Pro-Opiomelanocortin/genetics , Serotonin/pharmacology , Serotonin/physiology , Serotonin 5-HT2 Receptor Agonists/pharmacology , TRPC Cation Channels/antagonists & inhibitors
10.
Am J Physiol Regul Integr Comp Physiol ; 300(2): R428-36, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21123763

ABSTRACT

There is an expectation that repeated daily exposures to normobaric hypoxia (NH) will induce ventilatory acclimatization and lessen acute mountain sickness (AMS) and the exercise performance decrement during subsequent hypobaric hypoxia (HH) exposure. However, this notion has not been tested objectively. Healthy, unacclimatized sea-level (SL) residents slept for 7.5 h each night for 7 consecutive nights in hypoxia rooms under NH [n = 14, 24 ± 5 (SD) yr] or "sham" (n = 9, 25 ± 6 yr) conditions. The ambient percent O(2) for the NH group was progressively reduced by 0.3% [150 m equivalent (equiv)] each night from 16.2% (2,200 m equiv) on night 1 to 14.4% (3,100 m equiv) on night 7, while that for the ventilatory- and exercise-matched sham group remained at 20.9%. Beginning at 25 h after sham or NH treatment, all subjects ascended and lived for 5 days at HH (4,300 m). End-tidal Pco(2), O(2) saturation (Sa(O(2))), AMS, and heart rate were measured repeatedly during daytime rest, sleep, or exercise (11.3-km treadmill time trial). From pre- to posttreatment at SL, resting end-tidal Pco(2) decreased (P < 0.01) for the NH (from 39 ± 3 to 35 ± 3 mmHg), but not for the sham (from 39 ± 2 to 38 ± 3 mmHg), group. Throughout HH, only sleep Sa(O(2)) was higher (80 ± 1 vs. 76 ± 1%, P < 0.05) and only AMS upon awakening was lower (0.34 ± 0.12 vs. 0.83 ± 0.14, P < 0.02) in the NH than the sham group; no other between-group rest, sleep, or exercise differences were observed at HH. These results indicate that the ventilatory acclimatization induced by NH sleep was primarily expressed during HH sleep. Under HH conditions, the higher sleep Sa(O(2)) may have contributed to a lessening of AMS upon awakening but had no impact on AMS or exercise performance for the remainder of each day.


Subject(s)
Acclimatization/physiology , Altitude Sickness/prevention & control , Altitude , Atmospheric Pressure , Exercise/physiology , Hypoxia/physiopathology , Sleep/physiology , Adult , Altitude Sickness/diagnosis , Altitude Sickness/epidemiology , Carbon Dioxide/blood , Erythropoietin/blood , Female , Heart Rate/physiology , Hematocrit , Hemoglobins/metabolism , Humans , Hydrocortisone/blood , Male , Norepinephrine/blood , Oxygen/blood , Oxygen Consumption/physiology , Partial Pressure , Physical Exertion/physiology , Pulmonary Gas Exchange/physiology , Pulmonary Ventilation/physiology , Young Adult
11.
J Neurosci ; 30(44): 14630-4, 2010 Nov 03.
Article in English | MEDLINE | ID: mdl-21048120

ABSTRACT

D-Fenfluramine (D-Fen) increases serotonin (5-HT) content in the synaptic cleft and exerts anorexigenic effects in animals and humans. However, the neural circuits that mediate these effects are not fully identified. To address this issue, we assessed the efficacy of D-Fen-induced hypophagia in mouse models with manipulations of several genes in selective populations of neurons. Expectedly, we found that global deletion of 5-HT 2C receptors (5-HT(2C)Rs) significantly attenuated D-Fen-induced anorexia. These anorexigenic effects were restored in mice with 5-HT(2C)Rs expressed only in pro-opiomelanocortin (POMC) neurons. Further, we found that deletion of melanocortin 4 receptors (MC4Rs), a downstream target of POMC neurons, abolished anorexigenic effects of D-Fen. Reexpression of MC4Rs only in SIM1 neurons in the hypothalamic paraventricular nucleus and neurons in the amygdala was sufficient to restore the hypophagic property of D-Fen. Thus, our results identify a neurochemically defined neural circuit through which D-Fen influences appetite and thereby indicate that this 5-HT(2C)R/POMC-MC4R/SIM1 circuit may yield a more refined target to exploit for weight loss.


Subject(s)
Anorexia/metabolism , Anorexia/physiopathology , Fenfluramine/pharmacology , Melanocortins/physiology , Serotonin/physiology , Animals , Anorexia/chemically induced , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neural Pathways/cytology , Neural Pathways/drug effects , Neural Pathways/metabolism , Pro-Opiomelanocortin/physiology , Receptor, Melanocortin, Type 4/deficiency , Receptor, Melanocortin, Type 4/genetics , Receptor, Serotonin, 5-HT2C/deficiency , Receptor, Serotonin, 5-HT2C/genetics , Serotonin/metabolism , Selective Serotonin Reuptake Inhibitors/pharmacology , Weight Loss/genetics , Weight Loss/physiology
12.
Nat Neurosci ; 13(12): 1457-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21037584

ABSTRACT

Mice lacking 5-HT 2C receptors (5-HT(2C)Rs) displayed hepatic insulin resistance, a phenotype normalized by re-expression of 5-HT(2C)Rs only in pro-opiomelanocortin (POMC) neurons. 5-HT(2C)R deficiency also abolished the anti-diabetic effects of meta-chlorophenylpiperazine (a 5-HT(2C)R agonist); these effects were restored when 5-HT(2C)Rs were re-expressed in POMC neurons. Our findings indicate that 5-HT(2C)Rs expressed by POMC neurons are physiologically relevant regulators of insulin sensitivity and glucose homeostasis in the liver.


Subject(s)
Gene Expression Regulation , Insulin Resistance/physiology , Liver/metabolism , Neurons/metabolism , Pro-Opiomelanocortin/biosynthesis , Receptor, Serotonin, 5-HT2C/biosynthesis , Animals , Brain Stem/metabolism , Glucose/metabolism , Homeostasis/physiology , Hypothalamus/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neurons/physiology , Pro-Opiomelanocortin/physiology , Receptor, Serotonin, 5-HT2C/genetics , Receptor, Serotonin, 5-HT2C/physiology
13.
Med Sci Sports Exerc ; 41(6): 1317-25, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19461532

ABSTRACT

PURPOSE: This study examined the effect of 1 wk of normobaric intermittent hypoxic exposure (IHE) combined with exercise training on endurance performance at a 4300-m altitude (HA). METHODS: Seventeen male lowlanders were divided into an IHE (n = 11) or SHAM (n = 6) group. Each completed cycle endurance testing consisting of two 20-min steady state (SS) exercise bouts (at 40% and 60% V O2peak) followed by a 10-min break and then a 720-kJ cycle time trial at HA before IHE or SHAM treatment (Pre-T). IHE treatment consisted of a 2-h rest at a PO2 of 90 mm Hg followed by two 25-min bouts of exercise at approximately 80% of peak HR at a PO2 of 110 mm Hg for 1 wk in a hypoxia room. SHAM treatment was identical except that the PO2 was 148 mm Hg for both rest and exercise. After IHE or SHAM treatment (Post-T), all completed a second cycle endurance test at HA. HR, arterial oxygen saturation (SaO2), and RPE were obtained from the 10th to the 15th minute during the two SS exercise bouts and every 5 min during the time trial. RESULTS: Seven volunteers in the IHE group could not finish the 720-kJ time trial either at Pre-T or at Post-T. Time trial analysis was limited, therefore, to the time to reach 360 kJ (halfway point) for all volunteers. From Pre-T to Post-T, there was no improvement in time trial performance (min +/- SE) in the IHE (62.0 +/- 4.8 to 63.7 +/- 5.2) or SHAM (60.9 +/- 6.3 to 54.2 +/- 6.8) group. There was no change from Pre-T to Post-T in HR, SaO2, and RPE during the two SS exercise bouts or time trial in either group. CONCLUSIONS: One week of IHE combined with exercise training does not improve endurance performance at a 4300-m altitude in male lowlanders.


Subject(s)
Altitude , Environmental Exposure/adverse effects , Hypoxia , Oxygen Consumption , Adult , Analysis of Variance , Bicycling , Exercise Test , Exercise Tolerance , Humans , Hyperbaric Oxygenation , Male , Single-Blind Method , Time Factors
14.
J Neurosci ; 28(50): 13640-8, 2008 Dec 10.
Article in English | MEDLINE | ID: mdl-19074037

ABSTRACT

The PI3K-Akt-FoxO1 pathway contributes to the actions of insulin and leptin in several cell types, including neurons in the CNS. However, identifying these actions in chemically identified neurons has proven difficult. To address this problem, we have developed a reporter mouse for monitoring PI3K-Akt signaling in specific populations of neurons, based on FoxO1 nucleocytoplasmic shuttling. The reporter, FoxO1 fused to green fluorescent protein (FoxO1GFP), is expressed under the control of a ubiquitous promoter that is silenced by a loxP flanked transcriptional blocker. Thus, the expression of the reporter in selected cells is dependent on the action of Cre recombinase. Using this model, we found that insulin treatment resulted in the nuclear exclusion of FoxO1GFP within POMC and AgRP neurons in a dose- and time-dependent manner. FoxO1GFP nuclear exclusion was also observed in POMC neurons following in vivo administration of insulin. In addition, leptin induced transient nuclear export of FoxO1GFP in POMC neurons in a dose dependent manner. Finally, insulin-induced nuclear export was impaired in POMC neurons by pretreatment with free fatty acids, a paradigm known to induce insulin resistance in peripheral insulin target tissues. Thus, our FoxO1GFP mouse provides a tool for monitoring the status of PI3K-Akt signaling in a cell-specific manner under physiological and pathophysiological conditions.


Subject(s)
Forkhead Transcription Factors/metabolism , Green Fluorescent Proteins/metabolism , Neurons/metabolism , Signal Transduction/physiology , Animals , Fatty Acids, Nonesterified/metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/genetics , Gene Knock-In Techniques , Green Fluorescent Proteins/genetics , Hypothalamus/metabolism , Image Processing, Computer-Assisted , Immunohistochemistry , In Situ Hybridization , Insulin/metabolism , Leptin/metabolism , Mice , Organ Culture Techniques , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism
15.
Neuron ; 60(4): 582-9, 2008 Nov 26.
Article in English | MEDLINE | ID: mdl-19038216

ABSTRACT

Drugs activating 5-hydroxytryptamine 2C receptors (5-HT2CRs) potently suppress appetite, but the underlying mechanisms for these effects are not fully understood. To tackle this issue, we generated mice with global 5-HT2CR deficiency (2C null) and mice with 5-HT2CRs re-expression only in pro-opiomelanocortin (POMC) neurons (2C/POMC mice). We show that 2C null mice predictably developed hyperphagia, hyperactivity, and obesity and showed attenuated responses to anorexigenic 5-HT drugs. Remarkably, all these deficiencies were normalized in 2C/POMC mice. These results demonstrate that 5-HT2CR expression solely in POMC neurons is sufficient to mediate effects of serotoninergic compounds on food intake. The findings also highlight the physiological relevance of the 5-HT2CR-melanocortin circuitry in the long-term regulation of energy balance.


Subject(s)
Energy Metabolism/genetics , Homeostasis/genetics , Hypothalamus/metabolism , Pro-Opiomelanocortin/metabolism , Receptor, Serotonin, 5-HT2C/genetics , Serotonin/metabolism , Animals , Appetite/drug effects , Appetite/genetics , Appetite Depressants/pharmacology , Appetite Regulation/genetics , Drug Resistance/genetics , Hyperphagia/genetics , Hyperphagia/metabolism , Hyperphagia/physiopathology , Hypothalamus/cytology , Mice , Mice, Knockout , Motor Activity/genetics , Neural Pathways/cytology , Neural Pathways/metabolism , Obesity/genetics , Obesity/metabolism , Obesity/physiopathology
16.
High Alt Med Biol ; 9(4): 281-7, 2008.
Article in English | MEDLINE | ID: mdl-19115911

ABSTRACT

The purpose of this study was to determine in sea-level residents if 6 to 7 consecutive days of normobaric intermittent hypoxic exposure (IHE) (hypoxia room: 2-h ambient PO2=90 mmHg sedentary and 1-h ambient PO2=110 mmHg exercising at 80+/-5% of maximum heart rate) improved sleep quality (awakenings per hour) and quantity at altitude (4300 m). We hypothesized that IHE would improve sleep arterial oxygen saturation (SaO2) levels and decrease desaturation events, thereby contributing to improvements in sleep quality and quantity during subsequent exposure to high altitude. Ten sea-level residents (mean+/-SE: 22+/-1 yr, 179+/-2 cm, 79+/-3 kg) were assigned to an IHE group and six to a SHAM group (20+/-0.5 yr, 180+/-3 cm, 77+/-4 kg). Sleep quantity, SaO2, and heart rate (HR) were monitored at sea level and during high altitude (i.e., 4300 m in a hypobaric chamber) before pretest (PRE-T) and 60 h after posttest (POST-T) for the last IHE or SHAM treatment. Over the 6 to 7 days of IHE, resting SaO2 increased from 75+/-1% to 81+/-3% in the IHE group, while the SHAM group remained at 98+/-1%. From PRE-T to POST-T at 4300-m exposure, both the IHE and SHAM groups had significantly higher sleep SaO2, fewer desaturation events per hour, and an increase in the percentage of time asleep while sleeping (sleep percent). The IHE group, but not the SHAM group, had significantly lower sleep HR and a trend to more awakenings during the POST-T 4300-m exposure. These results indicate that although IHE treatment induced significant ventilatory acclimatization, relative to the SHAM group, IHE did not further improve sleep SaO2 quality and quantity following rapid ascent to 4300 m. Rather, it is likely that the acquired ventilatory acclimatization was lost in the 60 h between the last IHE session and the POST-T altitude exposure.


Subject(s)
Acclimatization/physiology , Altitude , Atmosphere Exposure Chambers , Exercise/physiology , Hypoxia/physiopathology , Oxygen/blood , Sleep/physiology , Humans , Hyperbaric Oxygenation/methods , Male , Oxygen Consumption , Physical Exertion/physiology , Polysomnography , Reference Values , Single-Blind Method , Sleep Stages , Time Factors , Young Adult
17.
Nat Neurosci ; 11(9): 998-1000, 2008 Sep.
Article in English | MEDLINE | ID: mdl-19160495

ABSTRACT

The physiologic importance of GABAergic neurotransmission in hypothalamic neurocircuits is unknown. To examine the importance of GABA release from agouti-related protein (AgRP) neurons (which also release AgRP and neuropeptide Y), we generated mice with an AgRP neuron-specific deletion of vesicular GABA transporter. These mice are lean, resistant to obesity and have an attenuated hyperphagic response to ghrelin. Thus, GABA release from AgRP neurons is important in regulating energy balance.


Subject(s)
Agouti-Related Protein/metabolism , Energy Metabolism/physiology , Neurons/metabolism , Synapses/metabolism , gamma-Aminobutyric Acid/metabolism , 2-Amino-5-phosphonovalerate/pharmacology , 6-Cyano-7-nitroquinoxaline-2,3-dione/pharmacology , Absorptiometry, Photon/methods , Agouti-Related Protein/genetics , Analysis of Variance , Animals , Arcuate Nucleus of Hypothalamus/cytology , Body Weight/physiology , Eating/drug effects , Eating/physiology , Energy Metabolism/genetics , Excitatory Amino Acid Antagonists/pharmacology , Female , Ghrelin/pharmacology , Green Fluorescent Proteins/genetics , In Vitro Techniques , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/genetics , Male , Mice , Mice, Transgenic , Neurons/drug effects , Oxygen Consumption/genetics , Oxygen Consumption/physiology , Patch-Clamp Techniques , Sex Factors , Vesicular Inhibitory Amino Acid Transport Proteins/genetics , Vesicular Inhibitory Amino Acid Transport Proteins/metabolism
18.
J Comp Neurol ; 494(3): 528-48, 2006 Jan 20.
Article in English | MEDLINE | ID: mdl-16320257

ABSTRACT

Ghrelin is a hormone that stimulates growth hormone secretion and signals energy insufficiency via interaction with its receptor, the growth hormone secretagogue receptor (GHSR). The GHSR is located in both the central nervous system and the periphery. Its distribution in the CNS, as assessed by in situ hybridization histochemistry (ISHH), has been described previously in a few mammalian species, although these studies were limited by either the detail provided or the extent of the regions examined. In the present study, we systematically examined the distribution of GHSR mRNA in the adult rat and mouse brains and cervical spinal cords by using ISHH with novel cRNA probes specific for the mRNA encoding functional GHSR (the type 1a variant). We confirmed GHSR mRNA expression in several hypothalamic nuclei, many of which have long been recognized as playing roles in body weight and food intake. GHSR also was found in several other regions previously unknown to express GHSR mRNA, including many parasympathetic preganglionic neurons. Additionally, we found GHSR mRNA within all three components of the dorsal vagal complex, including the area postrema, the nucleus of the solitary tract, and the dorsal motor nucleus of the vagus. Finally, we examined the coexpression of GHSR with tyrosine hydroxylase and cholecystokinin and demonstrate a high degree of GHSR mRNA expression within dopaminergic, cholecystokinin-containing neurons of the substantia nigra and ventral tegmental area.


Subject(s)
Autonomic Fibers, Preganglionic/metabolism , Brain/metabolism , RNA, Messenger/metabolism , Receptors, G-Protein-Coupled/metabolism , Spinal Cord/metabolism , Animals , Brain/cytology , Cervical Vertebrae , Cholecystokinin/metabolism , Ganglia, Parasympathetic/metabolism , Gene Expression , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley , Receptors, G-Protein-Coupled/genetics , Receptors, Ghrelin , Specific Pathogen-Free Organisms , Spinal Cord/cytology , Tissue Distribution , Tyrosine 3-Monooxygenase/metabolism
19.
J Clin Invest ; 115(12): 3564-72, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16322794

ABSTRACT

Ghrelin is the endogenous ligand for the growth hormone secretagogue receptor (GHSR; ghrelin receptor). Since its discovery, accumulating evidence has suggested that ghrelin may play a role in signaling and reversing states of energy insufficiency. For example, ghrelin levels rise following food deprivation, and ghrelin administration stimulates feeding and increases body weight and adiposity. However, recent loss-of-function studies have raised questions regarding the physiological significance of ghrelin in regulating these processes. Here, we present results of a study using a novel GHSR-null mouse model, in which ghrelin administration fails to acutely stimulate food intake or activate arcuate nucleus neurons. We show that when fed a high-fat diet, both female and male GHSR-null mice eat less food, store less of their consumed calories, preferentially utilize fat as an energy substrate, and accumulate less body weight and adiposity than control mice. Similar effects on body weight and adiposity were also observed in female, but not male, GHSR-null mice fed standard chow. GHSR deletion also affected locomotor activity and levels of glycemia. These findings support the hypothesis that ghrelin-responsive pathways are an important component of coordinated body weight control. Moreover, our data suggest that ghrelin signaling is required for development of the full phenotype of diet-induced obesity.


Subject(s)
Diet , Obesity/genetics , Peptide Hormones/physiology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Adipose Tissue/metabolism , Alleles , Analysis of Variance , Animal Feed , Animals , Blood Glucose/metabolism , Blotting, Southern , Blotting, Western , Body Composition , Body Weight , Crosses, Genetic , DNA/metabolism , Female , Gene Deletion , Genetic Predisposition to Disease , Genotype , Ghrelin , Heterozygote , Homeostasis , Hyperglycemia/metabolism , Insulin-Like Growth Factor I/metabolism , Leptin/metabolism , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Models, Genetic , Neurons/metabolism , Obesity/metabolism , Peptide Hormones/chemistry , Phenotype , RNA, Messenger/metabolism , Receptors, Ghrelin , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombination, Genetic , Signal Transduction , Silver Staining , Time Factors
20.
Am J Physiol Regul Integr Comp Physiol ; 289(2): R418-R423, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15831766

ABSTRACT

Caloric deprivation inhibits reproduction, including copulatory behaviors, in female mammals. Decreases in metabolic fuel availability are detected in the hindbrain, and this information is relayed to the forebrain circuits controlling estrous behavior by neuropeptide Y (NPY) projections. In the forebrain, the nutritional inhibition of estrous behavior appears to be mediated by corticotropin-releasing factor (CRF) or urocortin-signaling systems. Intracerebroventricular (ICV) infusion of the CRF antagonist, astressin, prevents the suppression of lordosis by food deprivation and by NPY treatment in Syrian hamsters. These experiments sought to determine which CRF receptor type(s) is involved. ICV infusion of the CRF receptor subtype CRFR2-selective agonists urocortin 2 and 3 (UCN2, UCN3) inhibited sexual receptivity in hormone-primed, ovariectomized hamsters. Furthermore, the CRFR2-selective antagonist, astressin 2B, prevented the inhibition of estrous behavior by UCN2 and by NPY, consistent with a role for CRFR2. On the other hand, astressin 2B did not prevent the inhibition of behavior induced by 48-h food deprivation or ICV administration of CRF, a mixed CRFR1 and CRFR2 agonist, suggesting that activation of CRFR1 signaling is sufficient to inhibit sexual receptivity in hamsters. Although administration of CRFR1-selective antagonists (NBI-27914 and CP-154,526) failed to reverse the inhibition of receptivity by CRF treatment, we could not confirm their biological effectiveness in hamsters. The most parsimonious interpretation of these findings is that, although NPY inhibits estrous behavior via downstream CRFR2 signaling, food deprivation may exert its inhibition via both CRFR1 and CRFR2 and that redundant neuropeptide systems may be involved.


Subject(s)
Animal Nutritional Physiological Phenomena , Estrus , Receptors, Corticotropin-Releasing Hormone/physiology , Sexual Behavior, Animal/physiology , Animals , Corticotropin-Releasing Hormone/antagonists & inhibitors , Corticotropin-Releasing Hormone/pharmacology , Cricetinae , Female , Food Deprivation/physiology , Mesocricetus , Neuropeptide Y/antagonists & inhibitors , Peptide Fragments/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Receptors, Corticotropin-Releasing Hormone/agonists , Receptors, Corticotropin-Releasing Hormone/antagonists & inhibitors , Sexual Behavior, Animal/drug effects , Urocortins
SELECTION OF CITATIONS
SEARCH DETAIL