Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
J Med Chem ; 67(14): 11688-11700, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38960379

ABSTRACT

Amylin, a member of the calcitonin family, acts via amylin receptors in the hindbrain and hypothalamus to suppress appetite. Native ligands of these receptors are peptides with short half-lives. Conjugating fatty acids to these peptides can increase their half-lives. The long-acting human amylin analog, NN1213, was generated from structure-activity efforts optimizing solubility, stability, receptor affinity, and selectivity, as well as in vivo potency and clearance. In both rats and dogs, a single dose of NN1213 reduced appetite in a dose-dependent manner and with a long duration of action. Consistent with the effect on appetite, studies in obese rats demonstrated that daily NN1213 dosing resulted in a dose-dependent reduction in body weight over a 21-day period. Magnetic resonance imaging indicated that this was primarily driven by loss of fat mass. Based on these data, NN1213 could be considered an attractive option for weight management in the clinical setting.


Subject(s)
Islet Amyloid Polypeptide , Animals , Islet Amyloid Polypeptide/chemistry , Islet Amyloid Polypeptide/metabolism , Humans , Dogs , Rats , Structure-Activity Relationship , Male , Obesity/drug therapy , Body Weight/drug effects , Receptors, Islet Amyloid Polypeptide/metabolism
2.
Eur J Pharmacol ; 955: 175912, 2023 Sep 15.
Article in English | MEDLINE | ID: mdl-37454968

ABSTRACT

The glucose-dependent insulinotropic polypeptide (GIPR) and glucagon-like peptide (GLP-1R) receptor agonists are insulin secretagogues that have long been shown to improve glycemic control and dual agonists have demonstrated successful weight loss in the clinic. GIPR and GLP-1R populations are located in the dorsal vagal complex where receptor activity-modifying proteins (RAMPs) are also present. According to recent literature, RAMPs not only regulate the signaling of the calcitonin receptor, but also that of other class B G-protein coupled receptors, including members of the glucagon receptor family such as GLP-1R and GIPR. The aim of this study was to investigate whether the absence of RAMP1 and RAMP3 interferes with the action of GIPR and GLP-1R agonists on body weight maintenance and glucose control. To this end, WT and RAMP 1/3 KO mice were fed a 45% high fat diet for 22 weeks and were injected daily with GLP-1R agonist (2 nmol/kg/d; NN0113-2220), GIPR agonist (30 nmol/kg/d; NN0441-0329) or both for 3 weeks. While the mono-agonists exerted little to no body weight lowering and anorectic effects in WT or RAMP1/3 KO mice, but at the given doses, when both compounds were administered together, they synergistically reduced body weight, with a greater effect observed in KO mice. Finally, GLP-1R and GIP/GLP-1R agonist treatment led to improved glucose tolerance, but the absence of RAMPs resulted in an improvement of the HOMA-IR score. These data suggest that RAMPs may play a crucial role in modulating the pharmacological actions of GLP-1 and GIP receptors.


Subject(s)
Glucagon-Like Peptide-1 Receptor , Receptors, Gastrointestinal Hormone , Animals , Mice , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/agonists , Glucose/pharmacology , Receptors, Gastrointestinal Hormone/agonists
3.
Commun Biol ; 5(1): 946, 2022 09 10.
Article in English | MEDLINE | ID: mdl-36088386

ABSTRACT

Most metabolic studies on mice are performed at room temperature, although under these conditions mice, unlike humans, spend considerable energy to maintain core temperature. Here, we characterize the impact of housing temperature on energy expenditure (EE), energy homeostasis and plasma concentrations of appetite- and glucoregulatory hormones in normal-weight and diet-induced obese (DIO) C57BL/6J mice fed chow or 45% high-fat-diet, respectively. Mice were housed for 33 days at 22, 25, 27.5, and 30 °C in an indirect-calorimetry-system. We show that energy expenditure increases linearly from 30 °C towards 22 °C and is ~30% higher at 22 °C in both mouse models. In normal-weight mice, food intake counter-balances EE. In contrast, DIO mice do not reduce food intake when EE is lowered. By end of study, mice at 30 °C, therefore, had higher body weight, fat mass and plasma glycerol and triglycerides than mice at 22 °C. Dysregulated counterbalancing in DIO mice may result from increased pleasure-based eating.


Subject(s)
Energy Intake , Housing , Animals , Body Weight , Diet, High-Fat/adverse effects , Energy Metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Obesity/etiology , Obesity/metabolism , Temperature
4.
Int J Mol Sci ; 23(6)2022 Mar 17.
Article in English | MEDLINE | ID: mdl-35328681

ABSTRACT

Restoring the control of food intake is the key to obesity management and prevention. The arcuate nucleus (ARC) of the hypothalamus is extensively being studied as a potential anti-obesity target. Animal studies showed that neuropeptide FF (NPFF) reduces food intake by its action in neuropeptide Y (NPY) neurons of the hypothalamic ARC, but the detailed mode of action observed in human neurons is missing, due to the lack of a human-neuron-based model for pharmacology testing. Here, we validated and utilized a human-neural-stem-cell-based (hNSC) model of ARC to test the effects of NPFF on cellular pathways and neuronal activity. We found that in the human neurons, decreased cAMP levels by NPFF resulted in a reduced rate of cytoplasmic calcium oscillations, indicating an inhibition of ARC NPY neurons. This suggests the therapeutic potential of NPFFR2 in obesity. In addition, we demonstrate the use of human-stem-cell-derived neurons in pharmacological applications and the potential of this model to address functional aspects of human hypothalamic neurons.


Subject(s)
Neuropeptide Y , Oligopeptides , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Humans , Neurons/metabolism , Neuropeptide Y/metabolism , Neuropeptide Y/pharmacology , Obesity/metabolism , Oligopeptides/pharmacology
5.
Obesity (Silver Spring) ; 30(4): 841-857, 2022 04.
Article in English | MEDLINE | ID: mdl-35333444

ABSTRACT

New appetite-regulating antiobesity treatments such as semaglutide and agents under investigation such as tirzepatide show promise in achieving weight loss of 15% or more. Energy expenditure, fat oxidation, and lean mass preservation are important determinants of weight loss and weight-loss maintenance beyond appetite regulation. This review discusses prior failures in clinical development of weight-loss drugs targeting energy expenditure and explores novel strategies for targeting energy expenditure: mitochondrial proton leak, uncoupling, dynamics, and biogenesis; futile calcium and substrate cycling; leptin for weight maintenance; increased sympathetic nervous system activity; and browning of white fat. Relevant targets for preserving lean mass are also reviewed: growth hormone, activin type II receptor inhibition, and urocortin 2 and 3. We endorse moderate modulation of energy expenditure and preservation of lean mass in combination with efficient appetite reduction as a means of obtaining a significant, safe, and long-lasting weight loss. Furthermore, we suggest that the regulatory guidelines should be revisited to focus more on the quality of weight loss and its maintenance rather than the absolute weight loss. Commitment to this research focus both from a scientific and from a regulatory point of view could signal the beginning of the next era in obesity therapies.


Subject(s)
Appetite Regulation , Weight Loss , Appetite , Energy Metabolism/physiology , Humans , Obesity/drug therapy , Weight Loss/physiology
6.
Eur J Neurosci ; 54(3): 4863-4876, 2021 08.
Article in English | MEDLINE | ID: mdl-34189795

ABSTRACT

The role of receptor activity-modifying proteins (RAMPs) in modulating the pharmacological effects of an amylin receptor selective agonist (NN1213) or the dual amylin-calcitonin receptor agonist (DACRA), salmon calcitonin (sCT), was tested in three RAMP KO mouse models, RAMP1, RAMP3 and RAMP1/3 KO. Male wild-type (WT) and knockout (KO) littermate mice were fed a 45% high-fat diet for 20 weeks prior to the 3-week treatment period. A decrease in body weight after NN1213 was observed in all WT mice, whereas sCT had no effect. The absence of RAMP1 had no significant effect on NN1213 efficacy, and sCT was still inactive. However, the absence of RAMP3 impeded NN1213 efficacy but improved sCT efficacy. Similar results were observed in RAMP1/3 KO suggesting that the amylin receptor 3 (AMY3 = CTR + RAMP3) is necessary for NN1213's maximal action on body weight and food intake and that the lack of AMY3 allowed sCT to be active. These results suggest that the chronic use of DACRA such as sCT can have unfavourable effect on body weight loss in mice (which differs from the situation in rats), whereas the use of the amylin receptor selective agonist does not. AMY3 seems to play a crucial role in modulating the action of these two compounds, but in opposite directions. The assessment of a long-term effect of amylin and DACRA in different rodent models is necessary to understand potential physiological beneficial and unfavourable effects on weight loss before its transition to clinical trials.


Subject(s)
Amylin Receptor Agonists , Islet Amyloid Polypeptide , Amylin Receptor Agonists/pharmacology , Animals , Body Weight , Calcitonin , Eating , Male , Mice , Rats , Receptor Activity-Modifying Protein 1 , Receptor Activity-Modifying Protein 3 , Receptor Activity-Modifying Proteins , Receptors, Calcitonin
7.
J Immunotoxicol ; 18(1): 30-36, 2021 12.
Article in English | MEDLINE | ID: mdl-33570451

ABSTRACT

In development of peptide therapeutics, rodents are commonly-used preclinical models when screening compounds for efficacy endpoints in the early stages of discovery projects. During the screening process, some peptides administered subcutaneously to rodents caused injection site reactions manifesting as localized swelling. Screening by postmortem evaluations of injection site swelling as a marker for local subcutaneous histamine release, were conducted in rats to select drug candidates without this adverse effect. Histological analysis of skin samples revealed that the injection site reactions were concurrent with mast cell degranulation, resulting in histamine release. Mast cell activation can be mediated by MRGPRX2, a GPCR that induces a pseudo-allergenic immune response. The present study demonstrates that a commercially-available cell-based MRGPRX2 assay reliably identifies compounds that induce histamine release or localized edema in ex vivo human and rodent skin samples. In vitro screening was subsequently implemented using the MRGPRX2 assay as a substitute for postmortem injection site evaluation, thus achieving a significant reduction in animal use. Thus, in cases where injection site reactions are encountered during in vivo screening, to enable faster screening during the early drug discovery process, an MRGPRX2 in vitro assay can be used as an efficient, more ethical tool with human translational value for the development of safer pharmacotherapies for patients.


Subject(s)
Cell Degranulation , Receptors, Neuropeptide , Allergens , Animals , Humans , Mast Cells , Nerve Tissue Proteins , Rats , Receptors, G-Protein-Coupled
8.
Endocr Connect ; 9(8): 755-768, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32688339

ABSTRACT

The prevalence of non-alcoholic fatty liver disease (NAFLD) has increased dramatically worldwide and, subsequently, also the risk of developing non-alcoholic steatohepatitis (NASH), hepatic fibrosis, cirrhosis and cancer. Today, weight loss is the only available treatment, but administration of fibroblast growth factor 21 (FGF21) analogues have, in addition to weight loss, shown improvements on liver metabolic health but the mechanisms behind are not entirely clear. The aim of this study was to investigate the hepatic metabolic profile in response to FGF21 treatment. Diet-induced obese (DIO) mice were treated with s.c. administration of FGF21 or subjected to caloric restriction by switching from high fat diet (HFD) to chow to induce 20% weight loss and changes were compared to vehicle dosed DIO mice. Cumulative caloric intake was reduced by chow, while no differences were observed between FGF21 and vehicle dosed mice. The body weight loss in both treatment groups was associated with reduced body fat mass and hepatic triglycerides (TG), while hepatic cholesterol was slightly decreased by chow. Liver glycogen was decreased by FGF21 and increased by chow. The hepatic gene expression profiles suggest that FGF21 increased uptake of fatty acids and lipoproteins, channeled TGs toward the production of cholesterol and bile acid, reduced lipogenesis and increased hepatic glucose output. Furthermore, FGF21 appeared to reduce inflammation and regulate hepatic leptin receptor-a expression. In conclusion, FGF21 affected several metabolic pathways to reduce hepatic steatosis and improve hepatic health and markedly more genes than diet restriction (61 vs 16 out of 89 investigated genes).

9.
Basic Clin Pharmacol Toxicol ; 127(3): 163-177, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32363722

ABSTRACT

The prevalence of obesity and associated comorbidities such as type 2 diabetes and cardiovascular disease is increasing globally. Body-weight loss reduces the risk of morbidity and mortality in obese individuals, and thus, pharmacotherapies that induce weight loss can be of great value in improving the health and well-being of people living with obesity. Treatment with amylin and calcitonin receptor agonists reduces food intake and induces weight loss in several animal models, and a number of companies have started clinical testing for peptide analogues in the treatment of obesity and/or type 2 diabetes. Studies predominantly performed in rodent models show that amylin and the dual amylin/calcitonin receptor agonist salmon calcitonin achieve their metabolic effects by engaging areas in the brain associated with regulating homeostatic energy balance. In particular, signalling via neuronal circuits in the caudal hindbrain and the hypothalamus is implicated in mediating effects on food intake and energy expenditure. We review the current literature investigating the interaction of amylin/calcitonin receptor agonists with neurocircuits that induce the observed metabolic effects. Moreover, the status of drug development of amylin and calcitonin receptor agonists for the treatment of metabolic diseases is summarized.


Subject(s)
Amylin Receptor Agonists/pharmacology , Amylin Receptor Agonists/therapeutic use , Metabolic Diseases/drug therapy , Receptors, Calcitonin/agonists , Receptors, Calcitonin/therapeutic use , Animals , Energy Metabolism , Humans , Hypothalamus , Islet Amyloid Polypeptide/physiology , Leptin , Mice , Rats , Rhombencephalon
10.
JCI Insight ; 5(6)2020 03 26.
Article in English | MEDLINE | ID: mdl-32213703

ABSTRACT

Semaglutide, a glucagon-like peptide 1 (GLP-1) analog, induces weight loss, lowers glucose levels, and reduces cardiovascular risk in patients with diabetes. Mechanistic preclinical studies suggest weight loss is mediated through GLP-1 receptors (GLP-1Rs) in the brain. The findings presented here show that semaglutide modulated food preference, reduced food intake, and caused weight loss without decreasing energy expenditure. Semaglutide directly accessed the brainstem, septal nucleus, and hypothalamus but did not cross the blood-brain barrier; it interacted with the brain through the circumventricular organs and several select sites adjacent to the ventricles. Semaglutide induced central c-Fos activation in 10 brain areas, including hindbrain areas directly targeted by semaglutide, and secondary areas without direct GLP-1R interaction, such as the lateral parabrachial nucleus. Automated analysis of semaglutide access, c-Fos activity, GLP-1R distribution, and brain connectivity revealed that activation may involve meal termination controlled by neurons in the lateral parabrachial nucleus. Transcriptomic analysis of microdissected brain areas from semaglutide-treated rats showed upregulation of prolactin-releasing hormone and tyrosine hydroxylase in the area postrema. We suggest semaglutide lowers body weight by direct interaction with diverse GLP-1R populations and by directly and indirectly affecting the activity of neural pathways involved in food intake, reward, and energy expenditure.


Subject(s)
Body Weight/drug effects , Brain/drug effects , Glucagon-Like Peptides/pharmacology , Neural Pathways/drug effects , Animals , Eating/drug effects , Energy Metabolism/drug effects , Glucagon-Like Peptide-1 Receptor/drug effects , Mice , Rats
11.
Neuropharmacology ; 167: 107987, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32035146

ABSTRACT

The amylin receptor (AMY) and calcitonin receptor (CTR) agonists induce acute suppression of food intake in rodents by binding to receptors in the area postrema (AP) and potentially by targeting arcuate (ARC) neurons directly. Salmon calcitonin (sCT) induces more potent, longer lasting anorectic effects compared to amylin. We thus aimed to investigate whether AMY/CTR agonists target key neuronal populations in the ARC, and whether differing brain distribution patterns could mediate the observed differences in efficacy with sCT and amylin treatment. Brains were examined by whole brain 3D imaging and confocal microscopy following subcutaneous administration of fluorescently labelled peptides to mice. We found that sCT, but not amylin, internalizes into a subset of ARC NPY neurons, along with an unknown subset of ARC, AP and dorsal vagal motor nucleus cells. ARC POMC neurons were not targeted. Furthermore, amylin and sCT displayed similar distribution patterns binding to receptors in the AP, the organum vasculosum of the lamina terminalis (OVLT) and the ARC. Amylin distributed within the median eminence with only specs of sCT being present in this region, however amylin was only detectable 10 minutes after injection while sCT displayed a residence time of up to 2 hours post injection. We conclude that AMY/CTR agonists bind to receptors in a subset of ARC NPY neurons and in circumventricular organs. Furthermore, the more sustained and greater anorectic efficacy of sCT compared to rat amylin is not attributable to differences in brain distribution patterns but may more likely be explained by greater potency at both the CTR and AMY.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Calcitonin/metabolism , Calcium-Regulating Hormones and Agents/metabolism , Neurons/metabolism , Neuropeptide Y/metabolism , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Calcitonin/administration & dosage , Calcium-Regulating Hormones and Agents/administration & dosage , Cell Line , Cricetinae , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Protein Binding/physiology , Rats
12.
Dis Model Mech ; 12(11)2019 11 22.
Article in English | MEDLINE | ID: mdl-31704726

ABSTRACT

Parkinson's disease (PD) is a basal ganglia movement disorder characterized by progressive degeneration of the nigrostriatal dopaminergic system. Immunohistochemical methods have been widely used for characterization of dopaminergic neuronal injury in animal models of PD, including the MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) mouse model. However, conventional immunohistochemical techniques applied to tissue sections have inherent limitations with respect to loss of 3D resolution, yielding insufficient information on the architecture of the dopaminergic system. To provide a more comprehensive and non-biased map of MPTP-induced changes in central dopaminergic pathways, we used iDISCO immunolabeling, light-sheet fluorescence microscopy (LSFM) and deep-learning computational methods for whole-brain three-dimensional visualization and automated quantitation of tyrosine hydroxylase (TH)-positive neurons in the adult mouse brain. Mice terminated 7 days after acute MPTP administration demonstrated widespread alterations in TH expression. Compared to vehicle controls, MPTP-dosed mice showed a significant loss of TH-positive neurons in the substantia nigra pars compacta and ventral tegmental area. Also, MPTP dosing reduced overall TH signal intensity in basal ganglia nuclei, i.e. the substantia nigra, caudate-putamen, globus pallidus and subthalamic nucleus. In contrast, increased TH signal intensity was predominantly observed in limbic regions, including several subdivisions of the amygdala and hypothalamus. In conclusion, mouse whole-brain 3D imaging is ideal for unbiased automated counting and densitometric analysis of TH-positive cells. The LSFM-deep learning pipeline tracked brain-wide changes in catecholaminergic pathways in the MPTP mouse model of PD, and may be applied for preclinical characterization of compounds targeting dopaminergic neurotransmission.


Subject(s)
Brain/diagnostic imaging , Disease Models, Animal , Imaging, Three-Dimensional/methods , Neurons/enzymology , Parkinson Disease/diagnostic imaging , Tyrosine 3-Monooxygenase/analysis , Animals , Deep Learning , MPTP Poisoning/diagnostic imaging , Mice , Microscopy, Fluorescence , Motor Skills , Parkinson Disease/enzymology
13.
Nat Med ; 23(10): 1158-1166, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28846099

ABSTRACT

Growth differentiation factor 15 (GDF15; also known as MIC-1) is a divergent member of the TGF-ß superfamily and is associated with body-weight regulation in humans and rodents. However, the cognate receptor of GDF15 is unknown. Here we show that GDF15 binds specifically to GDNF family receptor α-like (GFRAL) with high affinity, and that GFRAL requires association with the coreceptor RET to elicit intracellular signaling in response to GDF15 stimulation. We also found that GDF15-mediated reductions in food intake and body weight of mice with obesity were abolished in GFRAL-knockout mice. We further found that GFRAL expression was limited to hindbrain neurons and not present in peripheral tissues, which suggests that GDF15-GFRAL-mediated regulation of food intake is by a central mechanism. Lastly, given that GDF15 did not increase energy expenditure in treated mice with obesity, the anti-obesity actions of the cytokine are likely driven primarily by a reduction in food intake.


Subject(s)
Eating/drug effects , Energy Metabolism/drug effects , Glial Cell Line-Derived Neurotrophic Factor Receptors/drug effects , Growth Differentiation Factor 15/pharmacology , Obesity/metabolism , Weight Loss/drug effects , Animals , Eating/genetics , Energy Metabolism/genetics , Flow Cytometry , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , HEK293 Cells , Humans , In Vitro Techniques , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Surface Plasmon Resonance , Weight Loss/genetics
14.
J Cell Biol ; 167(5): 915-24, 2004 Dec 06.
Article in English | MEDLINE | ID: mdl-15569710

ABSTRACT

Thyroid hormone 3,5,3'-tri-iodothyronine (T3) binds and activates thyroid hormone receptors (TRs). Here, we present evidence for a nontranscriptional regulation of Ca2+ signaling by T3-bound TRs. Treatment of Xenopus thyroid hormone receptor beta subtype A1 (xTRbetaA1) expressing oocytes with T3 for 10 min increased inositol 1,4,5-trisphosphate (IP3)-mediated Ca2+ wave periodicity. Coexpression of TRbetaA1 with retinoid X receptor did not enhance regulation. Deletion of the DNA binding domain and the nuclear localization signal of the TRbetaA1 eliminated transcriptional activity but did not affect the ability to regulate Ca2+ signaling. T3-bound TRbetaA1 regulation of Ca2+ signaling could be inhibited by ruthenium red treatment, suggesting that mitochondrial Ca2+ uptake was required for the mechanism of action. Both xTRbetaA1 and the homologous shortened form of rat TRalpha1 (rTRalphaDeltaF1) localized to the mitochondria and increased O2 consumption, whereas the full-length rat TRalpha1 did neither. Furthermore, only T3-bound xTRbetaA1 and rTRalphaDeltaF1 affected Ca2+ wave activity. We conclude that T3-bound mitochondrial targeted TRs acutely modulate IP3-mediated Ca2+ signaling by increasing mitochondrial metabolism independently of transcriptional activity.


Subject(s)
Calcium Signaling/physiology , Calcium/metabolism , Mitochondria/metabolism , Receptors, Thyroid Hormone/metabolism , Triiodothyronine/metabolism , Animals , Binding Sites/physiology , Calcium Signaling/drug effects , Cell Respiration/drug effects , Cell Respiration/genetics , DNA-Binding Proteins/genetics , Genes, Regulator/genetics , Inositol 1,4,5-Trisphosphate/metabolism , Ligands , Mitochondria/drug effects , Mitochondria/genetics , Oocytes , Protein Structure, Tertiary/genetics , Rats , Receptors, Thyroid Hormone/drug effects , Receptors, Thyroid Hormone/genetics , Retinoid X Receptors/genetics , Retinoid X Receptors/metabolism , Ruthenium Red/pharmacology , Thyroid Hormone Receptors alpha/metabolism , Thyroid Hormone Receptors beta/drug effects , Thyroid Hormone Receptors beta/genetics , Thyroid Hormone Receptors beta/metabolism , Triiodothyronine/pharmacology , Up-Regulation/drug effects , Up-Regulation/genetics , Xenopus laevis
15.
Endocrinology ; 145(6): 2594-603, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14976145

ABSTRACT

Hormonal control of metabolic rate can be important in regulating the imbalance between energy intake and expenditure that underlies the development of obesity. In mice fed a high-fat diet, human fibroblast growth factor 19 (FGF19) increased metabolic rate [1.53 +/- 0.06 liters O(2)/h.kg(0.75) (vehicle) vs. 1.93 +/- 0.05 liters O(2)/h.kg(0.75) (FGF19); P < 0.001] and decreased respiratory quotient [0.82 +/- 0.01 (vehicle) vs. 0.80 +/- 0.01 (FGF19); P < 0.05]. In contrast to the vehicle-treated mice that gained weight (0.14 +/- 0.05 g/mouse.d), FGF19-treated mice lost weight (-0.13 +/- 0.03 g/mouse.d; P < 0.001) without a significant change in food intake. Furthermore, in addition to a reduction in weight gain, treatment with FGF19 prevented or reversed the diabetes that develops in mice made obese by genetic ablation of brown adipose tissue or genetic absence of leptin. To explore the mechanisms underlying the FGF19-mediated increase in metabolic rate, we profiled the FGF19-induced gene expression changes in the liver and brown fat. In brown adipose tissue, chronic exposure to FGF19 led to a gene expression profile that is consistent with activation of this tissue. We also found that FGF19 acutely increased liver expression of the leptin receptor (1.8-fold; P < 0.05) and decreased the expression of acetyl coenzyme A carboxylase 2 (0.6-fold; P < 0.05). The gene expression changes were consistent with the experimentally determined increase in fat oxidation and decrease in liver triglycerides. Thus, FGF19 is able to increase metabolic rate concurrently with an increase in fatty acid oxidation.


Subject(s)
Diabetes Mellitus/etiology , Diabetes Mellitus/physiopathology , Diet , Fibroblast Growth Factors/pharmacology , Leptin/deficiency , Recombinant Proteins/pharmacology , Acetyl-CoA Carboxylase/metabolism , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/physiopathology , Animals , Body Weight/drug effects , Diabetes Mellitus/metabolism , Gene Expression/drug effects , Humans , Liver/drug effects , Liver/metabolism , Metabolism/drug effects , Mice , Mice, Transgenic , Obesity , Receptors, Cell Surface/metabolism , Receptors, Leptin
16.
Endocrinology ; 143(5): 1741-7, 2002 May.
Article in English | MEDLINE | ID: mdl-11956156

ABSTRACT

The fibroblast growth factors (FGFs), and the corresponding receptors, are implicated in more than just the regulation of epithelial cell proliferation and differentiation. Specifically, FGF23 is a regulator of serum inorganic phosphate levels, and mice deficient in FGF receptor-4 have altered cholesterol metabolism. The recently described FGF19 is unusual in that it is nonmitogenic and appears to interact only with FGF receptor-4. Here, we report that FGF19 transgenic mice had a significant and specific reduction in fat mass that resulted from an increase in energy expenditure. Further, the FGF19 transgenic mice did not become obese or diabetic on a high fat diet. The FGF19 transgenic mice had increased brown adipose tissue mass and decreased liver expression of acetyl coenzyme A carboxylase 2, providing two mechanisms by which FGF19 may increase energy expenditure. Consistent with the reduction in expression of acetyl CoA carboxylase 2, liver triglyceride levels were reduced.


Subject(s)
Adipose Tissue/physiology , Energy Metabolism/genetics , Fibroblast Growth Factors/biosynthesis , Fibroblast Growth Factors/genetics , Adipose Tissue/drug effects , Adipose Tissue, Brown/metabolism , Animals , Body Temperature/genetics , Body Temperature/physiology , Body Weight/genetics , Body Weight/physiology , Calorimetry, Indirect , Diabetes Mellitus/metabolism , Diet , Dietary Fats/pharmacology , Energy Metabolism/drug effects , Fibroblast Growth Factor-23 , Glucose/metabolism , Glucose Tolerance Test , Humans , Liver/metabolism , Mice , Mice, Transgenic , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...