Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nat Commun ; 14(1): 3076, 2023 05 29.
Article in English | MEDLINE | ID: mdl-37248237

ABSTRACT

Coupling the release of pituitary hormones to the developmental stage of the oocyte is essential for female fertility. It requires estrogen to restrain kisspeptin (KISS1)-neuron pulsatility in the arcuate hypothalamic nucleus, while also exerting a surge-like effect on KISS1-neuron activity in the AVPV hypothalamic nucleus. However, a mechanistic basis for this region-specific effect has remained elusive. Our genomic analysis in female mice demonstrate that some processes, such as restraint of KISS1-neuron activity in the arcuate nucleus, may be explained by region-specific estrogen receptor alpha (ERα) DNA binding at gene regulatory regions. Furthermore, we find that the Kiss1-locus is uniquely regulated in these hypothalamic nuclei, and that the nuclear receptor co-repressor NR0B1 (DAX1) restrains its transcription specifically in the arcuate nucleus. These studies provide mechanistic insight into how ERα may control the KISS1-neuron, and Kiss1 gene expression, to couple gonadotropin release to the developmental stage of the oocyte.


Subject(s)
DAX-1 Orphan Nuclear Receptor , Estrogen Receptor alpha , Hypothalamus , Kisspeptins , Animals , Female , Mice , Arcuate Nucleus of Hypothalamus/metabolism , Estradiol/metabolism , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Hypothalamus/metabolism , Kisspeptins/genetics , Kisspeptins/metabolism , DAX-1 Orphan Nuclear Receptor/genetics , DAX-1 Orphan Nuclear Receptor/metabolism
2.
Cell Rep Med ; 3(11): 100810, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36384093

ABSTRACT

Glucagon analogs show promise as components of next-generation, multi-target, anti-obesity therapeutics. The biology of chronic glucagon treatment, in particular, its ability to induce energy expenditure and weight loss, remains poorly understood. Using a long-acting glucagon analog, G108, we demonstrate that glucagon-mediated body weight loss is intrinsically linked to the hypoaminoacidemia associated with its known amino acid catabolic action. Mechanistic studies reveal an energy-consuming response to low plasma amino acids in G108-treated mice, prevented by dietary amino acid supplementation and mimicked by a rationally designed low amino acid diet. Therefore, low plasma amino acids are a pre-requisite for G108-mediated energy expenditure and weight loss. However, preventing hypoaminoacidemia with additional dietary protein does not affect the ability of G108 to improve glycemia or hepatic steatosis in obese mice. These studies provide a mechanism for glucagon-mediated weight loss and confirm the hepatic glucagon receptor as an attractive molecular target for metabolic disease therapeutics.


Subject(s)
Glucagon , Weight Loss , Mice , Animals , Glucagon/metabolism , Energy Metabolism/physiology , Receptors, Glucagon/metabolism , Mice, Obese , Amino Acids/pharmacology
3.
Sci Rep ; 11(1): 22577, 2021 11 19.
Article in English | MEDLINE | ID: mdl-34799628

ABSTRACT

Glucagon receptor agonists show promise as components of next generation metabolic syndrome pharmacotherapies. However, the biology of glucagon action is complex, controversial, and likely context dependent. As such, a better understanding of chronic glucagon receptor (GCGR) agonism is essential to identify and mitigate potential clinical side-effects. Herein we present a novel, long-acting glucagon analogue (GCG104) with high receptor-specificity and potent in vivo action. It has allowed us to make two important observations about the biology of sustained GCGR agonism. First, it causes weight loss in mice by direct receptor signalling at the level of the liver. Second, subtle changes in GCG104-sensitivity, possibly due to interindividual variation, may be sufficient to alter its effects on metabolic parameters. Together, these findings confirm the liver as a principal target for glucagon-mediated weight loss and provide new insights into the biology of glucagon analogues.


Subject(s)
Anti-Obesity Agents/pharmacology , Glucagon/pharmacology , Hepatocytes/drug effects , Liver/drug effects , Receptors, Glucagon/agonists , Weight Loss/drug effects , Animals , Anti-Obesity Agents/pharmacokinetics , Biological Variation, Population , Dose-Response Relationship, Drug , Female , Glucagon/analogs & derivatives , Glucagon/pharmacokinetics , HEK293 Cells , Hepatocytes/metabolism , Humans , Liver/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Rats, Wistar , Receptors, Glucagon/genetics , Receptors, Glucagon/metabolism , Signal Transduction
4.
Psychoneuroendocrinology ; 129: 105244, 2021 07.
Article in English | MEDLINE | ID: mdl-33975151

ABSTRACT

Gamma-aminobutyric acid (GABA) is a key inhibitory neurotransmitter that has been implicated in the aetiology of common mood and behavioural disorders. By employing proton magnetic resonance spectroscopy in man, we demonstrate that administration of the reproductive neuropeptide, kisspeptin, robustly decreases GABA levels in the limbic system of the human brain; specifically the anterior cingulate cortex (ACC). This finding defines a novel kisspeptin-activated GABA pathway in man, and provides important mechanistic insights into the mood and behaviour-altering effects of kisspeptin seen in rodents and humans. In addition, this work has therapeutic implications as it identifies GABA-signalling as a potential target for the escalating development of kisspeptin-based therapies for common reproductive disorders of body and mind.


Subject(s)
Brain , Kisspeptins , gamma-Aminobutyric Acid , Brain/metabolism , Humans , Kisspeptins/metabolism , gamma-Aminobutyric Acid/metabolism
5.
Mol Metab ; 51: 101242, 2021 09.
Article in English | MEDLINE | ID: mdl-33933675

ABSTRACT

OBJECTIVE: Glucagon-like peptide-1 and glucagon receptor (GLP-1R/GCGR) co-agonism can maximise weight loss and improve glycaemic control in type 2 diabetes and obesity. In this study, we investigated the cellular and metabolic effects of modulating the balance between G protein and ß-arrestin-2 recruitment at GLP-1R and GCGR using oxyntomodulin (OXM)-derived co-agonists. This strategy has been previously shown to improve the duration of action of GLP-1R mono-agonists by reducing target desensitisation and downregulation. METHODS: Dipeptidyl dipeptidase-4 (DPP-4)-resistant OXM analogues were generated and assessed for a variety of cellular readouts. Molecular dynamic simulations were used to gain insights into the molecular interactions involved. In vivo studies were performed in mice to identify the effects on glucose homeostasis and weight loss. RESULTS: Ligand-specific reductions in ß-arrestin-2 recruitment were associated with slower GLP-1R internalisation and prolonged glucose-lowering action in vivo. The putative benefits of GCGR agonism were retained, with equivalent weight loss compared to the GLP-1R mono-agonist liraglutide despite a lesser degree of food intake suppression. The compounds tested showed only a minor degree of biased agonism between G protein and ß-arrestin-2 recruitment at both receptors and were best classified as partial agonists for the two pathways measured. CONCLUSIONS: Diminishing ß-arrestin-2 recruitment may be an effective way to increase the therapeutic efficacy of GLP-1R/GCGR co-agonists. These benefits can be achieved by partial rather than biased agonism.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide-1 Receptor/agonists , Hypoglycemic Agents/pharmacology , Peptides/pharmacology , Receptors, Glucagon/agonists , Animals , Blood Glucose/analysis , Blood Glucose/drug effects , Cells, Cultured , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Disease Models, Animal , HEK293 Cells , Hepatocytes , Humans , Hypoglycemic Agents/therapeutic use , Islets of Langerhans , Liraglutide/pharmacology , Liraglutide/therapeutic use , Male , Mice , Oxyntomodulin/genetics , Peptides/genetics , Peptides/therapeutic use , Primary Cell Culture , Rats , Weight Loss/drug effects , beta-Arrestin 2/metabolism
6.
Diabetologia ; 64(4): 850-864, 2021 04.
Article in English | MEDLINE | ID: mdl-33492421

ABSTRACT

AIMS/HYPOTHESIS: Variants close to the VPS13C/C2CD4A/C2CD4B locus are associated with altered risk of type 2 diabetes in genome-wide association studies. While previous functional work has suggested roles for VPS13C and C2CD4A in disease development, none has explored the role of C2CD4B. METHODS: CRISPR/Cas9-induced global C2cd4b-knockout mice and zebrafish larvae with c2cd4a deletion were used to study the role of this gene in glucose homeostasis. C2 calcium dependent domain containing protein (C2CD)4A and C2CD4B constructs tagged with FLAG or green fluorescent protein were generated to investigate subcellular dynamics using confocal or near-field microscopy and to identify interacting partners by mass spectrometry. RESULTS: Systemic inactivation of C2cd4b in mice led to marked, but highly sexually dimorphic changes in body weight and glucose homeostasis. Female C2cd4b mice displayed unchanged body weight compared with control littermates, but abnormal glucose tolerance (AUC, p = 0.01) and defective in vivo, but not in vitro, insulin secretion (p = 0.02). This was associated with a marked decrease in follicle-stimulating hormone levels as compared with wild-type (WT) littermates (p = 0.003). In sharp contrast, male C2cd4b null mice displayed essentially normal glucose tolerance but an increase in body weight (p < 0.001) and fasting blood glucose (p = 0.003) after maintenance on a high-fat and -sucrose diet vs WT littermates. No metabolic disturbances were observed after global inactivation of C2cd4a in mice, or in pancreatic beta cell function at larval stages in C2cd4a null zebrafish. Fasting blood glucose levels were also unaltered in adult C2cd4a-null fish. C2CD4B and C2CD4A were partially localised to the plasma membrane, with the latter under the control of intracellular Ca2+. Binding partners for both included secretory-granule-localised PTPRN2/phogrin. CONCLUSIONS/INTERPRETATION: Our studies suggest that C2cd4b may act centrally in the pituitary to influence sex-dependent circuits that control pancreatic beta cell function and glucose tolerance in rodents. However, the absence of sexual dimorphism in the impact of diabetes risk variants argues for additional roles for C2CD4A or VPS13C in the control of glucose homeostasis in humans. DATA AVAILABILITY: The datasets generated and/or analysed during the current study are available in the Biorxiv repository ( www.biorxiv.org/content/10.1101/2020.05.18.099200v1 ). RNA-Seq (GSE152576) and proteomics (PXD021597) data have been deposited to GEO ( www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE152576 ) and ProteomeXchange ( www.ebi.ac.uk/pride/archive/projects/PXD021597 ) repositories, respectively.


Subject(s)
Blood Glucose/metabolism , Diabetes Mellitus, Type 2/genetics , Homeostasis/genetics , Insulin-Secreting Cells/metabolism , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , Biomarkers/blood , Blood Glucose/genetics , Female , Follicle Stimulating Hormone/blood , Genotype , Humans , Insulin/blood , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Pituitary Gland/metabolism , Sex Characteristics , Weight Gain , Zebrafish/blood , Zebrafish/genetics , Zebrafish Proteins/blood , Zebrafish Proteins/genetics
7.
J Clin Endocrinol Metab ; 106(4): e1837-e1848, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33075807

ABSTRACT

CONTEXT: The hormone kisspeptin has crucial and well-characterized roles in reproduction. Emerging data from animal models also suggest that kisspeptin has important metabolic effects including modulation of food intake. However, to date there have been no studies exploring the effects of kisspeptin on brain responses to food stimuli in humans. OBJECTIVE: This work aims to investigate the effects of kisspeptin administration on brain responses to visual food stimuli and psychometric parameters of appetite, in healthy men. DESIGN: A double-blinded, randomized, placebo-controlled, crossover study was conducted. PARTICIPANTS: Participants included 27 healthy, right-handed, eugonadal men (mean ± SEM: age 26.5 ± 1.1 years; body mass index 23.9 ± 0.4 kg/m2). INTERVENTION: Participants received an intravenous infusion of 1 nmol/kg/h of kisspeptin or rate-matched vehicle over 75 minutes. MAIN OUTCOME MEASURES: Measurements included change in brain activity on functional magnetic resonance imaging in response to visual food stimuli and change in psychometric parameters of appetite, during kisspeptin administration compared to vehicle. RESULTS: Kisspeptin administration at a bioactive dose did not affect brain responses to visual food stimuli or psychometric parameters of appetite compared to vehicle. CONCLUSIONS: This is the first study in humans investigating the effects of kisspeptin on brain regions regulating appetite and demonstrates that peripheral administration of kisspeptin does not alter brain responses to visual food stimuli or psychometric parameters of appetite in healthy men. These data provide key translational insights to further our understanding of the interaction between reproduction and metabolism.


Subject(s)
Appetite/drug effects , Brain/drug effects , Kisspeptins/pharmacology , Adult , Brain/diagnostic imaging , Brain/physiology , Cross-Over Studies , Double-Blind Method , Food , Healthy Volunteers , Humans , Infusions, Intravenous , Kisspeptins/administration & dosage , Magnetic Resonance Imaging , Male , Nerve Net/diagnostic imaging , Nerve Net/drug effects , Photic Stimulation , Psychometrics , Reward , United Kingdom
8.
JCI Insight ; 5(3)2020 02 13.
Article in English | MEDLINE | ID: mdl-32051344

ABSTRACT

Successful reproduction is a fundamental physiological process that relies on the integration of sensory cues of attraction with appropriate emotions and behaviors and the reproductive axis. However, the factors responsible for this integration remain largely unexplored. Using functional neuroimaging, hormonal, and psychometric analyses, we demonstrate that the reproductive hormone kisspeptin enhances brain activity in response to olfactory and visual cues of attraction in men. Furthermore, the brain regions enhanced by kisspeptin correspond to areas within the olfactory and limbic systems that govern sexual behavior and perception of beauty as well as overlap with its endogenous expression pattern. Of key functional and behavioral significance, we observed that kisspeptin was most effective in men with lower sexual quality-of-life scores. As such, our results reveal a previously undescribed attraction pathway in humans activated by kisspeptin and identify kisspeptin signaling as a new therapeutic target for related reproductive and psychosexual disorders.


Subject(s)
Brain/physiology , Cues , Kisspeptins/physiology , Sexual Behavior/physiology , Smell/physiology , Vision, Ocular/physiology , Adult , Cross-Over Studies , Double-Blind Method , Female , Humans , Kisspeptins/metabolism , Male , Placebos , Quality of Life , Sexual Dysfunctions, Psychological/physiopathology , Signal Transduction
9.
Sci Rep ; 10(1): 1703, 2020 02 03.
Article in English | MEDLINE | ID: mdl-32015477

ABSTRACT

The nuclear hormone receptor Dax1 functions during development as a testes-determining gene. However, the phenotype of male mice lacking Dax1 is strain-dependent due to the background-specific abundance of male-determining Sry gene-transcripts. We hypothesised that inter-individual variation in Sry mRNA-abundance would result in a spectrum of phenotypes even within-strain. We found that while all XY C57BL/6J mice lacking Dax1 presented as phenotypic females, there was a marked inter-individual variability in measures of fertility. Indeed, we report rare occasions where sex-reversed mice had measures of fertility comparable to those in control females. On two occasions, these sex-reversed XY mice were able to give birth to live offspring following mating to stud-males. As such, this work documents within-strain variability in phenotypes of XY mice lacking Dax1, and reports for the first time a complete sex-reversal capable of achieving live birth in these mice.


Subject(s)
DAX-1 Orphan Nuclear Receptor/genetics , Disorders of Sex Development/genetics , Sex Determination Processes/physiology , Sex-Determining Region Y Protein/genetics , Testis/physiology , Animals , Biological Variation, Individual , Female , Fertility , Genetic Background , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Parturition , Phenotype
10.
Mol Metab ; 30: 221-229, 2019 12.
Article in English | MEDLINE | ID: mdl-31767173

ABSTRACT

OBJECTIVE: Coupling metabolic and reproductive pathways is essential for the survival of species. However, the functions of steroidogenic enzymes expressed in metabolic tissues are largely unknown. METHODS AND RESULTS: Here, we show that in the liver, the classical steroidogenic enzyme Cyp17a1 forms an essential nexus for glucose and ketone metabolism during feed-fast cycles. Both gain- and loss-of-function approaches are used to show that hepatic Cyp17a1 is induced by fasting, catalyzes the production of at least one hormone-ligand (DHEA) for the nuclear receptor PPARα, and is ultimately required for maintaining euglycemia and ketogenesis during nutrient deprivation. The feedback-loop that terminates Cyp17a1-PPARα activity, and re-establishes anabolic liver metabolism during re-feeding is mapped to postprandial bile acid-signaling, involving the receptors FXR, SHP and LRH-1. CONCLUSIONS: Together, these findings represent a novel paradigm of homeostatic control in which nutritional cues feed-forward on to metabolic pathways by influencing extragonadal steroidogenesis.


Subject(s)
Liver/metabolism , PPAR alpha/metabolism , Steroid 17-alpha-Hydroxylase/metabolism , Animals , Bile Acids and Salts/metabolism , Glucose/metabolism , HEK293 Cells , Hepatocytes/metabolism , Homeostasis , Humans , Ketones/metabolism , Lipogenesis , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxidation-Reduction , Receptors, Cytoplasmic and Nuclear , Signal Transduction , Steroid 17-alpha-Hydroxylase/physiology
11.
Nat Commun ; 9(1): 1602, 2018 04 23.
Article in English | MEDLINE | ID: mdl-29686402

ABSTRACT

Glucagon-like peptide-1 receptor (GLP-1R) activation promotes insulin secretion from pancreatic beta cells, causes weight loss, and is an important pharmacological target in type 2 diabetes (T2D). Like other G protein-coupled receptors, the GLP-1R undergoes agonist-mediated endocytosis, but the functional and therapeutic consequences of modulating GLP-1R endocytic trafficking have not been clearly defined. Here, we investigate a series of biased GLP-1R agonists with variable propensities for GLP-1R internalization and recycling. Compared to a panel of FDA-approved GLP-1 mimetics, compounds that retain GLP-1R at the plasma membrane produce greater long-term insulin release, which is dependent on a reduction in ß-arrestin recruitment and faster agonist dissociation rates. Such molecules elicit glycemic benefits in mice without concomitant increases in signs of nausea, a common side effect of GLP-1 therapies. Our study identifies a set of agents with specific GLP-1R trafficking profiles and the potential for greater efficacy and tolerability as T2D treatments.


Subject(s)
Diabetes Mellitus, Type 2/drug therapy , Glucagon-Like Peptide-1 Receptor/agonists , Hypoglycemic Agents/pharmacology , Insulin/metabolism , Animals , Blood Glucose/drug effects , CHO Cells , Cell Membrane/drug effects , Cell Membrane/metabolism , Cricetulus , Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/pathology , Endocytosis/drug effects , Glucagon-Like Peptide 1/metabolism , Glucagon-Like Peptide-1 Receptor/metabolism , HEK293 Cells , Humans , Hypoglycemic Agents/therapeutic use , Insulin/genetics , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Nausea/chemically induced , Nausea/epidemiology , Primary Cell Culture , Protein Transport/drug effects , RNA, Small Interfering/metabolism , Treatment Outcome
12.
Cell Metab ; 23(3): 427-40, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26959184

ABSTRACT

FGF21 plays a central role in energy, lipid, and glucose homeostasis. To characterize the pharmacologic effects of FGF21, we administered a long-acting FGF21 analog, PF-05231023, to obese cynomolgus monkeys. PF-05231023 caused a marked decrease in food intake that led to reduced body weight. To assess the effects of PF-05231023 in humans, we conducted a placebo-controlled, multiple ascending-dose study in overweight/obese subjects with type 2 diabetes. PF-05231023 treatment resulted in a significant decrease in body weight, improved plasma lipoprotein profile, and increased adiponectin levels. Importantly, there were no significant effects of PF-05231023 on glycemic control. PF-05231023 treatment led to dose-dependent changes in multiple markers of bone formation and resorption and elevated insulin-like growth factor 1. The favorable effects of PF-05231023 on body weight support further evaluation of this molecule for the treatment of obesity. Longer studies are needed to assess potential direct effects of FGF21 on bone in humans.


Subject(s)
Anti-Obesity Agents/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Fibroblast Growth Factors/pharmacology , Obesity/drug therapy , Adolescent , Adult , Aged , Animals , Anti-Obesity Agents/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Blood Glucose , Body Weight/drug effects , Diabetes Mellitus, Type 2/blood , Drug Evaluation, Preclinical , Female , Fibroblast Growth Factors/therapeutic use , Gene Expression/drug effects , Humans , Insulin/blood , Lipid Metabolism/drug effects , Macaca fascicularis , Male , Middle Aged , Obesity/blood , Subcutaneous Fat/drug effects , Subcutaneous Fat/metabolism , Weight Loss , Young Adult
13.
Mol Endocrinol ; 30(4): 469-78, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26974035

ABSTRACT

Androgen and estrogen biosynthesis in mammals requires the 17,20-lyase activity of cytochrome P450 17A1 (steroid 17-hydroxylase/17,20-lyase). Maximal 17,20-lyase activity in vitro requires the presence of cytochrome b5 (b5), and rare cases of b5 deficiency in human beings causes isolated 17,20-lyase deficiency. To study the consequences of conditional b5 removal from testicular Leydig cells in an animal model, we generated Cyb5(flox/flox):Sf1-Cre (LeyKO) mice. The LeyKO male mice had normal body weights, testis and sex organ weights, and fertility compared with littermates. Basal serum and urine steroid profiles of LeyKO males were not significantly different than littermates. In contrast, marked 17-hydroxyprogesterone accumulation (100-fold basal) and reduced testosterone synthesis (27% of littermates) were observed after human chorionic gonadotropin stimulation in LeyKO animals. Testis homogenates from LeyKO mice showed reduced 17,20-lyase activity and a 3-fold increased 17-hydroxylase to 17,20-lyase activity ratio, which were restored to normal upon addition of recombinant b5. We conclude that Leydig cell b5 is required for maximal androgen synthesis and to prevent 17-hydroxyprogesterone accumulation in the mouse testis; however, the b5-independent 17,20-lyase activity of mouse steroid 17-hydroxylase/17,20-lyase is sufficient for normal male genital development and fertility. LeyKO male mice are a good model for the biochemistry but not the physiology of isolated 17,20-lyase deficiency in human beings.


Subject(s)
Cytochromes b5/genetics , Leydig Cells/enzymology , Steroid 17-alpha-Hydroxylase/metabolism , 17-alpha-Hydroxyprogesterone/blood , Animals , Cytochromes b5/metabolism , Female , Fertility , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Phenotype
14.
Cell Metab ; 23(2): 344-9, 2016 Feb 09.
Article in English | MEDLINE | ID: mdl-26724861

ABSTRACT

Fibroblast growth factor 21 (FGF21) is a hormone induced by various metabolic stresses, including ketogenic and high-carbohydrate diets, that regulates energy homeostasis. In humans, SNPs in and around the FGF21 gene have been associated with macronutrient preference, including carbohydrate, fat, and protein intake. Here we show that FGF21 administration markedly reduces sweet and alcohol preference in mice and sweet preference in cynomolgus monkeys. In mice, these effects require the FGF21 co-receptor ß-Klotho in the central nervous system and correlate with reductions in dopamine concentrations in the nucleus accumbens. Since analogs of FGF21 are currently undergoing clinical evaluation for the treatment of obesity and type 2 diabetes, our findings raise the possibility that FGF21 administration could affect nutrient preference and other reward behaviors in humans.


Subject(s)
Alcohols/pharmacology , Fibroblast Growth Factors/metabolism , Food Preferences/drug effects , Taste/drug effects , Animals , Central Nervous System/drug effects , Central Nervous System/metabolism , Dopamine/metabolism , Haplorhini , Humans , Male , Mice, Inbred C57BL , Saccharin/pharmacology , Signal Transduction/drug effects
15.
Curr Opin Pharmacol ; 25: 30-5, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26531325

ABSTRACT

Considerable effort is currently being devoted to understanding the physiological and pharmacological action of the endocrine fibroblast growth factors (FGFs). These three proteins (FGF15/19, FGF21 and FGF23) act in a tissue-specific manner through a membrane-complex consisting of an FGF-receptor and α/ßKlotho. FGF15/19 is produced in the intestine and regulates postprandial liver metabolism and gallbladder filling. FGF21 is largely liver-derived and co-ordinates adaptive changes in response to nutritional and physiological stresses. FGF23 signals from the bone to the kidney to maintain phosphate homeostasis. In pharmacological settings, FGF15/19, FGF21, and the prototypical FGF1, potentially represent novel treatments for obesity and diabetes. This review summarises the recent advances in our understanding of the biology of these important metabolic regulators.


Subject(s)
Fibroblast Growth Factor 1/metabolism , Fibroblast Growth Factors/metabolism , Metabolism , Animals , Fibroblast Growth Factor 1/pharmacology , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/pharmacology , Homeostasis/drug effects , Humans , Metabolism/drug effects , Models, Biological
16.
Trends Endocrinol Metab ; 26(1): 22-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25476453

ABSTRACT

Fibroblast growth factors (FGFs) 15/19 and 21 belong to a subfamily of FGFs that function as hormones. Produced in response to specific nutritional cues, they act on overlapping sets of cell surface receptors composed of classic FGF receptors in complex with ßKlotho, and regulate metabolism and related processes during periods of fluctuating energy availability. Pharmacologically, both FGF15/19 and FGF21 cause weight loss and improve both insulin-sensitivity and lipid parameters in rodent and primate models of metabolic disease. Recently, FGF21 was shown to have similar effects in obese patients with type 2 diabetes. We discuss here emerging concepts in FGF15/19 and FGF21 tissue-specific actions and critically assess their putative role as candidate targets for treating metabolic disease.


Subject(s)
Fibroblast Growth Factors/physiology , Metabolic Diseases/metabolism , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Animals , Diabetes Mellitus, Type 2/metabolism , Fibroblast Growth Factors/pharmacology , Humans , Liver/drug effects , Liver/metabolism , Obesity/metabolism , Organ Specificity
17.
Mol Endocrinol ; 29(2): 213-23, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25495872

ABSTRACT

Hormones such as fibroblast growth factor 21 (FGF21) and glucocorticoids (GCs) play crucial roles in coordinating the adaptive starvation response. Here we examine the interplay between these hormones. It was previously shown that FGF21 induces corticosterone levels in mice by acting on the brain. We now show that this induces the expression of genes required for GC synthesis in the adrenal gland. FGF21 also increases corticosterone secretion from the adrenal in response to ACTH. We further show that the relationship between FGF21 and GCs is bidirectional. GCs induce Fgf21 expression in the liver by acting on the GC receptor (GR). The GR binds in a ligand-dependent manner to a noncanonical GR response element located approximately 4.4 kb upstream of the Fgf21 transcription start site. The GR cooperates with the nuclear fatty acid receptor, peroxisome proliferator-activated receptor-α, to stimulate Fgf21 transcription. GR and peroxisome proliferator-activated receptor-α ligands have additive effects on Fgf21 expression both in vivo and in primary cultures of mouse hepatocytes. We conclude that FGF21 and GCs regulate each other's production in a feed-forward loop and suggest that this provides a mechanism for bypassing negative feedback on the hypothalamic-pituitary-adrenal axis to allow sustained gluconeogenesis during starvation.


Subject(s)
Fibroblast Growth Factors/metabolism , Glucocorticoids/pharmacology , Adrenal Cortex/cytology , Adrenal Cortex/metabolism , Adrenocorticotropic Hormone/pharmacology , Animals , Base Pairing , Binding Sites , Chromatin/metabolism , Corticosterone/biosynthesis , Dexamethasone/pharmacology , Genetic Loci , Humans , Liver/drug effects , Liver/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , PPAR alpha/metabolism , Protein Binding/drug effects , Receptors, Glucocorticoid/metabolism , Transcription Initiation Site , Transcriptional Activation/drug effects
18.
Cell Metab ; 20(4): 670-7, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25130400

ABSTRACT

The mechanism by which pharmacologic administration of the hormone FGF21 increases energy expenditure to cause weight loss in obese animals is unknown. Here we report that FGF21 acts centrally to exert its effects on energy expenditure and body weight in obese mice. Using tissue-specific knockout mice, we show that ßKlotho, the obligate coreceptor for FGF21, is required in the nervous system for these effects. FGF21 stimulates sympathetic nerve activity to brown adipose tissue through a mechanism that depends on the neuropeptide corticotropin-releasing factor. Our findings provide an unexpected mechanistic explanation for the strong pharmacologic effects of FGF21 on energy expenditure and weight loss in obese animals.


Subject(s)
Energy Metabolism/drug effects , Fibroblast Growth Factors/pharmacology , Sympathetic Nervous System/drug effects , Weight Loss/drug effects , Adipose Tissue, Brown/metabolism , Animals , Corticotropin-Releasing Hormone/genetics , Corticotropin-Releasing Hormone/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Hypothalamus/metabolism , Klotho Proteins , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Mice, Transgenic , RNA, Messenger/metabolism , Sympathetic Nervous System/metabolism , Thermogenesis/genetics
19.
Nat Med ; 19(9): 1153-6, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23933983

ABSTRACT

Preventing reproduction during nutritional deprivation is an adaptive process that is conserved and essential for the survival of species. In mammals, the mechanisms that inhibit fertility during starvation are complex and incompletely understood. Here we show that exposure of female mice to fibroblast growth factor 21 (FGF21), a fasting-induced hepatokine, mimics infertility secondary to starvation. Mechanistically, FGF21 acts on the suprachiasmatic nucleus (SCN) in the hypothalamus to suppress the vasopressin-kisspeptin signaling cascade, thereby inhibiting the proestrus surge in luteinizing hormone. Mice lacking the FGF21 co-receptor, ß-Klotho, in the SCN are refractory to the inhibitory effect of FGF21 on female fertility. Thus, FGF21 defines an important liver-neuroendocrine axis that modulates female reproduction in response to nutritional challenge.


Subject(s)
Fibroblast Growth Factors/metabolism , Infertility, Female/metabolism , Membrane Proteins/metabolism , Reproduction , Starvation/metabolism , Animals , Energy Metabolism , Female , Hypothalamus , Kisspeptins/antagonists & inhibitors , Kisspeptins/metabolism , Klotho Proteins , Luteinizing Hormone/biosynthesis , Luteinizing Hormone/metabolism , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Proestrus/physiology , Signal Transduction , Suprachiasmatic Nucleus , Vasopressins/antagonists & inhibitors , Vasopressins/metabolism
20.
Nat Med ; 19(9): 1147-52, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23933984

ABSTRACT

Fibroblast growth factor 21 (FGF21) is a hepatokine that acts as a global starvation signal to modulate fuel partitioning and metabolism and repress growth; however, the site of action of these diverse effects remains unclear. FGF21 signals through a heteromeric cell-surface receptor composed of one of three FGF receptors (FGFR1c, FGFR2c or FGFR3c) in complex with ß-Klotho, a single-pass transmembrane protein that is enriched in metabolic tissues. Here we show that in addition to its known effects on peripheral metabolism, FGF21 increases systemic glucocorticoid levels, suppresses physical activity and alters circadian behavior, which are all features of the adaptive starvation response. These effects are mediated through ß-Klotho expression in the suprachiasmatic nucleus of the hypothalamus and the dorsal vagal complex of the hindbrain. Mice lacking the gene encoding ß-Klotho (Klb) in these regions are refractory to these effects, as well as those on metabolism, insulin and growth. These findings demonstrate a crucial role for the nervous system in mediating the diverse physiologic and pharmacologic actions of FGF21.


Subject(s)
Circadian Rhythm/physiology , Energy Metabolism , Fibroblast Growth Factors/metabolism , Membrane Proteins/metabolism , Nervous System/metabolism , Animals , Glucocorticoids/metabolism , Hypothalamus/metabolism , Klotho Proteins , Male , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Motor Activity , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Signal Transduction , Starvation , Suprachiasmatic Nucleus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...