Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Liver Int ; 43(9): 1890-1900, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37208943

RESUMO

BACKGROUND AND AIMS: Sleeve gastrectomy (VSG) leads to improvement in hepatic steatosis, associated with weight loss. The aims of this study were to investigate whether VSG leads to weight-loss independent improvements in liver steatosis in mice with diet-induced obesity (DIO); and to metabolically and transcriptomically profile hepatic changes in mice undergoing VSG. METHODS: Mice with DIO were treated with VSG, sham surgery with subsequent food restriction to weight-match to the VSG group (Sham-WM), or sham surgery with return to unrestricted diet (Sham-Ad lib). Hepatic steatosis, glucose tolerance, insulin and glucagon resistance, and hepatic transcriptomics were investigated at the end of the study period and treatment groups were compared with mice undergoing sham surgery only (Sham-Ad lib). RESULTS: VSG led to much greater improvement in liver steatosis than Sham-WM (liver triglyceride mg/mg 2.5 ± 0.1, 2.1 ± 0.2, 1.6 ± 0.1 for Sham-AL, Sham-WM and VSG respectively; p = 0.003). Homeostatic model assessment of insulin resistance was improved following VSG only (51.2 ± 8.8, 36.3 ± 5.3, 22.3 ± 6.1 for Sham-AL, Sham-WM and VSG respectively; p = 0.03). The glucagon-alanine index, a measure of glucagon resistance, fell with VSG but was significantly increased in Sham-WM (9.8 ± 1.7, 25.8 ± 4.6 and 5.2 ± 1.2 in Sham Ad-lib, Sham-WM and VSG respectively; p = 0.0003). Genes downstream of glucagon receptor signalling which govern fatty acid synthesis (Acaca, Acacb, Me1, Acly, Fasn and Elovl6) were downregulated following VSG but upregulated in Sham-WM. CONCLUSIONS: Changes in glucagon sensitivity may contribute to weight-loss independent improvements in hepatic steatosis following VSG.


Assuntos
Fígado Gorduroso , Glucagon , Camundongos , Animais , Glicemia , Redução de Peso , Obesidade/complicações , Obesidade/cirurgia , Fígado Gorduroso/complicações , Gastrectomia/efeitos adversos
2.
Diabetologia ; 64(4): 850-864, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33492421

RESUMO

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.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus Tipo 2/genética , Homeostase/genética , Células Secretoras de Insulina/metabolismo , Proteínas Nucleares/genética , Fatores de Transcrição/genética , Animais , Biomarcadores/sangue , Glicemia/genética , Feminino , Hormônio Foliculoestimulante/sangue , Genótipo , Humanos , Insulina/sangue , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Hipófise/metabolismo , Caracteres Sexuais , Aumento de Peso , Peixe-Zebra/sangue , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/sangue , Proteínas de Peixe-Zebra/genética
3.
Nat Commun ; 14(1): 3076, 2023 05 29.
Artigo em Inglês | MEDLINE | ID: mdl-37248237

RESUMO

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.


Assuntos
Receptor Nuclear Órfão DAX-1 , Receptor alfa de Estrogênio , Hipotálamo , Kisspeptinas , Animais , Feminino , Camundongos , Núcleo Arqueado do Hipotálamo/metabolismo , Estradiol/metabolismo , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Estrogênios/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/genética , Kisspeptinas/metabolismo , Receptor Nuclear Órfão DAX-1/genética , Receptor Nuclear Órfão DAX-1/metabolismo
4.
Cell Rep Med ; 3(11): 100810, 2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36384093

RESUMO

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.


Assuntos
Glucagon , Redução de Peso , Camundongos , Animais , Glucagon/metabolismo , Metabolismo Energético/fisiologia , Receptores de Glucagon/metabolismo , Camundongos Obesos , Aminoácidos/farmacologia
5.
Hepatology ; 52(4): 1341-9, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20842631

RESUMO

UNLABELLED: Pregnancy alters bile acid homeostasis and can unmask cholestatic disease in genetically predisposed but otherwise asymptomatic individuals. In this report, we show that normal pregnant mice have raised hepatic bile acid levels in the presence of procholestatic gene expression. The nuclear receptor farnesoid X receptor (FXR) regulates the transcription of the majority of these genes, and we show that both ablation and activation of Fxr prevent the accumulation of hepatic bile acids during pregnancy. These observations suggest that the function of Fxr may be perturbed during gestation. In subsequent in vitro experiments, serum from pregnant mice and humans was found to repress expression of the Fxr target gene, small heterodimer partner (Shp), in liver-derived Fao cells. Estradiol or estradiol metabolites may contribute to this effect because coincubation with the estrogen receptor (ER) antagonist fulvestrant (ICI 182780) abolished the repressive effects on Shp expression. Finally, we report that ERα interacts with FXR in an estradiol-dependent manner and represses its function in vitro. CONCLUSION: Ligand-activated ERα may inhibit FXR function during pregnancy and result in procholestatic gene expression and raised hepatic bile acid levels. We propose that this could cause intrahepatic cholestasis of pregnancy in genetically predisposed individuals.


Assuntos
Ácidos e Sais Biliares/metabolismo , Fígado/metabolismo , Prenhez/fisiologia , Receptores Citoplasmáticos e Nucleares/fisiologia , Animais , Estradiol/análogos & derivados , Estradiol/farmacologia , Receptor alfa de Estrogênio/antagonistas & inibidores , Receptor alfa de Estrogênio/metabolismo , Feminino , Fulvestranto , Perfilação da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Gravidez , Prenhez/sangue , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/biossíntese
6.
Sci Rep ; 11(1): 22577, 2021 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-34799628

RESUMO

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.


Assuntos
Fármacos Antiobesidade/farmacologia , Glucagon/farmacologia , Hepatócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Receptores de Glucagon/agonistas , Redução de Peso/efeitos dos fármacos , Animais , Fármacos Antiobesidade/farmacocinética , Variação Biológica da População , Relação Dose-Resposta a Droga , Feminino , Glucagon/análogos & derivados , Glucagon/farmacocinética , Células HEK293 , Hepatócitos/metabolismo , Humanos , Fígado/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ratos Wistar , Receptores de Glucagon/genética , Receptores de Glucagon/metabolismo , Transdução de Sinais
7.
Psychoneuroendocrinology ; 129: 105244, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33975151

RESUMO

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.


Assuntos
Encéfalo , Kisspeptinas , Ácido gama-Aminobutírico , Encéfalo/metabolismo , Humanos , Kisspeptinas/metabolismo , Ácido gama-Aminobutírico/metabolismo
8.
J Clin Endocrinol Metab ; 106(4): e1837-e1848, 2021 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-33075807

RESUMO

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.


Assuntos
Apetite/efeitos dos fármacos , Encéfalo/efeitos dos fármacos , Kisspeptinas/farmacologia , Adulto , Encéfalo/diagnóstico por imagem , Encéfalo/fisiologia , Estudos Cross-Over , Método Duplo-Cego , Alimentos , Voluntários Saudáveis , Humanos , Infusões Intravenosas , Kisspeptinas/administração & dosagem , Imageamento por Ressonância Magnética , Masculino , Rede Nervosa/diagnóstico por imagem , Rede Nervosa/efeitos dos fármacos , Estimulação Luminosa , Psicometria , Recompensa , Reino Unido
9.
Mol Metab ; 51: 101242, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-33933675

RESUMO

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.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Hipoglicemiantes/farmacologia , Peptídeos/farmacologia , Receptores de Glucagon/agonistas , Animais , Glicemia/análise , Glicemia/efeitos dos fármacos , Células Cultivadas , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/diagnóstico , Modelos Animais de Doenças , Células HEK293 , Hepatócitos , Humanos , Hipoglicemiantes/uso terapêutico , Ilhotas Pancreáticas , Liraglutida/farmacologia , Liraglutida/uso terapêutico , Masculino , Camundongos , Oxintomodulina/genética , Peptídeos/genética , Peptídeos/uso terapêutico , Cultura Primária de Células , Ratos , Redução de Peso/efeitos dos fármacos , beta-Arrestina 2/metabolismo
10.
Am J Physiol Gastrointest Liver Physiol ; 298(2): G151-8, 2010 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19815629

RESUMO

Rodents undergo gestational hepatomegaly to meet the increased metabolic demands on the maternal liver during pregnancy. This is an important physiological process, but the mechanisms and signals driving pregnancy-induced liver growth are not known. Here, we show that liver growth during pregnancy precedes maternal body weight gain, is proportional to fetal number, and is a result of hepatocyte hypertrophy associated with cell-cycle progression, polyploidy, and altered expression of cell-cycle regulators p53, Cyclin-D1, and p27. Because circulating reproductive hormones and bile acids are raised in normal pregnant women and can cause liver growth in rodents, these compounds are candidates for the signal driving gestational liver enlargement in rodents. Administration of pregnancy levels of reproductive hormones was not sufficient to cause liver growth, but mouse pregnancy was associated with increased serum bile acid levels. It is known that the bile acid sensor Fxr is required for normal recovery from partial hepatectomy, and we demonstrate that Fxr(-/-) mice undergo gestational liver growth by adaptive hepatocyte hyperplasia. This is the first identification of any component that is required to maintain the normal mechanisms of gestational hepatomegaly and also implicates Fxr in a physiologically normal process that involves control of the hepatocyte cell cycle. Understanding pregnancy-induced hepatocyte hypertrophy in mice could suggest mechanisms for safely increasing functional liver capacity in women during increased metabolic demand.


Assuntos
Regeneração Hepática/fisiologia , Fígado/fisiologia , Prenhez/fisiologia , Receptores Citoplasmáticos e Nucleares/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Colatos/metabolismo , Colatos/farmacologia , Receptor Constitutivo de Androstano , Ciclina D1/genética , Ciclina D1/metabolismo , Metabolismo Energético/fisiologia , Feminino , Hepatócitos/citologia , Hepatócitos/efeitos dos fármacos , Hepatócitos/fisiologia , Fígado/citologia , Fígado/crescimento & desenvolvimento , Camundongos , Camundongos Mutantes , Ovariectomia , Poliploidia , Gravidez , Fase S/efeitos dos fármacos , Fase S/fisiologia
11.
Drug Metab Dispos ; 38(1): 143-9, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19797606

RESUMO

The intestinal-derived secondary bile acid (BA) lithocholic acid (LCA) is hepatotoxic and is implicated in the pathogenesis of cholestatic diseases. LCA is an endogenous ligand of the xenobiotic nuclear receptor pregnane X receptor (PXR), but there is currently no consensus on the respective roles of hepatic and intestinal PXR in mediating protection against LCA in vivo. Under the conditions reported here, we show that mice lacking Pxr are resistant to LCA-mediated hepatotoxicity. This unexpected phenotype is found in association with enhanced urinary BA excretion and elevated basal expression of drug metabolism enzymes and the hepatic sulfate donor synthesis enzyme Papss2 in Pxr(-/-) mice. By subsequently comparing molecular responses to dietary and intraperitoneal administration of LCA, we made two other significant observations: 1) LCA feeding induces intestinal, but not hepatic, drug-metabolizing enzymes in a largely Pxr-independent manner; and 2) in contrast to LCA feeding, bypassing first-pass gut transit by intraperitoneal administration of LCA did induce hepatic detoxification machinery and in a Pxr-dependent manner. These data reconcile important discrepancies in the reported molecular responses to this BA and suggest that Pxr plays only a limited role in mediating responses to gut-derived LCA. Furthermore, the route of administration must be considered in the future planning and interpretation of experiments designed to assess hepatic responses to BAs, orally administered pharmaceuticals, and dietary toxins.


Assuntos
Inativação Metabólica/fisiologia , Mucosa Intestinal/metabolismo , Ácido Litocólico/farmacocinética , Fígado/metabolismo , Receptores de Esteroides/metabolismo , Administração Oral , Alanina Transaminase/sangue , Animais , Hidrocarboneto de Aril Hidroxilases/genética , Ácidos e Sais Biliares/sangue , Ácidos e Sais Biliares/metabolismo , Ácidos e Sais Biliares/urina , Peso Corporal/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/sangue , Doença Hepática Induzida por Substâncias e Drogas/patologia , Citocromo P-450 CYP3A/genética , Sistema Enzimático do Citocromo P-450/genética , Sistema Enzimático do Citocromo P-450/metabolismo , Família 2 do Citocromo P450 , Indução Enzimática/efeitos dos fármacos , Feminino , Expressão Gênica/efeitos dos fármacos , Injeções Intraperitoneais , Intestinos/efeitos dos fármacos , Ácido Litocólico/administração & dosagem , Ácido Litocólico/farmacologia , Fígado/efeitos dos fármacos , Fígado/patologia , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana Transportadoras/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexos Multienzimáticos/genética , Receptor de Pregnano X , Esteroide Hidroxilases/genética , Sulfato Adenililtransferase/genética , Sulfotransferases/genética
12.
Sci Rep ; 10(1): 1703, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-32015477

RESUMO

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.


Assuntos
Receptor Nuclear Órfão DAX-1/genética , Transtornos do Desenvolvimento Sexual/genética , Processos de Determinação Sexual/fisiologia , Proteína da Região Y Determinante do Sexo/genética , Testículo/fisiologia , Animais , Variação Biológica Individual , Feminino , Fertilidade , Patrimônio Genético , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Parto , Fenótipo
13.
JCI Insight ; 5(3)2020 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-32051344

RESUMO

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.


Assuntos
Encéfalo/fisiologia , Sinais (Psicologia) , Kisspeptinas/fisiologia , Comportamento Sexual/fisiologia , Olfato/fisiologia , Visão Ocular/fisiologia , Adulto , Estudos Cross-Over , Método Duplo-Cego , Feminino , Humanos , Kisspeptinas/metabolismo , Masculino , Placebos , Qualidade de Vida , Disfunções Sexuais Psicogênicas/fisiopatologia , Transdução de Sinais
14.
J Clin Invest ; 130(12): 6739-6753, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33196464

RESUMO

BACKGROUNDKisspeptin is a key regulator of hypothalamic gonadotropin-releasing hormone (GnRH) neurons and is essential for reproductive health. A specific kisspeptin receptor (KISS1R) agonist could significantly expand the potential clinical utility of therapeutics targeting the kisspeptin pathway. Herein, we investigate the effects of a KISS1R agonist, MVT-602, in healthy women and in women with reproductive disorders.METHODSWe conducted in vivo and in vitro studies to characterize the action of MVT-602 in comparison with native kisspeptin-54 (KP54). We determined the pharmacokinetic and pharmacodynamic properties of MVT-602 (doses 0.01 and 0.03 nmol/kg) versus KP54 (9.6 nmol/kg) in the follicular phase of healthy women (n = 9), and in women with polycystic ovary syndrome (PCOS; n = 6) or hypothalamic amenorrhea (HA; n = 6). Further, we investigated their effects on KISS1R-mediated inositol monophosphate (IP1) and Ca2+ signaling in cell lines and on action potential firing of GnRH neurons in brain slices.RESULTSIn healthy women, the amplitude of luteinizing hormone (LH) rise was similar to that after KP54, but peaked later (21.4 vs. 4.7 hours; P = 0.0002), with correspondingly increased AUC of LH exposure (169.0 vs. 38.5 IU∙h/L; P = 0.0058). LH increases following MVT-602 were similar in PCOS and healthy women, but advanced in HA (P = 0.004). In keeping with the clinical data, MVT-602 induced more potent signaling of KISS1R-mediated IP1 accumulation and a longer duration of GnRH neuron firing than KP54 (115 vs. 55 minutes; P = 0.0012).CONCLUSIONTaken together, these clinical and mechanistic data identify MVT-602 as having considerable therapeutic potential for the treatment of female reproductive disorders.TRIAL REGISTRATIONInternational Standard Randomised Controlled Trial Number (ISRCTN) Registry, ISRCTN21681316.FUNDINGNational Institute for Health Research and NIH.


Assuntos
Amenorreia , Sinalização do Cálcio/efeitos dos fármacos , Kisspeptinas/administração & dosagem , Fragmentos de Peptídeos/administração & dosagem , Síndrome do Ovário Policístico , Receptores de Kisspeptina-1/agonistas , Adolescente , Adulto , Amenorreia/sangue , Amenorreia/tratamento farmacológico , Amenorreia/patologia , Linhagem Celular , Feminino , Humanos , Hipotálamo/metabolismo , Hipotálamo/patologia , Hormônio Luteinizante/sangue , Síndrome do Ovário Policístico/sangue , Síndrome do Ovário Policístico/tratamento farmacológico , Síndrome do Ovário Policístico/patologia , Receptores de Kisspeptina-1/metabolismo
15.
Mol Metab ; 30: 221-229, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31767173

RESUMO

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.


Assuntos
Fígado/metabolismo , PPAR alfa/metabolismo , Esteroide 17-alfa-Hidroxilase/metabolismo , Animais , Ácidos e Sais Biliares/metabolismo , Glucose/metabolismo , Células HEK293 , Hepatócitos/metabolismo , Homeostase , Humanos , Cetonas/metabolismo , Lipogênese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Oxirredução , Receptores Citoplasmáticos e Nucleares , Transdução de Sinais , Esteroide 17-alfa-Hidroxilase/fisiologia
16.
Nat Commun ; 9(1): 1602, 2018 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-29686402

RESUMO

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.


Assuntos
Diabetes Mellitus Tipo 2/tratamento farmacológico , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Hipoglicemiantes/farmacologia , Insulina/metabolismo , Animais , Glicemia/efeitos dos fármacos , Células CHO , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Cricetulus , Diabetes Mellitus Experimental , Diabetes Mellitus Tipo 2/sangue , Diabetes Mellitus Tipo 2/patologia , Endocitose/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Células HEK293 , Humanos , Hipoglicemiantes/uso terapêutico , Insulina/genética , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Náusea/induzido quimicamente , Náusea/epidemiologia , Cultura Primária de Células , Transporte Proteico/efeitos dos fármacos , RNA Interferente Pequeno/metabolismo , Resultado do Tratamento
17.
Mol Endocrinol ; 30(4): 469-78, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26974035

RESUMO

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.


Assuntos
Citocromos b5/genética , Células Intersticiais do Testículo/enzimologia , Esteroide 17-alfa-Hidroxilase/metabolismo , 17-alfa-Hidroxiprogesterona/sangue , Animais , Citocromos b5/metabolismo , Feminino , Fertilidade , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo
18.
Cell Metab ; 23(3): 427-40, 2016 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-26959184

RESUMO

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.


Assuntos
Fármacos Antiobesidade/farmacologia , Anticorpos Monoclonais Humanizados/farmacologia , Diabetes Mellitus Tipo 2/tratamento farmacológico , Fatores de Crescimento de Fibroblastos/farmacologia , Obesidade/tratamento farmacológico , Adolescente , Adulto , Idoso , Animais , Fármacos Antiobesidade/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Glicemia , Peso Corporal/efeitos dos fármacos , Diabetes Mellitus Tipo 2/sangue , Avaliação Pré-Clínica de Medicamentos , Feminino , Fatores de Crescimento de Fibroblastos/uso terapêutico , Expressão Gênica/efeitos dos fármacos , Humanos , Insulina/sangue , Metabolismo dos Lipídeos/efeitos dos fármacos , Macaca fascicularis , Masculino , Pessoa de Meia-Idade , Obesidade/sangue , Gordura Subcutânea/efeitos dos fármacos , Gordura Subcutânea/metabolismo , Redução de Peso , Adulto Jovem
19.
Cell Metab ; 23(2): 344-9, 2016 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-26724861

RESUMO

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.


Assuntos
Álcoois/farmacologia , Fatores de Crescimento de Fibroblastos/metabolismo , Preferências Alimentares/efeitos dos fármacos , Paladar/efeitos dos fármacos , Animais , Sistema Nervoso Central/efeitos dos fármacos , Sistema Nervoso Central/metabolismo , Dopamina/metabolismo , Haplorrinos , Humanos , Masculino , Camundongos Endogâmicos C57BL , Sacarina/farmacologia , Transdução de Sinais/efeitos dos fármacos
20.
Curr Opin Pharmacol ; 25: 30-5, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26531325

RESUMO

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.


Assuntos
Fator 1 de Crescimento de Fibroblastos/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Metabolismo , Animais , Fator 1 de Crescimento de Fibroblastos/farmacologia , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/farmacologia , Homeostase/efeitos dos fármacos , Humanos , Metabolismo/efeitos dos fármacos , Modelos Biológicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA