Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Med ; 24(9): 1395-1406, 2018 09.
Article in English | MEDLINE | ID: mdl-30150719

ABSTRACT

Metformin is a first-line drug for the treatment of individuals with type 2 diabetes, yet its precise mechanism of action remains unclear. Metformin exerts its antihyperglycemic action primarily through lowering hepatic glucose production (HGP). This suppression is thought to be mediated through inhibition of mitochondrial respiratory complex I, and thus elevation of 5'-adenosine monophosphate (AMP) levels and the activation of AMP-activated protein kinase (AMPK), though this proposition has been challenged given results in mice lacking hepatic AMPK. Here we report that the AMP-inhibited enzyme fructose-1,6-bisphosphatase-1 (FBP1), a rate-controlling enzyme in gluconeogenesis, functions as a major contributor to the therapeutic action of metformin. We identified a point mutation in FBP1 that renders it insensitive to AMP while sparing regulation by fructose-2,6-bisphosphate (F-2,6-P2), and knock-in (KI) of this mutant in mice significantly reduces their response to metformin treatment. We observe this during a metformin tolerance test and in a metformin-euglycemic clamp that we have developed. The antihyperglycemic effect of metformin in high-fat diet-fed diabetic FBP1-KI mice was also significantly blunted compared to wild-type controls. Collectively, we show a new mechanism of action for metformin and provide further evidence that molecular targeting of FBP1 can have antihyperglycemic effects.


Subject(s)
Fructose-Bisphosphatase/metabolism , Glucose/biosynthesis , Liver/enzymology , Metformin/pharmacology , Adenosine Monophosphate/pharmacology , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/pharmacology , Animals , Base Sequence , Chickens , Disease Models, Animal , Fructose-Bisphosphatase/chemistry , Fructose-Bisphosphatase/genetics , Glucose Intolerance/pathology , Homeostasis/drug effects , Humans , Hypoglycemia/pathology , Liver/drug effects , Mice, Inbred C57BL , Mutation/genetics , Obesity/pathology , Prodrugs/chemistry , Ribonucleotides/pharmacology
2.
Diabetes Obes Metab ; 20(9): 2264-2273, 2018 09.
Article in English | MEDLINE | ID: mdl-29752759

ABSTRACT

AIMS: To test the hypothesis that brown adipose tissue (BAT) is a metformin target tissue by investigating in vivo uptake of [11 C]-metformin tracer in mice and studying in vitro effects of metformin on cultured human brown adipocytes. MATERIALS AND METHODS: Tissue-specific uptake of metformin was assessed in mice by PET/CT imaging after injection of [11 C]-metformin. Human brown adipose tissue was obtained from elective neck surgery and metformin transporter expression levels in human and murine BAT were determined by qPCR. Oxygen consumption in metformin-treated human brown adipocyte cell models was assessed by Seahorse XF technology. RESULTS: Injected [11 C]-metformin showed avid uptake in the murine interscapular BAT depot. Metformin exposure in BAT was similar to hepatic exposure. Non-specific inhibition of the organic cation transporter (OCT) protein by cimetidine administration eliminated BAT exposure to metformin, demonstrating OCT-mediated uptake. Gene expression profiles of OCTs in BAT revealed ample OCT3 expression in both human and mouse BAT. Incubation of a human brown adipocyte cell models with metformin reduced cellular oxygen consumption in a dose-dependent manner. CONCLUSION: These results support BAT as a putative metformin target.


Subject(s)
Adipose Tissue, Brown/drug effects , Hypoglycemic Agents/pharmacokinetics , Metformin/pharmacokinetics , Oxygen Consumption/drug effects , Animals , Cimetidine/administration & dosage , Dose-Response Relationship, Drug , Humans , Mice , Octamer Transcription Factor-3/metabolism , Organic Cation Transport Proteins/metabolism , Positron Emission Tomography Computed Tomography , Transcriptome
3.
Diabetes ; 65(6): 1724-30, 2016 06.
Article in English | MEDLINE | ID: mdl-26993065

ABSTRACT

Metformin is the most commonly prescribed oral antidiabetic drug, with well-documented beneficial preventive effects on diabetic complications. Despite being in clinical use for almost 60 years, the underlying mechanisms for metformin action remain elusive. Organic cation transporters (OCT), including multidrug and toxin extrusion proteins (MATE), are essential for transport of metformin across membranes, but tissue-specific activity of these transporters in vivo is incompletely understood. Here, we use dynamic positron emission tomography with [(11)C]-labeled metformin ([(11)C]-metformin) in mice to investigate the role of OCT and MATE in a well-established target tissue, the liver, and a putative target of metformin, the small intestine. Ablation of OCT1 and OCT2 significantly reduced the distribution of metformin in the liver and small intestine. In contrast, inhibition of MATE1 with pyrimethamine caused accumulation of metformin in the liver but did not affect distribution in the small intestine. The demonstration of OCT-mediated transport into the small intestine provides evidence of direct effects of metformin in this tissue. OCT and MATE have important but separate roles in uptake and elimination of metformin in the liver, but this is not due to changes in biliary secretion. [(11)C]-Metformin holds great potential as a tool to determine the pharmacokinetic properties of metformin in clinical studies.


Subject(s)
Hypoglycemic Agents/pharmacokinetics , Intestine, Small/metabolism , Liver/metabolism , Metformin/pharmacokinetics , Animals , Biological Transport , Mice , Organic Cation Transport Proteins/metabolism , Positron-Emission Tomography/methods
4.
Drug Discov Today ; 19(7): 999-1002, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24637044

ABSTRACT

Regular physical exercise has undisputed health benefits in the prevention and the treatment of many diseases. Understanding the mechanisms that regulate adaptations to exercise training therefore has obvious clinical perspectives. Several lines of evidence suggest that the AMP-activated protein kinase (AMPK) has a central role as a master metabolic regulator in skeletal muscle. Exercise is a potent activator of AMPK, and AMPK signaling can play a key part in regulating protein turnover during and after exercise training.


Subject(s)
Adaptation, Physiological/physiology , Adenylate Kinase/physiology , Exercise/physiology , Muscle, Skeletal/enzymology , Animals , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...