Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Elife ; 122023 01 17.
Article in English | MEDLINE | ID: mdl-36648330

ABSTRACT

Analogues of the hepatokine fibroblast growth factor 21 (FGF21) are in clinical development for type 2 diabetes and nonalcoholic steatohepatitis (NASH) treatment. Although their glucose-lowering and insulin-sensitizing effects have been largely unraveled, the mechanisms by which they alleviate liver injury have only been scarcely addressed. Here, we aimed to unveil the mechanisms underlying the protective effects of FGF21 on NASH using APOE*3-Leiden.CETP mice, a well-established model for human-like metabolic diseases. Liver-specific FGF21 overexpression was achieved in mice, followed by administration of a high-fat high-cholesterol diet for 23 weeks. FGF21 prevented hepatic lipotoxicity, accompanied by activation of thermogenic tissues and attenuation of adipose tissue inflammation, improvement of hyperglycemia and hypertriglyceridemia, and upregulation of hepatic programs involved in fatty acid oxidation and cholesterol removal. Furthermore, FGF21 inhibited hepatic inflammation, as evidenced by reduced Kupffer cell (KC) activation, diminished monocyte infiltration, and lowered accumulation of monocyte-derived macrophages. Moreover, FGF21 decreased lipid- and scar-associated macrophages, which correlated with less hepatic fibrosis as demonstrated by reduced collagen accumulation. Collectively, hepatic FGF21 overexpression limits hepatic lipotoxicity, inflammation, and fibrogenesis. Mechanistically, FGF21 blocks hepatic lipid influx and accumulation through combined endocrine and autocrine signaling, respectively, which prevents KC activation and lowers the presence of lipid- and scar-associated macrophages to inhibit fibrogenesis.


High-calorie modern diets have contributed to growing rates of obesity-linked diseases. One such disease is non-alcoholic steatohepatitis or NASH for short, which affects about 5% of adults in the United States. The livers of people with this condition accumulate fat, become inflamed, and develop scar tissue. People with NASH are also at increased risk of developing liver cancer, type 2 diabetes, and heart disease. Currently, no drugs are available to treat the condition and prevent such severe complications. Previous research has shown the liver produces a stress hormone, called FGF21, in response to fat accumulation. This hormone boosts fat burning and so helps to reduce excess fat in the liver. Drugs that mimic FGF21 have already been developed for type 2 diabetes. But so far, it was unclear if such drugs could also help reduce liver inflammation and scarring in patients with NASH. Liu et al. show that increasing the production of FGF21 in mice with a NASH-like condition reduces fat accumulation, liver inflammation, and scarring. In the experiments, the researchers used gene therapy to ramp up FGF21 production in the livers of mice that develop obesity and a NASH-like condition when fed a high-fat diet for 23 weeks. Increasing FGF21 production prevented the mice from developing obesity while on the high fat diet by making the body burn more fat in the liver and brown fat tissue. The treatment also reduced inflammation and prevented scarring by reducing the number and activity of immune cells in the liver. Increasing the production of the stress hormone FGF21 prevents diet-induced obesity and NASH in mice fed a high-fat diet. More studies are necessary to determine if using gene therapy to increase FGF21 may also cause weight loss and could reverse liver damage in mice that already have NASH. If this approach is effective in mice, it may be tested in humans, a process that may take several years. If human studies are successful, FGF21-boosting therapy might provide a new treatment approach for obesity or NASH.


Subject(s)
Diabetes Mellitus, Type 2 , Non-alcoholic Fatty Liver Disease , Mice , Humans , Animals , Non-alcoholic Fatty Liver Disease/prevention & control , Non-alcoholic Fatty Liver Disease/metabolism , Diabetes Mellitus, Type 2/metabolism , Macrophage Activation , Cicatrix/pathology , Liver/metabolism , Inflammation/pathology , Diet, High-Fat , Cholesterol/metabolism , Lipids , Mice, Inbred C57BL , Disease Models, Animal
2.
Sci Transl Med ; 14(668): eabh1316, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36288279

ABSTRACT

Circadian rhythms play a critical role in regulating metabolism, including daily cycles of feeding/fasting. Glucokinase (GCK) is central for whole-body glucose homeostasis and oscillates according to a circadian clock. GCK activators (GKAs) effectively reduce hyperglycemia, but their use is also associated with hypoglycemia, hyperlipidemia, and hepatic steatosis. Given the circadian rhythmicity and natural postprandial activation of GCK, we hypothesized that GKA treatment would benefit from being timed specifically during feeding periods. Acute treatment of obese Zucker rats with the GKA AZD1656 robustly increased flux into all major metabolic pathways of glucose disposal, enhancing glucose elimination. Four weeks of continuous AZD1656 treatment of obese Zucker rats improved glycemic control; however, hepatic steatosis and inflammation manifested. In contrast, timing AZD1656 to feeding periods robustly reduced hepatic steatosis and inflammation in addition to improving glycemia, whereas treatment timed to fasting periods caused overall detrimental metabolic effects. Mechanistically, timing AZD1656 to feeding periods diverted newly synthesized lipid toward direct VLDL secretion rather than intrahepatic storage. In line with increased hepatic insulin signaling, timing AZD1656 to feeding resulted in robust activation of AKT, mTOR, and SREBP-1C after glucose loading, pathways known to regulate VLDL secretion and hepatic de novo lipogenesis. In conclusion, intermittent AZD1656 treatment timed to feeding periods promotes glucose disposal when needed the most, restores metabolic flexibility and hepatic insulin sensitivity, and thereby avoids hepatic steatosis. Thus, chronotherapeutic approaches may benefit the development of GKAs and other drugs acting on metabolic targets.


Subject(s)
Fatty Liver , Glucokinase , Rats , Animals , Rats, Zucker , Glucokinase/metabolism , Hypoglycemic Agents/therapeutic use , Sterol Regulatory Element Binding Protein 1/metabolism , Sterol Regulatory Element Binding Protein 1/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Insulin/pharmacology , Glucose/metabolism , Obesity/drug therapy , Obesity/metabolism , Liver/metabolism , Chronotherapy , Inflammation/metabolism , TOR Serine-Threonine Kinases/metabolism , Lipids
3.
Mol Ther Nucleic Acids ; 28: 500-513, 2022 Jun 14.
Article in English | MEDLINE | ID: mdl-35592498

ABSTRACT

Fibroblast growth factor 21 (FGF21) is a promising therapeutic agent for treatment of type 2 diabetes (T2D) and non-alcoholic steatohepatitis (NASH). We show that therapeutic levels of FGF21 were achieved following subcutaneous (s.c.) administration of mRNA encoding human FGF21 proteins. The efficacy of mRNA was assessed following 2-weeks repeated s.c. dosing in diet-induced obese (DIO), mice which resulted in marked decreases in body weight, plasma insulin levels, and hepatic steatosis. Pharmacokinetic/pharmacodynamic (PK/PD) modelling of several studies in both lean and DIO mice showed that mRNA encoding human proteins provided improved therapeutic coverage over recombinant dosed proteins in vivo. This study is the first example of s.c. mRNA therapy showing pre-clinical efficacy in a disease-relevant model, thus, showing the potential for this modality in the treatment of chronic diseases, including T2D and NASH.

4.
J Lipid Res ; 63(3): 100176, 2022 03.
Article in English | MEDLINE | ID: mdl-35120993

ABSTRACT

Sodium-glucose cotransporter 2 (SGLT2) inhibitors have been shown to increase ketone bodies in patients with type 2 diabetes; however, the underlying mechanisms have not been fully elucidated. Here we examined the effect of the SGLT2 inhibitor dapagliflozin (1 mg/kg/day, formulated in a water, PEG400, ethanol, propylene glycol solution, 4 weeks) on lipid metabolism in obese Zucker rats. Fasting FFA metabolism was assessed in the anesthetized state using a [9,10-3H(N)]-palmitic acid tracer by estimating rates of plasma FFA appearance (Ra), whole-body FFA oxidation (Rox), and nonoxidative disposal (Rst). In the liver, clearance (Kß-ox) and flux (Rß-ox) of FFA into ß-oxidation were estimated using [9,10-3H]-(R)-bromopalmitate/[U-14C]palmitate tracers. As expected, dapagliflozin induced glycosuria and a robust antidiabetic effect; treatment reduced fasting plasma glucose and insulin, lowered glycated hemoglobin, and increased pancreatic insulin content compared with vehicle controls. Dapagliflozin also increased plasma FFA, Ra, Rox, and Rst with enhanced channeling toward oxidation versus storage. In the liver, there was also enhanced channeling of FFA to ß-oxidation, with increased Kß-ox, Rß-ox and tissue acetyl-CoA, compared with controls. Finally, dapagliflozin increased hepatic HMG-CoA and plasma ß-hydroxybutyrate, consistent with a specific enhancement of ketogenesis. Since ketogenesis has not been directly measured, we cannot exclude an additional contribution of impaired ketone body clearance to the ketosis. In conclusion, this study provides evidence that the dapagliflozin-induced increase in plasma ketone bodies is driven by the combined action of FFA mobilization from adipose tissue and diversion of hepatic FFA toward ß-oxidation.


Subject(s)
Diabetes Mellitus, Type 2 , Ketosis , Sodium-Glucose Transporter 2 Inhibitors , Animals , Benzhydryl Compounds , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/metabolism , Fatty Acids, Nonesterified , Glucosides , Humans , Insulin/metabolism , Ketone Bodies/metabolism , Ketosis/chemically induced , Ketosis/metabolism , Liver/metabolism , Rats , Rats, Zucker , Sodium-Glucose Transporter 2 Inhibitors/adverse effects , Sodium-Glucose Transporter 2 Inhibitors/metabolism
5.
Cardiovasc Res ; 118(2): 489-502, 2022 01 29.
Article in English | MEDLINE | ID: mdl-33693480

ABSTRACT

AIMS: Fibroblast growth factor (FGF) 21, a key regulator of energy metabolism, is currently evaluated in humans for treatment of type 2 diabetes and non-alcoholic steatohepatitis. However, the effects of FGF21 on cardiovascular benefit, particularly on lipoprotein metabolism in relation to atherogenesis, remain elusive. METHODS AND RESULTS: Here, the role of FGF21 in lipoprotein metabolism in relation to atherosclerosis development was investigated by pharmacological administration of a half-life extended recombinant FGF21 protein to hypercholesterolaemic APOE*3-Leiden.CETP mice, a well-established model mimicking atherosclerosis initiation and development in humans. FGF21 reduced plasma total cholesterol, explained by a reduction in non-HDL-cholesterol. Mechanistically, FGF21 promoted brown adipose tissue (BAT) activation and white adipose tissue (WAT) browning, thereby enhancing the selective uptake of fatty acids from triglyceride-rich lipoproteins into BAT and into browned WAT, consequently accelerating the clearance of the cholesterol-enriched remnants by the liver. In addition, FGF21 reduced body fat, ameliorated glucose tolerance and markedly reduced hepatic steatosis, related to up-regulated hepatic expression of genes involved in fatty acid oxidation and increased hepatic VLDL-triglyceride secretion. Ultimately, FGF21 largely decreased atherosclerotic lesion area, which was mainly explained by the reduction in non-HDL-cholesterol as shown by linear regression analysis, decreased lesion severity, and increased atherosclerotic plaque stability index. CONCLUSION: FGF21 improves hypercholesterolaemia by accelerating triglyceride-rich lipoprotein turnover as a result of activating BAT and browning of WAT, thereby reducing atherosclerotic lesion severity and increasing atherosclerotic lesion stability index. We have thus provided additional support for the clinical use of FGF21 in the treatment of atherosclerotic cardiovascular disease.


Subject(s)
Anticholesteremic Agents/pharmacology , Atherosclerosis/prevention & control , Cholesterol/blood , Fibroblast Growth Factors/pharmacology , Hypercholesterolemia/drug therapy , Plaque, Atherosclerotic , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Adipose Tissue, Brown/pathology , Adipose Tissue, White/drug effects , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Adiposity/drug effects , Animals , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Atherosclerosis/blood , Atherosclerosis/genetics , Atherosclerosis/pathology , Biomarkers/blood , Disease Models, Animal , Energy Metabolism/drug effects , Hypercholesterolemia/blood , Hypercholesterolemia/genetics , Hypercholesterolemia/pathology , Lipid Metabolism/drug effects , Lipoproteins, VLDL/blood , Liver/drug effects , Liver/metabolism , Liver/pathology , Mice, Transgenic , Recombinant Proteins/pharmacology , Triglycerides/blood
6.
STAR Protoc ; 2(3): 100731, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34409309

ABSTRACT

Ketohexokinase (KHK) catalyzes the first step of fructose metabolism. Inhibitors of KHK enzymatic activity are being evaluated in clinical trials for the treatment of non-alcoholic fatty liver disease (NAFLD) and diabetes. Here, we present a luminescence-based protocol to quantify KHK activity. The accuracy of this technique has been validated using knockdown and overexpression of KHK in vivo and in vitro. The specificity of the assay has been verified using 3-O-methyl-D-fructose, a non-metabolizable analog of fructose, heat inactivation of hexokinases, and depletion of potassium. For complete details on the use of this protocol, please refer to Damen et al. (2021).


Subject(s)
Enzyme Assays/methods , Fructokinases/metabolism , Fructose/metabolism , Luminescent Measurements/methods , Animals , Carbohydrate Metabolism , Fructokinases/antagonists & inhibitors , Hepatocytes/enzymology , Hepatocytes/metabolism , Humans , Luminescence , Mice , Non-alcoholic Fatty Liver Disease
7.
J Clin Endocrinol Metab ; 105(11)2020 11 01.
Article in English | MEDLINE | ID: mdl-32830851

ABSTRACT

CONTEXT: Insulin resistance (IR) is a risk factor for type 2 diabetes, diabetic kidney disease, cardiovascular disease and nonalcoholic steatohepatitis. Biliopancreatic diversion (BPD) is the most effective form of bariatric surgery for improving insulin sensitivity. OBJECTIVE: To identify plasma proteins correlating with the early restoration of insulin sensitivity after BPD. DESIGN: Prospective single-center study including 20 insulin-resistant men with morbid obesity scheduled for BPD. Patient characteristics and blood samples were repeatedly collected from baseline up to 4 weeks postsurgery. IR was assessed by homeostatic model assessment for insulin resistance (HOMA-IR), Matsuda Index, and by studying metabolic profiles during meal tolerance tests. Unbiased proteomic analysis was performed to identify plasma proteins altered by BPD. Detailed plasma profiles were made on a selected set of proteins by targeted multiple reaction monitoring mass spectrometry (MRM/MS). Changes in plasma proteome were evaluated in relation to metabolic and inflammatory changes. RESULTS: BPD resulted in improved insulin sensitivity and reduced body weight. Proteomic analysis identified 29 proteins that changed following BPD. Changes in plasma levels of afamin, apolipoprotein A-IV (ApoA4), and apolipoprotein A-II (ApoA2) correlated significantly with changes in IR. CONCLUSION: Circulating levels of afamin, ApoA4, and ApoA2 were associated with and may contribute to the rapid improvement in insulin sensitivity after BPD.


Subject(s)
Biliopancreatic Diversion , Insulin Resistance/physiology , Obesity, Morbid/blood , Adult , Blood Glucose/metabolism , Body Mass Index , Glucose Tolerance Test , Humans , Male , Middle Aged , Obesity, Morbid/surgery , Proteomics
8.
JCI Insight ; 2(19)2017 10 05.
Article in English | MEDLINE | ID: mdl-28978803

ABSTRACT

GPR81 is a receptor for the metabolic intermediate lactate with an established role in regulating adipocyte lipolysis. Potentially novel GPR81 agonists were identified that suppressed fasting plasma free fatty acid levels in rodents and in addition improved insulin sensitivity in mouse models of insulin resistance and diabetes. Unexpectedly, the agonists simultaneously induced hypertension in rodents, including wild-type, but not GPR81-deficient mice. Detailed cardiovascular studies in anesthetized dogs showed that the pressor effect was associated with heterogenous effects on vascular resistance among the measured tissues: increasing in the kidney while remaining unchanged in hindlimb and heart. Studies in rats revealed that the pressor effect could be blocked, and the renal resistance effect at least partially blocked, with pharmacological antagonism of endothelin receptors. In situ hybridization localized GPR81 to the microcirculation, notably afferent arterioles of the kidney. In conclusion, these results provide evidence for a potentially novel role of GPR81 agonism in blood pressure control and regulation of renal vascular resistance including modulation of a known vasoeffector mechanism, the endothelin system. In addition, support is provided for the concept of fatty acid lowering as a means of improving insulin sensitivity.


Subject(s)
Receptors, G-Protein-Coupled/agonists , Vascular Resistance/physiology , Adipocytes/drug effects , Adipocytes/metabolism , Animals , Arterioles/metabolism , Diabetes Mellitus, Experimental/prevention & control , Dogs , Dose-Response Relationship, Drug , Endothelins/physiology , Fatty Acids, Nonesterified/blood , Hypertension/chemically induced , Insulin Resistance , Lipolysis/drug effects , Male , Mice, Obese , Rats, Wistar , Receptors, Endothelin/physiology , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/metabolism , Renal Artery/physiopathology , Vascular Resistance/drug effects
9.
Chem Biol ; 21(11): 1486-96, 2014 Nov 20.
Article in English | MEDLINE | ID: mdl-25459661

ABSTRACT

Inhibition of AMP deaminase (AMPD) holds the potential to elevate intracellular adenosine and AMP levels and, therefore, to augment adenosine signaling and activation of AMP-activated protein kinase (AMPK). To test the latter hypothesis, novel AMPD pan inhibitors were synthesized and explored using a panel of in vitro, ex vivo, and in vivo models focusing on confirming AMPD inhibitory potency and the potential of AMPD inhibition to improve glucose control in vivo. Repeated dosing of selected inhibitors did not improve glucose control in insulin-resistant or diabetic rodent disease models. Mice with genetic deletion of the muscle-specific isoform Ampd1 did not showany favorable metabolic phenotype despite being challenged with high-fat diet feeding. Therefore, these results do not support the development of AMPD inhibitors for the treatment of type 2 diabetes.


Subject(s)
AMP Deaminase/antagonists & inhibitors , Diabetes Mellitus, Experimental/enzymology , Enzyme Inhibitors/chemistry , Obesity/enzymology , Small Molecule Libraries/chemistry , AMP Deaminase/genetics , AMP Deaminase/metabolism , Animals , Blood Glucose/analysis , Cells, Cultured , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Diet, High-Fat , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Female , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/enzymology , Insulin/blood , Insulin Resistance , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Obesity/drug therapy , Obesity/metabolism , Obesity/pathology , Protein Binding , Rats , Rats, Sprague-Dawley , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Small Molecule Libraries/pharmacology , Small Molecule Libraries/therapeutic use
10.
PLoS One ; 9(12): e113328, 2014.
Article in English | MEDLINE | ID: mdl-25486018

ABSTRACT

The current study extends previously reported PPARα agonist WY 14,643 (30 µmol/kg/day for 4 weeks) effects on circulating amino acid concentrations in rats fed a 48% saturated fat diet. Steady-state tracer experiments were used to examine in vivo kinetic mechanisms underlying altered plasma serine, glycine and arginine levels. Urinary urea and creatinine excretion were measured to assess whole-body amino acid catabolism. WY 14,643 treated animals demonstrated reduced efficiency to convert food consumed to body weight gain while liver weight was increased compared to controls. WY 14,643 raised total amino acid concentration (38%), largely explained by glycine, serine and threonine increases. 3H-glycine, 14C-serine and 14C-arginine tracer studies revealed elevated rates of appearance (Ra) for glycine (45.5 ± 5.8 versus 17.4 ± 2.7 µmol/kg/min) and serine (21.0 ± 1.4 versus 12.0 ± 1.0) in WY 14,643 versus control. Arginine was substantially decreased (-62%) in plasma with estimated Ra reduced from 3.1 ± 0.3 to 1.2 ± 0.2 µmol/kg/min in control versus WY 14,643. Nitrogen excretion over 24 hours was unaltered. Hepatic arginase activity was substantially decreased by WY 14,643 treatment. In conclusion, PPARα agonism potently alters metabolism of several specific amino acids in the rat. The changes in circulating levels of serine, glycine and arginine reflected altered fluxes into the plasma rather than changes in clearance or catabolism. This suggests that PPARα has an important role in modulating serine, glycine and arginine de novo synthesis.


Subject(s)
Arginine/blood , Glycine/blood , PPAR alpha/agonists , Pyrimidines/pharmacology , Serine/blood , Animals , Arginase/metabolism , Body Weight , Calorimetry, Indirect , Liver/metabolism , Male , Nitrogen/urine , Pyrimidines/pharmacokinetics , Rats
11.
PPAR Res ; 2013: 305347, 2013.
Article in English | MEDLINE | ID: mdl-24285952

ABSTRACT

METABOLIC FLEXIBILITY WAS ASSESSED IN MALE ZUCKER RATS: lean controls, obese controls, and obese rats treated with the dual peroxisome proliferator activated receptor (PPAR) α/γ agonist, tesaglitazar, 3 µ mol/kg/day for 3 weeks. Whole body glucose disposal rate (R d ) and hepatic glucose output (HGO) were assessed under basal fasting and hyperinsulinemic isoglycemic clamp conditions using [3,(3)H]glucose. Indices of tissue specific glucose utilization (R g ') were measured at basal, physiological, and supraphysiological levels of insulinemia using 2-deoxy-D-[2,6-(3)H]glucose. Finally, whole body and tissue specific FFA and glucose utilization and metabolic fate were evaluated under basal and hyperinsulinemic conditions using a combination of [U-(13)C]glucose, 2-deoxy-D-[U-(14)C]glucose, [U-(14)C]palmitate, and [9,10-(3)H]-(R)-bromopalmitate. Tesaglitazar improved whole body insulin action by greater suppression of HGO and stimulation of R d compared to obese controls. This involved increased insulin stimulation of R g ' in fat and skeletal muscle as well as increased glycogen synthesis. Tesaglitazar dramatically improved insulin mediated suppression of plasma FFA level, whole body turnover (R fa ), and muscle, liver, and fat utilization. At basal insulin levels, tesaglitazar failed to lower HGO or R fa compared to obese controls. In conclusion, the results demonstrate that tesaglitazar has a remarkable ability to improve insulin mediated control of glucose and FFA fluxes in obese Zucker rats.

12.
Dose Response ; 7(3): 247-69, 2009 Apr 08.
Article in English | MEDLINE | ID: mdl-19809543

ABSTRACT

The objective of this investigation was to use a pharmacokinetic (PK)/pharmacodynamic (PD) approach to describe and evaluate a PK model of nicotinic acid (NiAc) in guinea pigs and a PD feedback model of changes in non-esterified fatty acid (NEFA) concentrations in rats following multiple intravenous infusions of NiAc at different rates and durations of inhouse and literature (NEFA after extravascular NiAc dosing) data. Serial arterial blood samples were taken for evaluation of NiAc exposure in guinea pigs and NEFA in rats. The biophase kinetics of NiAc was assumed to impact on NEFA turnover with feedback incorporated via an inhibitory moderator compartment. The response acted linearly on the production of moderator, which then acted inversely on the turnover rate of response. The potency, expressed as the amount of NiAc in the biophase causing a 50 % inhibitory effect (ID(50)), was 6.5 nmol +/- 31 % and the half-life of response (t(1/2, kout)) 2 min +/- 18 %. The half-life of tolerance (t(1/2, ktol)) was 9 min +/- 27 %. The model can be used to provide information about factors that determine the time course of NEFA response following different rates and routes of administration of NiAc or NiAc analogues.

13.
Endocrinology ; 145(6): 2680-6, 2004 Jun.
Article in English | MEDLINE | ID: mdl-14988384

ABSTRACT

IL-6 is produced and released in large amounts from skeletal muscle during prolonged exercise in both mice and humans, but there are few data indicating the biological significance of this. IL-6 exerts metabolic effects such as stimulating energy expenditure and reducing body fat mass. We have now investigated the effects of IL-6 deficiency on exercise endurance and energy expenditure in preobese and obese IL-6-deficient (IL-6(-/-)) mice. Four-month-old preobese and 7-month-old obese IL-6(-/-) male mice backcrossed to C57BL/6 and their littermate controls were exercised on a treadmill, and energy expenditure was measured as oxygen consumption with the use of indirect calorimetry. The preobese IL-6(-/-) mice were significantly leaner than the control mice, whereas the older IL-6(-/-) mice, as expected, had developed obesity. Resting young, but not older, IL-6(-/-) mice had an elevated respiratory exchange ratio (RER), indicating that they oxidize carbohydrates rather than fat for energy utilization. During exercise, the young and older IL-6(-/-) mice had a reduced endurance and a progressive decrease in oxygen consumption compared with control mice. There was no difference in RER in young IL-6(-/-) mice, whereas RER was enhanced in older IL-6(-/-), mice during exercise. In summary, IL-6(-/-) mice have reduced endurance and energy expenditure during exercise, suggesting that IL-6 is necessary for normal exercise capacity.


Subject(s)
Interleukin-6/metabolism , Obesity/physiopathology , Physical Endurance/physiology , Animals , Body Composition , Energy Metabolism , Glycogen/blood , Interleukin-6/deficiency , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity , Obesity/blood , Obesity/metabolism , Pulmonary Gas Exchange
14.
Expert Opin Biol Ther ; 3(7): 1061-70, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14519071

ABSTRACT

Interleukin (IL)-6 is a multifunctional immune-modulating cytokine that has been suggested to have important functions in glucose and lipid metabolism. It is secreted from adipose tissue during resting conditions and from muscle during strenuous exercise. Recently, the authors reported that mice deficient of IL-6 develop mature-onset obesity, which was reversed by IL-6 replacement. The IL-6-deficient mice had increased glucose levels and decreased glucose tolerance, and blood lipids were increased in females. Furthermore, it was found that intracerebroventricular (ICV) IL-6 treatment acutely increased energy expenditure in rats and led to loss of fat mass following prolonged treatment, without causing symptoms of sickness behaviour or increased levels of acute-phase reactants. Thus, these data indicate a role for IL-6 in the regulation of energy homeostasis in rodents. In humans, several single nucleotide polymorphisms in the IL-6 gene promoter are known, one of which (174 C) is associated with reduced IL-6 transcription as well as decreased basal metabolic rate and insulin sensitivity in healthy male subjects. Furthermore, it was found that IL-6 levels in cerebrospinal fluid in obese humans were inversely correlated with more severe obesity, suggesting that severe obesity is coupled to a relative central IL-6 deficiency. Taken together, these data suggest that endogenous IL-6 has antiobesity effects and, therefore, it is possible that low endogenous IL-6 production contributes to obesity in humans.


Subject(s)
Interleukin-6/therapeutic use , Obesity/drug therapy , Adipose Tissue/physiology , Animals , Arteriosclerosis/drug therapy , Body Composition/genetics , Body Composition/physiology , Central Nervous System/drug effects , Central Nervous System/physiology , Clinical Trials as Topic , Humans , Insulin Resistance/physiology , Interleukin-6/administration & dosage , Interleukin-6/genetics , Interleukin-6/physiology , Promoter Regions, Genetic/genetics
15.
J Clin Endocrinol Metab ; 88(9): 4379-83, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12970313

ABSTRACT

Recently, we demonstrated that intracerebroventricular injection of IL-6 increases energy expenditure and decreases body fat in rodents. Therefore, IL-6 may play a role in appetite and body weight control in the central nervous system. In the present study we evaluated cerebrospinal fluid (CSF) and serum IL-6 levels in humans in relation to body fat content and to CSF and serum levels of leptin. Thirty-two healthy overweight/obese male subjects with a body mass index range of 29.3-36.0 kg/m(2) were studied. Total and sc body fat were measured by dual energy x-ray absorptiometry and computed tomography, respectively. CSF IL-6 levels were in some individuals higher than serum IL-6 levels and correlated negatively with total body weight, sc and total body fat. In contrast, CSF leptin levels were 30-60 times lower than serum leptin levels and correlated positively with serum leptin, body weight, sc and total body fat. Furthermore, there was a negative correlation between CSF IL-6 and leptin. In conclusion, CSF IL-6 differs in many ways from CSF leptin. CSF IL-6 may be locally produced rather than serum derived, and body fat-regulating regions in the central nervous system may be exposed to insufficient IL-6 levels in more severe obesity.


Subject(s)
Adipose Tissue/anatomy & histology , Body Composition/physiology , Central Nervous System/metabolism , Interleukin-6/cerebrospinal fluid , Obesity/cerebrospinal fluid , Absorptiometry, Photon , Adult , Body Weight/physiology , Enzyme-Linked Immunosorbent Assay , Humans , Leptin/blood , Leptin/cerebrospinal fluid , Male , Middle Aged , Obesity/pathology , Serum Albumin/metabolism
16.
Growth Horm IGF Res ; 13 Suppl A: S28-32, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12914723

ABSTRACT

We conducted an experimental study examining the site and mechanism of action of the anti-obesity effect of interleukin-6 (IL-6) in mice and rats. We used dual energy X-ray absorptiometry (DEXA) and computerized tomography to investigate the body composition of mice with knockout of the IL-6 gene and wild-type control mice. Rats were treated with IL-6 or vehicle through intracerebroventricular (ICV) cannulae. Energy expenditure was measured as oxygen consumption by indirect calorimetry in metabolic chambers. Results showed that the mice lacking IL-6 increased in body weight compared with wild-type mice from 6 months of age onward, although there was no marked difference in food intake between the pre-obese IL-6 knockout mice and the wild-type mice. IL-6 given as a single ICV injection to rats stimulated oxygen consumption; whereas, the same doses were ineffective when given peripherally. Chronic ICV IL-6 treatment decreased body weight and fat mass in rodents. Administration of IL-6 may decrease fat mass in mice and rats by stimulating energy expenditure at the CNS level, possibly in the hypothalamus.


Subject(s)
Anti-Obesity Agents/pharmacology , Interleukin-6/pharmacology , Adipose Tissue/drug effects , Animals , Blood Glucose/metabolism , Body Weight/drug effects , Brain/drug effects , Energy Metabolism/drug effects , Injections, Intraventricular , Interleukin-6/physiology , Mice , Mice, Knockout , Obesity/drug therapy , Obesity/metabolism , Oxygen Consumption/drug effects , Rats
17.
J Bone Miner Res ; 17(11): 1977-87, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12412805

ABSTRACT

Insulin-like growth factor (IGF) I is an important regulator of both skeletal growth and adult bone metabolism. To better understand the relative importance of systemic IGF-I versus locally expressed IGF-I we have developed a transgenic mouse model with inducible specific IGF-I gene inactivation in the liver (LI-IGF-I-/-). These mice are growing normally up to 12 weeks of age but have a disturbed carbohydrate and lipid metabolism. In this study, the long-term effects of liver-specific IGF-I inactivation on skeletal growth and adult bone metabolism were investigated. The adult (week 8-55) axial skeletal growth was decreased by 24% in the LI-IGF-I-/- mice whereas no major reduction of the adult appendicular skeletal growth was seen. The cortical cross-sectional bone area, as measured in the middiaphyseal region of the long bones, was decreased in old LI-IGF-I-/- mice. This reduction in the amount of cortical bone was caused mainly by decreased periosteal circumference and was associated with a weaker bone determined by a decrease in ultimate load. In contrast, the amount of trabecular bone was not decreased in the LI-IGF-I-/- mice. DNA microarray analysis of 30-week-old LI-IGF-I-/- and control mice indicated that only four genes were regulated in bone whereas approximately 40 genes were regulated in the liver, supporting the hypothesis that liver-derived IGF-I is of minor importance for adult bone metabolism. In summary, liver-derived IGF-I exerts a small but significant effect on cortical periosteal bone growth and on adult axial skeletal growth while it is not required for the maintenance of the trabecular bone in adult mice.


Subject(s)
Bone and Bones/metabolism , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Liver/physiology , Absorptiometry, Photon , Animals , Biomarkers/blood , Bone Density , Bone Development/genetics , Bone Resorption , Gene Expression Regulation, Developmental , Mechanics , Mice , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Organ Specificity
18.
Biochem Biophys Res Commun ; 293(1): 560-5, 2002 Apr 26.
Article in English | MEDLINE | ID: mdl-12054638

ABSTRACT

Recently we found that interleukin-6 (IL-6) knockout mice develop mature-onset obesity and that a single intracerebroventricular (ICV) injection of IL-6 increases energy expenditure. In the present study we investigated if chronic ICV treatment with IL-6 can suppress body fat mass. IL-6 was injected ICV daily for two weeks to rats fed a high-fat diet. IL-6 treatment but not saline treatment decreased body weight by 8.4% and decreased the relative weights of mesenteric and retroperitoneal fat pads. Consistent with this, circulating leptin levels were decreased by 40% after IL-6 treatment but not after saline treatment. Average food intake per day was decreased in the IL-6 treated group compared to the saline treated rats. IL-6 treatment did not change hepatic expression of the acute-phase protein haptoglobin, serum levels of insulin or insulin-like growth factor-I, or the weights of the heart, liver, kidneys, adrenals, and spleen. We conclude that centrally administered IL-6 can decrease body fat in rats without causing acute-phase reaction.


Subject(s)
Adipose Tissue/drug effects , Interleukin-6/pharmacology , Obesity/prevention & control , Animals , Body Weight/drug effects , Energy Intake , Haptoglobins/metabolism , Injections, Intraventricular , Interleukin-6/administration & dosage , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Knockout , Organ Size/drug effects , Rats , Rats, Wistar
19.
Nat Med ; 8(1): 75-9, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11786910

ABSTRACT

The immune-modulating cytokine interleukin-6 (IL-6) is expressed both in adipose tissue and centrally in hypothalamic nuclei that regulate body composition. We investigated the impact of loss of IL-6 on body composition in mice lacking the gene encoding IL-6 (Il6-/- mice) and found that they developed mature-onset obesity that was partly reversed by IL-6 replacement. The obese Il6-/- mice had disturbed carbohydrate and lipid metabolism, increased leptin levels and decreased responsiveness to leptin treatment. To investigate the possible mechanism and site of action of the anti-obesity effect of IL-6, we injected rats centrally and peripherally with IL-6 at low doses. Intracerebroventricular, but not intraperitoneal IL-6 treatment increased energy expenditure. In conclusion, centrally acting IL-6 exerts anti-obesity effects in rodents.


Subject(s)
Interleukin-6/deficiency , Obesity/genetics , Age Factors , Animals , Blood Glucose/analysis , Body Composition , Corticosterone/blood , Eating/physiology , Energy Metabolism/physiology , Female , Glucose Tolerance Test , Injections, Intraventricular , Interleukin-6/genetics , Interleukin-6/pharmacology , Leptin/blood , Lipids/blood , Male , Mice , Mice, Mutant Strains , Obesity/etiology , Sex Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...