Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Prog Neurobiol ; 240: 102652, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38955325

ABSTRACT

Psychotic disorders entail intricate conditions marked by disruptions in cognition, perception, emotions, and social behavior. Notably, psychotic patients who use cannabis tend to show less severe deficits in social behaviors, such as the misinterpretation of social cues and the inability to interact with others. However, the biological underpinnings of this epidemiological interaction remain unclear. Here, we used the NMDA receptor blocker phencyclidine (PCP) to induce psychotic-like states and to study the impact of adolescent cannabinoid exposure on social behavior deficits and synaptic transmission changes in hippocampal area CA2, a region known to be active during social interactions. In particular, adolescent mice underwent 7 days of subchronic treatment with the synthetic cannabinoid, WIN 55, 212-2 (WIN) followed by one injection of PCP. Using behavioral, biochemical, and electrophysiological approaches, we showed that PCP persistently reduced sociability, decreased GAD67 expression in the hippocampus, and induced GABAergic deficits in proximal inputs from CA3 and distal inputs from the entorhinal cortex (EC) to CA2. Notably, WIN exposure during adolescence specifically restores adult sociability deficits, the expression changes in GAD67, and the GABAergic impairments in the EC-CA2 circuit, but not in the CA3-CA2 circuit. Using a chemogenetic approach to target EC-CA2 projections, we demonstrated the involvement of this specific circuit on sociability deficits. Indeed, enhancing EC-CA2 transmission was sufficient to induce sociability deficits in vehicle-treated mice, but not in animals treated with WIN during adolescence, suggesting a mechanism by which adolescent cannabinoid exposure rescues sociability deficits caused by enhanced EC-CA2 activity in adult mice.

2.
Neurobiol Dis ; 176: 105942, 2023 01.
Article in English | MEDLINE | ID: mdl-36473591

ABSTRACT

Cyclin-dependent kinase 5 (CDK5) is a serine/threonine kinase that has emerged as a key regulator of neurotransmission in complex cognitive processes. Its expression is altered in treated schizophrenia patients, and cannabinoids modulate CDK5 levels in the brain of rodents. However, the role of this kinase, and its interaction with cannabis use in first-episode psychosis (FEP) patients is still not known. Hence, we studied the expression changes of CDK5 and its signaling partner, postsynaptic density protein 95 (PSD95) in olfactory neuroepithelial (ON) cells of FEP patients with (FEP/c) and without (FEP/nc) prior cannabis use, and in a dual-hit mouse model of psychosis. In this model, adolescent mice were exposed to the cannabinoid receptor 1 agonist (CB1R) WIN-55,212-2 (WIN: 1 mg/kg) during 21 days, and to the N-methyl-d-aspartate receptor (NMDAR) blocker phencyclidine (PCP: 10 mg/kg) during 10 days. FEP/c showed less social functioning deficits, lower CDK5 and higher PSD95 levels than FEP/nc. These changes correlated with social skills, but not cognitive deficits. Consistently, exposure of ON cells from FEP/nc patients to WIN in vitro reduced CDK5 levels. Convergent results were obtained in mice, where PCP by itself induced more sociability deficits, and PSD95/CDK5 alterations in the prefrontal cortex and hippocampus than exposure to PCP-WIN. In addition, central blockade of CDK5 activity with roscovitine in PCP-treated mice restored both sociability impairments and PSD95 levels. We provide translational evidence that increased CDK5 could be an early indicator of psychosis associated with social deficits, and that this biomarker is modulated by prior cannabis use.


Subject(s)
Cannabinoids , Psychotic Disorders , Schizophrenia , Mice , Animals , Cyclin-Dependent Kinase 5/metabolism , Psychotic Disorders/drug therapy , Phencyclidine/pharmacology , Cannabinoid Receptor Agonists , Disks Large Homolog 4 Protein
3.
Nat Metab ; 2(12): 1427-1442, 2020 12.
Article in English | MEDLINE | ID: mdl-33199895

ABSTRACT

Adipose tissue macrophages (ATMs) display tremendous heterogeneity depending on signals in their local microenvironment and contribute to the pathogenesis of obesity. The phosphoinositide 3-kinase (PI3K) signalling pathway, antagonized by the phosphatase and tensin homologue (PTEN), is important for metabolic responses to obesity. We hypothesized that fluctuations in macrophage-intrinsic PI3K activity via PTEN could alter the trajectory of metabolic disease by driving distinct ATM populations. Using mice harbouring macrophage-specific PTEN deletion or bone marrow chimeras carrying additional PTEN copies, we demonstrate that sustained PI3K activity in macrophages preserves metabolic health in obesity by preventing lipotoxicity. Myeloid PI3K signalling promotes a beneficial ATM population characterized by lipid uptake, catabolism and high expression of the scavenger macrophage receptor with collagenous structure (MARCO). Dual MARCO and myeloid PTEN deficiencies prevent the generation of lipid-buffering ATMs, reversing the beneficial actions of elevated myeloid PI3K activity in metabolic disease. Thus, macrophage-intrinsic PI3K signalling boosts metabolic health by driving ATM programmes associated with MARCO-dependent lipid uptake.


Subject(s)
Adipose Tissue/metabolism , Lipid Metabolism/genetics , Macrophages/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Immunologic/metabolism , Signal Transduction , Adipocytes/pathology , Adipose Tissue/pathology , Animals , Bone Marrow Transplantation , Cell Differentiation , Chimera , Glucose Tolerance Test , Lipidomics , Macrophages/pathology , Metabolic Diseases/metabolism , Mice , Mice, Inbred C57BL , Obesity/metabolism , Obesity/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Receptors, Immunologic/genetics , Signal Transduction/genetics
4.
Am J Physiol Endocrinol Metab ; 319(1): E203-E216, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32516027

ABSTRACT

Studies suggest the gut microbiota contributes to the development of obesity and metabolic syndrome. Exercise alters microbiota composition and diversity and is protective of these maladies. We tested whether the protective metabolic effects of exercise are mediated through fecal components through assessment of body composition and metabolism in recipients of fecal microbiota transplantation (FMT) from exercise-trained (ET) mice fed normal or high-energy diets. Donor C57BL/6J mice were fed a chow or high-fat, high-sucrose diet (HFHS) for 4 wk to induce obesity and glucose intolerance. Mice were divided into sedentary (Sed) or ET groups (6 wk treadmill-based ET) while maintaining their diets, resulting in four donor groups: chow sedentary (NC-Sed) or ET (NC-ET) and HFHS sedentary (HFHS-Sed) or ET (HFHS-ET). Chow-fed recipient mice were gavaged with feces from the respective donor groups weekly, creating four groups (NC-Sed-R, NC-ET-R, HFHS-Sed-R, HFHS-ET-R), and body composition and metabolism were assessed. The HFHS diet led to glucose intolerance and obesity in the donors, whereas exercise training (ET) restrained adiposity and improved glucose tolerance. No donor group FMT altered recipient body composition. Despite unaltered adiposity, glucose levels were disrupted when challenged in mice receiving feces from HFHS-fed donors, irrespective of donor-ET status, with a decrease in insulin-stimulated glucose clearance into white adipose tissue and large intestine and specific changes in the recipient's microbiota composition observed. FMT can transmit HFHS-induced disrupted glucose metabolism to recipient mice independently of any change in adiposity. However, the protective metabolic effect of ET on glucose metabolism is not mediated through fecal factors.


Subject(s)
Diet, High-Fat , Dietary Sucrose , Fecal Microbiota Transplantation , Glucose Intolerance/microbiology , Obesity/microbiology , Physical Conditioning, Animal , Sedentary Behavior , Adiposity , Animals , Gastrointestinal Microbiome , Glucose/metabolism , Glucose Intolerance/metabolism , Male , Mice , Obesity/metabolism , Random Allocation
5.
FASEB J ; 34(4): 5697-5714, 2020 04.
Article in English | MEDLINE | ID: mdl-32141144

ABSTRACT

Type 2 diabetes (T2D) manifests from inadequate glucose control due to insulin resistance, hypoinsulinemia, and deteriorating pancreatic ß-cell function. The pro-inflammatory factor Activin has been implicated as a positive correlate of severity in T2D patients, and as a negative regulator of glucose uptake by skeletal muscle, and of pancreatic ß-cell phenotype in mice. Accordingly, we sought to determine whether intervention with the Activin antagonist Follistatin can ameliorate the diabetic pathology. Here, we report that an intravenous Follistatin gene delivery intervention with tropism for striated muscle reduced the serum concentrations of Activin B and improved glycemic control in the db/db mouse model of T2D. Treatment reversed the hyperglycemic progression with a corresponding reduction in the percentage of glycated-hemoglobin to levels similar to lean, healthy mice. Follistatin gene delivery promoted insulinemia and abundance of insulin-positive pancreatic ß-cells, even when treatment was administered to mice with advanced diabetes, supporting a mechanism for improved glycemic control associated with maintenance of functional ß-cells. Our data demonstrate that single-dose intravascular Follistatin gene delivery can ameliorate the diabetic progression and improve prognostic markers of disease. These findings are consistent with other observations of Activin-mediated mechanisms exerting deleterious effects in models of obesity and diabetes, and suggest that interventions that attenuate Activin signaling could help further understanding of T2D and the development of novel T2D therapeutics.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 2/therapy , Follistatin/genetics , Gene Transfer Techniques , Genetic Therapy , Glycemic Control , Hyperglycemia/therapy , Administration, Intravenous , Animals , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Follistatin/administration & dosage , Hyperglycemia/genetics , Insulin Resistance , Mice
6.
Nature ; 574(7776): 63-68, 2019 10.
Article in English | MEDLINE | ID: mdl-31554967

ABSTRACT

The gp130 receptor cytokines IL-6 and CNTF improve metabolic homeostasis but have limited therapeutic use for the treatment of type 2 diabetes. Accordingly, we engineered the gp130 ligand IC7Fc, in which one gp130-binding site is removed from IL-6 and replaced with the LIF-receptor-binding site from CNTF, fused with the Fc domain of immunoglobulin G, creating a cytokine with CNTF-like, but IL-6-receptor-dependent, signalling. Here we show that IC7Fc improves glucose tolerance and hyperglycaemia and prevents weight gain and liver steatosis in mice. In addition, IC7Fc either increases, or prevents the loss of, skeletal muscle mass by activation of the transcriptional regulator YAP1. In human-cell-based assays, and in non-human primates, IC7Fc treatment results in no signs of inflammation or immunogenicity. Thus, IC7Fc is a realistic next-generation biological agent for the treatment of type 2 diabetes and muscle atrophy, disorders that are currently pandemic.


Subject(s)
Cytokine Receptor gp130/metabolism , Cytokines/chemical synthesis , Cytokines/therapeutic use , Diabetes Mellitus, Type 2/drug therapy , Immunoglobulin G/therapeutic use , Recombinant Fusion Proteins/therapeutic use , Adaptor Proteins, Signal Transducing/metabolism , Animals , Binding, Competitive , Cytokines/chemistry , Diabetes Mellitus, Type 2/metabolism , Drug Design , Fatty Liver/prevention & control , Glucose Tolerance Test , Humans , Hyperglycemia/drug therapy , Hyperglycemia/metabolism , Incretins/metabolism , Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Male , Mice , Muscle, Skeletal/drug effects , Obesity/metabolism , Pancreas/metabolism , Phosphoproteins/metabolism , Protein Engineering , Receptors, Interleukin-6/metabolism , Signal Transduction , Transcription Factors , Weight Gain/drug effects , YAP-Signaling Proteins
7.
Am J Physiol Endocrinol Metab ; 317(4): E597-E604, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31386565

ABSTRACT

It has been suggested that interleukin-6 (IL-6) produced by adipocytes in obesity leads to liver insulin resistance, although this hypothesis has never been definitively tested. Accordingly, we did so by generating adipocyte-specific IL-6-deficient (AdipoIL-6-/-) mice and studying them in the context of diet-induced and genetic obesity. Mice carrying two floxed alleles of IL-6 (C57Bl/6J) were crossed with Cre recombinase-overexpressing mice driven by the adiponectin promoter to generate AdipoIL-6-/- mice. AdipoIL-6-/- and floxed littermate controls were fed a standard chow or high-fat diet (HFD) for 16 wk and comprehensively metabolically phenotyped. In addition to a diet-induced obesity model, we also examined the role of adipocyte-derived IL-6 in a genetic model of obesity and insulin resistance by crossing the AdipoIL-6-/- mice with leptin-deficient (ob/ob) mice. As expected, mice on HFD and ob/ob mice displayed marked weight gain and increased fat mass compared with chow-fed and ob/+ (littermate control) animals, respectively. However, deletion of IL-6 from adipocytes in either model had no effect on glucose tolerance or fasting hyperinsulinemia. We concluded that adipocyte-specific IL-6 does not contribute to whole body glucose intolerance in obese mice.


Subject(s)
Adipocytes/metabolism , Glucose Intolerance/genetics , Interleukin-6/genetics , Obesity/genetics , Weight Gain/genetics , Adiponectin/biosynthesis , Adiponectin/genetics , Adiposity/genetics , Animals , Body Composition/genetics , Diet, High-Fat , Glucose Intolerance/etiology , Insulin Resistance/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology , Obesity/complications , Obesity/metabolism
8.
Diabetes Obes Metab ; 20(8): 1928-1936, 2018 08.
Article in English | MEDLINE | ID: mdl-29652108

ABSTRACT

AIMS: The induction of heat shock protein 72 (Hsp72) via heating, genetic manipulation or pharmacological activation is metabolically protective in the setting of obesity-induced insulin resistance across mammalian species. In this study, we set out to determine whether the overexpression of Hsp72, specifically in skeletal muscle, can protect against high-fat diet (HFD)-induced obesity and insulin resistance. MATERIALS AND METHODS: An Adeno-Associated Viral vector (AAV), designed to overexpress Hsp72 in skeletal muscle only, was used to study the effects of increasing Hsp72 levels on various metabolic parameters. Two studies were conducted, the first with direct intramuscular (IM) injection of the AAV:Hsp72 into the tibialis anterior hind-limb muscle and the second with a systemic injection to enable body-wide skeletal muscle transduction. RESULTS: IM injection of the AAV:Hsp72 significantly improved skeletal muscle insulin-stimulated glucose clearance in treated hind-limb muscles, as compared with untreated muscles of the contralateral leg when mice were fed an HFD. Despite this finding, systemic administration of AAV:Hsp72 did not improve body composition parameters such as body weight, fat mass or percentage body fat, nor did it lead to an improvement in fasting glucose levels or glucose tolerance. Furthermore, no differences were observed for other metabolic parameters such as whole-body oxygen consumption, energy expenditure or physical activity levels. CONCLUSIONS: At the levels of Hsp72 over-expression reported herein, skeletal muscle-specific Hsp72 overexpression via IM injection has the capacity to increase insulin-stimulated glucose clearance in this muscle. However, upon systemic injection, which results in lower muscle Hsp72 overexpression, no beneficial effects on whole-body metabolism are observed.


Subject(s)
Energy Metabolism/drug effects , Glucose Intolerance/prevention & control , HSP72 Heat-Shock Proteins/metabolism , Hypoglycemic Agents/therapeutic use , Insulin Resistance , Insulin/therapeutic use , Muscle, Skeletal/drug effects , Absorption, Physiological/drug effects , Animals , Brain/drug effects , Brain/metabolism , Diet, High-Fat/adverse effects , Gene Transfer Techniques , Glucose/metabolism , Glucose Intolerance/blood , Glucose Intolerance/etiology , Glucose Intolerance/metabolism , HSP72 Heat-Shock Proteins/genetics , Male , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Skeletal/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Obesity/etiology , Obesity/metabolism , Obesity/physiopathology , Organ Specificity , Pilot Projects , Rats
9.
Cell Metab ; 27(5): 1096-1110.e5, 2018 May 01.
Article in English | MEDLINE | ID: mdl-29681442

ABSTRACT

Chronic inflammation is a hallmark of obesity and is linked to the development of numerous diseases. The activation of toll-like receptor 4 (TLR4) by long-chain saturated fatty acids (lcSFAs) is an important process in understanding how obesity initiates inflammation. While experimental evidence supports an important role for TLR4 in obesity-induced inflammation in vivo, via a mechanism thought to involve direct binding to and activation of TLR4 by lcSFAs, several lines of evidence argue against lcSFAs being direct TLR4 agonists. Using multiple orthogonal approaches, we herein provide evidence that while loss-of-function models confirm that TLR4 does, indeed, regulate lcSFA-induced inflammation, TLR4 is not a receptor for lcSFAs. Rather, we show that TLR4-dependent priming alters cellular metabolism, gene expression, lipid metabolic pathways, and membrane lipid composition, changes that are necessary for lcSFA-induced inflammation. These results reconcile previous discordant observations and challenge the prevailing view of TLR4's role in initiating obesity-induced inflammation.


Subject(s)
Inflammation/metabolism , Macrophages/metabolism , Obesity/metabolism , Palmitates/metabolism , Toll-Like Receptor 4/metabolism , Animals , Humans , Inflammation/etiology , Macrophages/cytology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Obesity/complications , Signal Transduction
10.
Cell Metab ; 27(1): 237-251.e4, 2018 01 09.
Article in English | MEDLINE | ID: mdl-29320704

ABSTRACT

Exercise stimulates the release of molecules into the circulation, supporting the concept that inter-tissue signaling proteins are important mediators of adaptations to exercise. Recognizing that many circulating proteins are packaged in extracellular vesicles (EVs), we employed quantitative proteomic techniques to characterize the exercise-induced secretion of EV-contained proteins. Following a 1-hr bout of cycling exercise in healthy humans, we observed an increase in the circulation of over 300 proteins, with a notable enrichment of several classes of proteins that compose exosomes and small vesicles. Pulse-chase and intravital imaging experiments suggested EVs liberated by exercise have a propensity to localize in the liver and can transfer their protein cargo. Moreover, by employing arteriovenous balance studies across the contracting human limb, we identified several novel candidate myokines, released into circulation independently of classical secretion. These data identify a new paradigm by which tissue crosstalk during exercise can exert systemic biological effects.


Subject(s)
Exercise/physiology , Extracellular Vesicles/metabolism , Organ Specificity , Proteomics , Adult , Animals , Chromatography, High Pressure Liquid , Cytokines/metabolism , Endocytosis , Exosomes/metabolism , Female , Glycolysis , Humans , Intravital Microscopy , Isotope Labeling , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Nanotechnology , Proteome/metabolism , Tandem Mass Spectrometry
11.
Obesity (Silver Spring) ; 23(9): 1845-55, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26250514

ABSTRACT

OBJECTIVE: Adipose inflammation and dysfunction underlie metabolic obesity. Exercise improves glycemic control and metabolic indices, but effects on adipose function and inflammation are less clear. Accordingly, it was hypothesized that exercise improves adipose morphometry to reduce adipose inflammation in hyperphagic obese mice. METHODS: Alms1 mutant foz/foz mice housed in pairs were fed an atherogenic or chow diet; half the cages were fitted with a computer-monitored wheel for voluntary exercise. Insulin-induced AKT-phosphorylation, adipocyte size distribution, and inflammatory recruitment were studied in visceral versus subcutaneous depots, and severity of fatty liver disease was determined. RESULTS: Exercise prevented obesity and diabetes development in chow-fed foz/foz mice and delayed their onset in atherogenic-fed counterparts. Insulin-stimulated phospho-AKT levels in muscle were improved with exercise, but not in adipose or liver. Exercise suppressed adipose inflammatory recruitment, particularly in visceral adipose, associated with an increased number of small adipocyte subpopulations, and enhanced expression of beige adipocyte factor PRDM16 in subcutaneous fat. In atherogenic-fed foz/foz mice liver, exercise suppressed development of nonalcoholic steatohepatitis and related liver fibrosis. CONCLUSIONS: Exercise confers metabo-protective effects in atherogenic-fed hyperphagic mice by preventing early onset of obesity and diabetes in association with enhanced muscle insulin sensitivity, improved adipose morphometry, and suppressed adipose and liver inflammation.


Subject(s)
Diabetes Mellitus, Experimental/complications , Inflammation/complications , Non-alcoholic Fatty Liver Disease/therapy , Obesity/complications , Physical Conditioning, Animal/methods , Animals , Mice , Mice, Obese , Non-alcoholic Fatty Liver Disease/complications
12.
Cell Metab ; 21(3): 403-16, 2015 Mar 03.
Article in English | MEDLINE | ID: mdl-25738456

ABSTRACT

Interleukin-6 (IL-6) plays a paradoxical role in inflammation and metabolism. The pro-inflammatory effects of IL-6 are mediated via IL-6 "trans-signaling," a process where the soluble form of the IL-6 receptor (sIL-6R) binds IL-6 and activates signaling in inflammatory cells that express the gp130 but not the IL-6 receptor. Here we show that trans-signaling recruits macrophages into adipose tissue (ATM). Moreover, blocking trans-signaling with soluble gp130Fc protein prevents high-fat diet (HFD)-induced ATM accumulation, but does not improve insulin action. Importantly, however, blockade of IL-6 trans-signaling, unlike complete ablation of IL-6 signaling, does not exacerbate obesity-induced weight gain, liver steatosis, or insulin resistance. Our data identify the sIL-6R as a critical chemotactic signal for ATM recruitment and suggest that selectively blocking IL-6 trans-signaling may be a more favorable treatment option for inflammatory diseases, compared with current treatments that completely block the action of IL-6 and negatively impact upon metabolic homeostasis.


Subject(s)
Adipose Tissue/metabolism , Diet, High-Fat/adverse effects , Insulin Resistance/physiology , Interleukin-6/metabolism , Macrophages/metabolism , Macrophages/physiology , Signal Transduction/physiology , Adipose Tissue/physiology , Animals , Cytokine Receptor gp130/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Interleukin-6/metabolism
13.
Am J Physiol Endocrinol Metab ; 308(9): E778-91, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25714675

ABSTRACT

The accumulation of lipid at ectopic sites, including the skeletal muscle and liver, is a common consequence of obesity and is associated with tissue-specific and whole body insulin resistance. Exercise is well known to improve insulin resistance by mechanisms not completely understood. We performed lipidomic profiling via mass spectrometry in liver and skeletal muscle samples from exercise-trained mice to decipher the lipid changes associated with exercise-induced improvements in whole body glucose metabolism. Obesity and insulin resistance were induced in C57BL/6J mice by high-fat feeding for 4 wk. Mice then underwent an exercise training program (treadmill running) 5 days/wk (Ex) for 4 wk or remained sedentary (Sed). Compared with Sed, Ex displayed improved (P < 0.01) whole body metabolism as measured via an oral glucose tolerance test. Deleterious lipid species such as diacylglycerol (P < 0.05) and cholesterol esters (P < 0.01) that accumulate with high-fat feeding were decreased in the liver of trained mice. Furthermore, the ratio of phosphatidylcholine (PC) to phosphatidylethanolamine (PE) (the PC/PE ratio), which is associated with membrane integrity and linked to hepatic disease progression, was increased by training (P < 0.05). These findings occurred without corresponding changes in the skeletal muscle lipidome. A concomitant decrease (P < 0.05) was observed for the fatty acid transporters CD36 and FATP4 in the liver, suggesting that exercise stimulates a coordinated reduction in fatty acid entry into hepatocytes. Given the important role of the liver in the regulation of whole body glucose homeostasis, hepatic lipid regression may be a key component by which exercise can improve metabolism.


Subject(s)
Diet, High-Fat/adverse effects , Fatty Liver/etiology , Fatty Liver/prevention & control , Lipid Metabolism , Liver/metabolism , Metabolome , Physical Conditioning, Animal/physiology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adiposity/drug effects , Animals , Dietary Fats/pharmacology , Fatty Liver/metabolism , Glucose Intolerance/etiology , Glucose Intolerance/metabolism , Hyperinsulinism/etiology , Hyperinsulinism/metabolism , Lipid Metabolism/drug effects , Liver/drug effects , Male , Metabolome/drug effects , Mice , Mice, Inbred C57BL , Oxidative Phosphorylation/drug effects
14.
PLoS One ; 9(11): e111524, 2014.
Article in English | MEDLINE | ID: mdl-25375135

ABSTRACT

Alterations in the immune cell profile and the induction of inflammation within adipose tissue are a hallmark of obesity in mice and humans. Dual-specificity phosphatase 2 (DUSP2) is widely expressed within the immune system and plays a key role promoting immune and inflammatory responses dependent on mitogen-activated protein kinase (MAPK) activity. We hypothesised that the absence of DUSP2 would protect mice against obesity-associated inflammation and insulin resistance. Accordingly, male and female littermate mice that are either wild-type (wt) or homozygous for a germ-line null mutation of the dusp2 gene (dusp2-/-) were fed either a standard chow diet (SCD) or high fat diet (HFD) for 12 weeks prior to metabolic phenotyping. Compared with mice fed the SCD, all mice consuming the HFD became obese, developed glucose intolerance and insulin resistance, and displayed increased macrophage recruitment and markers of inflammation in epididymal white adipose tissue. The absence of DUSP2, however, had no effect on the development of obesity or adipose tissue inflammation. Whole body insulin sensitivity in male mice was unaffected by an absence of DUSP2 in response to either the SCD or HFD; however, HFD-induced insulin resistance was slightly, but significantly, reduced in female dusp2-/- mice. In conclusion, DUSP2 plays no role in regulating obesity-associated inflammation and only a minor role in controlling insulin sensitivity following HFD in female, but not male, mice. These data indicate that rather than DUSP2 being a pan regulator of MAPK dependent immune cell mediated inflammation, it appears to differentially regulate inflammatory responses that have a MAPK component.


Subject(s)
Dual Specificity Phosphatase 2/genetics , Glucose Intolerance/genetics , Inflammation/genetics , Insulin Resistance/genetics , Obesity/genetics , Adipose Tissue/metabolism , Animals , Blood Glucose/metabolism , Diet, High-Fat , Dual Specificity Phosphatase 2/metabolism , Female , Glucose Intolerance/metabolism , Glucose Tolerance Test , Inflammation/etiology , Inflammation/metabolism , Insulin/blood , Male , Mice , Mutation , Obesity/complications , Obesity/metabolism , Sex Factors
15.
Nat Immunol ; 15(5): 423-30, 2014 May.
Article in English | MEDLINE | ID: mdl-24681566

ABSTRACT

Obesity and resistance to insulin are closely associated with the development of low-grade inflammation. Interleukin 6 (IL-6) is linked to obesity-associated inflammation; however, its role in this context remains controversial. Here we found that mice with an inactivated gene encoding the IL-6Rα chain of the receptor for IL-6 in myeloid cells (Il6ra(Δmyel) mice) developed exaggerated deterioration of glucose homeostasis during diet-induced obesity, due to enhanced resistance to insulin. Tissues targeted by insulin showed increased inflammation and a shift in macrophage polarization. IL-6 induced expression of the receptor for IL-4 and augmented the response to IL-4 in macrophages in a cell-autonomous manner. Il6ra(Δmyel) mice were resistant to IL-4-mediated alternative polarization of macrophages and exhibited enhanced susceptibility to lipopolysaccharide (LPS)-induced endotoxemia. Our results identify signaling via IL-6 as an important determinant of the alternative activation of macrophages and assign an unexpected homeostatic role to IL-6 in limiting inflammation.


Subject(s)
Endotoxemia/immunology , Insulin Resistance , Interleukin-6/metabolism , Macrophage Activation , Macrophages/immunology , Obesity/immunology , Animals , Cells, Cultured , Humans , Insulin Resistance/genetics , Insulin Resistance/immunology , Interleukin-4/immunology , Interleukin-6/genetics , Lipopolysaccharides/immunology , Macrophage Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation/genetics , Receptors, Interleukin-6/genetics , Signal Transduction/genetics
16.
Diabetes ; 63(6): 1881-94, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24430435

ABSTRACT

Induction of heat shock protein (HSP)72 protects against obesity-induced insulin resistance, but the underlying mechanisms are unknown. Here, we show that HSP72 plays a pivotal role in increasing skeletal muscle mitochondrial number and oxidative metabolism. Mice overexpressing HSP72 in skeletal muscle (HSP72Tg) and control wild-type (WT) mice were fed either a chow or high-fat diet (HFD). Despite a similar energy intake when HSP72Tg mice were compared with WT mice, the HFD increased body weight, intramuscular lipid accumulation (triacylglycerol and diacylglycerol but not ceramide), and severe glucose intolerance in WT mice alone. Whole-body VO2, fatty acid oxidation, and endurance running capacity were markedly increased in HSP72Tg mice. Moreover, HSP72Tg mice exhibited an increase in mitochondrial number. In addition, the HSP72 coinducer BGP-15, currently in human clinical trials for type 2 diabetes, also increased mitochondrial number and insulin sensitivity in a rat model of type 2 diabetes. Together, these data identify a novel role for activation of HSP72 in skeletal muscle. Thus, the increased oxidative metabolism associated with activation of HSP72 has potential clinical implications not only for type 2 diabetes but also for other disorders where mitochondrial function is compromised.


Subject(s)
Cell Respiration , Diabetes Mellitus, Type 2/metabolism , HSP72 Heat-Shock Proteins/metabolism , Insulin Resistance , Mitochondria, Muscle/metabolism , Obesity/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Blood Glucose , Blotting, Western , Body Weight , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/physiopathology , Diet, High-Fat , Energy Metabolism , Fatty Acids/metabolism , Leptin/metabolism , Male , Mice , Muscle, Skeletal/metabolism , Obesity/genetics , Obesity/physiopathology , Oxidation-Reduction , Oxidative Phosphorylation , Peroxisome Proliferator-Activated Receptors/metabolism , Rats , Real-Time Polymerase Chain Reaction , Sirtuin 1/metabolism
17.
Diabetes ; 62(9): 3064-74, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23670974

ABSTRACT

Circulating interleukin (IL)-18 is elevated in obesity, but paradoxically causes hypophagia. We hypothesized that IL-18 may attenuate high-fat diet (HFD)-induced insulin resistance by activating AMP-activated protein kinase (AMPK). We studied mice with a global deletion of the α-isoform of the IL-18 receptor (IL-18R(-/-)) fed a standard chow or HFD. We next performed gain-of-function experiments in skeletal muscle, in vitro, ex vivo, and in vivo. We show that IL-18 is implicated in metabolic homeostasis, inflammation, and insulin resistance via mechanisms involving the activation of AMPK in skeletal muscle. IL-18R(-/-) mice display increased weight gain, ectopic lipid deposition, inflammation, and reduced AMPK signaling in skeletal muscle. Treating myotubes or skeletal muscle strips with IL-18 activated AMPK and increased fat oxidation. Moreover, in vivo electroporation of IL-18 into skeletal muscle activated AMPK and concomitantly inhibited HFD-induced weight gain. In summary, IL-18 enhances AMPK signaling and lipid oxidation in skeletal muscle implicating IL-18 in metabolic homeostasis.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Insulin Resistance/physiology , Interleukin-18/metabolism , Muscle, Skeletal/enzymology , Weight Gain/physiology , AMP-Activated Protein Kinases/genetics , Animals , Body Composition/genetics , Body Composition/physiology , Calorimetry, Indirect , Female , Insulin Resistance/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Receptors, Interleukin-18/deficiency , Receptors, Interleukin-18/genetics , Weight Gain/genetics
18.
PLoS One ; 7(6): e39100, 2012.
Article in English | MEDLINE | ID: mdl-22701749

ABSTRACT

Obesity is associated with a state of chronic low grade inflammation that plays an important role in the development of insulin resistance. Tumor progression locus 2 (Tpl2) is a serine/threonine mitogen activated protein kinase kinase kinase (MAP3K) involved in regulating responses to specific inflammatory stimuli. Here we have used mice lacking Tpl2 to examine its role in obesity-associated insulin resistance. Wild type (wt) and tpl2(-/-) mice accumulated comparable amounts of fat and lean mass when fed either a standard chow diet or two different high fat (HF) diets containing either 42% or 59% of energy content derived from fat. No differences in glucose tolerance were observed between wt and tpl2(-/-) mice on any of these diets. Insulin tolerance was similar on both standard chow and 42% HF diets, but was slightly impaired in tpl2(-/-) mice fed the 59% HFD. While gene expression markers of macrophage recruitment and inflammation were increased in the white adipose tissue of HF fed mice compared with standard chow fed mice, no differences were observed between wt and tpl2(-/-) mice. Finally, a HF diet did not increase Tpl2 expression nor did it activate Extracellular Signal-Regulated Kinase 1/2 (ERK1/2), the MAPK downstream of Tpl2. These findings argue that Tpl2 does not play a non-redundant role in obesity-associated metabolic dysfunction.


Subject(s)
Inflammation/physiopathology , Insulin Resistance/physiology , MAP Kinase Kinase Kinases/deficiency , Obesity/complications , Proto-Oncogene Proteins/deficiency , Signal Transduction/physiology , Adipose Tissue, White/metabolism , Animals , Biomarkers/metabolism , Blotting, Western , Body Composition/physiology , DNA Primers/genetics , Diet, High-Fat , Gene Expression Profiling , Inflammation/etiology , MAP Kinase Kinase Kinases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Proto-Oncogene Proteins/genetics
19.
Transfusion ; 52(11): 2339-47, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22452709

ABSTRACT

BACKGROUND: The fear for adverse effects of blood donation on subsequent exercise may prevent physically active people from donating. We studied the impact of a standard blood bank donation (i.e., 450-mL blood withdrawal) on the thermoregulatory and cardiovascular responses to prolonged exercise in the heat. STUDY DESIGN AND METHODS: Eight moderately trained, heat-acclimated males cycled for 1 hour at 60% in a hot environment (34.9±0.6 °C) on four occasions: 1) 2 days before blood donation (CON), 2) 2 hours after donation (DON), 3) 2 days after donation (2 DAYS), and 4) 7 days after donation (7 DAYS). RESULTS: Two-thirds of the blood volume withdrawn was endogenously restored before exercise in the DON trial (p<0.05). DON started with increased preexercise rectal temperature (TRE; 0.42±0.1 °C above CON; p<0.05), which resulted in high levels of hyperthermia (i.e., 39.0±0.2 °C) after 1 hour of exercise. Skin temperature (34.5±0.1 °C) and sweat rate (1.15±0.1 L/h) were not affected by DON. However, DON lowered the skin blood flow:TRE relationship and elevated heart rate (HR) above CON (12±4 beats/min; p<0.05) maintaining cardiac output. After 2 DAYS, TRE and HR were restored to CON levels while cardiac output increased above CON (6%; p<0.05) in association with reduced hemoglobin concentration (i.e., peak hemodilution). CONCLUSION: A blood bank donation increases preexercise TRE. Subsequent exercise in a hot environment results in high levels of hyperthermia and HR. These thermoregulatory and cardiovascular perturbations observed during exercise disappear 2 days after donation.


Subject(s)
Acclimatization/physiology , Blood Donors , Body Temperature Regulation/physiology , Exercise/physiology , Fever/physiopathology , Adult , Blood Pressure/physiology , Body Temperature/physiology , Carbohydrate Metabolism/physiology , Cardiac Output/physiology , Fever/blood , Heart Rate/physiology , Hemoglobins/metabolism , Humans , Lactic Acid/blood , Male , Oxygen Consumption/physiology , Skin Temperature/physiology , Sweating/physiology , Young Adult
20.
Eur J Appl Physiol ; 109(5): 973-81, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20349316

ABSTRACT

To determine if the increases in rectal temperature (T(REC)) during exercise in the heat at a given percent of VO2peak depend on a subject's aerobic fitness level. On three occasions, 10 endurance-trained (Tr) and 10 untrained (UTr) subjects (VO2peak: 60 +/- 6 vs. 44 +/- 3 mL kg(-1) min(-1), P < 0.05) cycled in a hot-dry environment (36 +/- 1 degrees C; 25 +/- 2% humidity, airflow 2.5 m s(-1)) at three workloads (40, 60, and 80% VO2peak). At the same percent of VO2peak, on average, Tr had 28 +/- 5% higher heat production but also higher skin blood flow (29 +/- 3%) and sweat rate (20 +/- 7%; P = 0.07) and lower skin temperature (0.5 degrees C; P < 0.05). Pre-exercise T(REC) was lower in the Tr subjects (37.4 +/- 0.2 vs. 37.6 +/- 0.2; P < 0.05) but similar to the UTr at the end of 40 and 60% VO2peak trials. Thus, exercise T(REC) increased more in the Tr group than in the UTr group (0.6 +/- 0.1 vs. 0.3 +/- 0.1 degrees C at 40% VO2peak and 1.0 +/- 0.1 vs. 0.6 +/- 0.3 degrees C at 60% VO2peak; P < 0.05). At 80% VO2peak not only the increase in T(REC) (1.7 +/- 0.1 vs. 1.3 +/- 0.3 degrees C) but also the final T(REC) was larger in Tr than in UTr subjects (39.15 +/- 0.1 vs. 38.85 +/- 0.1 degrees C; P < 0.05). During exercise in the heat at the same relative intensity, aerobically trained individuals have a larger rise in T(REC) than do the untrained ones which renders them more hyperthermic after high-intensity exercise.


Subject(s)
Body Temperature/physiology , Exercise/physiology , Hot Temperature , Physical Fitness/physiology , Adolescent , Adult , Female , Humans , Male , Oxygen Consumption/physiology , Physical Endurance/physiology , Rectum/physiology , Regional Blood Flow/physiology , Skin/blood supply , Skin Temperature/physiology , Sweating/physiology , Water-Electrolyte Balance/physiology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL