Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Biochim Biophys Acta Mol Basis Dis ; 1870(3): 167029, 2024 03.
Article in English | MEDLINE | ID: mdl-38325224

ABSTRACT

High fructose diets are associated with an increased risk of liver cancer. Previous studies in mice suggest increased lipogenesis is a key mechanism linking high fructose diets to liver tumour growth. However, these studies administered fructose to mice at supraphysiological levels. The aim of this study was to determine whether liver tumour growth and lipogenesis were altered in mice fed fructose at physiological levels. To test this, we injected male C57BL/6 mice with the liver carcinogen diethylnitrosamine and then fed them diets without fructose or fructose ranging from 10 to 20 % total calories. Results showed mice fed diets with ≥15 % fructose had significantly increased liver tumour numbers (2-4-fold) and total tumour burden (∼7-fold) vs mice fed no-fructose diets. However, fructose-associated tumour burden was not associated with lipogenesis. Conversely, unbiased metabolomic analyses revealed bile acids were elevated in the sera of mice fed a 15 % fructose diet vs mice fed a no-fructose diet. Using a syngeneic ectopic liver tumour model, we show that ursodeoxycholic acid, which decreases systemic bile acids, significantly reduced liver tumour growth in mice fed the 15 % fructose diet but not mice fed a no-fructose diet. These results point to a novel role for systemic bile acids in mediating liver tumour growth associated with a high fructose diet. Overall, our study shows fructose intake at or above normal human consumption (≥15 %) is associated with increased liver tumour numbers and growth and that modulating systemic bile acids inhibits fructose-associated liver tumour growth in mice.


Subject(s)
Bile Acids and Salts , Liver Neoplasms , Humans , Mice , Male , Animals , Fructose/adverse effects , Mice, Inbred C57BL , Liver Neoplasms/chemically induced
2.
Diabetes ; 73(3): 374-384, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-37870907

ABSTRACT

Excess body fat is a risk factor for metabolic diseases and is a leading preventable cause of morbidity and mortality worldwide. There is a strong need to find new treatments that decrease the burden of obesity and lower the risk of obesity-related comorbidities, including cardiovascular disease and type 2 diabetes. Pharmacologic mitochondrial uncouplers represent a potential treatment for obesity through their ability to increase nutrient oxidation. Herein, we report the in vitro and in vivo characterization of compound SHD865, the first compound to be studied in vivo in a newly discovered class of imidazolopyrazine mitochondrial uncouplers. SHD865 is a derivative of the furazanopyrazine uncoupler BAM15. SHD865 is a milder mitochondrial uncoupler than BAM15 that results in a lower maximal respiration rate. In a mouse model of diet-induced adiposity, 6-week treatment with SHD865 completely restored normal body composition and glucose tolerance to levels like those of chow-fed controls, without altering food intake. SHD865 treatment also corrected liver steatosis and plasma hyperlipidemia to normal levels comparable with chow-fed controls. SHD865 has maximal oral bioavailability in rats and slow clearance in human microsomes and hepatocytes. Collectively, these data identify the potential of imidazolopyrazine mitochondrial uncouplers as drug candidates for the treatment of obesity-related disorders.


Subject(s)
Diabetes Mellitus, Type 2 , Glucose Intolerance , Mice , Rats , Humans , Animals , Adiposity , Glucose Intolerance/drug therapy , Glucose Intolerance/metabolism , Diabetes Mellitus, Type 2/metabolism , Obesity/etiology , Liver/metabolism , Diet, High-Fat/adverse effects , Mice, Inbred C57BL
3.
J Med Chem ; 66(6): 3876-3895, 2023 03 23.
Article in English | MEDLINE | ID: mdl-36882080

ABSTRACT

Small-molecule mitochondrial uncouplers are gaining recognition as potential therapeutics for metabolic diseases such as obesity, diabetes, and nonalcoholic steatohepatitis (NASH). Specifically, heterocycles derived from BAM15, a potent and mitochondria-selective uncoupler, have yielded promising preclinical candidates that are efficacious in animal models of obesity and NASH. In this study, we report the structure-activity relationship studies of 6-amino-[1,2,5]oxadiazolo[3,4-b]pyridin-5-ol derivatives. Using oxygen consumption rate as a readout of mitochondrial uncoupling, we established 5-hydroxyoxadiazolopyridines as mild uncouplers. In particular, SHM115, which contains a pentafluoro aniline, had an EC50 value of 17 µM and exhibited 75% oral bioavailability. SHM115 treatment increased the energy expenditure and lowered the body fat mass in two diet-induced obesity mouse models, including an obesity prevention model and an obesity reversal model. Taken together, our findings demonstrate the therapeutic potential of mild mitochondrial uncouplers for the prevention of diet-induced obesity.


Subject(s)
Non-alcoholic Fatty Liver Disease , Mice , Animals , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/prevention & control , Mitochondria/metabolism , Obesity/drug therapy , Obesity/metabolism , Diet , Oxygen Consumption
4.
Mol Metab ; 63: 101543, 2022 09.
Article in English | MEDLINE | ID: mdl-35811051

ABSTRACT

OBJECTIVE: Adipose tissue is a critical regulator of energy balance that must rapidly shift its metabolism between fasting and feeding to maintain homeostasis. Adenosine has been characterized as an important regulator of adipocyte metabolism primarily through its actions on A1 adenosine receptors (A1R). We sought to understand the role A1R plays specifically in adipocytes during fasting and feeding to regulate glucose and lipid metabolism. METHODS: We used Adora1 floxed mice with an inducible, adiponectin-Cre to generate FAdora1-/- mice, where F designates a fat-specific deletion of A1R. We used these FAdora1-/- mice along with specific agonists and antagonists of A1R to investigate changes in adenosine signaling within adipocytes between the fasted and fed state. RESULTS: We found that the adipose tissue response to adenosine is not static, but changes dynamically according to nutrient conditions through the insulin-Akt-FOXO1 axis. We show that under fasted conditions, FAdora1-/- mice had impairments in the suppression of lipolysis by insulin on normal chow and impaired glucose tolerance on high-fat diet. FAdora1-/- mice also exhibited a higher lipolytic response to isoproterenol than WT controls when fasted, however this difference was lost after a 4-hour refeeding period. We demonstrate that FOXO1 binds to the A1R promoter, and refeeding leads to a rapid downregulation of A1R transcript and desensitization of adipocytes to A1R agonism. Obesity also desensitizes adipocyte A1R, and this is accompanied by a disruption of cyclical changes in A1R transcription between fasting and refeeding. CONCLUSIONS: We propose that FOXO1 drives high A1R expression under fasted conditions to limit excess lipolysis during stress and augment insulin action upon feeding. Subsequent downregulation of A1R under fed conditions leads to desensitization of these receptors in adipose tissue. This regulation of A1R may facilitate reentrance into the catabolic state upon fasting.


Subject(s)
Adipose Tissue , Lipolysis , Adenosine/metabolism , Adipose Tissue/metabolism , Animals , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Insulin/metabolism , Lipolysis/physiology , Mice , Receptors, Purinergic P1/metabolism
5.
Bioorg Med Chem Lett ; 73: 128912, 2022 10 01.
Article in English | MEDLINE | ID: mdl-35907607

ABSTRACT

We report new mitochondrial uncouplers derived from the conversion of [1,2,5]oxadiazolo[3,4-b]pyrazines to 1H-imidazo[4,5-b]pyrazines. The in situ Fe-mediated reduction of the oxadiazole fragment followed by cyclization gave access to imidazopyrazines in moderate to good yields. A selection of orthoesters also allowed functionalization on the 2-position of the imidazole ring. This method afforded a variety of imidazopyrazine derivatives with varying substitution on the 2, 5 and 6 positions. Our studies suggest that both a 2-trifluoromethyl group and N-methylation are crucial for mitochondrial uncoupling capacity.


Subject(s)
Mitochondria , Pyrazines , Cyclization , Mitochondria/metabolism , Oxadiazoles/metabolism , Pyrazines/metabolism
6.
Metabolism ; 117: 154724, 2021 04.
Article in English | MEDLINE | ID: mdl-33548253

ABSTRACT

AIMS: Mitochondrial uncouplers decrease caloric efficiency and have potential therapeutic benefits for the treatment of obesity and related metabolic disorders. Herein we investigate the metabolic and physiologic effects of a recently identified small molecule mitochondrial uncoupler named SHC517 in a mouse model of diet-induced obesity. METHODS: SHC517 was administered as an admixture in food. The effect of SHC517 on in vivo energy expenditure and respiratory quotient was determined by indirect calorimetry. A dose-finding obesity prevention study was performed by starting SHC517 treatment concomitant with high fat diet for a period of 12 days. An obesity reversal study was performed by feeding mice western diet for 4 weeks prior to SHC517 treatment for 7 weeks. Biochemical assays were used to determine changes in glucose, insulin, triglycerides, and cholesterol. SHC517 concentrations were determined by mass spectrometry. RESULTS: SHC517 increased lipid oxidation without affecting body temperature. SHC517 prevented diet-induced obesity when administered at 0.05% and 0.1% w/w in high fat diet and reversed established obesity when tested at the 0.05% dose. In the obesity reversal model, SHC517 restored adiposity to levels similar to chow-fed control mice without affecting food intake or lean body mass. SHC517 improved glucose tolerance and fasting glucose levels when administered in both the obesity prevention and obesity reversal modes. CONCLUSIONS: SHC517 is a mitochondrial uncoupler with potent anti-obesity and insulin sensitizing effects in mice. SHC517 reversed obesity without altering food intake or compromising lean mass, effects that are highly sought-after in anti-obesity therapeutics.


Subject(s)
Eating/drug effects , Mitochondria/drug effects , Obesity/drug therapy , Small Molecule Libraries/pharmacology , Adiposity/drug effects , Animals , Body Weight/drug effects , Calorimetry, Indirect/methods , Diet, High-Fat/adverse effects , Diet, Western/adverse effects , Energy Metabolism/drug effects , Glucose/metabolism , Insulin/metabolism , Insulin Resistance/physiology , Lipid Metabolism/drug effects , Male , Metabolic Diseases/drug therapy , Metabolic Diseases/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Obesity/metabolism
7.
J Med Chem ; 63(11): 6203-6224, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32392051

ABSTRACT

Small molecule mitochondrial uncouplers have recently garnered great interest for their potential in treating nonalcoholic steatohepatitis (NASH). In this study, we report the structure-activity relationship profiling of a 6-amino[1,2,5]oxadiazolo[3,4-b]pyrazin-5-ol core, which utilizes the hydroxy moiety as the proton transporter across the mitochondrial inner membrane. We demonstrate that a wide array of substituents is tolerated with this novel scaffold that increased cellular metabolic rates in vitro using changes in oxygen consumption rate as a readout. In particular, compound SHS4121705 (12i) displayed an EC50 of 4.3 µM in L6 myoblast cells and excellent oral bioavailability and liver exposure in mice. In the STAM mouse model of NASH, administration of 12i at 25 mg kg-1 day-1 lowered liver triglyceride levels and improved liver markers such as alanine aminotransferase, NAFLD activity score, and fibrosis. Importantly, no changes in body temperature or food intake were observed. As potential treatment of NASH, mitochondrial uncouplers show promise for future development.


Subject(s)
Pyrazines/chemistry , Alanine Transaminase/metabolism , Aniline Compounds/chemistry , Animals , Cell Line , Diet, High-Fat , Disease Models, Animal , Half-Life , Liver/drug effects , Liver/metabolism , Mice , Mitochondria/metabolism , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/metabolism , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , Oxygen Consumption/drug effects , Pyrazines/pharmacokinetics , Pyrazines/pharmacology , Pyrazines/therapeutic use , Rats , Structure-Activity Relationship , Triglycerides/metabolism , Uncoupling Protein 1/chemistry , Uncoupling Protein 1/metabolism
8.
Nat Commun ; 11(1): 2397, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32409697

ABSTRACT

Obesity is a health problem affecting more than 40% of US adults and 13% of the global population. Anti-obesity treatments including diet, exercise, surgery and pharmacotherapies have so far failed to reverse obesity incidence. Herein, we target obesity with a pharmacotherapeutic approach that decreases caloric efficiency by mitochondrial uncoupling. We show that a recently identified mitochondrial uncoupler BAM15 is orally bioavailable, increases nutrient oxidation, and decreases body fat mass without altering food intake, lean body mass, body temperature, or biochemical and haematological markers of toxicity. BAM15 decreases hepatic fat, decreases inflammatory lipids, and has strong antioxidant effects. Hyperinsulinemic-euglycemic clamp studies show that BAM15 improves insulin sensitivity in multiple tissue types. Collectively, these data demonstrate that pharmacologic mitochondrial uncoupling with BAM15 has powerful anti-obesity and insulin sensitizing effects without compromising lean mass or affecting food intake.


Subject(s)
Diamines/administration & dosage , Insulin Resistance , Mitochondria/drug effects , Obesity/drug therapy , Oxadiazoles/administration & dosage , Pyrazines/administration & dosage , Adipose Tissue/drug effects , Adipose Tissue/metabolism , Administration, Oral , Animals , Blood Glucose/analysis , Body Temperature/drug effects , Body Weight/drug effects , Diamines/adverse effects , Diet, Western/adverse effects , Disease Models, Animal , Dose-Response Relationship, Drug , Glucose Clamp Technique , Humans , Liver/drug effects , Liver/metabolism , Male , Membrane Potential, Mitochondrial/drug effects , Mice , Mitochondria/metabolism , Obesity/blood , Obesity/etiology , Obesity/metabolism , Oxadiazoles/adverse effects , Oxidative Stress/drug effects , Pyrazines/adverse effects
9.
Bioorg Med Chem Lett ; 30(8): 127057, 2020 04 15.
Article in English | MEDLINE | ID: mdl-32113842

ABSTRACT

Mitochondrial protonophores transport protons through the mitochondrial inner membrane into the matrix to uncouple nutrient oxidation from ATP production thereby decreasing the proton motive force. Mitochondrial uncouplers have beneficial effects of decrease reactive oxygen species generation and have the potential for treating diseases such as obesity, neurodegenerative diseases, non-alcoholic fatty liver disease (NAFLD), diabetes, and many others. In this study, we report the structure-activity relationship profile of the pyrazine scaffold bearing substituted aniline rings. Our work indicates that a trifluoromethyl group is best at the para position while the trifluoromethoxy group is preferred in the meta position of the aniline rings of 2,3-substituted pyrazines. As proton transport and cycling requires the formation of a negative charge that has to traverse the mitochondrial membrane, a stabilizing internal hydrogen bond is a key feature for efficient mitochondrial uncoupling activity.


Subject(s)
Aniline Compounds/pharmacology , Mitochondria/drug effects , Pyrazines/pharmacology , Uncoupling Agents/pharmacology , Aniline Compounds/chemistry , Dose-Response Relationship, Drug , Molecular Structure , Pyrazines/chemistry , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Structure-Activity Relationship , Uncoupling Agents/chemistry
10.
J Med Chem ; 63(5): 2511-2526, 2020 03 12.
Article in English | MEDLINE | ID: mdl-32017849

ABSTRACT

Small molecule mitochondrial uncouplers are emerging as a new class of molecules for the treatment of nonalcoholic steatohepatitis. We utilized BAM15, a potent protonophore that uncouples the mitochondria without depolarizing the plasma membrane, as a lead compound for structure-activity profiling. Using oxygen consumption rate as an assay for determining uncoupling activity, changes on the 5- and 6-position of the oxadiazolopyrazine core were introduced. Our studies suggest that unsymmetrical aniline derivatives bearing electron withdrawing groups are preferred compared to the symmetrical counterparts. In addition, alkyl substituents are not tolerated, and the N-H proton of the aniline ring is responsible for the protonophore activity. In particular, compound 10b had an EC50 value of 190 nM in L6 myoblast cells. In an in vivo model of NASH, 10b decreased liver triglyceride levels and showed improvement in fibrosis, inflammation, and plasma ALT. Taken together, our studies indicate that mitochondrial uncouplers have potential for the treatment of NASH.


Subject(s)
Diamines/therapeutic use , Mitochondria, Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Pyrazines/therapeutic use , Uncoupling Agents/therapeutic use , Animals , Diamines/chemistry , Diamines/pharmacology , Liver/drug effects , Liver/metabolism , Male , Mice, Inbred C57BL , Mitochondria, Liver/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Oxadiazoles/therapeutic use , Oxygen Consumption/drug effects , Pyrazines/chemistry , Pyrazines/pharmacology , Uncoupling Agents/chemistry , Uncoupling Agents/pharmacology
11.
Cancers (Basel) ; 10(9)2018 Sep 05.
Article in English | MEDLINE | ID: mdl-30189621

ABSTRACT

Rodent models of liver tumorigenesis have reproducibly shown that dietary sugar intake is a powerful driver of liver tumor initiation and growth. In contrast, dietary sugar restriction with ketogenic diets or calorie restriction generally prevents liver tumor formation. Ketogenic diet is viewed positively as a therapeutic adjuvant; however, most ketogenic diet studies described to date have been performed in prevention mode rather than treatment mode. Therefore, it remains unclear whether a ketogenic diet can be administered in late stages of disease to stall or reverse liver tumor growth. To model the clinically relevant treatment mode, we administered a ketogenic diet to mice after liver tumor initiation and monitored tumor growth by magnetic resonance imaging (MRI). Male C57BL/6 mice were injected with diethylnitrosamine (DEN) at 2 weeks of age and fed a chow diet until 39 weeks of age, when they underwent MRI imaging to detect liver tumors. Mice were then randomised into two groups and fed either a chow diet or switched to a ketogenic diet from 40⁻48 weeks of age. Serial MRIs were performed at 44 and 48 weeks of age. All mice had tumors at study completion and there were no differences in total tumor burden between diet groups. Although a ketogenic diet has marked protective effects against DEN-induced liver tumourigenesis in this mouse model, these data demonstrate that ketogenic diet cannot stop the progression of established liver tumors.

12.
Sci Rep ; 7(1): 127, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28273928

ABSTRACT

Hepatic glucose production (HGP) is required to maintain normoglycemia during fasting. Glucagon is the primary hormone responsible for increasing HGP; however, there are many additional hormone and metabolic factors that influence glucagon sensitivity. In this study we report that the bioactive lipid lysophosphatidic acid (LPA) regulates hepatocyte glucose production by antagonizing glucagon-induced expression of the gluconeogenic enzyme phosphoenolpyruvate carboxykinase (PEPCK). Treatment of primary hepatocytes with exogenous LPA blunted glucagon-induced PEPCK expression and glucose production. Similarly, knockout mice lacking the LPA-degrading enzyme phospholipid phosphate phosphatase type 1 (PLPP1) had a 2-fold increase in endogenous LPA levels, reduced PEPCK levels during fasting, and decreased hepatic gluconeogenesis in response to a pyruvate challenge. Mechanistically, LPA antagonized glucagon-mediated inhibition of STAT3, a transcriptional repressor of PEPCK. Importantly, LPA did not blunt glucagon-stimulated glucose production or PEPCK expression in hepatocytes lacking STAT3. These data identify a novel role for PLPP1 activity and hepatocyte LPA levels in glucagon sensitivity via a mechanism involving STAT3.


Subject(s)
Glucagon/metabolism , Gluconeogenesis , Hepatocytes/metabolism , Lysophospholipids/metabolism , Phosphatidate Phosphatase/metabolism , STAT3 Transcription Factor/metabolism , Animals , Glucagon/administration & dosage , Glucose/biosynthesis , Mice , Mice, Knockout , Phosphoenolpyruvate Carboxykinase (GTP)/metabolism , STAT3 Transcription Factor/genetics
13.
Nat Commun ; 8: 14689, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28290443

ABSTRACT

The metabolic pathway of de novo lipogenesis is frequently upregulated in human liver tumours, and its upregulation is associated with poor prognosis. Blocking lipogenesis in cultured liver cancer cells is sufficient to decrease cell viability; however, it is not known whether blocking lipogenesis in vivo can prevent liver tumorigenesis. Herein, we inhibit hepatic lipogenesis in mice by liver-specific knockout of acetyl-CoA carboxylase (ACC) genes and treat the mice with the hepatocellular carcinogen diethylnitrosamine (DEN). Unexpectedly, mice lacking hepatic lipogenesis have a twofold increase in tumour incidence and multiplicity compared to controls. Metabolomics analysis of ACC-deficient liver identifies a marked increase in antioxidants including NADPH and reduced glutathione. Importantly, supplementing primary wild-type hepatocytes with glutathione precursors improves cell survival following DEN treatment to a level indistinguishable from ACC-deficient primary hepatocytes. This study shows that lipogenesis is dispensable for liver tumorigenesis in mice treated with DEN, and identifies an important role for ACC enzymes in redox regulation and cell survival.


Subject(s)
Acetyl-CoA Carboxylase/genetics , Carcinogenesis/metabolism , Carcinoma, Hepatocellular/metabolism , Cell Survival/genetics , Lipogenesis/genetics , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms/metabolism , Liver/metabolism , Acetyl-CoA Carboxylase/metabolism , Alkylating Agents/toxicity , Animals , Antioxidants , Carcinogenesis/drug effects , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Cell Line, Tumor , Cell Survival/drug effects , Diethylnitrosamine/toxicity , Glutathione/metabolism , Hep G2 Cells , Humans , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Liver/drug effects , Liver Neoplasms/genetics , Liver Neoplasms, Experimental/chemically induced , Liver Neoplasms, Experimental/genetics , Metabolomics , Mice , Mice, Knockout , NADP/metabolism
14.
Metabolism ; 68: 150-162, 2017 03.
Article in English | MEDLINE | ID: mdl-28183447

ABSTRACT

BACKGROUND: Obese individuals present with an increased inflammatory tone as compared to healthy, normal-weight individuals, which is associated with insulin resistance. One factor hypothesized to contribute to increased inflammation in obese and diabetic states is elevated blood endotoxin levels, a condition known as metabolic endotoxemia. In non-obese and insulin sensitive individuals, circulating endotoxin concentrations fluctuate over the course of the day with elevations in the post-prandial state that return to baseline levels in the post-absorptive state. Evidence suggests that high-fat feeding alters these fluctuations causing endotoxin levels to remain high throughout the day. The effects of alterations in endotoxin levels on glucose metabolism are not clearly understood. PURPOSE/PROCEDURES: The goal of this study was to determine the effects of both short-term and long-term increases in endotoxin (lipopolysaccharide, LPS) of a low magnitude on the glucose tolerance and insulin signaling in a human primary cell line as well as the effects of short-term endotoxin treatments on glucose homeostasis in a C57/Bl6 mouse model. First, we tested the hypothesis that short-term low-dose endotoxin treatments would augment insulin signaling and glycogen synthesis while long-term treatments would be disruptive in the cell culture model. Second, we examined if these short-term low dose treatments of endotoxin would contribute to similar improvements in whole-body glucose homeostasis in a mouse model. MAIN FINDINGS: Contrary to our initial hypothesis, short-term endotoxin treatment had no effect on insulin signaling or glycogen synthesis, however long-term treatment indeed decreased glycogen synthesis (P<.05). Interestingly, short-term endotoxin treatment resulted in significant improvements in glucose homeostasis in the mouse model (P<.01); which is believed to be at least partly attributed to an inhibitory action of LPS on liver glucose production. CONCLUSIONS: This research shows that low-magnitude, short-term changes in LPS can have significant effects on whole body glucose metabolism and this likely occurs through its direct actions on the liver. Additional studies are necessary to understand the mechanisms responsible for altered glucose metabolism in response to low magnitude changes in LPS levels.


Subject(s)
Endotoxins/pharmacology , Glucose/metabolism , Homeostasis/drug effects , Lipopolysaccharides/pharmacology , Animals , Cell Line , Gluconeogenesis/drug effects , Glucose Tolerance Test , Glucose Transporter Type 4/metabolism , Glycogen/biosynthesis , Humans , Insulin/physiology , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Signal Transduction/drug effects
15.
Sci Rep ; 6: 22292, 2016 Feb 29.
Article in English | MEDLINE | ID: mdl-26924712

ABSTRACT

Overnutrition can promote liver cancer in mice and humans that have liver damage caused by alcohol, viruses, or carcinogens. However, the mechanism linking diet to increased liver tumorigenesis remains unclear in the context of whether tumorigenesis is secondary to obesity, or whether nutrients like sugar or fat drive tumorigenesis independent of obesity. In male mice, liver tumor burden was recently found to correlate with sugar intake, independent of dietary fat intake and obesity. However, females are less susceptible to developing liver cancer than males, and it remains unclear how nutrition affects tumorigenesis in females. Herein, female mice were exposed to the liver carcinogen diethylnitrosamine (DEN) and fed diets with well-defined sugar and fat content. Mice fed diets with high sugar content had the greatest liver tumor incidence while dietary fat intake was not associated with tumorigenesis. Diet-induced postprandial hyperglycemia and fasting hyperinsulinemia significantly correlated with tumor incidence, while tumor incidence was not associated with obesity and obesity-related disorders including liver steatosis, glucose intolerance, or elevated serum levels of estrogen, ALT, and lipids. These results simplify the pathophysiology of diet-induced liver tumorigenesis by focusing attention on the role of sugar metabolism and reducing emphasis on the complex milieu associated with obesity.


Subject(s)
Dietary Sucrose , Liver Neoplasms/etiology , Adiposity , Animal Feed , Animals , Body Weight , Carcinogens/toxicity , Dietary Fats , Diethylnitrosamine/adverse effects , Fatty Liver/etiology , Fatty Liver/pathology , Glucose Tolerance Test , Humans , Incidence , Insulin/metabolism , Liver Neoplasms/epidemiology , Liver Neoplasms/pathology , Mice , Risk Factors , Sex Factors
16.
Am J Physiol Endocrinol Metab ; 310(4): E276-88, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26625902

ABSTRACT

The related Rab GTPase-activating proteins (Rab GAPs) AS160 and Tbc1d1 regulate the trafficking of the glucose transporter GLUT4 that controls glucose uptake in muscle and fat cells and glucose homeostasis. AS160- and Tbc1d1-deficient mice exhibit different adipocyte- and skeletal muscle-specific defects in glucose uptake, GLUT4 expression and trafficking, and glucose homeostasis. A recent study analyzed male mice with simultaneous deletion of AS160 and Tbc1d1 (AS160(-/-)/Tbc1d1(-/-) mice). Herein, we describe abnormalities in male and female AS160(-/-)/Tbc1d1(-/-) mice on another strain background. We confirm the earlier observation that GLUT4 expression and glucose uptake defects of single-knockout mice join in AS160(-/-)/Tbc1d1(-/-) mice to affect all skeletal muscle and adipose tissues. In large mixed fiber-type skeletal muscles, changes in relative basal GLUT4 plasma membrane association in AS160(-/-) and Tbc1d1(-/-) mice also combine in AS160(-/-)/Tbc1d1(-/-) mice. However, we found different glucose uptake abnormalities in isolated skeletal muscles and adipocytes than reported previously, resulting in different interpretations of how AS160 and Tbc1d1 regulate GLUT4 translocation to the cell surface. In support of a larger role for AS160 in glucose homeostasis, in contrast with the previous study, we find similarly impaired glucose and insulin tolerance in AS160(-/-)/Tbc1d1(-/-) and AS160(-/-) mice. However, in vivo glucose uptake abnormalities in AS160(-/-)/Tbc1d1(-/-) skeletal muscles differ from those observed previously in AS160(-/-) mice, indicating additional defects due to Tbc1d1 deletion. Similar to AS160- and Tbc1d1-deficient mice, AS160(-/-)/Tbc1d1(-/-) mice show sex-specific abnormalities in glucose and energy homeostasis. In conclusion, our study supports nonredundant functions for AS160 and Tbc1d1.


Subject(s)
Adipocytes/metabolism , Adipose Tissue/metabolism , Blood Glucose/metabolism , Energy Metabolism/genetics , GTPase-Activating Proteins/genetics , Glucose Transporter Type 4/metabolism , Homeostasis/genetics , Muscle, Skeletal/metabolism , Animals , Female , Glucose/metabolism , Insulin/metabolism , Insulin Resistance , Male , Mice , Mice, Knockout , Muscle Fibers, Skeletal/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Sex Factors
17.
Mol Metab ; 4(9): 610-8, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26413467

ABSTRACT

OBJECTIVE: Defective glucose uptake in adipocytes leads to impaired metabolic homeostasis and insulin resistance, hallmarks of type 2 diabetes. Extracellular ATP-derived nucleotides and nucleosides are important regulators of adipocyte function, but the pathway for controlled ATP release from adipocytes is unknown. Here, we investigated whether Pannexin 1 (Panx1) channels control ATP release from adipocytes and contribute to metabolic homeostasis. METHODS: We assessed Panx1 functionality in cultured 3T3-L1 adipocytes and in adipocytes isolated from murine white adipose tissue by measuring ATP release in response to known activators of Panx1 channels. Glucose uptake in cultured 3T3-L1 adipocytes was measured in the presence of Panx1 pharmacologic inhibitors and in adipocytes isolated from white adipose tissue from wildtype (WT) or adipocyte-specific Panx1 knockout (AdipPanx1 KO) mice generated in our laboratory. We performed in vivo glucose uptake studies in chow fed WT and AdipPanx1 KO mice and assessed insulin resistance in WT and AdipPanx1 KO mice fed a high fat diet for 12 weeks. Panx1 channel function was assessed in response to insulin by performing electrophysiologic recordings in a heterologous expression system. Finally, we measured Panx1 mRNA in human visceral adipose tissue samples by qRT-PCR and compared expression levels with glucose levels and HOMA-IR measurements in patients. RESULTS: Our data show that adipocytes express functional Pannexin 1 (Panx1) channels that can be activated to release ATP. Pharmacologic inhibition or selective genetic deletion of Panx1 from adipocytes decreased insulin-induced glucose uptake in vitro and in vivo and exacerbated diet-induced insulin resistance in mice. Further, we identify insulin as a novel activator of Panx1 channels. In obese humans Panx1 expression in adipose tissue is increased and correlates with the degree of insulin resistance. CONCLUSIONS: We show that Panx1 channel activity regulates insulin-stimulated glucose uptake in adipocytes and thus contributes to control of metabolic homeostasis.

18.
Am J Physiol Endocrinol Metab ; 309(3): E233-45, 2015 Aug 01.
Article in English | MEDLINE | ID: mdl-26015432

ABSTRACT

Tbc1d1 is a Rab GTPase-activating protein (GAP) implicated in regulating intracellular retention and cell surface localization of the glucose transporter GLUT4 and thus glucose uptake in a phosphorylation-dependent manner. Tbc1d1 is most abundant in skeletal muscle but is expressed at varying levels among different skeletal muscles. Previous studies with male Tbc1d1-deficient (Tbc1d1(-/-)) mice on standard and high-fat diets established a role for Tbc1d1 in glucose, lipid, and energy homeostasis. Here we describe similar, but also additional abnormalities in male and female Tbc1d1(-/-) mice. We corroborate that Tbc1d1 loss leads to skeletal muscle-specific and skeletal muscle type-dependent abnormalities in GLUT4 expression and glucose uptake in female and male mice. Using subcellular fractionation, we show that Tbc1d1 controls basal intracellular GLUT4 retention in large skeletal muscles. However, cell surface labeling of extensor digitorum longus muscle indicates that Tbc1d1 does not regulate basal GLUT4 cell surface exposure as previously suggested. Consistent with earlier observations, female and male Tbc1d1(-/-) mice demonstrate increased energy expenditure and skeletal muscle fatty acid oxidation. Interestingly, we observe sex-dependent differences in in vivo phenotypes. Female, but not male, Tbc1d1(-/-) mice have decreased body weight and impaired glucose and insulin tolerance, but only male Tbc1d1(-/-) mice show increased lipid clearance after oil gavage. We surmise that similar changes at the tissue level cause differences in whole-body metabolism between male and female Tbc1d1(-/-) mice and between male Tbc1d1(-/-) mice in different studies due to variations in body composition and nutrient handling.


Subject(s)
Energy Metabolism , GTPase-Activating Proteins/metabolism , Glucose Transporter Type 4/metabolism , Glucose/metabolism , Homeostasis , Lipid Metabolism , Muscle, Skeletal/metabolism , Animals , Biological Transport , Female , GTPase-Activating Proteins/genetics , Gene Deletion , Gene Expression Regulation , Glucose Transporter Type 4/genetics , Insulin Resistance , Male , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Organ Specificity , Protein Transport , Sex Characteristics
19.
Am J Physiol Endocrinol Metab ; 303(10): E1273-86, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23011063

ABSTRACT

Tight control of glucose uptake in skeletal muscles and adipocytes is crucial to glucose homeostasis and is mediated by regulating glucose transporter GLUT4 subcellular distribution. In cultured cells, Rab GAP AS160 controls GLUT4 intracellular retention and release to the cell surface and consequently regulates glucose uptake into cells. To determine AS160 function in GLUT4 trafficking in primary skeletal muscles and adipocytes and investigate its role in glucose homeostasis, we characterized AS160 knockout (AS160(-/-)) mice. We observed increased and normal basal glucose uptake in isolated AS160(-/-) adipocytes and soleus, respectively, while insulin-stimulated glucose uptake was impaired and GLUT4 expression decreased in both. No such abnormalities were found in isolated AS160(-/-) extensor digitorum longus muscles. In plasma membranes isolated from AS160(-/-) adipose tissue and gastrocnemius/quadriceps, relative GLUT4 levels were increased under basal conditions and remained the same after insulin treatment. Concomitantly, relative levels of cell surface-exposed GLUT4, determined with a glucose transporter photoaffinity label, were increased in AS160(-/-) adipocytes and normal in AS160(-/-) soleus under basal conditions. Insulin augmented cell surface-exposed GLUT4 in both. These observations suggest that AS160 is essential for GLUT4 intracellular retention and regulation of glucose uptake in adipocytes and skeletal muscles in which it is normally expressed. In vivo studies revealed impaired insulin tolerance in the presence of normal (male) and impaired (female) glucose tolerance. Concurrently, insulin-elicited increases in glucose disposal were abolished in all AS160(-/-) skeletal muscles and liver but not in AS160(-/-) adipose tissues. This suggests AS160 as a target for differential manipulation of glucose homeostasis.


Subject(s)
Adipocytes/metabolism , GTPase-Activating Proteins/deficiency , Glucose Transporter Type 4/metabolism , Glucose/metabolism , Insulin/metabolism , Muscle, Skeletal/metabolism , Animals , Female , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Glucose Tolerance Test , Homeostasis , Immunoblotting , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/chemistry , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
20.
FEBS Lett ; 583(12): 2142-7, 2009 Jun 18.
Article in English | MEDLINE | ID: mdl-19500580

ABSTRACT

Interfering RNA was used to suppress the expression of the genes At1g06680 and At2g30790 in Arabidopsis thaliana, which encode the PsbP-1 and PsbP-2 proteins, respectively, of Photosystem II. A phenotypic series of transgenic plants was recovered that expressed intermediate and low amounts of PsbP. Earlier we had documented significant alterations in a variety of Photosystem II parameters in these plant lines [Yi, X., Liu, H., Hargett, S. R., Frankel, L. K., Bricker, T. M. (2007). The PsbP protein is required for photosystem II complex assembly/stability and photoautotrophy in Arabidopsis thaliana. J. Biol. Chem. 34, 24833-24841]. In this communication, we document extensive defects in the thylakoid membrane architecture of these plants. Interestingly, strong interfering RNA suppression of the genes encoding the PsbQ protein (At4g21280 and At4g05180) was found to have no effect on the architecture of thylakoid membranes.


Subject(s)
Arabidopsis Proteins/metabolism , Arabidopsis/metabolism , Arabidopsis/ultrastructure , Photosystem II Protein Complex/metabolism , Thylakoids/metabolism , Thylakoids/ultrastructure , Arabidopsis/genetics , Arabidopsis Proteins/antagonists & inhibitors , Arabidopsis Proteins/genetics , Microscopy, Electron, Transmission , Phenotype , Photosystem II Protein Complex/antagonists & inhibitors , Photosystem II Protein Complex/genetics , Plants, Genetically Modified , RNA Interference
SELECTION OF CITATIONS
SEARCH DETAIL
...