Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 37
1.
J Biol Chem ; 299(12): 105405, 2023 Dec.
Article En | MEDLINE | ID: mdl-38229396

Gestational diabetes mellitus (GDM) is characterized by glucose intolerance in pregnant women without a previous diagnosis of diabetes. While the etiology of GDM remains elusive, the close association of GDM with increased maternal adiposity and advanced gestational age implicates insulin resistance as a culpable factor for the pathogenesis of GDM. Pregnancy is accompanied by the physiological induction of insulin resistance in the mother secondary to maternal weight gain. This effect serves to spare blood glucose for the fetus. To overcome insulin resistance, maternal ß-cells are conditioned to release more insulin into the blood. Such an adaptive response, termed ß-cell compensation, is essential for maintaining normal maternal metabolism. ß-cell compensation culminates in the expansion of ß-cell mass and augmentation of ß-cell function, accounting for increased insulin synthesis and secretion. As a result, a vast majority of mothers are protected from developing GDM during pregnancy. In at-risk pregnant women, ß-cells fail to compensate for maternal insulin resistance, contributing to insulin insufficiency and GDM. However, gestational ß-cell compensation ensues in early pregnancy, prior to the establishment of insulin resistance in late pregnancy. How ß-cells compensate for pregnancy and what causes ß-cell failure in GDM are subjects of investigation. In this mini-review, we will provide clinical and preclinical evidence that ß-cell compensation is pivotal for overriding maternal insulin resistance to protect against GDM. We will highlight key molecules whose functions are critical for integrating gestational hormones to ß-cell compensation for pregnancy. We will provide mechanistic insights into ß-cell decompensation in the etiology of GDM.


Diabetes, Gestational , Insulin Resistance , Insulin-Secreting Cells , Female , Humans , Pregnancy , Blood Glucose/metabolism , Diabetes, Gestational/pathology , Glucose Tolerance Test , Insulin , Insulin-Secreting Cells/physiology
2.
J Clin Invest ; 132(14)2022 07 15.
Article En | MEDLINE | ID: mdl-35700043

Hepatic inflammation is culpable for the evolution of asymptomatic steatosis to nonalcoholic steatohepatitis (NASH). Hepatic inflammation results from abnormal macrophage activation. We found that FoxO1 links overnutrition to hepatic inflammation by regulating macrophage polarization and activation. FoxO1 was upregulated in hepatic macrophages, correlating with hepatic inflammation, steatosis, and fibrosis in mice and patients with NASH. Myeloid cell conditional FoxO1 knockout skewed macrophage polarization from proinflammatory M1 to the antiinflammatory M2 phenotype, accompanied by a reduction in macrophage infiltration in liver. These effects mitigated overnutrition-induced hepatic inflammation and insulin resistance, contributing to improved hepatic metabolism and increased energy expenditure in myeloid cell FoxO1-knockout mice on a high-fat diet. When fed a NASH-inducing diet, myeloid cell FoxO1-knockout mice were protected from developing NASH, culminating in a reduction in hepatic inflammation, steatosis, and fibrosis. Mechanistically, FoxO1 counteracts Stat6 to skew macrophage polarization from M2 toward the M1 signature to perpetuate hepatic inflammation in NASH. FoxO1 appears to be a pivotal mediator of macrophage activation in response to overnutrition and a therapeutic target for ameliorating hepatic inflammation to stem the disease progression from benign steatosis to NASH.


Forkhead Box Protein O1 , Non-alcoholic Fatty Liver Disease , Overnutrition , Animals , Diet, High-Fat/adverse effects , Disease Models, Animal , Fibrosis , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Inflammation/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/chemically induced , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/prevention & control , Overnutrition/pathology
3.
J Biol Chem ; 295(20): 7003-7017, 2020 05 15.
Article En | MEDLINE | ID: mdl-32273342

Cholelithiasis is one of the most prevalent gastroenterological diseases and is characterized by the formation of gallstones in the gallbladder. Both clinical and preclinical data indicate that obesity, along with comorbidity insulin resistance, is a predisposing factor for cholelithiasis. Forkhead box O1 (FoxO1) is a key transcription factor that integrates insulin signaling with hepatic metabolism and becomes deregulated in the insulin-resistant liver, contributing to dyslipidemia in obesity. To gain mechanistic insights into how insulin resistance is linked to cholelithiasis, here we determined FoxO1's role in bile acid homeostasis and its contribution to cholelithiasis. We hypothesized that hepatic FoxO1 deregulation links insulin resistance to impaired bile acid metabolism and cholelithiasis. To address this hypothesis, we used the FoxO1LoxP/LoxP-Albumin-Cre system to generate liver-specific FoxO1-knockout mice. FoxO1-knockout mice and age- and sex-matched WT littermates were fed a lithogenic diet, and bile acid metabolism and gallstone formation were assessed in these animals. We showed that FoxO1 affected bile acid homeostasis by regulating hepatic expression of key enzymes in bile acid synthesis and in biliary cholesterol and phospholipid secretion. Furthermore, FoxO1 inhibited hepatic expression of the bile acid receptor farnesoid X receptor and thereby counteracted hepatic farnesoid X receptor signaling. Nonetheless, hepatic FoxO1 depletion neither affected the onset of gallstone disease nor impacted the disease progression, as FoxO1-knockout and control mice of both sexes had similar gallstone weights and incidence rates. These results argue against the notion that FoxO1 is a link between insulin resistance and cholelithiasis.


Bile Acids and Salts/metabolism , Forkhead Box Protein O1/metabolism , Gallstones/metabolism , Insulin Resistance , Signal Transduction , Animals , Bile Acids and Salts/genetics , Cholesterol/genetics , Cholesterol/metabolism , Female , Forkhead Box Protein O1/genetics , Gallstones/genetics , Gene Deletion , Gene Expression Regulation , Liver , Male , Mice , Mice, Transgenic , Organ Specificity , Phospholipids/genetics , Phospholipids/metabolism , Receptors, Cytoplasmic and Nuclear/biosynthesis , Receptors, Cytoplasmic and Nuclear/genetics
4.
Diabetologia ; 62(12): 2325-2339, 2019 12.
Article En | MEDLINE | ID: mdl-31511929

AIMS/HYPOTHESIS: Absent in melanoma 2 (AIM2) is a cytosolic sensor for double-stranded DNA and a tumour suppressor. Binding of double-stranded DNA to AIM2 forms the AIM2 inflammasome, leading to activation of caspase-1 and production of IL-1ß and IL-18. Although inflammasome-independent effects of AIM2 have been reported, its role in energy metabolism is unknown. We aimed to evaluate the effect of AIM2 in energy metabolism and glucose homeostasis. METHODS: Male and female whole body Aim2 knockout (Aim2-/-) mice were used in the current study. Body weight, food intake, body composition, energy expenditure, fasting blood glucose levels, GTT and body temperature were measured at indicated time points. RNA sequencing was carried out on gonadal white adipose tissue (gWAT) in 14-month-old female mice. mRNA and protein levels in tissues were analysed by quantitative real-time PCR and immunoblot. Immune cell infiltration in gWAT was examined by flow cytometry. Stromal vascular fractions isolated from gWAT were used to investigate adipocyte differentiation. RESULTS: Male and female Aim2-/- mice were obese compared with wild-type controls from 7 weeks of age until 51 weeks of age, with increased adiposity in both subcutaneous and visceral fat depots. While there were no differences in food intake, Aim2-/- mice demonstrated decreased energy expenditure and impaired brown adipose tissue function compared with wild-type controls. Fasting glucose and insulin levels were elevated, and Aim2-/- mice were glucose intolerant on intraperitoneal GTT. RNA sequencing revealed marked upregulation of the IFN-inducible gene Ifi202b, which encodes protein 202 (p202) and elevated inflammatory signalling in gWAT of Aim2-/- mice. Increased infiltration of total and Ly6Clow monocytes was noted at 8 weeks of age in gWAT, before the onset of obesity and insulin resistance. Ifi202b knockdown blocked adipogenesis in stromal vascular fractions and reduced inflammation in bone marrow-derived macrophages, demonstrating a key role of p202 in mediating the increased adipogenesis and inflammation in Aim2-/- mice. CONCLUSIONS/INTERPRETATION: These results demonstrate a fundamental role for AIM2 in energy metabolism, inflammation and insulin resistance. Our studies establish a novel link between the innate immunity proteins, AIM2 and p202, and metabolism.


Adipogenesis/genetics , Adipose Tissue, White/metabolism , DNA-Binding Proteins/metabolism , Inflammation/metabolism , Insulin Resistance/genetics , Obesity/metabolism , Adipose Tissue, Brown/metabolism , Adiposity/genetics , Animals , Blood Glucose/metabolism , Body Temperature/genetics , DNA-Binding Proteins/genetics , Eating/genetics , Energy Metabolism/genetics , Fasting/metabolism , Female , Inflammation/genetics , Male , Mice , Mice, Knockout , Obesity/genetics
5.
Aging (Albany NY) ; 11(12): 4125-4144, 2019 06 25.
Article En | MEDLINE | ID: mdl-31246177

Decreased forkhead box O1 (FoxO1) activity induces hyperlipidemia and increased PPARγ, leading to hyperlipidemia in association with endoplasmic reticulum (ER) stress. In the liver, aging and comorbidities such as hyperlipidemia and diabetes significantly influence a wide variety of steatosis, but the underlying mechanisms are complex and remain elusive.To establish the modulatory role of FoxO1 and the functional consequences of its altered interaction with PPARγ in the present study, we utilized a cell culture system, aged rats and diabetic db/db mice.We found that, under ER stress, FoxO1 induces PPARγ-mediated lipid accumulation in aged rat livers. Our data showed that the FoxO1-induced hepatic lipid accumulation was negatively regulated by Akt signaling. PPARγ, a key lipogenesis transcription factor, was increased in aged liver, resulting in lipid accumulation via hepatic ER stress under hyperglycemic conditions. We further demonstrated that loss of FoxO1 causes a decline in PPARγ expression and reduces lipid accumulation. In addition, the interaction between FoxO1 and PPARγ was shown to induce hepatic steatosis in aging and db/db mice.We provide evidence that, in aged rats, FoxO1 interaction with PPARγ promotes hepatic steatosis, due to hyperglycemia-induced ER stress, which causes an impairment in Akt signaling, such in aging-related diabetes.


Aging , Endoplasmic Reticulum Stress , Fatty Liver , Nerve Tissue Proteins/metabolism , PPAR gamma/metabolism , Animals , Cell Line, Tumor , Glucose/metabolism , Lipid Metabolism , Male , Nerve Tissue Proteins/genetics , PPAR gamma/genetics , Rats , Rats, Sprague-Dawley
6.
Am J Pathol ; 189(7): 1413-1422, 2019 07.
Article En | MEDLINE | ID: mdl-31054988

Obesity is a major risk factor for type 2 diabetes because of chronic hepatic inflammation and resultant insulin resistance. Hepatocyte growth factor (HGF) is responsible for resetting hepatic homeostasis after injury following activation by urokinase-type plasminogen activator (u-PA; encoded by the PLAU gene). Plasminogen activator inhibitor type-1 (PAI-1; encoded by the SERPINE1 gene), a u-PA inhibitor and antifibrinolytic agent, is often elevated in obesity and is linked to cardiovascular events. We hypothesized that, in addition to its role in preventing fibrinolysis, elevated PAI-1 inhibits HGF's activation by u-PA and the resultant anti-inflammatory and hepatoprotective properties. Wild-type and PAI-1 knockout (KO) mice on a high-fat diet both became significantly heavier than lean controls; however, the obese KO mice demonstrated improved glucose metabolism compared with wild-type mice. Obese KO mice also exhibited an increase in conversion of latent single-chain HGF to active two-chain HGF, coinciding with an increase in the phosphorylation of the HGF receptor (HGFR or MET, encoded by the MET gene), as well as dampened inflammation. These results strongly suggest that, in addition to its other functions, PAI-mediated inhibition of HGF activation prohibits the resolution of inflammation in the context of obesity-induced type 2 diabetes.


Diabetes Mellitus, Type 2/metabolism , Obesity/metabolism , Plasminogen Activator Inhibitor 1/metabolism , Animals , Diabetes Mellitus, Type 2/chemically induced , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Dietary Fats/adverse effects , Dietary Fats/pharmacology , Hepatocyte Growth Factor/genetics , Hepatocyte Growth Factor/metabolism , Inflammation/chemically induced , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Knockout , Obesity/chemically induced , Obesity/genetics , Obesity/pathology , Plasminogen Activator Inhibitor 1/genetics , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism
7.
Redox Biol ; 24: 101184, 2019 06.
Article En | MEDLINE | ID: mdl-30974318

FoxO has been proposed to play a role in the promotion of insulin resistance, and inflammation. FoxO is a pro-inflammatory transcription factor that is a key mediator of generation of inflammatory cytokines such as IL-1ß in the liver. However, the detailed association of FoxO6 with insulin resistance and age-related inflammation has not been fully documented. Here, we showed that FoxO6 was elevated in the livers of aging rats and obese mice that exhibited insulin resistance. In addition, virus-mediated FoxO6 activation led to insulin resistance in mice with a notable increase in PAR2 and inflammatory signaling in the liver. On the other hand, FoxO6-KO mice showed reduced PAR2 signaling with a decrease in inflammatory cytokine expression and elevated insulin signaling. Because FoxO6 is closely associated with abnormal production of IL-1ß in the liver, we focused on the FoxO6/IL-1ß/PAR2 axis to further examine mechanisms underlying FoxO6-mediated insulin resistance and inflammation in the liver. In vitro experiments showed that FoxO6 directly binds to and elevates IL-1ß expression. In turn, IL-1ß treatment elevated the protein levels of PAR2 with a significant decrease in hepatic insulin signaling, whereas PAR2-siRNA treatment abolished these effects. However, PAR2-siRNA treatment had no effect on IL-1ß expression induced by FoxO6, indicating that IL-1ß may not be downstream of PAR2. Taken together, we assume that FoxO6-mediated IL-1ß is involved in hepatic inflammation and insulin resistance via TF/PAR2 pathway in the liver.


Forkhead Transcription Factors/genetics , Inflammation/etiology , Inflammation/metabolism , Insulin Resistance , Interleukin-1beta/metabolism , Liver/metabolism , Aging/genetics , Aging/metabolism , Animals , Cytokines/metabolism , Diabetes Mellitus, Experimental , Forkhead Transcription Factors/metabolism , Gene Expression , Hep G2 Cells , Humans , Insulin/metabolism , Male , Mice , Models, Biological , Obesity/etiology , Obesity/metabolism , Protein Binding , Receptor, PAR-2/metabolism , Signal Transduction
8.
Antioxidants (Basel) ; 6(4)2017 Nov 01.
Article En | MEDLINE | ID: mdl-29104232

Oxidative stress and persistent inflammation are exaggerated through chronic over-nutrition and a sedentary lifestyle, resulting in insulin resistance. In type 2 diabetes (T2D), impaired insulin signaling leads to hyperglycemia and long-term complications, including metabolic liver dysfunction, resulting in non-alcoholic fatty liver disease (NAFLD). The manganese metalloporphyrin superoxide dismustase (SOD) mimetic, manganese (III) meso-tetrakis (N-ethylpyridinium-2-yl) porphyrin (MnP), is an oxidoreductase known to scavenge reactive oxygen species (ROS) and decrease pro-inflammatory cytokine production, by inhibiting nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) activation. We hypothesized that targeting oxidative stress-induced inflammation with MnP would assuage liver complications and enhance insulin sensitivity and glucose tolerance in a high-fat diet (HFD)-induced mouse model of T2D. During 12 weeks of feeding, we saw significant improvements in weight, hepatic steatosis, and biomarkers of liver dysfunction with redox modulation by MnP treatment in HFD-fed mice. Additionally, MnP treatment improved insulin sensitivity and glucose tolerance, while reducing serum insulin and leptin levels. We attribute these effects to redox modulation and inhibition of hepatic NF-κB activation, resulting in diminished ROS and pro-inflammatory cytokine production. This study highlights the importance of controlling oxidative stress and secondary inflammation in obesity-mediated insulin resistance and T2D. Our data confirm the role of NF-κB-mediated inflammation in the development of T2D, and demonstrate the efficacy of MnP in preventing the progression to disease by specifically improving liver pathology and hepatic insulin resistance in obesity.

9.
Endocrinology ; 158(11): 4093-4104, 2017 11 01.
Article En | MEDLINE | ID: mdl-28938414

Estrogen sulfotransferase catalyzes the sulfoconjugation and deactivation of estrogens. Previously, we showed that loss of Est in male ob/ob mice, but not in female ob/ob mice, exacerbated the diabetic phenotype, but the underlying mechanism was unclear. In this study, we show that transgenic reconstitution of Est in the adipose tissue, but not in the liver, attenuated diabetic phenotype in Est-deficient ob/ob mice (obe mice). Mechanistically, adipose reconstitution of Est in obe mice (oae mice) resulted in reduced local and systemic inflammation, improved insulin sensitivity, and increased energy expenditure. At the molecular level, adipose induction of lipocalin-2 (Lcn2) in oae males may have contributed to the inhibition of inflammation because the level of Lcn2 was negatively associated with tumor necrosis factor (Tnf) α expression, and treatment of differentiated adipocytes with Lcn2 antagonized Tnfα-responsive inhibition of insulin signaling. The metabolic benefit of adipose reconstitution of Est was sex specific, because adipose reconstitution of Est in obe females had little effect. Interestingly, despite their improved metabolic functions, obe male mice with reconstituted Est in their adipose tissue failed to ameliorate the impairment of the structure and function of the pancreatic islets. In summary, our study uncovers a crucial adipose- and male-specific role of Est in maintaining the whole-body energy homeostasis.


Energy Metabolism/genetics , Insulin Resistance/genetics , Sulfotransferases/physiology , 3T3-L1 Cells , Adipose Tissue/metabolism , Adiposity/genetics , Animals , Cells, Cultured , Female , Homeostasis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Mice, Transgenic , Organ Specificity/genetics , Sex Factors
10.
J Endocrinol ; 233(2): R67-R79, 2017 05.
Article En | MEDLINE | ID: mdl-28213398

The forkhead box O family consists of FoxO1, FoxO3, FoxO4 and FoxO6 proteins in mammals. Expressed ubiquitously in the body, the four FoxO isoforms share in common the amino DNA-binding domain, known as 'forkhead box' domain. They mediate the inhibitory action of insulin or insulin-like growth factor on key functions involved in cell metabolism, growth, differentiation, oxidative stress, senescence, autophagy and aging. Genetic mutations in FoxO genes or abnormal expression of FoxO proteins are associated with metabolic disease, cancer or altered lifespan in humans and animals. Of the FoxO family, FoxO6 is the least characterized member and is shown to play pivotal roles in the liver, skeletal muscle and brain. Altered FoxO6 expression is associated with the pathogenesis of insulin resistance, dietary obesity and type 2 diabetes and risk of neurodegeneration disease. FoxO6 is evolutionally divergent from other FoxO isoforms. FoxO6 mediates insulin action on target genes in a mechanism that is fundamentally different from other FoxO members. Here, we focus our review on the role of FoxO6, in contrast with other FoxO isoforms, in health and disease. We review the distinctive mechanism by which FoxO6 integrates insulin signaling to hepatic glucose and lipid metabolism. We highlight the importance of FoxO6 dysregulation in the dual pathogenesis of fasting hyperglycemia and hyperlipidemia in diabetes. We review the role of FoxO6 in memory consolidation and its contribution to neurodegeneration disease and aging. We discuss the potential therapeutic option of pharmacological FoxO6 inhibition for improving glucose and lipid metabolism in diabetes.


Forkhead Transcription Factors/metabolism , Insulin/metabolism , Lipid Metabolism/physiology , Signal Transduction/physiology , Animals , Forkhead Transcription Factors/genetics , Gene Expression Regulation/physiology , Insulin/genetics , Mammals , Mutation , Protein Isoforms
11.
J Biol Chem ; 292(9): 3692-3705, 2017 03 03.
Article En | MEDLINE | ID: mdl-28115523

Nonalcoholic fatty liver disease (NAFLD), characterized by excessive fat accumulation in liver, is prevalent in obesity. Genetic factors that link obesity to NAFLD remain obscure. Apolipoprotein C3 (APOC3) is a lipid-binding protein with a pivotal role in triglyceride metabolism. Humans with APOC3 gain-of-function mutations and mice with APOC3 overproduction are associated with hypertriglyceridemia. Nonetheless, it remains controversial whether APOC3 is culpable for diet-induced NAFLD. To address this fundamental issue, we fed APOC3-transgenic and wild-type littermates a high fructose diet or high fat diet, followed by determination of the effect of APOC3 on hepatic lipid metabolism and inflammation and the progression of NAFLD. To gain mechanistic insight into NAFLD, we determined the impact of APOC3 on hepatic triglyceride synthesis and secretion versus fatty acid oxidation. APOC3-transgenic mice were hypertriglyceridemic, culminating in marked elevation of triglycerides, cholesterols, and non-esterified fatty acids in plasma. Despite the prevailing hypertriglyceridemia, APOC3-transgenic mice, relative to wild-type littermates, had similar weight gain and hepatic lipid content without alterations in hepatic expression of key genes involved in triglyceride synthesis and secretion and fatty acid oxidation. APOC3-transgenic and wild-type mice had similar Kupffer cell content without alterations in hepatic expression of pro- and anti-inflammatory cytokines. APOC3 neither exacerbated diet-induced adiposity nor aggravated the degree of steatosis in high fructose or high fat-fed APOC3-transgenic mice. These effects ensued independently of weight gain even after 10-month high fat feeding. We concluded that APOC3, whose dysregulation is liable for hypertriglyceridemia, is not a predisposing factor for linking overnutrition to NAFLD in obesity.


Apolipoprotein C-III/genetics , Apolipoprotein C-III/metabolism , Gene Expression Regulation , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Body Weight , Cholesterol/metabolism , Cytokines/metabolism , Diet, High-Fat , Female , Fructose/chemistry , Genetic Predisposition to Disease , Glucose Tolerance Test , Hypertriglyceridemia/metabolism , Inflammation , Insulin Resistance/genetics , Lipid Metabolism/drug effects , Lipids/chemistry , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Obesity/metabolism , Promoter Regions, Genetic , Transgenes , Triglycerides/metabolism , Weight Gain
12.
J Biol Chem ; 291(28): 14695-705, 2016 Jul 08.
Article En | MEDLINE | ID: mdl-27226540

Hypertriglyceridemia results from increased production and decreased clearance of triglyceride-rich very low-density lipoproteins, a pathological condition that accounts for heightened risk of ischemic vascular diseases in obesity and type 2 diabetes. Despite its intimate association with insulin resistance, whether hypertriglyceridemia constitutes an independent risk for beta cell dysfunction in diabetes is unknown. Answering this fundamental question is stymied by the fact that hypertriglyceridemia is intertwined with hyperglycemia and insulin resistance in obese and diabetic subjects. To circumvent this limitation, we took advantage of apolipoprotein C3 (ApoC3)-transgenic mice, a model with genetic predisposition to hypertriglyceridemia. We showed that ApoC3-transgenic mice, as opposed to age/sex-matched wild-type littermates, develop hypertriglyceridemia with concomitant elevations in plasma cholesterol and non-esterified fatty acid levels. Anti-insulin and anti-glucagon dual immunohistochemistry in combination with morphometric analysis revealed that ApoC3-transgenic and wild-type littermates had similar beta cell and alpha cell masses as well as islet size and architecture. These effects correlated with similar amplitudes of glucose-stimulated insulin secretion and similar degrees of postprandial glucose excursion in ApoC3-transgenic versus wild-type littermates. Oil Red O histology did not visualize lipid infiltration into islets, correlating with the lack of ectopic triglyceride and cholesterol depositions in the pancreata of ApoC3-transgenic versus wild-type littermates. ApoC3-transgenic mice, despite persistent hypertriglyceridemia, maintained euglycemia under both fed and fasting conditions without manifestation of insulin resistance and fasting hyperinsulinemia. Thus, hypertriglyceridemia per se is not an independent risk factor for beta cell dysfunction in ApoC3 transgenic mice.


Apolipoprotein C-III/metabolism , Diabetes Mellitus, Type 2/complications , Hypertriglyceridemia/complications , Insulin-Secreting Cells/pathology , Animals , Apolipoprotein C-III/genetics , Blood Glucose/analysis , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Female , Glucose/metabolism , Humans , Hypertriglyceridemia/blood , Hypertriglyceridemia/metabolism , Hypertriglyceridemia/pathology , Insulin Resistance , Insulin-Secreting Cells/metabolism , Lipid Metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Obesity
13.
Endocrinology ; 157(3): 1055-70, 2016 Mar.
Article En | MEDLINE | ID: mdl-26727107

ß-Cell compensation is an essential mechanism by which ß-cells increase insulin secretion for overcoming insulin resistance to maintain euglycemia in obesity. Failure of ß-cells to compensate for insulin resistance contributes to insulin insufficiency and overt diabetes. To understand the mechanism of ß-cell compensation, we characterized the role of forkhead box O1 (FoxO1) in ß-cell compensation in mice under physiological and pathological conditions. FoxO1 is a key transcription factor that serves as a nutrient sensor for integrating insulin signaling to cell metabolism, growth, and proliferation. We showed that FoxO1 improved ß-cell compensation via 3 distinct mechanisms by increasing ß-cell mass, enhancing ß-cell glucose sensing, and augmenting ß-cell antioxidative function. These effects accounted for increased glucose-stimulated insulin secretion and enhanced glucose tolerance in ß-cell-specific FoxO1-transgenic mice. When fed a high-fat diet, ß-cell-specific FoxO1-transgenic mice were protected from developing fat-induced glucose disorder. This effect was attributable to increased ß-cell mass and function. Furthermore, we showed that FoxO1 activity was up-regulated in islets, correlating with the induction of physiological ß-cell compensation in high-fat-induced obese C57BL/6J mice. These data characterize FoxO1 as a pivotal factor for orchestrating physiological adaptation of ß-cell mass and function to overnutrition and obesity.


Adaptation, Physiological/genetics , Forkhead Transcription Factors/genetics , Insulin Resistance/genetics , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Cell Line, Tumor , Chromatin Immunoprecipitation , Diet, High-Fat , Energy Metabolism , Forkhead Box Protein O1 , Forkhead Transcription Factors/metabolism , Glucose Tolerance Test , Glucose Transporter Type 2/metabolism , Homeodomain Proteins/metabolism , Humans , Immunoblotting , Insulin Secretion , Insulin-Secreting Cells/pathology , Islets of Langerhans/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Organ Size , Pancreas/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Trans-Activators/metabolism
14.
J Diabetes ; 8(3): 314-23, 2016 May.
Article En | MEDLINE | ID: mdl-25851734

BACKGROUND: Hypertriglyceridemia is a common lipid disorder that is characterized by elevated plasma levels of triglyceride (TG)-rich particles, such as very low-density lipoprotein (VLDL), in poorly controlled diabetes. The aim of the present study was to determine the potential therapeutic effect of hepatic insulin production on hypertriglyceridemia in mice. METHODS: Mice were induced diabetic and hypertriglyceridemic by streptozotocin (STZ) treatment. Using an adenovirus-mediated gene transfer approach, we delivered rat preproinsulin cDNA into the liver of diabetic mice and then determined plasma TG metabolism. To investigate the mechanism by which hepatic insulin improves TG metabolism, we determined hepatic expression of apolipoprotein C-III (ApoC-III), a structural moiety and functional inhibitor of VLDL-TG catabolism. RESULTS: Plasma VLDL-TG levels were markedly elevated in STZ-treated mice, and were accompanied by hyperglycemia and hypertriglyceridemia. These metabolic abnormalities were restored to near normal following hepatic insulin production in insulin vector-treated diabetic mice. In contrast, hypertriglyceridemia and hyperglycemia persisted in control vector-treated diabetic animals. Hepatic ApoC-III expression became deregulated secondary to insulin deficiency, contributing to impaired TG metabolism in diabetic mice. Hepatic insulin production suppressed excessive hepatic ApoC-III production to basal levels. CONCLUSION: Hepatic insulin production is efficacious in correcting hypertriglyceridemia associated with insulin deficiency in diabetic mice.


Diabetes Mellitus, Experimental/metabolism , Insulin/pharmacology , Lipid Metabolism/drug effects , Liver/metabolism , Protein Precursors/pharmacology , Animals , Apolipoprotein C-III/genetics , Apolipoprotein C-III/metabolism , Blotting, Western , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/pathology , Fatty Acids, Nonesterified/metabolism , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Immunoenzyme Techniques , Insulin/administration & dosage , Male , Mice , Mice, Inbred C57BL , Protein Precursors/administration & dosage , RNA, Messenger/genetics , Rats , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Triglycerides/metabolism
15.
Am J Physiol Endocrinol Metab ; 309(3): E283-92, 2015 Aug 01.
Article En | MEDLINE | ID: mdl-26058861

Humanin (HN) is an endogenous mitochondria-associated peptide that has been shown to protect against various Alzheimer's disease-associated insults, myocardial ischemia-reperfusion injury, and reactive oxygen species-induced cell death. We have shown previously that HN improves whole body glucose homeostasis by improving insulin sensitivity and increasing glucose-stimulated insulin secretion (GSIS) from the ß-cells. Here, we report that intraperitoneal treatment with one of HN analogs, HNG, decreases body weight gain, visceral fat, and hepatic triglyceride (TG) accumulation in high-fat diet-fed mice. The decrease in hepatic TG accumulation is due to increased activity of hepatic microsomal triglyceride transfer protein (MTTP) and increased hepatic TG secretion. Both intravenous (iv) and intracerebroventricular (icv) infusion of HNG acutely increase TG secretion from the liver. Vagotomy blocks the effect on both iv and icv HNG on TG secretion, suggesting that the effects of HNG on hepatic TG flux are centrally mediated. Our data suggest that HN is a new player in central regulation of peripheral lipid metabolism.


Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Models, Biological , Obesity/metabolism , Triglycerides/metabolism , Adiposity/drug effects , Animals , Anti-Obesity Agents/administration & dosage , Anti-Obesity Agents/pharmacology , Anti-Obesity Agents/therapeutic use , Carrier Proteins/agonists , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Central Nervous System Agents/administration & dosage , Central Nervous System Agents/pharmacology , Central Nervous System Agents/therapeutic use , Diet, High-Fat/adverse effects , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Infusions, Intravenous , Infusions, Intraventricular , Intra-Abdominal Fat/drug effects , Intra-Abdominal Fat/pathology , Intracellular Signaling Peptides and Proteins/administration & dosage , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/pharmacology , Intracellular Signaling Peptides and Proteins/therapeutic use , Liver/drug effects , Liver/pathology , Male , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/etiology , Obesity/pathology , Peptides/administration & dosage , Peptides/pharmacology , Peptides/therapeutic use , Rats, Sprague-Dawley , Reproducibility of Results , Triglycerides/blood , Vagotomy, Truncal
16.
J Biol Chem ; 290(25): 15581-15594, 2015 Jun 19.
Article En | MEDLINE | ID: mdl-25944898

Excessive endogenous glucose production contributes to fasting hyperglycemia in diabetes. FoxO6 is a distinct member of the FoxO subfamily. To elucidate the role of FoxO6 in hepatic gluconeogenesis and assess its contribution to the pathogenesis of fasting hyperglycemia in diabetes, we generated FoxO6 knock-out (FoxO6-KO) mice followed by determining the effect of FoxO6 loss-of-function on hepatic gluconeogenesis under physiological and pathological conditions. FoxO6 depletion attenuated hepatic gluconeogenesis and lowered fasting glycemia in FoxO6-KO mice. FoxO6-deficient primary hepatocytes were associated with reduced capacities to produce glucose in response to glucagon. When fed a high fat diet, FoxO6-KO mice exhibited significantly enhanced glucose tolerance and reduced blood glucose levels accompanied by improved insulin sensitivity. These effects correlated with attenuated hepatic gluconeogenesis in FoxO6-KO mice. In contrast, wild-type littermates developed fat-induced glucose intolerance with a concomitant induction of fasting hyperinsulinemia and hyperglycemia. Furthermore, FoxO6-KO mice displayed significantly diminished macrophage infiltration into liver and adipose tissues, correlating with the reduction of macrophage expression of C-C chemokine receptor 2 (CCR2), a factor that is critical for regulating macrophage recruitment in peripheral tissues. Our data indicate that FoxO6 depletion protected against diet-induced glucose intolerance and insulin resistance by attenuating hepatic gluconeogenesis and curbing macrophage infiltration in liver and adipose tissues in mice.


Dietary Fats/adverse effects , Forkhead Transcription Factors/metabolism , Gluconeogenesis/drug effects , Hyperglycemia/metabolism , Hyperinsulinism/metabolism , Liver/metabolism , Adipose Tissue/metabolism , Adipose Tissue/pathology , Animals , Cells, Cultured , Dietary Fats/pharmacology , Forkhead Transcription Factors/genetics , Gluconeogenesis/genetics , Hyperglycemia/chemically induced , Hyperglycemia/genetics , Hyperglycemia/pathology , Hyperglycemia/prevention & control , Hyperinsulinism/chemically induced , Hyperinsulinism/genetics , Hyperinsulinism/pathology , Hyperinsulinism/prevention & control , Liver/pathology , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Knockout , Receptors, CCR2/genetics , Receptors, CCR2/metabolism
17.
Mol Metab ; 4(1): 70-6, 2015 Jan.
Article En | MEDLINE | ID: mdl-25685692

OBJECTIVE: Type 1 diabetes results from autoimmune destruction of beta-cells in the pancreas. Our objective is to reconstitute a glucose-responsive system in the liver to regulate hepatic insulin production for improving glycemic control in type 1 diabetes. METHODS: We have cloned the glucose-responsive element (GRE) from the promoter of acetyl-CoA carboxylase (ACC), an enzyme that catalyzes the rate-limiting step in fatty acid synthesis in the liver in response to glucose. To increase the amplitude of glucose induction, we quadruplicated the GRE DNA by gene duplication. The resulting GRE multimer (4×GRE) was tested for its ability to drive rat proinsulin cDNA expression in hepatocytes and insulin-deficient diabetic mice. RESULTS: We showed that this GRE multimer-directed glucose-responsive system produced insulin in hepatocytes in a glucose-dependent manner. When delivered into the liver by adenovirus-mediated gene transfer, this glucose-responsive insulin production system was able to reverse hyperglycemia to a normal range without causing hypoglycemia after glucose challenge or overnight fasting. Insulin vector-treated diabetic mice exhibited significantly improved blood glucose profiles in response to glucose tolerance, correlating with insulin production in the liver. We recapitulated these findings in streptozotocin-induced diabetic CD1 mice and autoimmune non-obese diabetic mice. CONCLUSION: Our data characterized the GRE motif from the ACC promoter as a potent glucose-responsive element, and provided proof-of-concept that the 4×GRE-mediated hepatic insulin production is capable of correcting insulin deficiency and improving glycemic control in type 1 diabetes.

18.
Physiol Rep ; 2(3): e00247, 2014.
Article En | MEDLINE | ID: mdl-24760506

Abstract Apolipoprotein C-III (apoC-III) is not only predominantly synthesized by the liver but also by the small intestine. Because apoC-III is secreted from the intestine on the chylomicron along with lipid absorption, we questioned whether apoC-III might play a role in intestinal lipid absorption and/or transport. Using both wild-type (WT) and apoC-III transgenic (apoC-III Tg) mice, we showed that apoC-III Tg mice have decreased lymphatic lipid transport compared with WT mice in response to an intraduodenal infusion of radiolabeled lipid. This is associated with accumulation of radiolabeled lipids in the luminal compartment of the apoC-III Tg mice, indicating delayed lipid uptake from the lumen. The total amount of radioactive lipids in the mucosal compartment did not differ between apoC-III Tg and WT mice, but the lipid distribution analysis indicated a predominance of free fatty acids and monoacylglycerol in the mucosa of apoC-III Tg mice, implying impaired esterification capacity. Thus, the mechanisms underlying the reduced lymphatic lipid transport in apoC-III Tg mice involve both a delayed lipid uptake into enterocytes, as well as impaired esterification to form triglyceride in the mucosa. These data document a novel role for apoC-III in the uptake, re-esterification, and lymphatic transport of dietary lipids in the intestine.

19.
Endocrinology ; 155(4): 1255-67, 2014 Apr.
Article En | MEDLINE | ID: mdl-24437489

Excessive production of triglyceride-rich very low-density lipoproteins (VLDL-TG) contributes to hypertriglyceridemia in obesity and type 2 diabetes. To understand the underlying mechanism, we studied hepatic regulation of VLDL-TG production by (forkhead box O6) FoxO6, a forkhead transcription factor that integrates insulin signaling to hepatic metabolism. We showed that transgenic mice expressing a constitutively active FoxO6 allele developed hypertriglyceridemia, culminating in elevated VLDL-TG levels and impaired postprandial TG clearance. This effect resulted in part from increased hepatic VLDL-TG production. We recapitulated these findings in cultured HepG2 cells and human primary hepatocytes, demonstrating that FoxO6 promoted hepatic VLDL-TG secretion. This action correlated with the ability of FoxO6 to stimulate hepatic production of microsomal triglyceride transfer protein (MTP), a molecular chaperone that catalyzes the rate-limiting step in VLDL-TG assembly and secretion. FoxO6 was shown to bind to the MTP promoter and stimulate MTP promoter activity in HepG2 cells. This effect was inhibited by insulin, consistent with the ability of insulin to promote FoxO6 phosphorylation and disable FoxO6 DNA-binding activity. Mutations of the FoxO6 target site within the MTP promoter abrogated FoxO6-mediated induction of MTP promoter activity. Hepatic FoxO6 expression became deregulated in insulin-resistant mice with obesity and type 2 diabetes. FoxO6 inhibition in insulin-resistant liver suppressed hepatic MTP expression and curbed VLDL-TG overproduction, contributing to the amelioration of hypertriglyceridemia in obese and diabetic db/db mice. These results characterize FoxO6 as an important signaling molecule upstream of MTP for regulating hepatic VLDL-TG production.


Carrier Proteins/metabolism , Forkhead Transcription Factors/metabolism , Hypertriglyceridemia/metabolism , Insulin/metabolism , Lipoproteins, VLDL/metabolism , Liver/metabolism , Signal Transduction , Triglycerides/metabolism , Animals , Female , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Hep G2 Cells , Hepatocytes/cytology , Humans , Lipids/chemistry , Lipoprotein Lipase/metabolism , Male , Mice , Mice, Transgenic , PPAR gamma/metabolism , Phosphorylation , Promoter Regions, Genetic
20.
J Biol Chem ; 288(35): 25350-25361, 2013 Aug 30.
Article En | MEDLINE | ID: mdl-23888053

Hypertriglyceridemia is the most common lipid disorder in obesity and type 2 diabetes. It results from increased production and/or decreased clearance of triglyceride-rich lipoproteins. To better understand the pathophysiology of hypertriglyceridemia, we studied hepatic regulation of triglyceride metabolism by the activating transcription factor 4 (ATF4), a member of the basic leucine zipper-containing protein subfamily. We determined the effect of ATF4 on hepatic lipid metabolism in Atf4(-/-) mice fed regular chow or provided with free access to fructose drinking water. ATF4 depletion preferentially attenuated hepatic lipogenesis without affecting hepatic triglyceride production and fatty acid oxidation. This effect prevented excessive fat accumulation in the liver of Atf4(-/-) mice, when compared with wild-type littermates. To gain insight into the underlying mechanism, we showed that ATF4 depletion resulted in a significant reduction in hepatic expression of peroxisome proliferator-activated receptor-γ, a nuclear receptor that acts to promote lipogenesis in the liver. This effect was accompanied by a significant reduction in hepatic expression of sterol regulatory element-binding protein 1c (SREBP-1c), acetyl-CoA carboxylase, and fatty-acid synthase, three key functions in the lipogenic pathway in Atf4(-/-) mice. Of particular significance, we found that Atf4(-/-) mice, as opposed to wild-type littermates, were protected against the development of steatosis and hypertriglyceridemia in response to high fructose feeding. These data demonstrate that ATF4 plays a critical role in regulating hepatic lipid metabolism in response to nutritional cues.


Activating Transcription Factor 4 , Fructose/adverse effects , Hypertriglyceridemia , Lipid Metabolism , Liver/metabolism , Sweetening Agents/adverse effects , Acetyl-CoA Carboxylase/genetics , Acetyl-CoA Carboxylase/metabolism , Animals , Fatty Acid Synthase, Type I/genetics , Fatty Acid Synthase, Type I/metabolism , Fructose/pharmacology , Hypertriglyceridemia/chemically induced , Hypertriglyceridemia/genetics , Hypertriglyceridemia/metabolism , Hypertriglyceridemia/pathology , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Liver/pathology , Mice , Mice, Knockout , PPAR gamma/genetics , PPAR gamma/metabolism , Sterol Regulatory Element Binding Protein 1/genetics , Sterol Regulatory Element Binding Protein 1/metabolism , Sweetening Agents/pharmacology
...