Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Lipids ; 58(3): 107-115, 2023 05.
Article in English | MEDLINE | ID: mdl-36849669

ABSTRACT

We investigated the relationships between ceramide species concentrations in liver, plasma and very low-density lipoproteins (VLDL) particles of humans with obesity as well as the relationships between hepatic fat content and hepatic ceramide concentrations and proportional distribution. Twenty-five obese (body mass index >35 kg/m2 ) adults participated in this study. Plasma, VLDL and hepatocellular ceramide concentrations were measured by liquid chromatography/tandem mass spectrometry. The proportionate distribution of measured ceramide species differed between liver, whole plasma and the VLDL fraction. We found significant, positive correlations between the proportion of C14:0, C18:0, C20:0 and C24:1 ceramide in the liver and whole plasma (γ = 0.491, p = 0.013; γ = 0.573, p = 0.003; γ = 0.479, p = 0.015; γ = 0.716, p = 0.00006; respectively). In contrast, only the proportional contribution of C24:1 ceramide correlated positively between VLDL and liver (γ = 0.425, p = 0.013). The percent hepatic fat correlated positively with the proportion of C18:1, C18:0 and C20:0 hepatic ceramides (γ = 0.415, p = 0.039; γ = 0.426, p = 0.034; γ = 0.612, p = 0.001; respectively), but not with total hepatic ceramide concentration. The proportions of whole plasma ceramide subspecies, especially C14:0, C18:0, C20:0 and C24:1chain length, are reflective of those of hepatic ceramide subspecies in obese humans; these appear to be markers of hepatic ceramide species composition.


Subject(s)
Insulin Resistance , Obesity, Morbid , Humans , Adult , Ceramides , Lipoproteins, VLDL , Obesity , Liver , Lipoproteins, LDL
2.
Diabetes ; 71(3): 381-393, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34857544

ABSTRACT

The role of adipose tissue (AT) inflammation in AT function in humans is unclear. We tested whether AT macrophage (ATM) content, cytokine gene expression, and senescent cell burden (markers of AT inflammation) predict AT insulin resistance measured as the insulin concentration that suppresses lipolysis by 50% (IC50). We studied 86 volunteers with normal weight or obesity at baseline and a subgroup of 25 volunteers with obesity before and after weight loss. There was a strong positive relationship between IC50 and abdominal subcutaneous and femoral fat cell size (FCS). The positive, univariate relationships between IC50 and abdominal AT inflammatory markers CD68, CD14, CD206 ATM/100 adipocytes, senescent cells, IL-6, and TNF-α mRNA were not significant after adjustment for FCS. A 10% weight loss significantly reduced IC50; however, there was no reduction in adipose ATM content, senescent cells, or cytokine gene expression. Our study suggests that commonly used markers of AT inflammation are not causally linked to AT insulin resistance, whereas FCS is a strong predictor of AT insulin resistance with respect to lipolysis.


Subject(s)
Adipose Tissue/physiopathology , Inflammation/physiopathology , Insulin Resistance/physiology , Obesity/physiopathology , Abdominal Fat/pathology , Abdominal Fat/physiopathology , Adipocytes/pathology , Adipose Tissue/pathology , Adult , Blood Glucose/metabolism , Cell Size , Cellular Senescence , Cytokines/analysis , Cytokines/genetics , Female , Gene Expression , Humans , Inflammation/pathology , Insulin/blood , Macrophages/pathology , Male , Middle Aged , Obesity/pathology , Weight Loss/physiology
3.
Obesity (Silver Spring) ; 29(8): 1320-1327, 2021 08.
Article in English | MEDLINE | ID: mdl-34114359

ABSTRACT

OBJECTIVE: Adipose tissue (AT) senescence is associated with AT dysfunction in rodents, but little is known about human AT senescence. The study goal was to define the distribution of senescent cells in two subcutaneous depots and understand relationships with adiposity and inflammation. METHODS: Sixty-three volunteers (48 females) underwent abdominal and femoral subcutaneous fat biopsies. Fat cell size, senescent cells using senescence-associated ß-galactosidase staining per 100 nucleated cells (percentage), and mRNA expression of four cytokines were measured. RESULTS: There was a larger proportion of senescent cells in femoral than abdominal subcutaneous AT (mean difference 1.6% [95% CI: 0.98%-2.3%], p < 0.001), and the percentage of femoral AT senescent cells was greater in women than men (median 3.9% vs. 2.1%, p < 0.01). There was a positive correlation between senescence and fat cell size in abdominal (rs  = 0.44, p < 0.001) and femoral (rs  = 0.35, p = 0.007) AT depots. Abdominal AT tumor necrosis factor alpha (rs  = 0.49, p < 0.01) and interleukin-1ß (rs  = 0.44, p = 0.01) expression was positively correlated with abdominal, but not femoral, AT senescence. CONCLUSIONS: In human subcutaneous AT, there are more senescent cells in femoral than abdominal depots; abdominal AT senescent cells are more associated with inflammatory signals than femoral AT senescent cells.


Subject(s)
Adipose Tissue , Obesity , Adipose Tissue/metabolism , Adiposity , Cellular Senescence , Cross-Sectional Studies , Female , Humans , Male , Obesity/metabolism , Subcutaneous Fat, Abdominal
4.
Obesity (Silver Spring) ; 29(6): 1014-1021, 2021 06.
Article in English | MEDLINE | ID: mdl-33893721

ABSTRACT

OBJECTIVE: This study tested whether substrate concentrations or fatty acid storage proteins predict storage of endogenous lipids in visceral adipose tissue (VAT) and upper body subcutaneous adipose tissue (UBSQ) fat. METHODS: The day prior to surgery, 25 patients undergoing bariatric procedures received an infusion of autologous [1-14 C]triolein-labeled very low-density lipoprotein (VLDL) particles, and during surgery, they received a continuous [U-13 C]palmitate infusion/bolus [9,10-3 H]palmitate tracer. VAT and UBSQ fat were collected to measure VLDL-triglyceride (TG) storage, direct free fatty acid (FFA) storage rates, CD36 content, lipoprotein lipase (LPL), acyl-CoA synthetase, diacylglycerol acetyl-transferase, and glycerol-3-phosphate acyltransferase activities. RESULTS: Storage of VLDL-TG and FFA-palmitate in UBSQ and VAT was not different. Plasma palmitate concentrations correlated with palmitate storage rates in UBSQ and VAT (r = 0.46, P = 0.02 and r = 0.46, P = 0.02, respectively). In VAT, VLDL-TG storage was correlated with VLDL concentrations (r = 0.53, P < 0.009) and LPL (r = 0.42, P < 0.05). In UBSQ, VLDL-TG storage was correlated with LPL (r = 0.42, P < 0.05). CD36, acyl-CoA synthetase, glycerol-3-phosphate acyltransferase, and diacylglycerol acetyl-transferase were not correlated with VLDL-TG or palmitate storage. CONCLUSIONS: Adipose storage of VLDL-TG is predicted by VLDL-TG concentrations and LPL; FFA concentrations predict direct adipose tissue FFA storage rates.


Subject(s)
Fatty Acids/metabolism , Intra-Abdominal Fat/metabolism , Obesity, Morbid/metabolism , Subcutaneous Fat/metabolism , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Adolescent , Adult , Bariatric Surgery , Fatty Acids, Nonesterified/metabolism , Female , Humans , Intra-Abdominal Fat/pathology , Lipoproteins, VLDL/metabolism , Male , Middle Aged , Obesity, Morbid/pathology , Obesity, Morbid/surgery , Subcutaneous Fat/pathology , Triglycerides/metabolism , Young Adult
5.
J Clin Endocrinol Metab ; 106(4): e1631-e1646, 2021 03 25.
Article in English | MEDLINE | ID: mdl-33382888

ABSTRACT

PURPOSE: Skeletal muscle is the primary site for insulin-stimulated glucose disposal, and muscle insulin resistance is central to abnormal glucose metabolism in obesity. Whether muscle insulin signaling to the level of Akt/AS160 is intact in insulin-resistant obese humans is controversial. METHODS: We defined a linear range of insulin-stimulated systemic and leg glucose uptake in 14 obese and 14 nonobese volunteers using a 2-step insulin clamp (Protocol 1) and then examined the obesity-related defects in muscle insulin action in 16 nonobese and 25 obese male and female volunteers matched for fitness using a 1-step, hyperinsulinemic, euglycemic clamp coupled with muscle biopsies (Protocol 2). RESULTS: Insulin-stimulated glucose disposal (Si) was reduced by > 60% (P < 0.0001) in the obese group in Protocol 2; however, the phosphorylation of Akt and its downstream effector AS160 were not different between nonobese and obese groups. The increase in phosphorylation of Akt2 in response to insulin was positively correlated with Si for both the nonobese (r = 0.53, P = 0.03) and the obese (r = 0.55, P = 0.01) groups. Total muscle GLUT4 protein was 17% less (P < 0.05) in obese subjects. CONCLUSIONS: We suggest that reduced muscle glucose uptake in obesity is not due to defects in the insulin signaling pathway at the level of Akt/AS160, which suggests there remain significant gaps in our knowledge of muscle insulin resistance in obesity. Our data imply that models of acute lipotoxicity do not replicate the pathophysiology of obesity.


Subject(s)
Glucose/metabolism , Insulin/metabolism , Muscle, Skeletal/metabolism , Obesity/metabolism , Adult , Female , Humans , Male , Signal Transduction
6.
Metabolites ; 9(11)2019 Oct 28.
Article in English | MEDLINE | ID: mdl-31661783

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is a major public health problem worldwide. NAFLD ranges in severity from benign steatosis to nonalcoholic steatohepatitis (NASH), cirrhosis, and primary hepatocellular cancer (HCC). Obesity and type 2 diabetes mellitus (T2DM) are strongly associated with NAFLD, and the western diet (WD) is a major contributor to the onset and progression of these chronic diseases. Our aim was to use a lipidomic approach to identify potential lipid mediators of diet-induced NASH. We previously used a preclinical mouse (low density lipoprotein receptor null mouse, Ldlr -/-) model to assess transcriptomic mechanisms linked to WD-induced NASH and docosahexaenoic acid (DHA, 22:6, ω3)-mediated remission of NASH. This report used livers from the previous study to carry out ultra-high-performance liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) and high-performance liquid chromatography coupled with dynamic multi-reaction monitoring (HPLC-dMRM) to assess the impact of the WD and DHA on hepatic membrane lipid and oxylipin composition, respectively. Feeding mice the WD increased hepatic saturated and monounsaturated fatty acids and arachidonic acid (ARA, 20:4, ω6) in membrane lipids and suppressed ω3 polyunsaturated fatty acids (PUFA) in membrane lipids and ω3 PUFA-derived anti-inflammatory oxylipins. Supplementing the WD with DHA lowered hepatic ARA in membrane lipids and ARA-derived oxylipins and significantly increased hepatic DHA and its metabolites in membrane lipids, as well as C20-22 ω3 PUFA-derived oxylipins. NASH markers of inflammation and fibrosis were inversely associated with hepatic C20-22 ω3 PUFA-derived Cyp2C- and Cyp2J-generated anti-inflammatory oxylipins (false discovery rate adjusted p-value; q ≤ 0.026). Our findings suggest that dietary DHA promoted partial remission of WD-induced NASH, at least in part, by lowering hepatic pro-inflammatory oxylipins derived from ARA and increasing hepatic anti-inflammatory oxylipins derived from C20-22 ω3 PUFA.

7.
J Clin Endocrinol Metab ; 104(12): 6171-6181, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31408176

ABSTRACT

OBJECTIVE: Nonalcoholic fatty liver disease can lead to hepatic inflammation/damage. Understanding the physiological mechanisms that contribute to excess hepatic lipid accumulation may help identify effective treatments. DESIGN: We recruited 25 nondiabetic patients with severe obesity scheduled for bariatric surgery. To evaluate liver export of triglyceride fatty acids, we measured very-low-density lipoprotein (VLDL)-triglyceride secretion rates the day prior to surgery using an infusion of autologous [1-14C]triolein-labeled VLDL particles. Ketone body response to fasting and intrahepatic long-chain acylcarnitine concentrations were used as indices of hepatic fatty acid oxidation. We measured intraoperative hepatic uptake rates of plasma free fatty acids using a continuous infusion of [U-13C]palmitate, combined with a bolus dose of [9,10-3H]palmitate and carefully timed liver biopsies. Total intrahepatic lipids were measured in liver biopsy samples to determine fatty liver status. The hepatic concentrations and enrichment from [U-13C]palmitate in diacylglycerols, sphingolipids, and acyl-carnitines were measured using liquid chromatography/tandem mass spectrometry. RESULTS: Among study participants with fatty liver disease, intrahepatic lipid was negatively correlated with VLDL-triglyceride secretion rates (r = -0.92, P = 0.01) but unrelated to hepatic free fatty acid uptake or indices of hepatic fatty acid oxidation. VLDL-triglyceride secretion rates were positively correlated with hepatic concentrations of saturated diacylglycerol (r = 0.46, P = 0.02) and sphingosine-1-phosphate (r = 0.44, P = 0.03). CONCLUSION: We conclude that in nondiabetic humans with severe obesity, excess intrahepatic lipid is associated with limited export of triglyceride in VLDL particles rather than increased uptake of systemic free fatty acids.


Subject(s)
Fatty Acids/metabolism , Lipid Metabolism , Liver/metabolism , Obesity, Morbid/metabolism , Adolescent , Adult , Bariatric Surgery , Fatty Acids, Nonesterified/blood , Female , Humans , Lipoproteins, VLDL/blood , Liver/pathology , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Obesity, Morbid/complications , Obesity, Morbid/pathology , Obesity, Morbid/surgery , Sphingolipids/metabolism , Triglycerides/blood , Young Adult
8.
J Nutr ; 148(9): 1421-1425, 2018 09 01.
Article in English | MEDLINE | ID: mdl-30107536

ABSTRACT

Background: α-Cyclodextrin (α-CD), a soluble dietary fiber, may improve abnormal plasma lipids and promote weight loss. Preliminary evidence suggests that it may exert these effects by binding dietary fat and reducing absorption; this has not been tested in humans. Objective: The primary objective was to test whether supplemental α-CD increases fecal content of dietary lipid in humans. Methods: This was a randomized, double-blind, placebo-controlled, crossover study completed at the Mayo Clinic. Eight healthy volunteers, 5 premenopausal women and 3 men ages 23-54 y with body mass index (BMI; kg/m2) 18-27, underwent 2 separate study visits with a ≥2-wk washout period. The first morning of each visit volunteers consumed a standardized breakfast (14.5% protein, 27.5% fat, 60% carbohydrate, and 1.5 kcal/mL) containing [14C]tripalmitin and [3H]triolein with 2 g of α-CD or placebo, followed by 2 g of α-CD or placebo per meal for 2 more days. Volunteers consumed 100 g/d of dietary fat. Feces were collected for 72 h after the labeled breakfast to measure radiotracer content and total fecal fat. Radiotracer appearance in plasma TGs was measured at intervals after the labeled meal as a secondary outcome. Results: Virtually no 3H radiotracer, but an average of ∼20% of the 14C radiotracer was recovered in fecal lipids, with no difference between α-CD and placebo. Total fecal fat content and radiotracer appearance in postprandial plasma TGs did not differ between the α-CD and placebo treatments. Plasma appearance of 14C-TG was 37% ± 14% less (P < 0.0001) than 3H-TG. Conclusions: α-CD supplementation did not increase loss of dietary lipid in stool or total fecal fat compared with placebo in healthy adults. Greater stool loss and lesser appearance in plasma TGs of tripalmitin-derived [14C] compared with triolein-derived [3H] TGs imply different metabolic handling of these 2 dietary fat tracers. This trial was registered at www.clinicaltrials.gov as NCT03002168.


Subject(s)
Dietary Fats/pharmacokinetics , Feces/chemistry , alpha-Cyclodextrins/administration & dosage , Adult , Breakfast , Carbon Radioisotopes , Cross-Over Studies , Dietary Fats/analysis , Dietary Fats/metabolism , Dietary Fiber , Double-Blind Method , Female , Humans , Intestinal Absorption/drug effects , Male , Middle Aged , Placebos , Triglycerides/administration & dosage , Triglycerides/blood , Triglycerides/pharmacokinetics , Triolein/administration & dosage , Triolein/pharmacokinetics , Tritium , alpha-Cyclodextrins/metabolism
9.
Pharmacol Ther ; 181: 108-125, 2018 01.
Article in English | MEDLINE | ID: mdl-28723414

ABSTRACT

Obese and type 2 diabetic (T2DM) patients have a high prevalence of nonalcoholic fatty liver disease (NAFLD). NAFLD is a continuum of chronic liver diseases ranging from benign hepatosteatosis to nonalcoholic steatohepatitis (NASH), cirrhosis and primary hepatocellular cancer (HCC). Because of its strong association with the obesity epidemic, NAFLD is rapidly becoming a major public health concern worldwide. Surprisingly, there are no FDA approved NAFLD therapies; and current therapies focus on the co-morbidities associated with NAFLD, namely, obesity, hyperglycemia, dyslipidemia, and hypertension. The goal of this review is to provide background on the disease process, discuss human studies and preclinical models that have examined treatment options. We also provide an in-depth rationale for the use of dietary ω3 polyunsaturated fatty acid (ω3 PUFA) supplements as a treatment option for NAFLD. This focus is based on recent studies indicating that NASH patients and preclinical mouse models of NASH have low levels of hepatic C20-22 ω3 PUFA. This decline in hepatic PUFA may account for the major phenotypic features associated with NASH, including steatosis, inflammation and fibrosis. Finally, our discussion will address the strengths and limitations of ω3 PUFA supplements use in NAFLD therapy.


Subject(s)
Fatty Acids, Omega-3/therapeutic use , Non-alcoholic Fatty Liver Disease/diet therapy , Animals , Disease Models, Animal , Disease Progression , Humans
10.
PLoS One ; 12(4): e0173376, 2017.
Article in English | MEDLINE | ID: mdl-28422962

ABSTRACT

BACKGROUND: Nonalcoholic fatty liver disease (NAFLD) is a major public health concern in western societies. Nonalcoholic steatohepatitis (NASH), the progressive form of NAFLD, is characterized by hepatic steatosis, inflammation, oxidative stress and fibrosis. NASH is a risk factor for cirrhosis and hepatocellular carcinoma. NASH is predicted to be the leading cause of liver transplants by 2020. Despite this growing public health concern, there remain no Food and Drug Administration (FDA) approved NASH treatments. Using Ldlr -/- mice as a preclinical model of western diet (WD)-induced NASH, we previously established that dietary supplementation with docosahexaenoic acid (DHA, 22:6,ω3) attenuated WD-induced NASH in a prevention study. Herein, we evaluated the capacity of DHA supplementation of the WD and a low fat diet to fully reverse NASH in mice with pre-existing disease. METHODS: Ldlr -/- mice fed the WD for 22 wks developed metabolic syndrome (MetS) and a severe NASH phenotype, including obesity, dyslipidemia, hyperglycemia, hepatic steatosis, inflammation, fibrosis and low hepatic polyunsaturated fatty acid (PUFA) content. These mice were randomized to 5 groups: a baseline group (WDB, sacrificed at 22 wks) and 4 treatments: 1) WD + olive oil (WDO); 2) WD + DHA (WDD); 3) returned to chow + olive oil (WDChO); or 4) returned to chow + DHA (WDChD). The four treatment groups were maintained on their respective diets for 8 wks. An additional group was maintained on standard laboratory chow (Reference Diet, RD) for the 30-wk duration of the study. RESULTS: When compared to the WDB group, the WDO group displayed increased hepatic expression of genes linked to inflammation (Opn, Il1rn, Gdf15), hepatic fibrosis (collagen staining, Col1A1, Thbs2, Lox) reflecting disease progression. Mice in the WDD group, in contrast, had increased hepatic C20-22 ω3 PUFA and no evidence of NASH progression. MetS and NASH markers in the WDChO or WDChD groups were significantly attenuated and marginally different from the RD group, reflecting disease remission. CONCLUSION: While these studies establish that DHA supplementation of the WD blocks WD-induced NASH progression, DHA alone does not promote full remission of diet-induced MetS or NASH.


Subject(s)
Dietary Fats/administration & dosage , Docosahexaenoic Acids/administration & dosage , Non-alcoholic Fatty Liver Disease/diet therapy , Receptors, LDL/deficiency , Animals , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Diet, Western , Disease Models, Animal , Disease Progression , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Gene Expression Regulation , Growth Differentiation Factor 15/genetics , Growth Differentiation Factor 15/metabolism , Interleukin 1 Receptor Antagonist Protein/genetics , Interleukin 1 Receptor Antagonist Protein/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/physiopathology , Olive Oil/administration & dosage , Osteopontin/genetics , Osteopontin/metabolism , Protein-Lysine 6-Oxidase/genetics , Protein-Lysine 6-Oxidase/metabolism , Receptors, LDL/genetics , Thrombospondins/genetics , Thrombospondins/metabolism
11.
PLoS One ; 11(1): e0146942, 2016.
Article in English | MEDLINE | ID: mdl-26761430

ABSTRACT

BACKGROUND: Nonalcoholic fatty liver disease (NAFLD) is a major public health burden in western societies. The progressive form of NAFLD, nonalcoholic steatohepatitis (NASH), is characterized by hepatosteatosis, inflammation, oxidative stress, and hepatic damage that can progress to fibrosis and cirrhosis; risk factors for hepatocellular carcinoma. Given the scope of NASH, validating treatment protocols (i.e., low fat diets and weight loss) is imperative. METHODS: We evaluated the efficacy of two diets, a non-purified chow (NP) and purified (low-fat low-cholesterol, LFLC) diet to reverse western diet (WD)-induced NASH and fibrosis in Ldlr-/- mice. RESULTS: Mice fed WD for 22-24 weeks developed robust hepatosteatosis with mild fibrosis, while mice maintained on the WD an additional 7-8 weeks developed NASH with moderate fibrosis. Returning WD-fed mice to the NP or LFLC diets significantly reduced body weight and plasma markers of metabolic syndrome (dyslipidemia, hyperglycemia) and hepatic gene expression markers of inflammation (Mcp1), oxidative stress (Nox2), fibrosis (Col1A, LoxL2, Timp1) and collagen crosslinking (hydroxyproline). Time course analyses established that plasma triglycerides and hepatic Col1A1 mRNA were rapidly reduced following the switch from the WD to the LFLC diet. However, hepatic triglyceride content and fibrosis did not return to normal levels 8 weeks after the change to the LFLC diet. Time course studies further revealed a strong association (r2 ≥ 0.52) between plasma markers of inflammation (TLR2 activators) and hepatic fibrosis markers (Col1A, Timp1, LoxL2). Inflammation and fibrosis markers were inversely associated (r2 ≥ 0.32) with diet-induced changes in hepatic ω3 and ω6 polyunsaturated fatty acids (PUFA) content. CONCLUSION: These studies establish a temporal link between plasma markers of inflammation and hepatic PUFA and fibrosis. Low-fat low-cholesterol diets promote reversal of many, but not all, features associated with WD-induced NASH and fibrosis in Ldlr-/- mice.


Subject(s)
Diet, Fat-Restricted , Diet, Western/adverse effects , Non-alcoholic Fatty Liver Disease/genetics , Receptors, LDL/genetics , Animals , Body Weight , Carcinoma, Hepatocellular/metabolism , Dietary Fats/adverse effects , Disease Progression , Fatty Acids/metabolism , Fatty Acids, Unsaturated/metabolism , Fibrosis/pathology , Gene Expression Regulation , Inflammation/blood , Lipids/chemistry , Liver/metabolism , Liver Cirrhosis/pathology , Liver Neoplasms/pathology , Male , Mice , Mice, Knockout , Oxidative Stress , Risk Factors , Time Factors , Triglycerides/blood
12.
Proc Nutr Soc ; 75(1): 1-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26282529

ABSTRACT

The prevalence of non-alcoholic fatty liver disease (NAFLD) has increased in parallel with central obesity and is now the most common chronic liver disease in developed countries. NAFLD is defined as excessive accumulation of lipid in the liver, i.e. hepatosteatosis. The severity of NAFLD ranges from simple fatty liver (steatosis) to non-alcoholic steatohepatitis (NASH). Simple steatosis is relatively benign until it progresses to NASH, which is characterised by hepatic injury, inflammation, oxidative stress and fibrosis. Hepatic fibrosis is a risk factor for cirrhosis and primary hepatocellular carcinoma. Our studies have focused on the impact of diet on the onset and progression of NASH. We developed a mouse model of NASH by feeding Ldlr-/- mice a western diet (WD), a diet moderately high in saturated and trans-fat, sucrose and cholesterol. The WD induced a NASH phenotype in Ldlr-/- mice that recapitulates many of the clinical features of human NASH. We also assessed the capacity of the dietary n-3 PUFA, i.e. EPA (20 : 5,n-3) and DHA (22 : 6,n-3), to prevent WD-induced NASH in Ldlr-/- mice. Histologic, transcriptomic, lipidomic and metabolomic analyses established that DHA was equal or superior to EPA at attenuating WD-induced dyslipidemia and hepatic injury, inflammation, oxidative stress and fibrosis. Dietary n-3 PUFA, however, had no significant effect on WD-induced changes in body weight, body fat or blood glucose. These studies provide a molecular and metabolic basis for understanding the strengths and weaknesses of using dietary n-3 PUFA to prevent NASH in human subjects.

13.
Adv Nutr ; 6(6): 694-702, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26567194

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) has increased in parallel with central obesity, and its prevalence is anticipated to increase as the obesity epidemic remains unabated. NAFLD is now the most common cause of chronic liver disease in developed countries and is defined as excessive lipid accumulation in the liver, that is, hepatosteatosis. NAFLD ranges in severity from benign fatty liver to nonalcoholic steatohepatitis (NASH), and NASH is characterized by hepatic injury, inflammation, oxidative stress, and fibrosis. NASH can progress to cirrhosis, and cirrhosis is a risk factor for primary hepatocellular carcinoma (HCC). The prevention of NASH will lower the risk of cirrhosis and NASH-associated HCC. Our studies have focused on NASH prevention. We developed a model of NASH by using mice with the LDL cholesterol receptor gene ablated fed the Western diet (WD). The WD induces a NASH phenotype in these mice that is similar to that seen in humans and includes robust induction of hepatic steatosis, inflammation, oxidative stress, and fibrosis. With the use of transcriptomic, lipidomic, and metabolomic approaches, we examined the capacity of 2 dietary ω-3 (n-3) polyunsaturated fatty acids, eicosapentaenoic acid (20:5ω-3; EPA) and docosahexaenoic acid (22:6ω-3; DHA), to prevent WD-induced NASH. Dietary DHA was superior to EPA at attenuating WD-induced changes in plasma lipids and hepatic injury and at reversing WD effects on hepatic metabolism, oxidative stress, and fibrosis. The outcome of these studies suggests that DHA may be useful in preventing NASH and reducing the risk of HCC.


Subject(s)
Dietary Fats, Unsaturated/therapeutic use , Fatty Acids, Omega-3/therapeutic use , Liver Neoplasms/prevention & control , Non-alcoholic Fatty Liver Disease/prevention & control , Animals , Diet, Western/adverse effects , Dietary Fats, Unsaturated/administration & dosage , Disease Models, Animal , Docosahexaenoic Acids , Eicosapentaenoic Acid , Fatty Acids, Omega-3/administration & dosage , Fatty Liver , Hepatitis , Humans , Liver Cirrhosis , Mice , Mice, Knockout , Mice, Obese , Non-alcoholic Fatty Liver Disease/etiology , Obesity/complications , Obesity/epidemiology , Obesity/etiology , Oxidative Stress , Receptors, LDL/deficiency , Receptors, LDL/genetics , Risk Factors
14.
J Lipid Res ; 56(10): 1936-46, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26315048

ABSTRACT

DHA (22:6,ω3), but not EPA (20:5,ω3), attenuates Western diet (WD)-induced hepatic fibrosis in a Ldlr(-/-) mouse model of nonalcoholic steatohepatitis. We examined the molecular basis for the differential effect of dietary EPA and DHA on WD-induced hepatic fibrosis. DHA was more effective than EPA at preventing WD-induced effects on hepatic transcripts linked to fibrosis, including collagen 1A1 (Col1A1), transforming growth factor-ß (TGFß) signaling and proteins involved in remodeling the extracellular matrix, including metalloproteases, tissue inhibitors of metalloproteases, and lysyl oxidase subtypes. Examination of the TGFß pathway showed that mice fed the WD supplemented with either olive oil or EPA had a significant (≥2.5-fold) increase in hepatic nuclear abundance of phospho-mothers against decapentaplegic homolog (Smad)3 when compared with mice fed the reference diet (RD); Smad3 is a key regulator of Col1A1 expression in stellate cells. In contrast, mice fed the WD supplemented with DHA had no increase in phospho-Smad3 when compared with mice fed the RD. Changes in hepatic phospho-Smad3 nuclear content correlated with proCol1A1 mRNA and protein abundance. Pretreatment of human LX2 stellate cells with DHA, but not other unsaturated fatty acids, blocked TGFß1-mediated induction of Col1A1. In conclusion, DHA attenuates WD-induced fibrosis by targeting the TGFß-Smad3-Col1A1 pathway in stellate cells.


Subject(s)
Docosahexaenoic Acids/pharmacology , Eicosapentaenoic Acid/pharmacology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Animals , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Diet, Western , Dietary Supplements , Disease Models, Animal , Fatty Acids, Unsaturated/metabolism , Hepatic Stellate Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism
15.
J Lipid Res ; 55(7): 1448-64, 2014 07.
Article in English | MEDLINE | ID: mdl-24814977

ABSTRACT

Nonalcoholic fatty liver disease is a major public health concern in the obese and type 2 diabetic populations. The high-fat lard diet induces obesity and fatty liver in C57BL/6J mice and suppresses expression of the PPAR-target gene, FA elongase 5 (Elovl5). Elovl5 plays a key role in MUFA and PUFA synthesis. Increasing hepatic Elovl5 activity in obese mice lowered hepatic TGs and endoplasmic reticulum stress markers (X-box binding protein 1 and cAMP-dependent transcription factor 6α) and increased TG catabolism and fatty acyl carnitines. Increased hepatic Elovl5 activity did not increase hepatic capacity for ß-oxidation. Elovl5 effects on hepatic TG catabolism were linked to increased protein levels of adipocyte TG lipase (ATGL) and comparative gene identification 58 (CGI58). Elevated hepatic Elovl5 activity also induced the expression of some (pyruvate dehydrogenase kinase 4 and fibroblast growth factor 21), but not other cytochrome P450 4A10 (CYP4A10), PPAR-target genes. FA products of Elovl5 activity increased ATGL, but not CGI58, mRNA through PPARß-dependent mechanisms in human HepG2 cells. Treatment of mouse AML12 hepatocytes with the PPARß agonist (GW0742) decreased (14)C-18:2,n-6 in TGs but did not affect ß-oxidation. These studies establish that Elovl5 activity regulates hepatic levels of FAs controlling PPARß activity, ATGL expression, and TG catabolism, but not FA oxidation.


Subject(s)
Acetyltransferases/metabolism , Endoplasmic Reticulum Stress , Hepatocytes/metabolism , Liver/metabolism , Obesity/metabolism , Triglycerides/metabolism , Acetyltransferases/genetics , Animals , Fatty Acid Elongases , Hep G2 Cells , Hepatocytes/pathology , Humans , Lipase/genetics , Lipase/metabolism , Liver/pathology , Male , Mice , Obesity/genetics , Obesity/pathology , PPAR-beta/antagonists & inhibitors , PPAR-beta/genetics , PPAR-beta/metabolism , Thiazoles/pharmacology , Triglycerides/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...