Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
J Lipid Res ; 65(7): 100551, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39002195

ABSTRACT

Intestinal disease is one of the earliest manifestations of cystic fibrosis (CF) in children and is closely tied to deficits in growth and nutrition, both of which are directly linked to future mortality. Patients are treated aggressively with pancreatic enzyme replacement therapy and a high-fat diet to circumvent fat malabsorption, but this does not reverse growth and nutritional defects. We hypothesized that defects in chylomicron production could explain why CF body weights and nutrition are so resistant to clinical treatments. We used gold standard intestinal lipid absorption and metabolism approaches, including mouse mesenteric lymph cannulation, in vivo chylomicron secretion kinetics, transmission electron microscopy, small intestinal organoids, and chylomicron metabolism assays to test this hypothesis. In mice expressing the G542X mutation in cystic fibrosis transmembrane conductance regulator (CFTR-/- mice), we find that defective FFA trafficking across the epithelium into enterocytes drives a chylomicron formation defect. Furthermore, G542X mice secrete small, triglyceride-poor chylomicrons into the lymph and blood. These defective chylomicrons are cleared into extraintestinal tissues at ∼10-fold faster than WT chylomicrons. This defect in FFA absorption resulting in dysfunctional chylomicrons cannot be explained by steatorrhea or pancreatic insufficiency and is maintained in primary small intestinal organoids treated with micellar lipids. These studies suggest that the ultrahigh-fat diet that most people with CF are counselled to follow may instead make steatorrhea and malabsorption defects worse by overloading the absorptive capacity of the CF small intestine.


Subject(s)
Chylomicrons , Cystic Fibrosis , Cystic Fibrosis/metabolism , Cystic Fibrosis/pathology , Cystic Fibrosis/genetics , Animals , Chylomicrons/metabolism , Mice , Fatty Acids, Nonesterified/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis Transmembrane Conductance Regulator/deficiency , Exocrine Pancreatic Insufficiency/metabolism , Exocrine Pancreatic Insufficiency/genetics , Exocrine Pancreatic Insufficiency/pathology , Biological Transport , Humans , Intestinal Mucosa/metabolism
2.
J Lipid Res ; 63(11): 100284, 2022 11.
Article in English | MEDLINE | ID: mdl-36152881

ABSTRACT

The intestine plays a crucial role in regulating whole-body lipid metabolism through its unique function of absorbing dietary fat. In the small intestine, absorptive epithelial cells emulsify hydrophobic dietary triglycerides (TAGs) prior to secreting them into mesenteric lymphatic vessels as chylomicrons. Except for short- and medium-chain fatty acids, which are directly absorbed from the intestinal lumen into portal vasculature, the only way for an animal to absorb dietary TAG is through the chylomicron/mesenteric lymphatic pathway. Isolating intestinal lipoproteins, including chylomicrons, is extremely difficult in vivo because of the dilution of postprandial lymph in the peripheral blood. In addition, once postprandial lymph enters the circulation, chylomicron TAGs are rapidly hydrolyzed. To enhance isolation of large quantities of pure postprandial chylomicrons, we have modified the Tso group's highly reproducible gold-standard double-cannulation technique in rats to enable single-day surgery and lymph collection in mice. Our technique has a significantly higher survival rate than the traditional 2-day surgical model and allows for the collection of greater than 400 µl of chylous lymph with high postprandial TAG concentrations. Using this approach, we show that after an intraduodenal lipid bolus, the mesenteric lymph contains naïve CD4+ T-cell populations that can be quantified by flow cytometry. In conclusion, this experimental approach represents a quantitative tool for determining dietary lipid absorption, intestinal lipoprotein dynamics, and mesenteric immunity. Our model may also be a powerful tool for studies of antigens, the microbiome, pharmacokinetics, and dietary compound absorption.


Subject(s)
Chylomicrons , Lymphatic Vessels , Animals , Mice , Rats , Chylomicrons/metabolism , Dietary Fats/metabolism , Intestinal Absorption/physiology , Lipoproteins/metabolism , Lymph/metabolism , Lymphatic Vessels/metabolism , Lymphocytes/metabolism , Triglycerides/metabolism
3.
J Mol Cell Cardiol ; 144: 24-34, 2020 07.
Article in English | MEDLINE | ID: mdl-32418915

ABSTRACT

Cardiolipin (CL) is a unique tetra-acyl phospholipid localized to the inner mitochondrial membrane and essential for normal respiratory function. It has been previously reported that the failing human heart and several rodent models of cardiac pathology have a selective loss of CL. A rare genetic disease, Barth syndrome (BTHS), is similarly characterized by a cardiomyopathy due to reduced levels of cardiolipin. A mouse model of cardiolipin deficiency was recently developed by knocking-down the cardiolipin biosynthetic enzyme tafazzin (TAZ KD). These mice develop an age-dependent cardiomyopathy due to mitochondrial dysfunction. Since reduced mitochondrial capacity in the heart may promote the accumulation of lipids, we examined whether cardiolipin deficiency in the TAZ KD mice promotes the development of a lipotoxic cardiomyopathy. In addition, we investigated whether treatment with resveratrol, a small cardioprotective nutraceutical, attenuated the aberrant lipid accumulation and associated cardiomyopathy. Mice deficient in tafazzin and the wildtype littermate controls were fed a low-fat diet, or a high-fat diet with or without resveratrol for 16 weeks. In the absence of obesity, TAZ KD mice developed a hypertrophic cardiomyopathy characterized by reduced left-ventricle (LV) volume (~36%) and 30-50% increases in isovolumetric contraction (IVCT) and relaxation times (IVRT). The progression of cardiac hypertrophy with tafazzin-deficiency was associated with several underlying pathological processes including altered mitochondrial complex I mediated respiration, elevated oxidative damage (~50% increase in reactive oxygen species, ROS), the accumulation of triglyceride (~250%) as well as lipids associated with lipotoxicity (diacylglyceride ~70%, free-cholesterol ~44%, ceramide N:16-35%) compared to the low-fat fed controls. Treatment of TAZ KD mice with resveratrol maintained normal LV volumes and preserved systolic function of the heart. The beneficial effect of resveratrol on cardiac function was accompanied by a significant improvement in mitochondrial respiration, ROS production and oxidative damage to the myocardium. Resveratrol treatment also attenuated the development of cardiac steatosis in tafazzin-deficient mice through reduced de novo fatty acid synthesis. These results indicate for the first time that cardiolipin deficiency promotes the development of a hypertrophic lipotoxic cardiomyopathy. Furthermore, we determined that dietary resveratrol attenuates the cardiomyopathy by reducing ROS, cardiac steatosis and maintaining mitochondrial function.


Subject(s)
Cardiolipins/metabolism , Cardiomyopathy, Hypertrophic/etiology , Cardiomyopathy, Hypertrophic/metabolism , Disease Susceptibility , Lipid Metabolism , Animals , Biomarkers , Cardiomyopathy, Hypertrophic/diagnosis , Disease Models, Animal , Echocardiography , Electron Transport Complex I/metabolism , Heart Function Tests , Immunohistochemistry , Male , Mice , Mice, Transgenic , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria, Heart/drug effects , Mitochondria, Heart/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Resveratrol/pharmacology
4.
Am J Physiol Endocrinol Metab ; 315(5): E758-E770, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30086648

ABSTRACT

Accumulation of myeloid cells in the liver, notably dendritic cells (DCs) and monocytes/macrophages (MCs), is a major component of the metainflammation of obesity. However, the mechanism(s) stimulating hepatic DC/MC infiltration remain ill defined. Herein, we addressed the hypothesis that adipose tissue (AT) free fatty acids (FFAs) play a central role in the initiation of hepatic DC/MC accumulation, using a number of mouse models of altered FFA supply to the liver. In two models of acute FFA elevation (lipid infusion and fasting) hepatic DC/MC and triglycerides (TGs) but not AT DC/MC were increased without altering plasma cytokines (PCs; TNFα and monocyte chemoattractant protein 1) and with variable effects on oxidative stress (OxS) markers. However, fasting in mice with profoundly reduced AT lipolysis (AT-specific deletion of adipose TG lipase; AAKO) failed to elevate liver DC/MC, TG, or PC, but liver OxS increased. Livers of obese AAKO mice that are known to be resistant to steatosis were similarly protected from inflammation. In high-fat feeding studies of 1, 3, 6, or 20-wk duration, liver DC/MC accumulation dissociated from PC and OxS but tracked with liver TGs. Furthermore, decreasing OxS by ~80% in obese mice failed to decrease liver DC/MC. Therefore, FFA and more specifically AT-derived FFA stimulate hepatic DC/MC accumulation, thus recapitulating the pathology of the obese liver. In a number of cases the effects of FFA can be dissociated from OxS and PC but match well with liver TG, a marker of FFA oversupply.


Subject(s)
Adipose Tissue/metabolism , Fasting/metabolism , Fatty Acids, Nonesterified/metabolism , Liver/metabolism , Myeloid Cells/metabolism , Animals , Cytokines/blood , Diet, High-Fat , Fatty Acids, Nonesterified/pharmacology , Lipase/genetics , Lipase/metabolism , Lipolysis/physiology , Liver/drug effects , Mice , Mice, Knockout , Obesity/metabolism , Oxidative Stress/drug effects , Oxidative Stress/physiology , Triglycerides/metabolism
5.
Am J Physiol Endocrinol Metab ; 312(1): E11-E18, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27827807

ABSTRACT

Leptin has potent effects on lipid metabolism in a number of peripheral tissues. In liver, an acute leptin infusion (~120 min) stimulates hepatic fatty acid oxidation (~30%) and reduces triglycerides (TG, ~40%), effects that are dependent on phosphoinositol-3-kinase (PI3K) activity. In the current study we addressed the hypothesis that leptin actions on liver-resident immune cells are required for these metabolic effects. Myeloid cell-specific deletion of the leptin receptor (ObR) in mice or depletion of liver Kupffer cells (KC) in rats in vivo prevented the acute effects of leptin on liver lipid metabolism, while the metabolic effects of leptin were maintained in mice lacking ObR in hepatocytes. Notably, liver TG were elevated in both lean and obese myeloid cell ObR, but the degree of obesity and insulin resistance induced by a high-fat diet was similar to control mice. In isolated primary hepatocytes (HEP), leptin had no effects on HEP lipid metabolism and only weakly stimulated PI3K. However, the coculture of KC with HEP restored leptin action on HEP fatty acid metabolism and stimulation of HEP PI3K. Notably, leptin stimulated the release from KC of a number of cytokines. However, the exposure of HEP to these cytokines individually [granulocyte macrophage colony-stimulating factor, IL-1α, IL-1ß, IL-6, IL-10, and IL-18] or in combination had no effects on HEP lipid metabolism. Together, these data demonstrate a role for liver mononuclear cells in the regulation of liver lipid metabolism by leptin.


Subject(s)
Hepatocytes/metabolism , Kupffer Cells/physiology , Leptin/metabolism , Lipid Metabolism , Liver/metabolism , Triglycerides/metabolism , Animals , Cytokines/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interleukin-10/immunology , Interleukin-18/immunology , Interleukin-1alpha/immunology , Interleukin-1beta/immunology , Interleukin-6/immunology , Kupffer Cells/immunology , Kupffer Cells/metabolism , Mice , Mice, Inbred C57BL , Mice, Obese , Myeloid Cells/metabolism , Phosphatidylinositol 3-Kinase/metabolism , Rats , Rats, Wistar , Receptors, Leptin/genetics
6.
J Vis Exp ; (189)2022 11 30.
Article in English | MEDLINE | ID: mdl-36533833

ABSTRACT

Intestinal lipoproteins, especially triglyceride-rich chylomicrons, are a major driver of metabolism, inflammation, and cardiovascular diseases. However, isolating intestinal lipoproteins is very difficult in vivo because they are first secreted from the small intestine into the mesenteric lymphatics. Chylomicron-containing lymph then empties into the subclavian vein from the thoracic duct to deliver components of the meal to the heart, lungs, and, ultimately, whole-body circulation. Isolating naïve chylomicrons is impossible from blood since chylomicron triglyceride undergoes hydrolysis immediately upon interaction with lipoprotein lipase and other lipoprotein receptors in circulation. Therefore, the original 2-day lymph fistula procedure, described by Bollman et al. in rats, has historically been used to isolate fresh mesenteric lymph before its entry into the thoracic vein. That protocol has been improved upon and professionalized by the laboratory of Patrick Tso for the last 45 years, allowing for the analysis of these critical lipoproteins and secretions from the gut. The Tso lymph fistula procedure has now been updated and is presented here visually for the first time. This revised procedure is a single-day surgical technique for installing a duodenal feeding tube, cannulating the mesenteric lymph duct, and collecting lymph after a meal in conscious mice. The major benefits of these new techniques include the ability to reproducibly collect lymph from mice (which harnesses the power of genetic mouse models); the reduced anesthesia time for mice during the implantation of the duodenal infusion tube and the lymph cannula; the ability to continuously sample lymph throughout the feeding and post-prandial period; the ability to quantitatively measure hormones and cytokines before their dilution and enzymatic hydrolysis in blood; and the ability to collect large quantities of lymph for isolating intestinal lipoproteins. This technique is a powerful tool for directly and quantitatively measuring dietary nutrient absorption, intestinal lipoprotein synthesis, and chylomicron secretion.


Subject(s)
Chylomicrons , Dietary Fats , Animals , Mice , Rats , Chylomicrons/metabolism , Kinetics , Lymph , Triglycerides/metabolism , Lipoproteins/metabolism , Intestinal Absorption
7.
iScience ; 25(12): 105569, 2022 Dec 22.
Article in English | MEDLINE | ID: mdl-36465107

ABSTRACT

Growth differentiation factor 15 (GDF15) is a stress-induced secreted protein whose circulating levels are increased in the context of obesity. Recombinant GDF15 reduces body weight and improves glycemia in obese models, which is largely attributed to the central action of GDF15 to suppress feeding and reduce body weight. Despite these advances in knowledge, the tissue-specific sites of GDF15 production during obesity are unknown, and the effects of modulating circulating GDF15 levels on insulin sensitivity have not been evaluated directly. Here, we demonstrate that hepatocyte Gdf15 expression is sufficient for changes in circulating levels of GDF15 during obesity and that restoring Gdf15 expression specifically in hepatocytes of Gdf15 knockout mice results in marked improvements in hyperinsulinemia, hepatic insulin sensitivity, and to a lesser extent peripheral insulin sensitivity. These data support that liver hepatocytes are the primary source of circulating GDF15 in obesity.

8.
PLoS One ; 14(5): e0203101, 2019.
Article in English | MEDLINE | ID: mdl-31136580

ABSTRACT

Petite Integration Factor 1 (PIF1) is a multifunctional helicase present in nuclei and mitochondria. PIF1 knock out (KO) mice exhibit accelerated weight gain and decreased wheel running on a normal chow diet. In the current study, we investigated whether Pif1 ablation alters whole body metabolism in response to weight gain. PIF1 KO and wild type (WT) C57BL/6J mice were fed a Western diet (WD) rich in fat and carbohydrates before evaluation of their metabolic phenotype. Compared with weight gain-resistant WT female mice, WD-fed PIF1 KO females, but not males, showed accelerated adipose deposition, decreased locomotor activity, and reduced whole-body energy expenditure without increased dietary intake. Surprisingly, PIF1 KO females did not show obesity-induced alterations in fasting blood glucose and glucose clearance. WD-fed PIF1 KO females developed mild hepatic steatosis and associated changes in liver gene expression that were absent in weight-matched, WD-fed female controls, linking hepatic steatosis to Pif1 ablation rather than increased body weight. WD-fed PIF1 KO females also showed decreased expression of inflammation-associated genes in adipose tissue. Collectively, these data separated weight gain from inflammation and impaired glucose homeostasis. They also support a role for Pif1 in weight gain resistance and liver metabolic dysregulation during nutrient stress.


Subject(s)
DNA Helicases/deficiency , Diet, Western , Glucose/metabolism , Inflammation Mediators/metabolism , Weight Gain/genetics , Adipose Tissue/metabolism , Animals , Body Composition , Cholesterol/metabolism , Cytokines/metabolism , Energy Metabolism , Fatty Liver/metabolism , Fatty Liver/pathology , Female , Glucose Tolerance Test , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Knockout , Mitochondria/metabolism , Motor Activity
9.
Physiol Rep ; 7(8): e14043, 2019 04.
Article in English | MEDLINE | ID: mdl-31004398

ABSTRACT

Adropin is a liver- and brain-secreted peptide hormone with striking effects on fuel metabolism regulation in a number of tissues. Previous studies demonstrated that adropin secretion is decreased in obese mice subjected to a long-term high-fat diet (HFD), and that whole-body loss of adropin expression resulted in systemic insulin resistance. Treatment of obese mice with adropin improves glucose tolerance, which has been linked to increased glucose oxidation and inhibition of fatty acid utilization in isolated skeletal muscle homogenates. In this study, we used in vivo physiological measurements to determine how treatment of obese mice with adropin affects whole-body glucose metabolism. Treatment with adropin reduced fasting blood glucose and, as shown previously, increased glucose tolerance in HFD mice during standard glucose tolerance tests. Under hyperinsulinemic-euglycemic clamp conditions, adropin treatment led to a nonsignificant increase in whole-body insulin sensitivity, and a significant reduction in whole-body glucose uptake. Finally, we show that adropin treatment suppressed hepatic glucose production and improved hepatic insulin sensitivity. This correlated with reduced expression of fatty acid import proteins and gluconeogenic regulatory enzymes in the liver, suggesting that adropin treatment may impact the pathways that drive vital aspects of hepatic glucose metabolism.


Subject(s)
Anti-Obesity Agents/pharmacology , Blood Glucose/metabolism , Gluconeogenesis , Intercellular Signaling Peptides and Proteins/pharmacology , Liver/metabolism , Animals , Anti-Obesity Agents/therapeutic use , Diet, High-Fat/adverse effects , Insulin Resistance , Intercellular Signaling Peptides and Proteins/therapeutic use , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Obesity/drug therapy , Obesity/etiology
10.
Diabetes ; 68(6): 1221-1229, 2019 06.
Article in English | MEDLINE | ID: mdl-30936145

ABSTRACT

Systemic hyperuricemia (HyUA) in obesity/type 2 diabetes facilitated by elevated activity of xanthine oxidoreductase (XOR), which is the sole source of uric acid (UA) in mammals, has been proposed to contribute to the pathogenesis of insulin resistance/dyslipidemia in obesity. Here, the effects of hepatocyte-specific ablation of Xdh, the gene encoding XOR (HXO), and whole-body pharmacologic inhibition of XOR (febuxostat) on obesity-induced insulin resistance/dyslipidemia were assessed. Deletion of hepatocyte Xdh substantially lowered liver and plasma UA concentration. When exposed to an obesogenic diet, HXO and control floxed (FLX) mice became equally obese, but systemic HyUA was absent in HXO mice. Despite this, obese HXO mice became as insulin resistant and dyslipidemic as obese FLX mice. Similarly, febuxostat dramatically lowered plasma and tissue UA and XOR activity in obese wild-type mice without altering obesity-associated insulin resistance/dyslipidemia. These data demonstrate that hepatocyte XOR activity is a critical determinant of systemic UA homeostasis, that deletion of hepatocyte Xdh is sufficient to prevent systemic HyUA of obesity, and that neither prevention nor correction of HyUA improves insulin resistance/dyslipidemia in obesity. Thus, systemic HyUA, although clearly a biomarker of the metabolic abnormalities of obesity, does not appear to be causative.


Subject(s)
Glucose/metabolism , Hepatocytes/metabolism , Hyperuricemia/genetics , Lipid Metabolism , Obesity/metabolism , Uric Acid/metabolism , Xanthine Dehydrogenase/genetics , Animals , Diet, High-Fat , Fatty Acids, Nonesterified/metabolism , Febuxostat/pharmacology , Glucose Tolerance Test , Hepatocytes/drug effects , Hyperuricemia/metabolism , Mice , Triglycerides/metabolism , Xanthine Dehydrogenase/antagonists & inhibitors
11.
Physiol Rep ; 6(18): e13836, 2018 09.
Article in English | MEDLINE | ID: mdl-30251338

ABSTRACT

Mice with a deletion of the p50 subunit of the proinflammatory nuclear factor kappa B pathway (NF-κB p50) have reduced weight compared to wild-type control mice. However, the physiological underpinning of this phenotype remains unknown. This study addressed this issue. Compared to littermate controls, lean male p50 null mice (p50-/- ) had an increased metabolic rate (~20%) that was associated with increased skeletal muscle (SkM, ~35%), but not liver, oxidative metabolism. These metabolic alterations were accompanied by decreases in adiposity, and tissue and plasma triglyceride levels (all ~30%). Notably, there was a marked decrease in skeletal muscle, but not liver, DGAT2 gene expression (~70%), but a surprising reduction in muscle PPARα and CPT1 (both ~20%) gene expression. Exposure to a high-fat diet accentuated the diminished adiposity of p50-/- mice despite elevated caloric intake, whereas plasma triglycerides and free fatty acids (both ~30%), and liver (~40%) and SkM (~50%) triglyceride accumulation were again reduced compared to WT. Although SkM cytokine expression (IL-6 and TNFα, each ~100%) were increased in p50-/- mice, neither cytokine acutely increased SkM oxidative metabolism. We conclude that the reduced susceptibility to diet-induced obesity and dyslipidemia in p50-/- mice results from an increase in metabolic rate, which is associated with elevated skeletal muscle oxidative metabolism and decreased DGAT2 expression.


Subject(s)
Basal Metabolism/physiology , Inflammation Mediators/metabolism , Muscle, Skeletal/metabolism , NF-kappa B p50 Subunit/deficiency , Obesity/metabolism , Oxidative Stress/physiology , Animals , Diet, High-Fat/adverse effects , Diet, High-Fat/trends , Energy Metabolism/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Obesity/etiology , Obesity/prevention & control
12.
Physiol Rep ; 4(6)2016 Mar.
Article in English | MEDLINE | ID: mdl-27033445

ABSTRACT

In obesity, adipose tissue (AT) and liver are infiltrated with Th-1 polarized immune cells, which are proposed to play an important role in the pathogenesis of the metabolic abnormalities of obesity. Aging is also associated with increased adiposity, but the effects of this increase on inflammation and associated metabolic dysfunction are poorly understood. To address this issue, we assessed insulin resistance (IR) andATand liver immunophenotype in aged, lean (AL) and aged, obese (AO) mice, all of whom were maintained on a standard chow diet (11% fat diet) throughout their lives. For comparison, these variables were also assessed in young, lean (YL) and young diet-induced obese mice (41% fat diet,YO). Despite similar body weight and fat accumulation,YOmice were substantially moreIRand had greater liver steatosis compared toAOmice.YOalso had elevated infiltration of macrophages/dendritic cells inATand liver, but these increases were absent inAO Furthermore, liver immune cells ofYOwere more Th-1 polarized thenAO Notably, aging was associated with accumulation of T cells, but this occurred independent of obesity. Together, the data suggest that reduced inflammation inAOunderlies the improved insulin sensitivity and lowered steatosis compared toYO.


Subject(s)
Adipose Tissue , Aging , Diet, High-Fat , Fatty Liver/etiology , Insulin Resistance , Liver , Obesity/etiology , Adipose Tissue/immunology , Adipose Tissue/metabolism , Adiposity , Age Factors , Aging/blood , Aging/immunology , Animals , Dendritic Cells/immunology , Disease Models, Animal , Fatty Liver/blood , Fatty Liver/immunology , Immunophenotyping , Liver/immunology , Liver/metabolism , Macrophages/immunology , Male , Mice, Inbred C57BL , Obesity/blood , Obesity/immunology , Th1 Cells/immunology , Time Factors
13.
Diabetes ; 61(9): 2330-9, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22851575

ABSTRACT

Obesity-associated increases in adipose tissue (AT) CD11c(+) cells suggest that dendritic cells (DC), which are involved in the tissue recruitment and activation of macrophages, may play a role in determining AT and liver immunophenotype in obesity. This study addressed this hypothesis. With the use of flow cytometry, electron microscopy, and loss-and-gain of function approaches, the contribution of DC to the pattern of immune cell alterations and recruitment in obesity was assessed. In AT and liver there was a substantial, high-fat diet (HFD)-induced increase in DC. In AT, these increases were associated with crown-like structures, whereas in liver the increase in DC constituted an early and reversible response to diet. Notably, mice lacking DC had reduced AT and liver macrophages, whereas DC replacement in DC-null mice increased liver and AT macrophage populations. Furthermore, delivery of bone marrow-derived DC to lean wild-type mice increased AT and liver macrophage infiltration. Finally, mice lacking DC were resistant to the weight gain and metabolic abnormalities of an HFD. Together, these data demonstrate that DC are elevated in obesity, promote macrophage infiltration of AT and liver, contribute to the determination of tissue immunophenotype, and play a role in systemic metabolic responses to an HFD.


Subject(s)
Adipose Tissue/immunology , CD11c Antigen/immunology , Dendritic Cells/immunology , Liver/immunology , Macrophages/immunology , Obesity/immunology , Adipose Tissue/cytology , Animals , Diet, High-Fat , Liver/cytology , Male , Mice , Mice, Inbred C57BL
14.
Diabetes ; 59(2): 347-57, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19934001

ABSTRACT

OBJECTIVE: Increased activity of the innate immune system has been implicated in the pathogenesis of the dyslipidemia and insulin resistance associated with obesity and type 2 diabetes. In this study, we addressed the potential role of Kupffer cells (liver-specific macrophages, KCs) in these metabolic abnormalities. RESEARCH DESIGN AND METHODS: Rats were depleted of KCs by administration of gadolinium chloride, after which all animals were exposed to a 2-week high-fat or high-sucrose diet. Subsequently, the effects of these interventions on the development of hepatic insulin resistance and steatosis were assessed. In further studies, the effects of M1-polarized KCs on hepatocyte lipid metabolism and insulin sensitivity were addressed. RESULTS: As expected, a high-fat or high-sucrose diet induced steatosis and hepatic insulin resistance. However, these metabolic abnormalities were prevented when liver was depleted of KCs. In vitro, KCs recapitulated the in vivo effects of diet by increasing hepatocyte triglyceride accumulation and fatty acid esterification, and decreasing fatty acid oxidation and insulin responsiveness. To address the mechanisms(s) of KC action, we inhibited a panel of cytokines using neutralizing antibodies. Only neutralizing antibodies against tumor necrosis factor-alpha (TNFalpha) attenuated KC-induced alterations in hepatocyte fatty acid oxidation, triglyceride accumulation, and insulin responsiveness. Importantly, KC TNFalpha levels were increased by diet in vivo and in isolated M1-polarized KCs in vitro. CONCLUSIONS: These data demonstrate a role for liver macrophages in diet-induced alterations in hepatic lipid metabolism and insulin sensitivity, and suggest a role for these cells in the etiology of the metabolic abnormalities of obesity/type 2 diabetes.


Subject(s)
Fatty Liver/prevention & control , Insulin Resistance/physiology , Kupffer Cells/metabolism , Liver/metabolism , Liver/pathology , Adipose Tissue/pathology , Animals , Antigens, CD/genetics , Antigens, Differentiation/genetics , Antigens, Differentiation, Myelomonocytic/genetics , Dietary Fats/pharmacology , Energy Intake , Gadolinium/pharmacology , Glucose Clamp Technique , Hepatocytes/cytology , Hepatocytes/pathology , Immunohistochemistry , Kupffer Cells/drug effects , Kupffer Cells/pathology , Kupffer Cells/physiology , Male , Membrane Glycoproteins/genetics , Rats , Rats, Wistar , Sucrose/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL