Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Am J Physiol ; 270(1 Pt 1): G203-12, 1996 Jan.
Article in English | MEDLINE | ID: mdl-8772519

ABSTRACT

During glyceryl trioleate absorption in the rat, mucosal triacylglycerol (TG) fatty acids have been shown to consist of only 71% exogenous oleate. Chylomicron remnants are enriched with endogenous TG fatty acids, compared with their parent chylomicrons, which consist primarily of exogenous TG fatty acids. Because enterocytes have the apolipoprotein B-100/E receptor, this study was directed at determining whether the cells can take up and metabolize chylomicron remnants and, if so, whether this was receptor mediated. Isolated enterocytes were incubated with purified 3H-labeled chylomicron remnants. The remnants were shown to be taken up by the basolateral membrane, not the apical membrane. Remnant uptake was proportional to time and number of enterocytes, and saturation kinetics were observed. Nonradiolabeled remnants, human low-density lipoprotein (LDL), anti-LDL receptor antibody, and receptor-associated protein, an LDL-related receptor inhibitor, were all shown to compete for or reduce 3H-remnant uptake. Remnants taken up by the enterocytes could not be removed on incubation with excess human LDL. Uptake was shown to be greatest in the villus tips of the proximal intestine. These studies suggest that enterocytes take up chylomicron remnants by a receptor-mediated process from their basolateral membranes and that the remnants could provide a source of endogenous TG fatty acids for the enterocytes.


Subject(s)
Chylomicrons/metabolism , Intestinal Mucosa/metabolism , Receptors, LDL/metabolism , Alkaline Phosphatase/metabolism , Animals , Antibodies/immunology , Binding, Competitive , Cell Count , Cholesterol/metabolism , Humans , Intestines/cytology , Lipoproteins, LDL/metabolism , Microvilli/metabolism , Rats , Rats, Sprague-Dawley , Receptors, LDL/immunology , Time Factors , Tissue Distribution , Triglycerides/metabolism
2.
Cancer Res ; 51(11): 2803-7, 1991 Jun 01.
Article in English | MEDLINE | ID: mdl-2032220

ABSTRACT

Multiple cytokines stimulate hepatic lipogenesis in rodents. We have previously shown that lipogenic cytokines can be divided into 2 classes by their mechanism of action and their synergistic interactions. We now report the effects of interleukin 4, a cytokine known to inhibit the synthesis and action of other cytokines. Interleukin 4 by itself did not alter hepatic lipogenesis. However, interleukin 4 inhibited the characteristic stimulation of hepatic lipogenesis that is seen with tumor necrosis factor, interleukin 1, and interleukin 6. These 3 cytokines stimulate hepatic lipogenesis by the same mechanism, increasing hepatic levels of citrate, a key allosteric activator of acetyl CoA carboxylase, the rate-limiting enzyme of fatty acid synthesis. Interleukin 4 blocks the ability of tumor necrosis factor to increase hepatic citrate. In contrast, interleukin 4 does not block the stimulation of hepatic lipogenesis by interferon-alpha, a cytokine that increases hepatic lipogenesis by a mechanism other than increasing hepatic citrate levels. These results demonstrate that interleukin 4 can inhibit the metabolic action of selected cytokines, which provides strong support for our proposal that lipogenic cytokines operate through 2 distinct mechanisms of action and can therefore be divided into 2 separate classes based on their interactions. These results also emphasize the multiple relationships between the immune response and lipid metabolism.


Subject(s)
Interleukin-4/pharmacology , Lipids/biosynthesis , Liver/metabolism , Animals , Interferon Type I/antagonists & inhibitors , Interleukin-1/antagonists & inhibitors , Interleukin-6/antagonists & inhibitors , Male , Mice , Tumor Necrosis Factor-alpha/antagonists & inhibitors
3.
Arterioscler Thromb ; 11(3): 495-500, 1991.
Article in English | MEDLINE | ID: mdl-2029492

ABSTRACT

Infection and inflammation are associated with hypertriglyceridemia, which is thought to be mediated by cytokines. Previous studies at our laboratory and others have shown that tumor necrosis factor acutely increases serum triglyceride levels primarily by stimulating hepatic lipid synthesis and secretion. The role of interleukin-1 (IL-1), a cytokine that is also secreted by stimulated macrophages and that has many actions that overlap those of tumor necrosis factor, has not been studied in depth. The present study demonstrates that IL-1, at doses similar to those that cause fever and anorexia and that stimulate adrenocorticotropic hormone secretion, rapidly increases serum triglyceride levels; this elevation persists for at least 17 hours. Serum cholesterol levels are not altered by IL-1. Neither is the clearance of triglyceride-rich lipoproteins affected by IL-1. However, hepatic triglyceride secretion, measured by the Triton WR-1339 technique, is increased in IL-1-treated animals. Accompanying this stimulation in hepatic lipid secretion is an increase in de novo fatty acid synthesis in the liver. IL-1 does not increase serum free fatty acid and glycerol levels, suggesting that IL-1 does not stimulate lipolysis in vivo. Additionally, inhibition of lipolysis does not prevent the increase in serum triglyceride levels, providing further evidence that lipolysis does not play a crucial role in the increased hepatic lipid synthesis and secretion induced by IL-1. In contrast, tumor necrosis factor increases lipolysis, which contributes to the increase in serum triglycerides. That multiple cytokines rapidly elevate plasma triglyceride levels suggest that these changes in lipid metabolism may play an important role in the organism's response to infection and inflammation.


Subject(s)
Interleukin-1/pharmacology , Lipids/blood , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cholesterol/biosynthesis , Cholesterol/blood , Chylomicrons/metabolism , Fatty Acids/biosynthesis , Fatty Acids/blood , Fatty Acids, Nonesterified/blood , Glycerol/blood , Lipolysis , Liver/metabolism , Male , Rats , Rats, Inbred Strains , Triglycerides/blood , Triglycerides/metabolism
4.
Diabetes ; 39(12): 1569-74, 1990 Dec.
Article in English | MEDLINE | ID: mdl-1978829

ABSTRACT

Previous studies demonstrated that administration of tumor necrosis factor (TNF) to diabetic rats rapidly increases serum triglyceride levels and stimulates hepatic lipogenesis without affecting the activity of adipose tissue lipoprotein lipase or serum insulin levels. The purpose of this study was to determine the mechanism by which TNF increases serum triglyceride levels and stimulates hepatic fatty acid synthesis in diabetic animals. The maximal increase (approximately 2-fold) in serum triglyceride levels in diabetic rats is seen with a dose of 10 micrograms TNF/200 g body wt, and the half-maximal effect is observed with 5 micrograms TNF/200 g body wt. The clearance of labeled triglyceride-rich lipoproteins from the circulation is not affected by TNF administration (triglyceride t 1/2; diabetic vs. TNF-administered diabetic, 3.5 +/- 0.7 vs. 4.0 +/- 0.6 min, respectively; NS). The production of triglyceride, measured by the Triton WR-1339 technique, is increased twofold in diabetic animals after TNF administration. These results indicate that the rapid increase in serum triglyceride levels after TNF treatment is accounted for by increased hepatic lipoprotein secretion. TNF administration did not alter either the amount or activation state of hepatic acetyl-CoA carboxylase, a key regulatory enzyme in fatty acid synthesis. There was also no change in the hepatic levels of fatty acyl-CoA, an allosteric inhibitor of acetyl-CoA carboxylase. However, there was a 71% increase in hepatic citrate concentrations. Citrate is an allosteric activator of acetyl-CoA carboxylase, and changes in hepatic citrate concentrations have been shown to mediate changes in the rates of fatty acid synthesis.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Diabetes Mellitus, Experimental/blood , Lipoproteins, VLDL/blood , Triglycerides/blood , Tumor Necrosis Factor-alpha/physiology , 3-Hydroxybutyric Acid , Acetyl-CoA Carboxylase/metabolism , Acetyl-CoA Carboxylase/physiology , Animals , Blood Glucose/analysis , Fatty Acids/biosynthesis , Female , Hydroxybutyrates/blood , Male , Rats , Rats, Inbred Strains
5.
Endocrinology ; 127(5): 2247-52, 1990 Nov.
Article in English | MEDLINE | ID: mdl-2226313

ABSTRACT

Several studies have demonstrated that intestinal triglyceride production and secretion are increased in diabetic animals and may contribute to the hypertriglyceridemia that accompanies diabetes. There are three potential sources of fatty acids for intestinally derived triglycerides; de novo fatty acid synthesis, circulating free fatty acids, or dietarily derived fatty acids. Prior data have demonstrated that de novo cholesterol synthesis is increased in the small intestine of diabetic animals. The primary aim of the present study was to determine the effect of diabetes on small intestinal de novo fatty acid synthesis. We found that de novo fatty acid synthesis in the small intestine is increased approximately 2-fold in streptozotocin-induced diabetic rats. In contrast, hepatic fatty acid synthesis is decreased in the diabetic animals. The increase in intestinal fatty acid synthesis is observed in both the fed and fasting states. Limiting food intake by pair feeding prevents the diabetic-induced increase in small intestinal fatty acid synthesis, a finding similar to previous data on cholesterol synthesis. Thus, the increase in both small intestinal cholesterol and fatty acid synthesis is dependent on the increased food intake that accompanies poorly controlled diabetes. The present study indicates that increases in small intestinal de novo fatty acid synthesis in diabetic animals could play an important role in providing fatty acids for increased small intestinal triglyceride synthesis and secretion.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Fatty Acids/biosynthesis , Intestine, Small/metabolism , Animals , Diabetes Mellitus, Experimental/pathology , Fasting , Female , Hypertrophy , Intestine, Small/pathology , Lipid Metabolism , Liver/metabolism , Rats , Time Factors
6.
Cancer Res ; 50(14): 4233-8, 1990 Jul 15.
Article in English | MEDLINE | ID: mdl-2364381

ABSTRACT

The significance of potential second messengers as mediators of the metabolic effects of tumor necrosis factor (TNF) was explored by studying their role in stimulating hepatic lipogenesis. Platelet-activating factor and prostaglandins have previously been suggested to mediate some of the toxic effects of TNF. An inhibitor of platelet-activating factor (WEB 2086) and two inhibitors of the synthesis of prostaglandins (ibuprofen and aspirin) had no effect on the ability of TNF to increase hepatic lipogenesis or serum triglyceride levels in the rat. Another inhibitor of the toxic effects of TNF, pentoxifylline, also had no effect on lipid metabolism in the rat. Catecholamines are increased after TNF administration, but alpha- and beta-adrenergic blockade did not prevent the lipogenic effects of TNF. However, interleukin 6, a cytokine whose synthesis and secretion are induced by TNF, is able to acutely stimulate hepatic lipogenesis in mice. Interleukin 6 stimulates hepatic lipogenesis by increasing hepatic citrate concentrations, the same mechanism by which TNF stimulates hepatic lipogenesis. These data suggest that interleukin 6, but not platelet-activating factor, prostaglandins, or catecholamines, could potentially mediate the lipogenic effects of TNF.


Subject(s)
Interleukin-6/pharmacology , Lipids/biosynthesis , Liver/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Aspirin/pharmacology , Azepines/pharmacology , Cholesterol/biosynthesis , Cholesterol/metabolism , Fatty Acids/biosynthesis , Ibuprofen/pharmacology , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Oleic Acid , Oleic Acids/metabolism , Pentoxifylline/pharmacology , Phenoxybenzamine/pharmacology , Platelet Activating Factor/antagonists & inhibitors , Propranolol/pharmacology , Rats , Rats, Inbred Strains , Reference Values , Triazoles/pharmacology , Triglycerides/biosynthesis
7.
Endocrinology ; 127(1): 46-54, 1990 Jul.
Article in English | MEDLINE | ID: mdl-1972922

ABSTRACT

Tumor necrosis factor (TNF) increases serum triglycerides in rats by increasing de novo hepatic fatty acid synthesis and very low density lipoprotein production. We have recently shown that several other cytokines increase hepatic fatty acid synthesis in the mouse. We now explore the mechanism by which these cytokines increase de novo lipogenesis and the interactions between cytokines in fed mice. TNF administration results in increased hepatic levels of citrate, the primary allosteric activator of acetyl-CoA carboxylase, which is the major rate-limiting enzyme for fatty acid synthesis. The TNF-induced increase in citrate occurs within 15 min of administration, early enough to account for the acute rise in hepatic fatty acid synthesis seen by 30 min after TNF administration. IL-1, which also increases hepatic fatty acid synthesis, produces similar increases in hepatic citrate levels. In contrast, another potent stimulator of hepatic fatty acid synthesis, interferon-alpha (IFN alpha), has no effect on hepatic citrate levels. There were no acute effects of TNF or IL-1 on the activation state of acetyl-CoA carboxylase. A trend toward an increase in the activation state of acetyl-CoA carboxylase was seen after IFN alpha administration. Low doses of TNF and IL-1 given in combination show no synergy while maximal doses are not additive. In contrast, when a low dose of either TNF or IL-1 is combined with a low dose of IFN alpha, there is synergy in stimulating hepatic fatty acid synthesis. A maximal dose of TNF or IL-1 and a high dose of IFN alpha produce a further increase in hepatic fatty acid synthesis. These data support the concept that there are two classes of cytokines that stimulate hepatic fatty acid synthesis, those that can increase hepatic citrate levels and those that cannot.


Subject(s)
Interferon Type I/pharmacology , Interleukin-1/pharmacology , Lipids/biosynthesis , Liver/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Acetyl-CoA Carboxylase/metabolism , Acyl Coenzyme A/metabolism , Animals , Citrates/metabolism , Citric Acid , Fatty Acids/biosynthesis , Female , Liver/drug effects , Mice , Mice, Inbred C57BL
8.
Metabolism ; 39(6): 623-32, 1990 Jun.
Article in English | MEDLINE | ID: mdl-2352479

ABSTRACT

Previous studies have demonstrated that tumor necrosis factor (TNF) acutely increases serum triglyceride levels and stimulates hepatic lipid synthesis. In this study, we determined the effects of TNF on serum lipid levels and hepatic lipid synthesis in animals whose diets and feeding conditions were varied to induce changes in baseline serum lipid levels and/or rates of hepatic lipid synthesis. In animals studied at both the nadir and peak of the diurnal cycle of hepatic lipid synthesis, TNF acutely increases serum triglyceride levels, stimulates hepatic fatty acid synthesis, and increases the quantity of newly synthesized fatty acids found in the serum. Similarly, in animals ingesting either high-sucrose or cholesterol-enriched diets, TNF induces the characteristic rapid increase in serum triglyceride levels, hepatic fatty acid synthesis, and quantity of labeled fatty acids in the serum. In animals fed a diet high in triglycerides, using either corn oil or lard, TNF stimulates hepatic fatty acid synthesis and increases the quantity of newly synthesized fatty acids in the serum, but serum triglyceride levels do not change. However, TNF inhibits gastric emptying, which results in a marked decrease in fat absorption in TNF-treated animals. It is likely that a decrease in the dietary contribution to serum triglyceride levels during high-triglyceride feeding counterbalances the increased hepatic contribution induced by TNF treatment. In animals fasted before TNF administration there was no acute change in either serum lipid levels, hepatic fatty acid synthesis, or the quantity of labeled fatty acids in the serum. Thus, TNF stimulates hepatic fatty acid synthesis and increases serum triglyceride levels under many diverse dietary conditions, suggesting that there is a strong linkage between the immune system and lipid metabolism that is independent of most dietary manipulations and may be of fundamental importance in the body's response to infection.


Subject(s)
Diet , Lipids/biosynthesis , Liver/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cholesterol, Dietary/pharmacology , Deuterium , Fasting , Fatty Acids/blood , Intestine, Small/metabolism , Male , Rats , Rats, Inbred Strains , Stimulation, Chemical , Sucrose/administration & dosage , Sucrose/pharmacology , Water
9.
Endocrinology ; 125(1): 267-74, 1989 Jul.
Article in English | MEDLINE | ID: mdl-2661207

ABSTRACT

We have previously shown that administration of tumor necrosis factor-alpha (TNF alpha) to intact rats results in an acute (within 60-120 min) stimulation of hepatic fatty acid synthesis, which persists for an extended period. Hepatic cholesterol synthesis is also stimulated by 16-17 h after TNF alpha treatment. We now demonstrate, using intact mice, that stimulation of hepatic lipid synthesis is not solely the property of the cytokine TNF alpha. Incorporation of 3H2O into fatty acids in the liver was increased 60-120 min and 16-17 h after the administration of TNF beta, interleukin-1 (IL-1), and interferon-alpha (IFN alpha). TNF alpha, IL-1, and IFN alpha all rapidly stimulate hepatic fatty acid synthesis (within 0-30 min), with the peak occurring at 60-120 min. The half-maximal doses of TNF alpha (200 ng) and IL-1 (20 ng) that stimulate hepatic fatty acid synthesis are similar to those that induce fever, a well recognized biological effect of these cytokines. Additionally, hepatic cholesterol synthesis was increased 16-17 h after TNF beta, IL-1, and IFN gamma treatment. The present study demonstrates that multiple cytokines from different cell types which act through different receptors can stimulate hepatic fatty acid and cholesterol synthesis. Previous studies have shown that multiple cytokines can inhibit the synthesis and storage of fat in cultured adipose cells. Taken together, these data indicate that multiple signals to perturb lipid metabolism may be produced as a consequence of an immunological or inflammatory response.


Subject(s)
Biological Factors/pharmacology , Lipids/biosynthesis , Liver/metabolism , Animals , Biological Factors/administration & dosage , Cytokines , Dose-Response Relationship, Drug , Male , Mice , Stimulation, Chemical , Time Factors
10.
J Clin Invest ; 83(4): 1116-21, 1989 Apr.
Article in English | MEDLINE | ID: mdl-2703526

ABSTRACT

Tumor necrosis factor (TNF) administration produces an increase in plasma triglycerides that may be due to inhibition of adipose lipoprotein lipase activity and/or a stimulation of hepatic lipogenesis. We now report that TNF administration to insulinopenic diabetic rats increases serum triglycerides (2 h, 2.4-fold; 17 h, 4.3-fold). Adipose tissue lipoprotein lipase activity was markedly decreased in diabetic animals compared with controls and was not further inhibited by TNF. Incorporation of tritiated water into fatty acids in the liver was increased 45% 1-2 h after TNF and 87% at 16-17 h. These results indicate that the TNF-induced increase in circulating lipid levels can occur in the absence of a TNF-induced inhibition of adipose tissue lipoprotein lipase activity. Moreover, the clearance from the circulation of triglycerides in chylomicrons was similar in control and TNF-treated animals; these results provide further evidence that the removal of triglyceride-rich lipoproteins is not altered in the TNF-treated animals. Our data suggest that the TNF-induced stimulation of hepatic lipid synthesis may play an important role in the increase in serum triglycerides. In addition, TNF administration to diabetic animals leads to an elevation in serum glucose levels (73% at 17 h) without a change in serum insulin levels. Thus, TNF stimulation of hepatic lipogenesis is independent of changes in insulin.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Lipid Metabolism , Tumor Necrosis Factor-alpha/pharmacokinetics , Adipose Tissue/enzymology , Animals , Blood Glucose/analysis , Chylomicrons/metabolism , Diabetes Mellitus, Experimental/blood , Fasting , Female , Lipids/biosynthesis , Lipids/blood , Lipoprotein Lipase/metabolism , Male , Metabolic Clearance Rate/drug effects , Rats , Rats, Inbred Strains
11.
Gastroenterology ; 96(2 Pt 1): 307-13, 1989 Feb.
Article in English | MEDLINE | ID: mdl-2521331

ABSTRACT

Previous studies have demonstrated that a variety of conditions that result in an increase in food intake lead to an increase in small-intestinal cholesterol synthesis. In the present study, it was determined whether hyperphagia induces an increase in cholesterol synthesis in segments of the small intestine excluded from contact with the food stream and whether this increase would occur in bypassed segments of the proximal or mid-small intestine. In hyperphagic diabetic rats, cholesterol synthesis is increased 91% in the proximal portion of the small intestine excluded from contact with nutrients. In lactating rats, another model of hyperphagia, cholesterol synthesis is increased 2.4-fold in midintestinal segments excluded from contact with the food stream and 2.9-fold in segments of the proximal intestine that have been bypassed. These observations demonstrate that the hyperphagia-induced increase in small-intestinal cholesterol synthesis will occur in portions of the small intestine, even if contact with the food stream is prevented. In addition, this data demonstrated that the mass of the bypassed portion of the small intestine is increased in hyperphagic animals. In diabetic animals, the weight of the bypassed proximal intestine is increased 2.1-fold, whereas in lactating animals the mass is increased 50% in the bypassed midintestine and 74% in the bypassed proximal small intestine. In conclusion, the present study suggests that circulating or neurologic factors, or both, play a role in stimulating intestinal cholesterol synthesis in hyperphagic animals. These findings also suggest that indirect factors play a role in the increase in intestinal mass associated with hyperphagia.


Subject(s)
Cholesterol/biosynthesis , Hyperphagia/metabolism , Intestine, Small/metabolism , Animals , Diabetes Mellitus, Experimental/metabolism , Female , Jejunoileal Bypass/methods , Lactation , Pregnancy , Rats , Rats, Inbred Strains , Streptozocin
12.
Diabetes ; 38(2): 219-24, 1989 Feb.
Article in English | MEDLINE | ID: mdl-2914624

ABSTRACT

Cholesterol synthesis is increased two- to threefold in the small intestine of diabetic rats. We have observed, in three separate experiments, that the characteristic increase in small intestinal cholesterol synthesis (SICS) in diabetic rats was prevented by total gastrectomy. Food intake was increased twofold, and the small intestine hypertrophied in the gastrectomized diabetic animals. In normal animals, total gastrectomy resulted in only a very small increase in intestinal cholesterol synthesis. In hyperphagic lactating animals, total gastrectomy did not prevent the characteristic increase in SICS that is usually observed in this hyperphagic model. These results indicate that the effects of total gastrectomy on preventing an increase in SICS are relatively specific for the diabetic state. The mechanism by which total gastrectomy prevents the increase in intestinal cholesterol synthesis in diabetic animals is unknown. Vagotomy did not prevent the typical increase in intestinal synthesis in diabetic animals. Additionally, selectively removing either the antrum or fundus of the stomach did not prevent the increase in SICS in diabetic animals, indicating that the inhibition requires the removal of the entire stomach. It can be speculated that the stomach produces a substance that induces the increase in SICS observed in diabetic animals and that total gastrectomy removes this stimulatory substance.


Subject(s)
Cholesterol/biosynthesis , Diabetes Mellitus, Experimental/metabolism , Gastrectomy , Intestine, Small/metabolism , Animals , Blood Glucose/analysis , Eating , Female , Rats , Rats, Inbred Strains , Reference Values
13.
Biochem Biophys Res Commun ; 153(2): 576-82, 1988 Jun 16.
Article in English | MEDLINE | ID: mdl-3132913

ABSTRACT

TNF is cytotoxic to tumor cell lines but enhances growth of some nontransformed cells. Because animals administered TNF have an increase in liver size, we studied the [3H]thymidine incorporation into DNA in the liver of intact rats. A significant increase in [3H]thymidine incorporation is seen 20 hours following TNF administration and peaks at 24 hours. The lowest dose of TNF that increases DNA synthesis is 10 micrograms/200 g rat with a maximal increase occurring with 25 micrograms/200 g, considerably less than the dose required for maximally increasing plasma triglycerides. The increase in [3H]thymidine incorporation was shown to be due to an increase in DNA polymerase alpha activity (associated with the replication of DNA) rather than DNA polymerases beta (associated with DNA repair) plus gamma activity. These results indicate that TNF administration stimulates DNA replication in the liver of intact animals.


Subject(s)
DNA/biosynthesis , Liver/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Animals , Aphidicolin , DNA Repair/drug effects , DNA Replication/drug effects , Diterpenes/pharmacology , Dose-Response Relationship, Drug , Liver/metabolism , Liver Regeneration , Male , Rats , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...