Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 115
Filter
1.
Nutrients ; 12(6)2020 Jun 15.
Article in English | MEDLINE | ID: mdl-32549314

ABSTRACT

A high fructose intake exacerbates postprandial plasma triacylglycerol (TAG) concentration, an independent risk factor for cardiovascular disease, although it is unclear whether this is due to increased production or impaired clearance of triacylglycerol (TAG)-rich lipoproteins. We determined the in vivo acute effect of fructose on postprandial intestinal and hepatic lipoprotein TAG kinetics and de novo lipogenesis (DNL). Five overweight men were studied twice, 4 weeks apart. They consumed hourly mixed-nutrient drinks that were high-fructose (30% energy) or low-fructose (<2% energy) for 11 h. Oral 2H2O was administered to measure fasting and postprandial DNL. Postprandial chylomicron (CM)-TAG and very low-density lipoprotein (VLDL)-TAG kinetics were measured with an intravenous bolus of [2H5]-glycerol. CM and VLDL were separated by their apolipoprotein B content using antibodies. Plasma TAG (p < 0.005) and VLDL-TAG (p = 0.003) were greater, and CM-TAG production rate (PR, p = 0.046) and CM-TAG fractional catabolic rate (FCR, p = 0.073) lower when high-fructose was consumed, with no differences in VLDL-TAG kinetics. Insulin was lower (p = 0.005) and apoB48 (p = 0.039), apoB100 (p = 0.013) and non-esterified fatty acids (NEFA) (p = 0.013) were higher after high-fructose. Postprandial hepatic fractional DNL was higher than intestinal fractional DNL with high-fructose (p = 0.043) and low-fructose (p = 0.043). Fructose consumption had no effect on the rate of intestinal or hepatic DNL. We provide the first measurement of the rate of intestinal DNL in humans. Lower CM-TAG PR and CM-TAG FCR with high-fructose consumption suggests lower clearance of CM, rather than elevated production, may contribute to elevated plasma TAG, possibly due to lower insulin-mediated stimulation of lipoprotein lipase.


Subject(s)
Fatty Acids/biosynthesis , Fructose/administration & dosage , Intestines/drug effects , Lipogenesis/drug effects , Triglycerides/biosynthesis , Adult , Beverages , Chylomicrons/biosynthesis , Diet , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Lipoproteins, VLDL/biosynthesis , Liver/drug effects , Liver/metabolism , Male , Middle Aged , Postprandial Period/physiology , Triglycerides/blood
2.
Drug Dev Ind Pharm ; 46(4): 557-565, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32126844

ABSTRACT

Raloxifene hydrochloride (RH) suffers from low oral bioavailability due to its low water-solubility and first-pass metabolism. Therefore, a novel phospholipid complex of RH (RHPC) and a matrix dispersion based on phospholipid complex (RHPC-MD) were successfully prepared and optimized. Several methods were used to validate the formation of RHPC and RHPC-MD, such as differential scanning calorimetry, X-ray diffraction, scanning electron microscopy, transmission electron microscopy, infrared spectroscopy, particle size, and zeta potential, meanwhile, their octanol-water partition coefficient, solubility, and dissolution in vitro were also evaluated. To investigate the absorption mechanism of RHPC in vivo, the RHPC was administered to the chylomicron flow blockage rat model. Interestingly, as we expected, a significant reduction in RHPC absorption (67%) (**p< .01) in presence of cycloheximide (CXI) inhibitor was observed, thus confirming the RHPC could be absorbed by lymphatic transport in vivo. Pharmacokinetic studies revealed that the relative oral bioavailability of RHPC as well as RHPC-MD was 223% and 329%, respectively, when comparing with the commercial RH tablets. These outcomes suggested that the current study provided an attractive formulation to enhance the oral bioavailability of RH and stimulated to further research the absorption mechanism of RHPC in vivo.


Subject(s)
Bone Density Conservation Agents/administration & dosage , Phospholipids/chemistry , Raloxifene Hydrochloride/administration & dosage , Selective Estrogen Receptor Modulators/administration & dosage , Administration, Oral , Animals , Biological Availability , Bone Density Conservation Agents/chemistry , Bone Density Conservation Agents/pharmacokinetics , Calorimetry, Differential Scanning , Chylomicrons/biosynthesis , Cycloheximide/administration & dosage , Drug Liberation , Female , Humans , Intestinal Absorption/drug effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestine, Small/drug effects , Intestine, Small/metabolism , Models, Animal , Osteoporosis, Postmenopausal/drug therapy , Particle Size , Raloxifene Hydrochloride/chemistry , Raloxifene Hydrochloride/pharmacokinetics , Rats , Selective Estrogen Receptor Modulators/chemistry , Selective Estrogen Receptor Modulators/pharmacokinetics , Solubility , Tablets , X-Ray Diffraction
3.
Nutrients ; 11(6)2019 Jun 08.
Article in English | MEDLINE | ID: mdl-31181761

ABSTRACT

Postprandial lipemia, which is one of the main characteristics of the atherogenic dyslipidemia with fasting plasma hypertriglyceridemia, low high-density lipoprotein cholesterol and an increase of small and dense low-density lipoproteins is now considered a causal risk factor for atherosclerotic cardiovascular disease and all-cause mortality. Postprandial lipemia, which is mainly related to the increase in chylomicron production, is frequently elevated in individuals at high cardiovascular risk such as obese or overweight patients, type 2 diabetic patients and subjects with a metabolic syndrome who share an insulin resistant state. It is now well known that chylomicron production and thus postprandial lipemia is highly regulated by many factors such as endogenous factors: circulating factors such as hormones or free fatty acids, genetic variants, circadian rhythms, or exogenous factors: food components, dietary supplements and prescription drugs. In this review, we focused on the effect of nutrients, micronutrients and phytochemicals but also on food structure on chylomicron production and postprandial lipemia.


Subject(s)
Chylomicrons/biosynthesis , Diet , Feeding Behavior , Hyperlipidemias/etiology , Micronutrients/pharmacology , Postprandial Period , Cardiovascular Diseases/etiology , Cardiovascular Diseases/prevention & control , Humans , Hyperlipidemias/prevention & control , Metabolic Diseases/etiology , Metabolic Diseases/prevention & control , Micronutrients/analysis , Micronutrients/therapeutic use , Nutrients/analysis , Nutrients/pharmacology , Nutrients/therapeutic use , Phytochemicals/chemistry , Phytochemicals/pharmacology , Phytochemicals/therapeutic use , Plant Extracts/chemistry , Plant Extracts/pharmacology , Plant Extracts/therapeutic use
4.
Cell Mol Gastroenterol Hepatol ; 7(3): 487-501, 2019.
Article in English | MEDLINE | ID: mdl-30819663

ABSTRACT

Rapid and efficient digestion and absorption of dietary triglycerides and other lipids by the intestine, the packaging of those lipids into lipoprotein chylomicron (CM) particles, and their secretion via the lymphatic duct into the blood circulation are essential in maintaining whole-body lipid and energy homeostasis. Biosynthesis and assembly of CMs in enterocytes is a complex multistep process that is subject to regulation by intracellular signaling pathways as well as by hormones, nutrients, and neural factors extrinsic to the enterocyte. Dysregulation of this process has implications for health and disease, contributing to dyslipidemia and a potentially increased risk of atherosclerotic cardiovascular disease. There is increasing recognition that, besides intracellular regulation of CM assembly and secretion, regulation of postassembly pathways also plays important roles in CM secretion. This review examines recent advances in our understanding of the regulation of CM secretion in relation to mobilization of intestinal lipid stores, drawing particular attention to post-assembly regulatory mechanisms, including intracellular trafficking of triglycerides in enterocytes, CM mobilization from the lamina propria, and regulated transport of CM by intestinal lymphatics.


Subject(s)
Chylomicrons/metabolism , Animals , Biological Transport , Chylomicrons/biosynthesis , Dietary Fats/metabolism , Humans , Lipid Metabolism , Lymph/metabolism , Mucous Membrane/metabolism
5.
J Clin Invest ; 129(1): 281-295, 2019 01 02.
Article in English | MEDLINE | ID: mdl-30507612

ABSTRACT

The lipin phosphatidic acid phosphatase (PAP) enzymes are required for triacylglycerol (TAG) synthesis from glycerol 3-phosphate in most mammalian tissues. The 3 lipin proteins (lipin 1, lipin 2, and lipin 3) each have PAP activity, but have distinct tissue distributions, with lipin 1 being the predominant PAP enzyme in many metabolic tissues. One exception is the small intestine, which is unique in expressing exclusively lipin 2 and lipin 3. TAG synthesis in small intestinal enterocytes utilizes 2-monoacylglycerol and does not require the PAP reaction, making the role of lipin proteins in enterocytes unclear. Enterocyte TAGs are stored transiently as cytosolic lipid droplets or incorporated into lipoproteins (chylomicrons) for secretion. We determined that lipin enzymes are critical for chylomicron biogenesis, through regulation of membrane phospholipid composition and association of apolipoprotein B48 with nascent chylomicron particles. Lipin 2/3 deficiency caused phosphatidic acid accumulation and mammalian target of rapamycin complex 1 (mTORC1) activation, which were associated with enhanced protein levels of a key phospholipid biosynthetic enzyme (CTP:phosphocholine cytidylyltransferase α) and altered membrane phospholipid composition. Impaired chylomicron synthesis in lipin 2/3 deficiency could be rescued by normalizing phospholipid synthesis levels. These data implicate lipin 2/3 as a control point for enterocyte phospholipid homeostasis and chylomicron biogenesis.


Subject(s)
Chylomicrons/biosynthesis , Enterocytes/metabolism , Homeostasis , Phosphatidate Phosphatase/metabolism , Phospholipids/metabolism , Animals , Apolipoprotein B-48/genetics , Apolipoprotein B-48/metabolism , Chylomicrons/genetics , Enterocytes/cytology , Female , Lipid Droplets/metabolism , Male , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Knockout , Phosphatidate Phosphatase/genetics , Phospholipids/genetics , Triglycerides/biosynthesis , Triglycerides/genetics
6.
Arterioscler Thromb Vasc Biol ; 38(9): 2198-2206, 2018 09.
Article in English | MEDLINE | ID: mdl-30026275

ABSTRACT

Objective- Treatment with liraglutide, a GLP-1 (glucagon-like peptide-1) agonist, has been shown to reduce postprandial lipidemia, an important feature of diabetic dyslipidemia. However, the underlying mechanisms for this effect remain unknown. This prompted us to study the effect of liraglutide on the metabolism of ApoB48 (apolipoprotein B48). Approach and Results- We performed an in vivo kinetic study with stable isotopes (D8-valine) in the fed state in 10 patients with type 2 diabetes mellitus before treatment and 6 months after the initiation of treatment with liraglutide (1.2 mg/d). We also evaluated, in mice, the effect of a 1-week liraglutide treatment on postload triglycerides and analysed in vitro on jejunum, the direct effect of liraglutide on the expression of genes involved in the biosynthesis of chylomicron. In diabetic patients, liraglutide treatment induced a dramatic reduction of ApoB48 pool (65±38 versus 162±87 mg; P=0.005) because of a significant decrease in ApoB48 production rate (3.02±1.33 versus 6.14±4.27 mg kg-1 d-1; P=0.009) and a significant increase in ApoB48 fractional catabolic rate (5.12±1.35 versus 3.69±0.75 pool d-1; P=0.005). One-week treatment with liraglutide significantly reduced postload plasma triglycerides in mice and liraglutide, in vitro, reduced the expression of ApoB48, DGAT1 (diacylglycerol O-acyltransferase 1), and MTP (microsomal transfer protein) genes. Conclusions- We show that treatment with liraglutide induces a significant reduction of the ApoB48 pool because of both a reduction of ApoB48 production and an increase in ApoB48 catabolism. In vitro, liraglutide reduces the expression of genes involved in chylomicron synthesis. These effects might benefit cardiovascular health. Clinical Trial Registration- URL: https://www.clinicaltrials.gov . Unique identifier: NCT02721888.


Subject(s)
Apolipoprotein B-48/blood , Diabetes Mellitus, Type 2/complications , Hyperlipidemias/blood , Hyperlipidemias/drug therapy , Liraglutide/therapeutic use , Adipose Tissue/metabolism , Animals , Apolipoprotein B-48/drug effects , Apolipoprotein B-48/genetics , Carrier Proteins/genetics , Carrier Proteins/metabolism , Chylomicrons/biosynthesis , Diabetes Mellitus, Type 2/blood , Diacylglycerol O-Acyltransferase/genetics , Diacylglycerol O-Acyltransferase/metabolism , Female , Gene Expression , Humans , Hyperlipidemias/complications , Jejunum/metabolism , Lipoprotein Lipase/metabolism , Male , Mice, Inbred BALB C , Postprandial Period , Prospective Studies , Triglycerides/blood
7.
Compr Physiol ; 8(2): 493-507, 2018 03 26.
Article in English | MEDLINE | ID: mdl-29687890

ABSTRACT

Several proteins have been implicated in fatty acid (FA) transport by enterocytes including the scavenger receptor CD36 (SR-B2), the scavenger receptor B1 (SR-B1) a member of the CD36 family and the FA transport protein 4 (FATP4). Here, we review the regulation of enterocyte FA uptake and its function in lipid absorption including prechylomicron formation, assembly and transport. Emphasis is given to CD36, which is abundantly expressed along the digestive tract of rodents and humans and has been the most studied. We also address the pleiotropic functions of CD36 that go beyond lipid absorption and metabolism to include recent evidence of its impact on intestinal homeostasis and barrier maintenance. Areas of progress involving contribution of membrane phospholipid remodeling and of cytosolic FA-binding proteins, FABP1 and FABP2 to fat absorption will be covered. © 2018 American Physiological Society. Compr Physiol 8:493-507, 2018.


Subject(s)
CD36 Antigens/physiology , Intestinal Absorption/physiology , Lipid Metabolism/physiology , Animals , Biological Transport/physiology , Chylomicrons/biosynthesis , Digestive System/metabolism , Enterocytes/metabolism , Fatty Acids/metabolism , Homeostasis/physiology , Humans , Phospholipids/metabolism
8.
J Lipid Res ; 58(5): 853-865, 2017 05.
Article in English | MEDLINE | ID: mdl-28159868

ABSTRACT

Since its initial report in 2009, the intestinal enteroid culture system has been a powerful tool used to study stem cell biology and development in the gastrointestinal tract. However, a major question is whether enteroids retain intestinal function and physiology. There have been significant contributions describing ion transport physiology of human intestinal organoid cultures, as well as physiology of gastric organoids, but critical studies on dietary fat absorption and chylomicron synthesis in primary intestinal enteroids have not been undertaken. Here we report that primary murine enteroid cultures recapitulate in vivo intestinal lipoprotein synthesis and secretion, and reflect key aspects of the physiology of intact intestine in regard to dietary fat absorption. We also show that enteroids can be used to elucidate intestinal mechanisms behind CVD risk factors, including tissue-specific apolipoprotein functions. Using enteroids, we show that intestinal apoC-III overexpression results in the secretion of smaller, less dense chylomicron particles along with reduced triacylglycerol secretion from the intestine. This model significantly expands our ability to test how specific genes or genetic polymorphisms function in dietary fat absorption and the precise intestinal mechanisms that are critical in the etiology of metabolic disease.


Subject(s)
Absorption, Physicochemical , Apolipoprotein C-III/metabolism , Chylomicrons/biosynthesis , Dietary Fats/metabolism , Intestinal Mucosa/metabolism , Triglycerides/metabolism , Animals , Apolipoprotein C-III/genetics , Cell Differentiation , Chylomicrons/metabolism , Humans , Intestines/cytology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity
9.
Biochim Biophys Acta ; 1851(11): 1428-41, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26303578

ABSTRACT

The phospholipid (PL) requirement in fish is revealed by enhanced performance when larvae are provided PL-enriched diets. To elucidate the molecular mechanism underlying PL requirement in Atlantic salmon, Salmo salar, were fed a minimal PL diet and tissue samples from major lipid metabolic sites were dissected from fry and parr. In silico analysis and cloning techniques demonstrated that salmon possess a full set of enzymes for the endogenous production of PL. The gene expression data indicated that major PL biosynthetic genes of phosphatidylcholine (PtdCho), phosphatidylethanolamine (PtdEtn) and phosphatidylinositol (PtdIns) display lower expression in intestine during the early developmental stage (fry). This is consistent with the hypothesis that the intestine of salmon is immature at the early developmental stage with limited capacity for endogenous PL biosynthesis. The results also indicate that intact PtdCho, PtdEtn and PtdIns are required in the diet at this stage. PtdCho and sphingomyelin constitute the predominant PL in chylomicrons, involved in the transport of dietary lipids from the intestine to the rest of the body. As sphingomyelin can be produced from PtdCho in intestine of fry, our findings suggest that supplementation of dietary PtdCho alone during early developmental stages of Atlantic salmon would be sufficient to promote chylomicron formation. This would support efficient transport of dietary lipids, including PL precursors, from the intestine to the liver where biosynthesis of PtdEtn, PtdSer, and PtdIns is not compromised as in intestine facilitating efficient utilisation of dietary energy and the endogenous production of membrane PL for the rapidly growing and developing animal.


Subject(s)
CDPdiacylglycerol-Serine O-Phosphatidyltransferase/metabolism , Dietary Fats/metabolism , Fish Proteins/metabolism , Salmo salar/metabolism , Amino Acid Sequence , Animals , Biological Transport , CDPdiacylglycerol-Serine O-Phosphatidyltransferase/genetics , Chylomicrons/biosynthesis , Dietary Fats/administration & dosage , Fish Proteins/genetics , Gene Expression Regulation, Developmental , Intestinal Mucosa/metabolism , Intestines/growth & development , Larva/genetics , Larva/growth & development , Larva/metabolism , Liver/growth & development , Liver/metabolism , Molecular Sequence Annotation , Molecular Sequence Data , Phosphatidylcholines/biosynthesis , Phosphatidylethanolamines/biosynthesis , Phosphatidylinositols/biosynthesis , Salmo salar/genetics , Salmo salar/growth & development , Sequence Alignment , Sphingomyelins/biosynthesis
10.
Annu Rev Nutr ; 35: 265-94, 2015.
Article in English | MEDLINE | ID: mdl-25974693

ABSTRACT

Dietary lipids are efficiently absorbed by the small intestine, incorporated into triglyceride-rich lipoproteins (chylomicrons), and transported in the circulation to various tissues. Intestinal lipid absorption and mobilization and chylomicron synthesis and secretion are highly regulated processes. Elevated chylomicron production rate contributes to the dyslipidemia seen in common metabolic disorders such as insulin-resistant states and type 2 diabetes and likely increases the risk for atherosclerosis seen in these conditions. An in-depth understanding of the regulation of chylomicron production may provide leads for the development of drugs that could be of therapeutic utility in the prevention of dyslipidemia and atherosclerosis. Chylomicron secretion is subject to regulation by various factors, including diet, body weight, genetic variants, hormones, nutraceuticals, medications, and emerging interventions such as bariatric surgical procedures. In this review we discuss the regulation of chylomicron production, mechanisms that underlie chylomicron dysregulation, and potential avenues for future research.


Subject(s)
Chylomicrons/biosynthesis , Homeostasis/physiology , Atherosclerosis/blood , Cholesterol, Dietary/metabolism , Cholesterol, Dietary/pharmacology , Chylomicrons/blood , Chylomicrons/genetics , Circadian Rhythm , Diabetes Mellitus, Type 2/blood , Diet , Dietary Fats/metabolism , Dietary Fats/pharmacokinetics , Dietary Supplements , Gastrointestinal Microbiome/physiology , Hormones/physiology , Humans , Insulin Resistance , Intestinal Absorption , Intestinal Mucosa/metabolism , Lipid Metabolism/physiology , Nutritional Physiological Phenomena , Triglycerides/biosynthesis , Triglycerides/blood , Triglycerides/genetics
11.
Food Funct ; 6(5): 1726-35, 2015 May.
Article in English | MEDLINE | ID: mdl-25923344

ABSTRACT

Formulating healthy food rich in omega 3 fatty acids requires prior knowledge of the parameters influencing their bioavailability and their metabolic fate. In this context, we studied the effects of various emulsifiers widely used in the food industry, on the gastrointestinal lipolysis of flaxseed oil emulsions in an in vitro model and on the intestinal absorption and lymphatic secretion of alpha-linolenic acid (ALA) in rats. In vitro data showed that the emulsification of flaxseed oil with soya lecithin improved the gastric lipolysis of the oil (+30%), while the presence of Tween 80 or of sodium caseinate decreased it (-80% and -40%, respectively). The in vivo data demonstrated that the intestinal absorption and the lymphatic secretion of ALA were improved with soya lecithin (Cmax = 24 mg mL(-1)) and reduced in the presence of sodium caseinate (Cmax = 7 mg mL(-1)) compared to unemulsified flaxseed oil (Cmax = 16 mg mL(-1)); Tween 80 had no effect. In addition, the synthesized chylomicrons were notably larger and more numerous with soya lecithin whereas they were smaller in the presence of sodium caseinate (p < 0.05). This study shows that the intestinal bioavailability of ALA was increased by the emulsification of flaxseed oil with soya lecithin via an improved lipolysis, favouring the intestinal absorption of ALA and the secretion of many large chylomicrons in lymph.


Subject(s)
Chylomicrons/biosynthesis , Gastrointestinal Tract/metabolism , Lipolysis/drug effects , alpha-Linolenic Acid/chemistry , alpha-Linolenic Acid/pharmacokinetics , Animals , Biological Availability , Chemistry, Pharmaceutical , Emulsifying Agents/chemistry , Lecithins/chemistry , Linseed Oil/chemistry , Linseed Oil/pharmacokinetics , Male , Rats , Rats, Wistar , Glycine max/chemistry
12.
Biochimie ; 96: 37-47, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23958439

ABSTRACT

Today, it is well established that the development of obesity and associated diseases results, in part, from excessive lipid intake associated with a qualitative imbalance. Among the organs involved in lipid homeostasis, the small intestine is the least studied even though it determines lipid bioavailability and largely contributes to the regulation of postprandial hyperlipemia (triacylglycerols (TG) and free fatty acids (FFA)). Several Lipid-Binding Proteins (LBP) are expressed in the small intestine. Their supposed intestinal functions were initially based on what was reported in other tissues, and took no account of the physiological specificity of the small intestine. Progressively, the identification of regulating factors of intestinal LBP and the description of the phenotype of their deletion have provided new insights into cellular and molecular mechanisms involved in fat absorption. This review will discuss the physiological contribution of each LBP in the main steps of intestinal absorption of long-chain fatty acids (LCFA): uptake, trafficking and reassembly into chylomicrons (CM). Moreover, current data indicate that the small intestine is able to adapt its lipid absorption capacity to the fat content of the diet, especially through the coordinated induction of LBP. This adaptation requires the existence of a mechanism of intestinal lipid sensing. Emerging data suggest that the membrane LBP CD36 may operate as a lipid receptor that triggers an intracellular signal leading to the modulation of the expression of LBP involved in CM formation. This event could be the starting point for the optimized synthesis of large CM, which are efficiently degraded in blood. Better understanding of this intestinal lipid sensing might provide new approaches to decrease the prevalence of postprandial hypertriglyceridemia, which is associated with cardiovascular diseases, insulin resistance and obesity.


Subject(s)
Chylomicrons/biosynthesis , Dietary Fats/metabolism , Fatty Acid-Binding Proteins/physiology , Fatty Acids/metabolism , Lipid Metabolism , Animals , CD36 Antigens/physiology , Chylomicrons/metabolism , Enterocytes/metabolism , Humans , Intestinal Absorption , Intestine, Small/metabolism
13.
Diabetes ; 62(2): 373-81, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23028139

ABSTRACT

The glucagon-like peptides (GLP-1 and GLP-2) are processed from the proglucagon polypeptide and secreted in equimolar amounts but have opposite effects on chylomicron (CM) production, with GLP-1 significantly reducing and GLP-2 increasing postprandial chylomicronemia. In the current study, we evaluated the apparent paradoxical roles of GLP-1 and GLP-2 under physiological conditions in the Syrian golden hamster, a model with close similarity to humans in terms of lipoprotein metabolism. A short (30-min) intravenous infusion of GLP-2 resulted in a marked increase in postprandial apolipoprotein B48 (apoB48) and triglyceride (TG) levels in the TG-rich lipoprotein (TRL) fraction, whereas GLP-1 infusion decreased lipid absorption and levels of TRL-TG and apoB48. GLP-1 and GLP-2 coinfusion resulted in net increased lipid absorption and an increase in TRL-TG and apoB48. However, prolonged (120-min) coinfusion of GLP-1 and GLP-2 decreased postprandial lipemia. Blocking dipeptidyl peptidase-4 activity resulted in decreased postprandial lipemia. Interestingly, fructose-fed, insulin-resistant hamsters showed a more pronounced response, including possible hypersensitivity to GLP-2 or reduced sensitivity to GLP-1. In conclusion, under normal physiological conditions, the actions of GLP-2 predominate; however, when GLP-1 activity is sustained, the hypolipidemic action of GLP-1 predominates. Pharmacological inhibition of GLP-1 degradation tips the balance toward an inhibitory effect on intestinal production of atherogenic CM particles.


Subject(s)
Glucagon-Like Peptide 1/administration & dosage , Glucagon-Like Peptide 2/administration & dosage , Hyperlipidemias/metabolism , Insulin Resistance/physiology , Intestines/drug effects , Lipoproteins/biosynthesis , Postprandial Period/drug effects , Animals , Apolipoprotein B-48/biosynthesis , Chylomicrons/biosynthesis , Chylomicrons/drug effects , Cricetinae , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Fructose/administration & dosage , Hyperlipidemias/blood , Intestinal Absorption/drug effects , Lipid Metabolism/drug effects , Male , Mesocricetus , Triglycerides/blood , Triglycerides/metabolism
14.
Article in English | MEDLINE | ID: mdl-23238590

ABSTRACT

Two lines of rainbow trout divergently selected for muscle fat content, fat line (F) and lean line (L) were used to investigate the effect of genetic selection on digestion, intestinal nutrient transport and fatty acid bioconversion, in relation to dietary starch intake. This study involved a digestibility trial for 2 weeks using Cr(2)O(3) as inert marker, followed by a feeding trial for 4 weeks. For the entire duration, juvenile trout from the two lines were fed diets with or without gelatinized starch. Blood, pyloric ceca, midgut and hindgut were sampled at 24 h after the last meal. Transcripts of the proteins involved in nutrient transport and fatty acid bioconversion were abundant in the proximal intestine. GLUT2 transcripts were slightly higher in the F line ceca than in the L line. Dietary starch intake did not enhance the transcription of intestinal glucose transporters, SGLT1 and GLUT2; but it was associated with the higher expression of ApoA1 and PepT1 in the midgut. Significantly, the F line exhibited higher intestinal mRNA levels of MTP, ApoA4, Elovl2, Elovl5 and D6D than the L line, linked to chylomicron assembly and fatty acid bioconversion. Apparent digestibility coefficients of protein, lipid and starch were high in both lines, but not significantly different between them. In conclusion, we found a higher potential of chylomicron synthesis and fatty acid bioconversion in the intestine of F line, but no adaptive transcriptional response of glucose transporters to dietary starch and no genotypic differences in nutrient digestibility.


Subject(s)
Adipose Tissue , Fatty Acids, Unsaturated/biosynthesis , Muscles , Oncorhynchus mykiss , Selection, Genetic , Adipose Tissue/growth & development , Adipose Tissue/metabolism , Animals , Chylomicrons/biosynthesis , Diet , Digestion/genetics , Digestion/physiology , Glucose Transporter Type 2/metabolism , Intestinal Mucosa/metabolism , Muscles/metabolism , Muscles/physiology , Oncorhynchus mykiss/genetics , Oncorhynchus mykiss/growth & development , Oncorhynchus mykiss/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sodium-Glucose Transporter 1/metabolism , Starch/administration & dosage
15.
PLoS One ; 7(11): e49515, 2012.
Article in English | MEDLINE | ID: mdl-23145182

ABSTRACT

Elevated postprandial plasma triacylglycerol (TG) concentrations are commonly associated with obesity and the risk of cardiovascular disease. Dietary fat contributes to this condition through the production of chylomicrons. Carboxylesterases have been mainly studied for their role in drug metabolism, but recently they have been shown to participate in lipid metabolism; however, their role in intestinal lipid metabolism is unknown. Carboxylesterase1/esterase-x (Ces1/Es-x) deficient mice become obese, hyperlipidemic and develop hepatic steatosis even on standard chow diet. Here, we aimed to explore the role of Ces1/Es-x in intestinal lipid metabolism. Six-month old wild-type and Ces1/Es-x deficient mice were maintained on chow diet and intestinal lipid metabolism and plasma chylomicron clearance were analyzed. Along the intestine Ces1/Es-x protein is expressed only in proximal jejunum. Ablation of Ces1/Es-x expression results in postprandial hyperlipidemia due to increased secretion of chylomicrons. The secreted chylomicrons have aberrant protein composition, which results in their reduced clearance. In conclusion, Ces1/Es-x participates in the regulation of chylomicron assembly and secretion. Ces1/Es-x might act as a lipid sensor in enterocytes regulating chylomicron secretion rate. Ces1/Es-x might represent an attractive pharmacological target for the treatment of lipid abnormalities associated with obesity, insulin resistance and fatty liver disease.


Subject(s)
Carboxylic Ester Hydrolases/physiology , Chylomicrons/biosynthesis , Esterases/physiology , Animals , Carboxylic Ester Hydrolases/genetics , Carboxylic Ester Hydrolases/metabolism , Chylomicrons/blood , Chylomicrons/metabolism , Esterases/genetics , Esterases/metabolism , Female , Hyperlipidemias/genetics , Jejunum/metabolism , Lipid Metabolism/genetics , Mice
16.
Article in English | MEDLINE | ID: mdl-23030446

ABSTRACT

Apolipoprotein B48 (apoB48)-containing triglyceride-rich lipoproteins are atherogenic and therefore it is important to understand factors that regulate their metabolism in the intestine. Insulin resistant states are associated with increased intestinal output of apo B48, but the mechanistic studies explaining this overproduction have relied heavily on models of diet-induced insulin resistance. There is evidence that glucagon-like peptide (GLP) secretion is diminished in insulin resistant and diabetic states, which may have implications for postprandial lipid secretion. This review presents a survey of studies on GLPs and intestinal lipoprotein metabolism, along with some insights into the net physiological significance of GLP action in postprandial lipoprotein metabolism. Studies with GLP-1 receptor agonists and dipeptidyl peptidase-IV inhibitors have indicated that GLP-1 in pharmacological and physiological doses can inhibit intestinal TRL production. On the other hand, GLP-2 has an acute stimulatory effect on intestinal apoB48 secretion. Modulating GLP action may serve as a strategy to improve postprandial dyslipidemia in insulin resistant states.


Subject(s)
Chylomicrons/biosynthesis , Glucagon-Like Peptides/metabolism , Intestinal Mucosa/metabolism , Animals , Humans
17.
J Lipid Res ; 53(12): 2643-55, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22993231

ABSTRACT

Intestinal apolipoprotein B (apoB) mRNA undergoes C-to-U editing, mediated by the catalytic deaminase apobec-1, which results in translation of apoB48. Apobec1(-/-) mice produce only apoB100 and secrete larger chylomicron particles than those observed in wild-type (WT) mice. Here we show that transgenic rescue of intestinal apobec-1 expression (Apobec1(Int/O)) restores C-to-U RNA editing of apoB mRNA in vivo, including the canonical site at position 6666 and also at approximately 20 other newly identified downstream sites present in WT mice. The small intestine of Apobec1(Int/O) mice produces only apoB48, and the liver produces only apoB100. Serum chylomicron particles were smaller in Apobec1(Int/O) mice compared with those from Apobec1(-/-) mice, and the predominant fraction of serum apoB48 in Apobec1(Int/O) mice migrated in lipoproteins smaller than chylomicrons, even when these mice were fed a high-fat diet. Because apoB48 arises exclusively from the intestine in Apobec1(Int/O) mice and intestinal apoB48 synthesis and secretion rates were comparable to WT mice, we were able to infer the major sites of origin of serum apoB48 in WT mice. Our findings imply that less than 25% of serum apoB48 in WT mice arises from the intestine, with the majority originating from the liver.


Subject(s)
Apolipoproteins B/genetics , Chylomicrons/biosynthesis , Cytidine Deaminase/deficiency , Intestine, Small/metabolism , RNA Editing , APOBEC-1 Deaminase , Animals , Apolipoproteins B/metabolism , Chromatography, High Pressure Liquid , Chylomicrons/blood , Chylomicrons/chemistry , Cytidine Deaminase/genetics , Cytidine Deaminase/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Electron , Organ Specificity , RNA, Messenger/genetics , RNA, Messenger/metabolism
18.
J Lipid Res ; 53(9): 1811-22, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22764088

ABSTRACT

Phosphatidylethanolamine is an important inner-leaflet phospholipid, and CTP:phosphoethanolamine cytidylyltransferase-Pcyt2 acts as the main regulator of the de novo phosphatidylethanolamine synthesis from ethanolamine and diacylglycerol. Complete deletion of the mouse Pcyt2 gene is embryonic lethal, and the single-allele deficiency leads to development of the metabolic syndrome phenotype, including liver steatosis, hypertriglyceridemia, obesity, and insulin resistance. This study aimed to specifically elucidate the mechanisms of hypertriglyceridemia in Pcyt2 heterozygous mice (Pcyt2(+/-)). Evidence here shows that unlike 8 week-old mice, 32 week- and 42 week-old Pcyt2(+/-) mice experience increased VLDL secretion and liver microsomal triglyceride transfer protein activity. Older Pcyt2(+/-) mice also demonstrate increased levels of postprandial plasma TAGs, increased stimulation of genes responsible for intestinal lipid absorption, transport and chylomicron secretion, and dramatically elevated plasma Angptl4, apoB-100, and apoB-48 content. In addition, plasma HL and LPL activities and TAG clearance following a lipid challenge were significantly reduced in Pcyt2(+/-) mice relative to control littermates. Collectively, these results establish that the hypertriglyceridemia that accompanies Pcyt2 deficiency is the result of multiple metabolic adaptations, including elevated hepatic and intestinal lipoprotein secretion and stimulated expression and/or activity of genes involved in lipid absorption and transport and lipoprotein assembly, together with reduced plasma TAG clearance and utilization with peripheral tissues.


Subject(s)
Hypertriglyceridemia/enzymology , RNA Nucleotidyltransferases/deficiency , Angiopoietin-Like Protein 4 , Angiopoietins/genetics , Animals , Apolipoproteins B/blood , Carrier Proteins/metabolism , Chylomicrons/biosynthesis , Chylomicrons/metabolism , Fatty Acids/metabolism , Gene Expression Regulation, Enzymologic , Homeostasis , Hypertriglyceridemia/blood , Hypertriglyceridemia/metabolism , Intestinal Absorption/genetics , Intestinal Mucosa/metabolism , Lipase/blood , Lipoprotein Lipase/blood , Lipoprotein Lipase/genetics , Liver/metabolism , Mice , Postprandial Period , Triglycerides/blood , Triglycerides/metabolism
20.
Am J Physiol Gastrointest Liver Physiol ; 302(9): G1043-52, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22345552

ABSTRACT

Ezetimibe is a cholesterol uptake inhibitor that targets the Niemann-Pick C1-like 1 cholesterol transporter. Ezetimibe treatment has been shown to cause significant decreases in plasma cholesterol levels in patients with hypercholesterolemia and familial hypercholesterolemia. A recent study in humans has shown that ezetimibe can decrease the release of atherogenic postprandial intestinal lipoproteins. In the present study, we evaluated the mechanisms by which ezetimibe treatment can lower postprandial apoB48-containing chylomicron particles, using a hyperlipidemic and insulin-resistant hamster model fed a diet rich in fructose and fat (the FF diet) and fructose, fat, and cholesterol (the FFC diet). Male Syrian Golden hamsters were fed either chow or the FF or FFC diet ± ezetimibe for 2 wk. After 2 wk, chylomicron production was assessed following intravenous triton infusion. Tissues were then collected and analyzed for protein and mRNA content. FFC-fed hamsters treated with ezetimibe showed improved glucose tolerance, decreased fasting insulin levels, and markedly reduced circulating levels of TG and cholesterol in both the LDL and VLDL fractions. Examination of triglyceride (TG)-rich lipoprotein (TRL) fractions showed that ezetimibe treatment reduced postprandial cholesterol content in TRL lipoproteins as well as reducing apoB48 content. Although ezetimibe did not decrease TRL-TG levels in FFC hamsters, ezetimibe treatment in FF hamsters resulted in decreases in TRL-TG. Jejunal apoB48 protein expression was lower in ezetimibe-treated hamsters. Reductions in jejunal protein levels of scavenger receptor type B-1 (SRB-1) and fatty acid transport protein 4 were also observed. In addition, ezetimibe-treated hamsters showed significantly lower jejunal mRNA expression of a number of genes involved in lipid synthesis and transport, including srebp-1c, sr-b1, ppar-γ, and abcg1. These data suggest that treatment with ezetimibe not only inhibits cholesterol uptake, but may also alter intestinal function to promote improved handling of dietary lipids and reduced chylomicron production. These, in turn, promote decreases in fasting and postprandial lipid levels and improvements in glucose homeostasis.


Subject(s)
Azetidines/administration & dosage , Blood Glucose/metabolism , Chylomicrons/biosynthesis , Disease Models, Animal , Glucose Tolerance Test , Metabolic Syndrome/drug therapy , Metabolic Syndrome/physiopathology , Animals , Anticholesteremic Agents/administration & dosage , Blood Glucose/drug effects , Cricetinae , Diet, High-Fat , Ezetimibe , Male , Mesocricetus , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL