Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
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
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 Lipid Res ; 63(11): 100278, 2022 11.
Article in English | MEDLINE | ID: mdl-36100090

ABSTRACT

The small intestinal epithelium has classically been envisioned as a conduit for nutrient absorption, but appreciation is growing for a larger and more dynamic role for enterocytes in lipid metabolism. Considerable gaps remain in our knowledge of this physiology, but it appears that the enterocyte's structural polarization dictates its behavior in fat partitioning, treating fat differently based on its absorption across the apical versus the basolateral membrane. In this review, we synthesize existing data and thought on this dual-track model of enterocyte fat metabolism through the lens of human integrative physiology. The apical track includes the canonical pathway of dietary lipid absorption across the apical brush-border membrane, leading to packaging and secretion of those lipids as chylomicrons. However, this track also reserves a portion of dietary lipid within cytoplasmic lipid droplets for later uses, including the "second-meal effect," which remains poorly understood. At the same time, the enterocyte takes up circulating fats across the basolateral membrane by mechanisms that may include receptor-mediated import of triglyceride-rich lipoproteins or their remnants, local hydrolysis and internalization of free fatty acids, or enterocyte de novo lipogenesis using basolaterally absorbed substrates. The ultimate destinations of basolateral-track fat may include fatty acid oxidation, structural lipid synthesis, storage in cytoplasmic lipid droplets, or ultimate resecretion, although the regulation and purposes of this basolateral track remain mysterious. We propose that the enterocyte integrates lipid flux along both of these tracks in order to calibrate its overall program of lipid metabolism.


Subject(s)
Chylomicrons , Enterocytes , Humans , Enterocytes/metabolism , Chylomicrons/metabolism , Lipid Metabolism , Dietary Fats/metabolism , Lipid Droplets/metabolism
4.
Nat Commun ; 13(1): 3799, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35778407

ABSTRACT

Atherosclerosis is a chronic inflammatory disease driven by hypercholesterolemia. During aging, T cells accumulate cholesterol, potentially affecting inflammation. However, the effect of cholesterol efflux pathways mediated by ATP-binding cassette A1 and G1 (ABCA1/ABCG1) on T cell-dependent age-related inflammation and atherosclerosis remains poorly understood. In this study, we generate mice with T cell-specific Abca1/Abcg1-deficiency on the low-density-lipoprotein-receptor deficient (Ldlr-/-) background. T cell Abca1/Abcg1-deficiency decreases blood, lymph node, and splenic T cells, and increases T cell activation and apoptosis. T cell Abca1/Abcg1-deficiency induces a premature T cell aging phenotype in middle-aged (12-13 months) Ldlr-/- mice, reflected by upregulation of senescence markers. Despite T cell senescence and enhanced T cell activation, T cell Abca1/Abcg1-deficiency decreases atherosclerosis and aortic inflammation in middle-aged Ldlr-/- mice, accompanied by decreased T cells in atherosclerotic plaques. We attribute these effects to T cell apoptosis downstream of T cell activation, compromising T cell functionality. Collectively, we show that T cell cholesterol efflux pathways suppress T cell apoptosis and senescence, and induce atherosclerosis in middle-aged Ldlr-/- mice.


Subject(s)
Atherosclerosis , T-Lymphocytes , Animals , Apoptosis , Atherosclerosis/genetics , Biological Transport , Immunologic Deficiency Syndromes , Inflammation , Mice , Thymus Gland/abnormalities
5.
J Biol Chem ; 296: 100193, 2021.
Article in English | MEDLINE | ID: mdl-33334888

ABSTRACT

Calcific aortic valve disease (CAVD) occurs when subpopulations of valve cells undergo specific differentiation pathways, promoting tissue fibrosis and calcification. Lipoprotein particles carry oxidized lipids that promote valvular disease, but low-density lipoprotein-lowering therapies have failed in clinical trials, and there are currently no pharmacological interventions available for this disease. Apolipoproteins are known promoters of atherosclerosis, but whether they possess pathogenic properties in CAVD is less clear. To search for a possible link, we assessed 12 apolipoproteins in nonfibrotic/noncalcific and fibrotic/calcific aortic valve tissues by proteomics and immunohistochemistry to understand if they were enriched in calcified areas. Eight apolipoproteins (apoA-I, apoA-II, apoA-IV, apoB, apoC-III, apoD, apoL-I, and apoM) were enriched in the calcific versus nonfibrotic/noncalcific tissues. Apo(a), apoB, apoC-III, apoE, and apoJ localized within the disease-prone fibrosa and colocalized with calcific regions as detected by immunohistochemistry. Circulating apoC-III on lipoprotein(a) is a potential biomarker of aortic stenosis incidence and progression, but whether apoC-III also induces aortic valve calcification is unknown. We found that apoC-III was increased in fibrotic and calcific tissues and observed within the calcification-prone fibrosa layer as well as around calcification. In addition, we showed that apoC-III induced calcification in primary human valvular cell cultures via a mitochondrial dysfunction/inflammation-mediated pathway. This study provides a first assessment of a broad array of apolipoproteins in CAVD tissues, demonstrates that specific apolipoproteins associate with valvular calcification, and implicates apoC-III as an active and modifiable driver of CAVD beyond its potential role as a biomarker.


Subject(s)
Aortic Valve Stenosis/metabolism , Aortic Valve/pathology , Apolipoprotein C-III/metabolism , Calcinosis/metabolism , Aortic Valve/metabolism , Aortic Valve Stenosis/pathology , Apolipoprotein C-III/analysis , Calcinosis/pathology , Cells, Cultured , Humans , Inflammation/metabolism , Inflammation/pathology , Mitochondria/metabolism , Mitochondria/pathology
6.
J Lipid Res ; 60(9): 1503-1515, 2019 09.
Article in English | MEDLINE | ID: mdl-31152000

ABSTRACT

Chylomicron metabolism is critical for determining plasma levels of triacylglycerols (TAGs) and cholesterol, both of which are risk factors for CVD. The rates of chylomicron secretion and remnant clearance are controlled by intracellular and extracellular factors, including apoC-III. We have previously shown that human apoC-III overexpression in mice (apoC-IIITg mice) decreases the rate of chylomicron secretion into lymph, as well as the TAG composition in chylomicrons. We now find that this decrease in chylomicron secretion is not due to the intracellular effects of apoC-III, but instead that primary murine enteroids are capable of taking up TAG from TAG-rich lipoproteins (TRLs) on their basolateral surface; and via Seahorse analyses, we find that mitochondrial respiration is induced by basolateral TRLs. Furthermore, TAG uptake into the enterocyte is inhibited when excess apoC-III is present on TRLs. In vivo, we find that dietary TAG is diverted from the cytosolic lipid droplets and driven toward mitochondrial FA oxidation when plasma apoC-III is high (or when basolateral substrates are absent). We propose that this pathway of basolateral lipid substrate transport (BLST) plays a physiologically relevant role in the maintenance of dietary lipid absorption and chylomicron secretion. Further, when apoC-III is in excess, it inhibits BLST and chylomicron secretion.


Subject(s)
Apolipoprotein C-III/metabolism , Chylomicrons/metabolism , Intestinal Mucosa/metabolism , Triglycerides/metabolism , Animals , Cholesterol/metabolism , Chromatography, Thin Layer , Female , Flow Cytometry , Lipoproteins/metabolism , Male , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission
7.
Methods Mol Biol ; 1576: 195-204, 2019.
Article in English | MEDLINE | ID: mdl-28929461

ABSTRACT

Since the initial report in 2009 by Sato and Clevers, primary enteroids have been of major interest in the fields of stem cell biology and gastrointestinal (GI) tract biology. More recently, we and others have made major inroads into the physiological relevance of these enteroid models and have shown that enteroids derived from the stomach, intestine, or colon recapitulate major functions of these tissues, namely, gastric acid secretion, lipid absorption and lipoprotein secretion, and ion transport. Here, we detail the isolation of stem cells from the small intestine and the culture and propagation of those stem cells into mature three-dimensional enteroids. We will also detail how we use enteroids to determine intestinal mechanisms behind dietary lipid absorption and lipoprotein secretion. The primary enteroid model is a powerful tool that significantly expands our ability to model GI tract function in vitro.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Cell Separation/methods , Dietary Fats/administration & dosage , Intestines/cytology , Lipid Metabolism , Stem Cells/cytology , Animals , Cells, Cultured , Intestines/physiology , Mice , Mice, Inbred C57BL , Stem Cells/metabolism
8.
Diabetologia ; 60(11): 2262-2273, 2017 11.
Article in English | MEDLINE | ID: mdl-28835988

ABSTRACT

AIM/HYPOTHESIS: Here, our aim was to examine whether VLDL and apolipoprotein (apo) CIII induce endoplasmic reticulum (ER) stress, inflammation and insulin resistance in skeletal muscle. METHODS: Studies were conducted in mouse C2C12 myotubes, isolated skeletal muscle and skeletal muscle from transgenic mice overexpressing apoCIII. RESULTS: C2C12 myotubes exposed to VLDL showed increased levels of ER stress and inflammatory markers whereas peroxisome proliferator-activated receptor γ co-activator 1α (PGC-1α) and AMP-activated protein kinase (AMPK) levels were reduced and the insulin signalling pathway was attenuated. The effects of VLDL were also observed in isolated skeletal muscle incubated with VLDL. The changes caused by VLDL were dependent on extracellular signal-regulated kinase (ERK) 1/2 since they were prevented by the ERK1/2 inhibitor U0126 or by knockdown of this kinase by siRNA transfection. ApoCIII mimicked the effects of VLDL and its effects were also blocked by ERK1/2 inhibition, suggesting that this apolipoprotein was responsible for the effects of VLDL. Skeletal muscle from transgenic mice overexpressing apoCIII showed increased levels of some ER stress and inflammatory markers and increased phosphorylated ERK1/2 levels, whereas PGC-1α levels were reduced, confirming apoCIII effects in vivo. Finally, incubation of myotubes with a neutralising antibody against Toll-like receptor 2 abolished the effects of apoCIII on ER stress, inflammation and insulin resistance, indicating that the effects of apoCIII were mediated by this receptor. CONCLUSIONS/INTERPRETATION: These results imply that elevated VLDL in diabetic states can contribute to the exacerbation of insulin resistance by activating ERK1/2 through Toll-like receptor 2.


Subject(s)
Apolipoprotein C-III/pharmacology , Cholesterol, VLDL/pharmacology , Insulin/metabolism , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Toll-Like Receptor 2/metabolism , Animals , Cell Line , Inflammation/drug therapy , Mice , Signal Transduction/drug effects
9.
Biochem Biophys Res Commun ; 491(3): 747-753, 2017 09 23.
Article in English | MEDLINE | ID: mdl-28739253

ABSTRACT

ApoC-III is a critical cardiovascular risk factor, and humans expressing null mutations in apoC-III are robustly protected from cardiovascular disease. Because of its critical role in elevating plasma lipids and CVD risk, hepatic apoC-III regulation has been studied at length. Considerably less is known about the factors that regulate intestinal apoC-III. In this work, we use primary murine enteroids, Caco-2 cells, and dietary studies in wild-type mice to show that intestinal apoC-III expression does not change in response to fatty acids, glucose, or insulin administration, in contrast to hepatic apoC-III. Intestinal apoC-III is not sensitive to changes in FoxO1 expression (which is itself very low in the intestine, as is FoxO1 target IGFBP-1), nor is intestinal apoC-III responsive to western diet, a significant contrast to hepatic apoC-III stimulation during western diet. These data strongly suggest that intestinal apoC-III is not a FoxO1 target and support the idea that apoC-III is not regulated coordinately with hepatic apoC-III, and establishes another key aspect of apoC-III that is unique in the intestine from the liver.


Subject(s)
Apolipoprotein C-III/metabolism , Forkhead Box Protein O1/metabolism , Gene Expression Regulation/physiology , Intestinal Mucosa/metabolism , Lipid Metabolism/physiology , Liver/metabolism , Animals , Caco-2 Cells , Diet, Western , Female , Glucose/metabolism , Humans , In Vitro Techniques , Insulin/metabolism , Male , Mice , Mice, Inbred C57BL , Organ Specificity/physiology
10.
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
11.
Am J Physiol Gastrointest Liver Physiol ; 311(4): G648-G654, 2016 10 01.
Article in English | MEDLINE | ID: mdl-27514481

ABSTRACT

It is not well understood how monosodium glutamate (MSG) affects gastrointestinal physiology, especially regarding the absorption and the subsequent transport of dietary lipids into lymph. Thus far, there is little information about how the ingestion of MSG affects the lipid lipolysis, uptake, intracellular esterification, and formation and secretion of chylomicrons. Using lymph fistula rats treated with the infusion of a 2% MSG solution before a continuous infusion of triglyceride, we show that MSG causes a significant decrease in both triglyceride and cholesterol secretion into lymph. Intriguingly, the diminished lymphatic transport of triglyceride and cholesterol was not caused by an accumulation of these labeled lipids in the intestinal lumen or in the intestinal mucosa. Rather, it is a result of increased portal transport in the animals fed acutely the lipid plus 2% MSG in the lipid emulsion. This is a first demonstration of MSG on intestinal lymphatic transport of lipids.


Subject(s)
Cholesterol/metabolism , Intestinal Absorption/drug effects , Intestinal Mucosa/metabolism , Lipolysis/drug effects , Lymphatic System/drug effects , Sodium Glutamate/pharmacology , Triglycerides/metabolism , Animals , Biological Transport/drug effects , Chylomicrons/metabolism , Intestinal Mucosa/drug effects , Lymphatic System/metabolism , Lymphatic Vessels/drug effects , Lymphatic Vessels/metabolism , Male , Rats , Rats, Sprague-Dawley , Triglycerides/pharmacology
12.
Am J Physiol Gastrointest Liver Physiol ; 310(10): G776-89, 2016 05 15.
Article in English | MEDLINE | ID: mdl-26968208

ABSTRACT

Dietary lipids are transported from the intestine through contractile lymphatics. Chronic lipid loads can adversely affect lymphatic function. However, the acute lymphatic pump response in the mesentery to a postprandial lipid meal has gone unexplored. In this study, we used the rat mesenteric collecting vessel as an in vivo model to quantify the effect of lipoproteins on vessel function. Lipid load was continuously monitored by using the intensity of a fluorescent fatty-acid analog, which we infused along with a fat emulsion through a duodenal cannula. The vessel contractility was simultaneously quantified. We demonstrated for the first time that collecting lymphatic vessels respond to an acute lipid load by reducing pump function. High lipid levels decreased contraction frequency and amplitude. We also showed a strong tonic response through a reduction in the end-diastolic and systolic diameters. We further characterized the changes in flow rate and viscosity and showed that both increase postprandially. In addition, shear-mediated Ca(2+) signaling in lymphatic endothelial cells differed when cultured with lipoproteins. Together these results show that the in vivo response could be both shear and lipid mediated and provide the first evidence that high postprandial lipid has an immediate negative effect on lymphatic function even in the acute setting.


Subject(s)
Dietary Fats/metabolism , Lymphatic Vessels/physiology , Muscle Contraction , Postprandial Period , Animals , Calcium Signaling , Cells, Cultured , Endothelial Cells/metabolism , Humans , Lymph/metabolism , Lymph/physiology , Lymphatic Vessels/cytology , Lymphatic Vessels/metabolism , Male , Muscle, Smooth/physiology , Rats , Rats, Sprague-Dawley , Viscosity
13.
Am J Physiol Gastrointest Liver Physiol ; 309(10): G807-15, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26336929

ABSTRACT

The incretin hormones, glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1), enhance postprandial insulin secretion, promote adipogenesis, and regulate gastrointestinal motility and food intake. To date, a consensus on how the incretin response is altered in obesity is lacking. We investigated the effects of chronic high-fat (HF) feeding on incretin secretion in the lymph fistula rat model. Male Sprague-Dawley rats (8 wk) were provided a semipurified AIN93M HF or low-fat (LF) diet ad libitum for 3 or 13 wk; a HF pair-fed (HF-PF) group was included as a control during the 3-wk feeding trial. Energy intake, body weight, and body composition were regularly monitored. At the culmination of the feeding period, an intestinal lymphatic duct cannula and duodenal infusion tube were installed. All animals were challenged with a 3-ml Ensure bolus (3.125 kcal/animal) to measure lymphatic incretin secretion. Despite a significantly higher energy intake, both the 3-wk and 13-wk HF-fed animals did not have an increase in body weight and only a slight increase in body fat compared with LF-fed rats. Following the duodenal Ensure challenge, the 3-wk and 13-wk HF-fed rats had significantly greater lymphatic GIP and GLP-1 secretion than the LF-fed animals. Additionally, the HF-PF group displayed a secretion profile similar to the HF-fed animals for GIP but a similar pattern to the LF-fed animals for GLP-1. The HF-PF data suggest that the increased GIP secretion is driven by the greater percentage of fat intake, whereas the increased GLP-1 secretion is driven by the excess caloric intake.


Subject(s)
Body Weight , Diet, High-Fat/methods , Gastric Inhibitory Polypeptide/metabolism , Glucagon-Like Peptide 1/metabolism , Incretins/metabolism , Insulin/metabolism , Obesity , Adipogenesis/physiology , Animals , Body Composition , Dietary Fats/metabolism , Disease Models, Animal , Gastrointestinal Motility/physiology , Insulin Secretion , Male , Obesity/metabolism , Obesity/physiopathology , Postprandial Period/physiology , Rats , Rats, Sprague-Dawley
14.
J Lipid Res ; 56(8): 1403-18, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25640749

ABSTRACT

The purpose of this review is to summarize our current understanding of the physiological roles of apoA-IV in metabolism, and to underscore the potential for apoA-IV to be a focus for new therapies aimed at the treatment of diabetes and obesity-related disorders. ApoA-IV is primarily synthesized by the small intestine, attached to chylomicrons by enterocytes, and secreted into intestinal lymph during fat absorption. In circulation, apoA-IV is associated with HDL and chylomicron remnants, but a large portion is lipoprotein free. Due to its anti-oxidative and anti-inflammatory properties, and because it can mediate reverse-cholesterol transport, proposed functions of circulating apoA-IV have been related to protection from cardiovascular disease. This review, however, focuses primarily on several properties of apoA-IV that impact other metabolic functions related to food intake, obesity, and diabetes. In addition to participating in triglyceride absorption, apoA-IV can act as an acute satiation factor through both peripheral and central routes of action. It also modulates glucose homeostasis through incretin-like effects on insulin secretion, and by moderating hepatic glucose production. While apoA-IV receptors remain to be conclusively identified, the latter modes of action suggest that this protein holds therapeutic promise for treating metabolic disease.


Subject(s)
Apolipoproteins A/metabolism , Metabolism , Animals , Bariatric Surgery , Gene Expression Regulation , Humans
15.
Curr Opin Endocrinol Diabetes Obes ; 22(2): 119-25, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25692924

ABSTRACT

PURPOSE OF REVIEW: The purpose of this article is to summarize the recent epidemiological, basic science, and pharmaceutical research linking apolipoprotein C-III (apoC-III) with the development and treatment of cardiovascular disease (CVD). RECENT FINDINGS: ApoC-III is an important emerging target linking hypertriglyceridemia with CVD. ApoC-III is a potent modulator of many established CVD risk factors, and is found on chylomicrons, very-low density lipoprotein, low-density lipoprotein, and high-density lipoprotein particles. Recent studies show that in humans, apoC-III levels are an independent risk factor for CVD, and its presence on lipoproteins may promote their atherogenicity. This year, two large-scale epidemiological studies have linked mutations in apoC-III with increased incidence of CVD and hypertriglyceridemia. ApoC-III raises plasma triglycerides through inhibition of lipoprotein lipase, stimulation of very-low density lipoprotein secretion, and is a novel factor in modulating intestinal triglyceride trafficking. ApoC-III also stimulates inflammatory processes in the vasculature and the pancreas. The combination of raising plasma triglycerides and independently stimulating inflammatory processes makes apoC-III a valuable target for reducing the residual CVD risk in patients already on statin therapy, or for whom triglycerides are poorly controlled. Clinical trials on apoC-III antisense oligonucleotides are in progress. SUMMARY: ApoC-III is a potent direct modulator of established CVD risk factors: plasma triglycerides and inflammation. Recent findings show that changes in apoC-III levels are directly associated with changes in cardiovascular risk and the atherogenicity of the lipoproteins on which apoC-III resides. Emerging roles of apoC-III include a role in directing the atherogenicity of high-density lipoprotein, intestinal dietary triglyceride trafficking, and modulating pancreatic ß-cell survival. The combination of these roles makes apoC-III an important therapeutic target for the management and prevention of CVD.


Subject(s)
Apolipoprotein C-III/blood , Cardiovascular Diseases/blood , Hypertriglyceridemia/blood , Animals , Biomarkers/blood , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/prevention & control , Humans , Hypertriglyceridemia/drug therapy , Hypertriglyceridemia/epidemiology , Hypolipidemic Agents/therapeutic use , Inflammation Mediators/blood , Risk Factors , Signal Transduction , Triglycerides/blood
16.
Am J Physiol Gastrointest Liver Physiol ; 308(6): G472-81, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25591862

ABSTRACT

Apolipoprotein A-IV (apoA-IV) is secreted by the small intestine on chylomicrons into intestinal lymph in response to fat absorption. Many physiological functions have been ascribed to apoA-IV, including a role in chylomicron assembly and lipid metabolism, a mediator of reverse-cholesterol transport, an acute satiety factor, a regulator of gastric function, and, finally, a modulator of blood glucose homeostasis. The purpose of this review is to update our current view of intestinal apoA-IV synthesis and secretion and the physiological roles of apoA-IV in lipid metabolism and energy homeostasis, and to underscore the potential for intestinal apoA-IV to serve as a therapeutic target for the treatment of diabetes and obesity-related disease.


Subject(s)
Apolipoproteins A/metabolism , Brain/metabolism , Energy Metabolism , Glucose/metabolism , Intestine, Small/metabolism , Lipid Metabolism , Satiety Response , Animals , Chylomicrons/metabolism , Eating , Feeding Behavior , Homeostasis , Humans , Intestinal Absorption , Signal Transduction
17.
Physiol Rep ; 2(3): e00247, 2014.
Article in English | MEDLINE | ID: mdl-24760506

ABSTRACT

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

18.
Regul Pept ; 189: 40-5, 2014 Feb 10.
Article in English | MEDLINE | ID: mdl-24583245

ABSTRACT

Using a newly developed in vivo model measuring glucagon-like peptide-1 (GLP-1) in gut lymphatics in mice, we quantified GLP-1 secretion in vivo after glucose versus fat ingestion with and without concomitant DPP-4 inhibition. The mesenteric lymphatic duct was cannulated in anesthetized C57BL6/J mice and lymph was collected in 30 min intervals. Glucose or fat emulsion (Intralipid®) (0.03, 0.1 or 0.3 kcal) with or without DPP-4-inhibition (NVP DPP728; 10 µmol/kg) was administered by gastric gavage. Basal intact GLP-1 levels were 0.37±0.04 pmol/l (n=61) in lymph compared to 0.07±0.03 in plasma (n=6; P=0.04) and basal DPP-4 activity was 4.7±0.3 pmol/min/µl in lymph (n=23) compared to 22.3±0.9 pmol/min/µl in plasma (n=8; P<0.001). Lymph flow increased from 1.2±0.1 µl/min to 2.3±02µl/min at 30 min after glucose and fat administration, with no difference between type of challenge or dose (n=81). Lymph GLP-1 levels increased calorie-dependently after both glucose and fat but with different time courses in that glucose induced a transient increase which had returned to baseline after 90 min whereas the lipid induced a sustained increase which was still elevated above baseline after 210 min. Lymph GLP-1 appearance during 210 min was two to three-fold higher after glucose (7.4±2.3 fmol at 0.3 kcal) than after isocaloric fat (2.9±0.8 fmol at 0.3 kcal; P<0.001). The slope between caloric load and lymph GLP-1 appearance was, however, identical after glucose and fat. We conclude that lymph GLP-1 is higher than plasma GLP-1 whereas lymph DPP-4 activity is lower than plasma DPP-4 activity and that both glucose and fat clearly stimulate GLP-1 secretion calorie-dependently in vivo but with different time courses.


Subject(s)
Glucagon-Like Peptide 1/blood , Glucagon-Like Peptide 1/metabolism , Glucose/metabolism , Animals , Dipeptidyl Peptidase 4/metabolism , Female , Lymph/metabolism , Mice , Mice, Inbred C57BL
19.
J Biol Chem ; 289(4): 2396-404, 2014 Jan 24.
Article in English | MEDLINE | ID: mdl-24311788

ABSTRACT

We showed recently that apoA-IV improves glucose homeostasis by enhancing pancreatic insulin secretion in the presence of elevated levels of glucose. Therefore, examined whether apolipoprotein A-IV (apoA-IV) also regulates glucose metabolism through the suppression of hepatic gluconeogenesis. The ability of apoA-IV to lower gluconeogenic gene expression and glucose production was measured in apoA-IV(-/-) and wild-type mice and primary mouse hepatocytes. The transcriptional regulation of Glc-6-Pase and phosphoenolpyruvate carboxykinase (PEPCK) by apoA-IV was determined by luciferase activity assay. Using bacterial two-hybrid library screening, NR1D1 was identified as a putative apoA-IV-binding protein. The colocalization and interaction between apoA-IV and NR1D1 were confirmed by immunofluorescence, in situ proximity ligation assay, and coimmunoprecipitation. Enhanced recruitment of NR1D1 and activity by apoA-IV to Glc-6-Pase promoter was verified with ChIP and a luciferase assay. Down-regulation of apoA-IV on gluconeogenic genes is mediated through NR1D1, as illustrated in cells with NR1D1 knockdown by siRNA. We found that apoA-IV suppresses the expression of PEPCK and Glc-6-Pase in hepatocytes; decreases hepatic glucose production; binds and activates nuclear receptor NR1D1 and stimulates NR1D1 expression; in cells lacking NR1D1, fails to inhibit PEPCK and Glc-6-Pase gene expression; and stimulates higher hepatic glucose production and higher gluconeogenic gene expression in apoA-IV(-/-) mice. We conclude that apoA-IV inhibits hepatic gluconeogenesis by decreasing Glc-6-Pase and PEPCK gene expression through NR1D1. This novel regulatory pathway connects an influx of energy as fat from the gut (and subsequent apoA-IV secretion) with inhibition of hepatic glucose production.


Subject(s)
Apolipoproteins A/metabolism , Gluconeogenesis/physiology , Glucose/biosynthesis , Hepatocytes/metabolism , Liver/metabolism , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Animals , Apolipoproteins A/genetics , Gene Expression Regulation, Enzymologic/physiology , Glucose/genetics , Glucose-6-Phosphatase/biosynthesis , Glucose-6-Phosphatase/genetics , HEK293 Cells , Hep G2 Cells , Hepatocytes/cytology , Humans , Liver/cytology , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group D, Member 1/genetics , Phosphoenolpyruvate Carboxykinase (GTP)/biosynthesis , Phosphoenolpyruvate Carboxykinase (GTP)/genetics , Promoter Regions, Genetic/physiology
20.
Am J Physiol Gastrointest Liver Physiol ; 304(12): G1128-35, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23599044

ABSTRACT

Apolipoprotein A-IV (apoA-IV) is synthesized by the intestine and secreted when dietary fat is absorbed and transported into lymph associated with chylomicrons. We have recently demonstrated that loss of apoA-IV increases chylomicron size and delays its clearance from the blood. There is still uncertainty, however, about the precise role of apoA-IV on the transport of dietary fat from the intestine into the lymph. ApoA-IV knockout (KO) mice do not have a gross defect in dietary lipid absorption, as measured by oral fat tolerance and fecal fat measurements. Here, using the in vivo lymph fistula mouse model, we show that the cumulative secretion of triglyceride (TG) into lymph in apoA-IV KO mice is very similar to that of wild-type (WT) mice. However, the apoA-IV KO mice do have subtle changes in TG accumulation in the intestinal mucosa during a 6-h continuous, but not bolus, infusion of lipid. There are no changes in the ratio of esterified to free fatty acids in the intestinal mucosa of the apoA-IV KO, however. When we extended these findings, by giving a higher dose of lipid (6 µmol/h) and for a longer infusion period (8 h), we found no effect of apoA-IV KO on intestinal TG absorption. This higher lipid infusion most certainly stresses the intestine, as we see a drastically lower absorption of TG (in both WT and KO mice); however, the loss of A-IV does not exacerbate this effect. This supports our hypothesis that apoA-IV is not required for TG absorption in the intestine. Our data suggest that the mechanisms by which the apoA-IV KO intestine responds to intestinal lipid may not be different from their WT counterparts. We conclude that apoA-IV is not required for normal lymphatic transport of TG.


Subject(s)
Apolipoproteins A/metabolism , Intestinal Absorption , Triglycerides/metabolism , Animals , Apolipoproteins A/genetics , Dietary Fats/metabolism , Fatty Acids, Nonesterified/metabolism , Fistula , Infusions, Parenteral , Intestinal Mucosa/metabolism , Kinetics , Lipids/administration & dosage , Lymph/chemistry , Lymph/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Triglycerides/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...