Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
JCI Insight ; 7(19)2022 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-36040803

RESUMEN

BackgroundApolipoprotein C-III (apoC-III) is a regulator of triglyceride (TG) metabolism, and due to its association with risk of cardiovascular disease, is an emergent target for pharmacological intervention. The impact of substantially lowering apoC-III on lipoprotein metabolism is not clear.MethodsWe investigated the kinetics of apolipoproteins B48 and B100 (apoB48 and apoB100) in chylomicrons, VLDL1, VLDL2, IDL, and LDL in patients heterozygous for a loss-of-function (LOF) mutation in the APOC3 gene. Studies were conducted in the postprandial state to provide a more comprehensive view of the influence of this protein on TG transport.ResultsCompared with non-LOF variant participants, a genetically determined decrease in apoC-III resulted in marked acceleration of lipolysis of TG-rich lipoproteins (TRLs), increased removal of VLDL remnants from the bloodstream, and substantial decrease in circulating levels of VLDL1, VLDL2, and IDL particles. Production rates for apoB48-containing chylomicrons and apoB100-containing VLDL1 and VLDL2 were not different between LOF carriers and noncarriers. Likewise, the rate of production of LDL was not affected by the lower apoC-III level, nor were the concentration and clearance rate of LDL-apoB100.ConclusionThese findings indicate that apoC-III lowering will have a marked effect on TRL and remnant metabolism, with possibly significant consequences for cardiovascular disease prevention.Trial registrationClinicalTrials.gov NCT04209816 and NCT01445730.FundingSwedish Heart-Lung Foundation, Swedish Research Council, ALF grant from the Sahlgrenska University Hospital, Novo Nordisk Foundation, Sigrid Juselius Foundation, Helsinki University Hospital Government Research funds, Finnish Heart Foundation, and Finnish Diabetes Research Foundation.


Asunto(s)
Enfermedades Cardiovasculares , Lipoproteínas VLDL , Apolipoproteína B-48/genética , Apolipoproteína B-48/metabolismo , Apolipoproteína C-III/genética , Apolipoproteína C-III/metabolismo , Enfermedades Cardiovasculares/genética , Proteínas Portadoras/genética , Quilomicrones/genética , Quilomicrones/metabolismo , Humanos , Lipoproteínas/metabolismo , Lipoproteínas VLDL/metabolismo , Mutación , Triglicéridos/metabolismo
2.
J Clin Invest ; 131(12)2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34128469

RESUMEN

Although tissue uptake of fatty acids from chylomicrons is primarily via lipoprotein lipase (LpL) hydrolysis of triglycerides (TGs), studies of patients with genetic LpL deficiency suggest additional pathways deliver dietary lipids to tissues. Despite an intact endothelial cell (EC) barrier, hyperchylomicronemic patients accumulate chylomicron-derived lipids within skin macrophages, leading to the clinical finding eruptive xanthomas. We explored whether an LpL-independent pathway exists for transfer of circulating lipids across the EC barrier. We found that LpL-deficient mice had a marked increase in aortic EC lipid droplets before and after a fat gavage. Cultured ECs internalized chylomicrons, which were hydrolyzed within lysosomes. The products of this hydrolysis fueled lipid droplet biogenesis in ECs and triggered lipid accumulation in cocultured macrophages. EC chylomicron uptake was inhibited by competition with HDL and knockdown of the scavenger receptor-BI (SR-BI). In vivo, SR-BI knockdown reduced TG accumulation in aortic ECs and skin macrophages of LpL-deficient mice. Thus, ECs internalize chylomicrons, metabolize them in lysosomes, and either store or release their lipids. This latter process may allow accumulation of TGs within skin macrophages and illustrates a pathway that might be responsible for creation of eruptive xanthomas.


Asunto(s)
Aorta/metabolismo , Quilomicrones/metabolismo , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Gotas Lipídicas/metabolismo , Triglicéridos/metabolismo , Xantomatosis/metabolismo , Animales , Aorta/patología , Quilomicrones/genética , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Células Endoteliales de la Vena Umbilical Humana/patología , Humanos , Gotas Lipídicas/patología , Lipoproteína Lipasa/deficiencia , Lipoproteína Lipasa/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Triglicéridos/genética , Xantomatosis/genética , Xantomatosis/patología
3.
Hepatology ; 70(4): 1168-1184, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31004524

RESUMEN

The mechanisms by which alterations in intestinal bile acid (BA) metabolism improve systemic glucose tolerance and hepatic metabolic homeostasis are incompletely understood. We examined metabolic adaptations in mice with conditional intestinal deletion of the abetalipoproteinemia (ABL) gene microsomal triglyceride transfer protein (Mttp-IKO), which blocks chylomicron assembly and impairs intestinal lipid transport. Mttp-IKO mice exhibit improved hepatic glucose metabolism and augmented insulin signaling, without weight loss. These adaptations included decreased BA excretion, increased pool size, altered BA composition, and increased fibroblast growth factor 15 production. Mttp-IKO mice absorb fructose normally but are protected against dietary fructose-induced hepatic steatosis, without weight loss or changes in energy expenditure. In addition, Mttp-IKO mice exhibit altered cecal microbial communities, both at baseline and following fructose feeding, including increased abundance of Bacteroides and Lactobacillus genera. Transplantation of cecal microbiota from chow-fed Mttp-IKO mice into antibiotic-treated wild-type recipients conferred transmissible protection against fructose-induced hepatic steatosis in association with a bloom in Akkermansia and increased Clostridium XIVa genera, whose abundance was positively correlated with fecal coprostanol and total neutral sterol excretion in recipient mice. However, antibiotic-treated Mttp-IKO mice were still protected against fructose-induced hepatic steatosis, suggesting that changes in microbiota are not required for this phenotype. Nevertheless, we found increased abundance of fecal Akkermansia from two adult ABL subjects with MTTP mutations compared to their heterozygous parents and within the range noted in six healthy control subjects. Furthermore, Akkermansia abundance across all subjects was positively correlated with fecal coprostanol excretion. Conclusion: The findings collectively suggest multiple adaptive pathways of metabolic regulation following blocked chylomicron assembly, including shifts in BA signaling and altered microbial composition that confer a transmissible phenotype.


Asunto(s)
Adaptación Fisiológica/genética , Quilomicrones/genética , Hígado Graso/metabolismo , Microbioma Gastrointestinal/genética , Metabolismo de los Lípidos/genética , Akkermansia , Animales , Ácidos y Sales Biliares/metabolismo , Transporte Biológico/genética , Proteínas Portadoras/metabolismo , Modelos Animales de Enfermedad , Hígado Graso/patología , Fructosa/farmacología , Prueba de Tolerancia a la Glucosa , Humanos , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Distribución Aleatoria , Sensibilidad y Especificidad , Transducción de Señal , Verrucomicrobia/patogenicidad
4.
J Clin Invest ; 129(1): 281-295, 2019 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-30507612

RESUMEN

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.


Asunto(s)
Quilomicrones/biosíntesis , Enterocitos/metabolismo , Homeostasis , Fosfatidato Fosfatasa/metabolismo , Fosfolípidos/metabolismo , Animales , Apolipoproteína B-48/genética , Apolipoproteína B-48/metabolismo , Quilomicrones/genética , Enterocitos/citología , Femenino , Gotas Lipídicas/metabolismo , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Noqueados , Fosfatidato Fosfatasa/genética , Fosfolípidos/genética , Triglicéridos/biosíntesis , Triglicéridos/genética
5.
Am J Physiol Gastrointest Liver Physiol ; 315(2): G177-G184, 2018 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-29698057

RESUMEN

The mechanisms underlying the oversecretion of apolipoprotein (apo)B-48-containing triglyceride-rich lipoproteins (TRL) in insulin-resistance (IR) states in humans remain to be fully understood. The objective of this study was to evaluate the association between the plasma levels of insulin and glucose and the intestinal expression of key genes involved in chylomicron metabolism in a large sample of nondiabetic men displaying various degrees of IR. Duodenal biopsies were obtained by gastroduodenoscopy in 127 men free of intestinal disease. Gene expression was measured using quantitative PCR in duodenal samples. Plasma insulin and glucose concentrations were measured in the fasting state. Postprandial TRL apoB-48 kinetics were measured using a primed-constant infusion of l-[5,5,5-D3]leucine for 12 h in a subgroup of 75 subjects maintained in a constant fed state. Plasma insulin levels were negatively associated with intestinal expression of ACS1 (standard ß = -0.20, P = 0.007), DGAT1 (ß = -0.18, P = 0.001), DGAT2 (ß = -0.20, P = 0.02), and MTP (ß = -0.27, P = 0.0005), whereas glucose levels were positively associated with MTP expression (ß = 0.15, P = 0.04) independent of age, BMI, waist circumference, dietary intake, and duodenal expression of SREBP1c. Insulin levels, but not glucose concentrations, were positively correlated with postprandial TRL apoB-48 production rate ( r = 0.24, P = 0.04) and pool size ( r = 0.27, P = 0.03). In conclusion, plasma insulin and glucose levels are differentially associated with the expression of key genes involved in chylomicron metabolism. These results suggest that alterations in intestinal lipoprotein metabolism associated with IR may be regulated by plasma levels of both insulin and glucose concurrently and are therefore likely modified by the onset of insulin insufficiency. NEW & NOTEWORTHY We demonstrate that plasma insulin and glucose levels are differentially associated with the expression of key genes involved in chylomicron metabolism in men. For instance, intestinal expression of MTP is negatively associated with plasma insulin concentrations and positively associated with plasma glucose concentrations. Alterations in intestinal lipoprotein metabolism associated with insulin resistance may be regulated by plasma levels of both insulin and glucose concurrently and are therefore likely modified by the onset of insulin insufficiency.


Asunto(s)
Glucemia/metabolismo , Quilomicrones , Expresión Génica/fisiología , Resistencia a la Insulina/genética , Insulina/sangre , Lipoproteínas/metabolismo , Adulto , Apolipoproteína B-48/genética , Proteínas Portadoras/genética , Quilomicrones/genética , Quilomicrones/metabolismo , Diacilglicerol O-Acetiltransferasa/genética , Duodeno/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Masculino , N-Acetilglucosaminiltransferasas/genética , Periodo Posprandial/fisiología
6.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(6): 600-614, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28249764

RESUMEN

Enterocytes, the absorptive cells of the small intestine, mediate efficient absorption of dietary fat (triacylglycerol, TAG). The digestive products of dietary fat are taken up by enterocytes, re-esterified into TAG, and packaged on chylomicrons (CMs) for secretion into blood or temporarily stored within cytoplasmic lipid droplets (CLDs). Altered enterocyte TAG distribution impacts susceptibility to high fat diet associated diseases, but molecular mechanisms directing TAG toward these fates are unclear. Two enzymes, acyl CoA: diacylglycerol acyltransferase 1 (Dgat1) and Dgat2, catalyze the final, committed step of TAG synthesis within enterocytes. Mice with intestine-specific overexpression of Dgat1 (Dgat1Int) or Dgat2 (Dgat2Int), or lack of Dgat1 (Dgat1-/-), were previously found to have altered intestinal TAG secretion and storage. We hypothesized that varying intestinal Dgat1 and Dgat2 levels alters TAG distribution in subcellular pools for CM synthesis as well as the morphology and proteome of CLDs. To test this we used ultrastructural and proteomic methods to investigate intracellular TAG distribution and CLD-associated proteins in enterocytes from Dgat1Int, Dgat2Int, and Dgat1-/- mice 2h after a 200µl oral olive oil gavage. We found that varying levels of intestinal Dgat1 and Dgat2 altered TAG pools involved in CM assembly and secretion, the number or size of CLDs present in enterocytes, and the enterocyte CLD proteome. Overall, these results support a model where Dgat1 and Dgat2 function coordinately to regulate the process of dietary fat absorption by preferentially synthesizing TAG for incorporation into distinct subcellular TAG pools in enterocytes.


Asunto(s)
Diacilglicerol O-Acetiltransferasa/metabolismo , Grasas de la Dieta/farmacología , Enterocitos/metabolismo , Gotas Lipídicas/metabolismo , Triglicéridos/metabolismo , Animales , Quilomicrones/genética , Quilomicrones/metabolismo , Diacilglicerol O-Acetiltransferasa/genética , Enterocitos/citología , Ratones , Ratones Noqueados , Triglicéridos/genética
8.
Klin Lab Diagn ; 61(6): 324-34, 2016 Jun.
Artículo en Ruso | MEDLINE | ID: mdl-30601622

RESUMEN

According phylogenetic theory of general pathology, necessity in supply of cells in vivo with exogenous fatty acids-substrates for gaining energy and physical-chemical parameters of fatty acids resulted in development (initially in single paracrine regulated cenosis of cells) of system of fatty acids transfer through hydrophilic inter-cellular medium in form of triglycerides in composition of hydrophobic chylomicrons -from enterocytes where they were developed to cells into which they were deposited. It is assumed that system of transfer of hydrophobic chylomicrons through aqueous inter-cellular medium was developed by anatomic combination of tubules of endoplasmic reticulum of enterocytes and the same tubules of cells of areolar tissue in paracrine regulated cenosis of cells of enterocytes with development of the most early in phylogenesis system of directed transfer, lymph flow. This occurrence became a prototype of lymphatic system; its first biological predestination is transferring of fatty acids to all cells escaping hydrophilic inter-cellular medium. With time, the lymphatic system all paracrin regulated cenosis of cells in vivo united in single system and became the foundation of development of organs and systems. The unification of epithelial and connective tissue cells of areolar tissue in single structure turned out to become the basis that at later stages of phylogenesis the lymphatic system began, under expressed convolution of its structure (development of lymphatic nodes), to combine: a) implementation ofprimary biological function of trophology (function of nutrition) with biological function of adaptation; b) later biological reactions of inherited and acquired immune defense at stages of phylogenesis and, probably, c) participation in hydrodynamics of distal sections of closed systemic blood circulation. The mechanisms of osmotic "pushing" of lymph have a lot of common with initiation of stream of cerebrospinal fluid of central nervous system and flow of primary urine in tubules of nephron.


Asunto(s)
Quilomicrones/genética , Ácidos Grasos/genética , Metabolismo de los Lípidos/genética , Triglicéridos/genética , Animales , Quilomicrones/metabolismo , Retículo Endoplásmico/metabolismo , Ácidos Grasos/metabolismo , Humanos , Leptina/genética , Leptina/metabolismo , Vasos Linfáticos/metabolismo , Vasos Linfáticos/patología , Comunicación Paracrina/genética , Triglicéridos/metabolismo
9.
Am J Physiol Gastrointest Liver Physiol ; 309(5): G350-9, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-26138463

RESUMEN

The small intestine contributes to diabetic dyslipidemia through the overproduction of apolipoprotein B48 (apoB48)-containing chylomicron particles. An important regulator of chylomicron generation is dietary lipid absorption, underlining the potential involvement of intestinal lipid transporters for developing dyslipidemia. Intestinal expression of scavenger receptor class B type I (SR-BI) has been found to be upregulated in animal models of insulin resistance. Here we characterized the potential importance of SR-BI in contributing to chylomicron production and postprandial hypertriglyceridemia in vivo. Postprandial triglyceride (TG)-rich lipoprotein (TRL) production was characterized in hamsters treated with the SR-BI inhibitor to block lipid transport-1 (BLT-1) under healthy conditions or conditions of diet-induced obesity and dyslipidemia. BLT-1 (1 mg/kg) or vehicle was administered acutely in chow-fed hamsters or gavaged twice daily over 10 days during high-fructose, high-fat, high-cholesterol (FFC) feeding. Effects of acute SR-BI inhibition by BLT-1 were confirmed in healthy fat-loaded rats. Finally, plasma lipid levels were compared between SR-BI(-/-) mice and their wild-type counterparts fed either chow or a 12-wk high-fat diet. Acute BLT-1 treatment reduced postprandial plasma and TRL TG levels in healthy hamsters and rats. Chronic BLT-1 treatment of FFC-fed hamsters blunted diet-induced weight gain and fasting hypertriglyceridemia, and lowered postprandial TRL-TG, -cholesterol, and -apoB48 levels. Finally, SR-BI(-/-) mice displayed lower plasma and TRL TG levels relative to wild type, and diet-induced weight gain and postprandial hypertriglyceridemia were hindered in SR-BI(-/-) mice. We conclude that intestinal SR-BI is a critical regulator of postprandial lipoprotein production, emphasizing its potential as a target for preventing diabetic dyslipidemia.


Asunto(s)
Quilomicrones/metabolismo , Intestino Delgado/metabolismo , Obesidad/metabolismo , Receptores Depuradores de Clase B/metabolismo , Animales , Quilomicrones/genética , Cricetinae , Dieta Alta en Grasa/efectos adversos , Dislipidemias/etiología , Dislipidemias/metabolismo , Masculino , Mesocricetus , Ratones , Ratones Endogámicos C57BL , Obesidad/etiología , Ratas , Ratas Sprague-Dawley , Receptores Depuradores de Clase B/genética , Triglicéridos/sangre , Triglicéridos/metabolismo
10.
Annu Rev Nutr ; 35: 265-94, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25974693

RESUMEN

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.


Asunto(s)
Quilomicrones/biosíntesis , Homeostasis/fisiología , Aterosclerosis/sangre , Colesterol en la Dieta/metabolismo , Colesterol en la Dieta/farmacología , Quilomicrones/sangre , Quilomicrones/genética , Ritmo Circadiano , Diabetes Mellitus Tipo 2/sangre , Dieta , Grasas de la Dieta/metabolismo , Grasas de la Dieta/farmacocinética , Suplementos Dietéticos , Microbioma Gastrointestinal/fisiología , Hormonas/fisiología , Humanos , Resistencia a la Insulina , Absorción Intestinal , Mucosa Intestinal/metabolismo , Metabolismo de los Lípidos/fisiología , Fenómenos Fisiológicos de la Nutrición , Triglicéridos/biosíntesis , Triglicéridos/sangre , Triglicéridos/genética
11.
J Lipid Res ; 56(3): 588-598, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25589507

RESUMEN

To determine the role of LPL for binding of lipoproteins to the vascular endothelium, and for the distribution of lipids from lipoproteins, four lines of induced mutant mice were used. Rat chylomicrons labeled in vivo with [(14)C]oleic acid (primarily in TGs, providing a tracer for lipolysis) and [(3)H]retinol (primarily in ester form, providing a tracer for the core lipids) were injected. TG label was cleared more rapidly than core label. There were no differences between the mouse lines in the rate at which core label was cleared. Two minutes after injection, about 5% of the core label, and hence chylomicron particles, were in the heart of WT mice. In mice that expressed LPL only in skeletal muscle, and had much reduced levels of LPL in the heart, binding of chylomicrons was reduced to 1%, whereas in mice that expressed LPL only in the heart, the binding was increased to over 10%. The same patterns of distribution were evident at 20 min when most of the label had been cleared. Thus, the amount of LPL expressed in muscle and heart governed both the binding of chylomicron particles and the assimilation of chylomicron lipids in the tissue.


Asunto(s)
Quilomicrones/metabolismo , Lipoproteína Lipasa/metabolismo , Músculo Esquelético/metabolismo , Miocardio/metabolismo , Animales , Quilomicrones/genética , Humanos , Lipoproteína Lipasa/genética , Ratones , Ratones Transgénicos , Especificidad de Órganos/genética , Ratas
12.
J Lipid Res ; 56(3): 489-501, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25231105

RESUMEN

The intestine plays a prominent role in the biosynthesis of triacylglycerol (triglyceride; TAG). Digested dietary TAG is repackaged in the intestine to form the hydrophobic core of chylomicrons, which deliver metabolic fuels, essential fatty acids, and other lipid-soluble nutrients to the peripheral tissues. By controlling the flux of dietary fat into the circulation, intestinal TAG synthesis can greatly impact systemic metabolism. Genes encoding many of the enzymes involved in TAG synthesis have been identified. Among TAG synthesis enzymes, acyl-CoA:monoacylglycerol acyltransferase 2 and acyl-CoA:diacylglycerol acyltransferase (DGAT)1 are highly expressed in the intestine. Their physiological functions have been examined in the context of whole organisms using genetically engineered mice and, in the case of DGAT1, specific inhibitors. An emerging theme from recent findings is that limiting the rate of TAG synthesis in the intestine can modulate gut hormone secretion, lipid metabolism, and systemic energy balance. The underlying mechanisms and their implications for humans are yet to be explored. Pharmacological inhibition of TAG hydrolysis in the intestinal lumen has been employed to combat obesity and associated disorders with modest efficacy and unwanted side effects. The therapeutic potential of inhibiting specific enzymes involved in intestinal TAG synthesis warrants further investigation.


Asunto(s)
Metabolismo Energético/fisiología , Absorción Intestinal/fisiología , Mucosa Intestinal/metabolismo , Metabolismo de los Lípidos/fisiología , Triglicéridos/biosíntesis , Animales , Quilomicrones/genética , Quilomicrones/metabolismo , Diacilglicerol O-Acetiltransferasa/biosíntesis , Diacilglicerol O-Acetiltransferasa/genética , Regulación Enzimológica de la Expresión Génica/fisiología , Humanos , Ratones , Triglicéridos/genética
13.
J Lipid Res ; 55(7): 1279-87, 2014 07.
Artículo en Inglés | MEDLINE | ID: mdl-24831470

RESUMEN

Cell death-inducing DFF45-like effector b (Cideb), an endoplasmic reticulum (ER)- and lipid droplet (LD)-associated protein, has been shown to play a critical role in maintaining hepatic lipid homeostasis by promoting the lipidation and maturation of VLDL particles. Here, we observed that Cideb is expressed in the jejunum and ileum sections of the small intestine, and its expression was induced by high-fat diet. Intragastric gavage with lipids resulted in the retention of lipids in the intestine in Cideb-deficient mice. In addition, we observed that mice with Cideb deficiency exhibited reduced intestinal chylomicron-TG secretion and increased lipid accumulation in the enterocytes. The sizes of chylomicrons secreted from the small intestine of Cideb-deficient mice were also smaller than those from wild-type mice. Furthermore, the overexpression of Cideb increased TG secretion and reduced lipid accumulation in the enterocyte-like Caco-2 cells. In addition, we proved that Cideb was localized to the ER and LDs and could interact with ApoB48 in Caco-2 cells. Overall, these data revealed that Cideb plays an important role in controlling intestinal chylomicron lipidation.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Quilomicrones/metabolismo , Intestino Delgado/metabolismo , Lipoilación/fisiología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Células CACO-2 , Quilomicrones/genética , Humanos , Ratones , Ratones Mutantes
14.
J Biol Chem ; 287(13): 10178-10188, 2012 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-22303004

RESUMEN

Native cytosol requires ATP to initiate the budding of the pre-chylomicron transport vesicle from intestinal endoplasmic reticulum (ER). When FABP1 alone is used, no ATP is needed. Here, we test the hypothesis that in native cytosol FABP1 is present in a multiprotein complex that prevents FABP1 binding to the ER unless the complex is phosphorylated. We found on chromatography of native intestinal cytosol over a Sephacryl S-100 HR column that FABP1 (14 kDa) eluted in a volume suggesting a 75-kDa protein complex that contained four proteins on an anti-FABP1 antibody pulldown. The FABP1-containing column fractions were chromatographed over an anti-FABP1 antibody adsorption column. Proteins co-eluted from the column were identified as FABP1, Sar1b, Sec13, and small VCP/p97-interactive protein by immunoblot, LC-MS/MS, and MALDI-TOF. The four proteins of the complex had a total mass of 77 kDa and migrated on native PAGE at 75 kDa. When the complex was incubated with intestinal ER, there was no increase in FABP1-ER binding. However, when the complex member Sar1b was phosphorylated by PKCζ and ATP, the complex completely disassembled into its component proteins that migrated at their monomer molecular weight on native PAGE. FABP1, freed from the complex, was now able to bind to intestinal ER and generate the pre-chylomicron transport vesicle (PCTV). No increase in ER binding or PCTV generation was observed in the absence of PKCζ or ATP. We conclude that phosphorylation of Sar1b disrupts the FABP1-containing four-membered 75-kDa protein complex in cytosol enabling it to bind to the ER and generate PCTV.


Asunto(s)
Quilomicrones/metabolismo , Retículo Endoplásmico/metabolismo , Proteínas de Unión a Ácidos Grasos/metabolismo , Proteínas de Unión al GTP Monoméricas/metabolismo , Precursores de Proteínas/metabolismo , Adenosina Trifosfato/genética , Adenosina Trifosfato/metabolismo , Animales , Transporte Biológico Activo/fisiología , Quilomicrones/genética , Citosol/metabolismo , Retículo Endoplásmico/genética , Proteínas de Unión a Ácidos Grasos/genética , Mucosa Intestinal/metabolismo , Masculino , Proteínas de Unión al GTP Monoméricas/genética , Fosforilación , Unión Proteica , Proteína Quinasa C-epsilon/genética , Proteína Quinasa C-epsilon/metabolismo , Precursores de Proteínas/genética , Ratas , Ratas Sprague-Dawley , Vesículas Secretoras
15.
Biochim Biophys Acta ; 1818(3): 530-6, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22119776

RESUMEN

Transintestinal cholesterol efflux (TICE) has been proposed to represent a non-hepatobiliary route of cholesterol secretion directly "from blood to gut" and to play a physiologically significant role in excretion of neutral sterols, but so far little is known about the proteins involved in the process. We have previously observed that apolipoprotein A-1 (apoA-1) synthesized by enterocytes of the small intestine is mainly secreted apically into the gut lumen during fasting where its assembly into chylomicrons and basolateral discharge is at a minimal level. In the present work we showed, both by immunomicroscopy and subcellular fractionation, that a fraction of the apically secreted apoA-1 in porcine small intestine was not released from the cell surface but instead deposited in the brush border. Cholesterol was detected in immunoisolated microvillar apoA-1, and it was partially associated with detergent resistant membranes (DRMs), indicative of localization in lipid raft microdomains. The apolipoprotein was not readily released from microvillar vesicles by high salt or by incubation with phosphatidylcholine-specific phospholipase C or trypsin, indicating a relatively firm attachment to the membrane bilayer. However, whole bile or taurocholate efficiently released apoA-1 at low concentrations that did not solubilize the transmembrane microvillar protein aminopeptidase N. Based on these findings and the well known role played by apoA-1 in extrahepatic cellular cholesterol removal and reverse cholesterol transport (RCT), we propose that brush border-deposited apoA-1 in the small intestine acts in TICE by mediating cholesterol efflux into the gut lumen.


Asunto(s)
Apolipoproteína A-I/metabolismo , Membrana Celular/metabolismo , Colesterol/metabolismo , Enterocitos/metabolismo , Intestino Delgado/metabolismo , Animales , Apolipoproteína A-I/genética , Transporte Biológico Activo/fisiología , Antígenos CD13/química , Antígenos CD13/genética , Antígenos CD13/metabolismo , Colesterol/genética , Quilomicrones/genética , Quilomicrones/metabolismo , Enterocitos/citología , Humanos , Microvellosidades/genética , Microvellosidades/metabolismo , Porcinos , Fosfolipasas de Tipo C/química , Fosfolipasas de Tipo C/genética , Fosfolipasas de Tipo C/metabolismo
16.
J Clin Endocrinol Metab ; 96(10): E1675-9, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21816778

RESUMEN

CONTEXT: GPIHBP1 is a new endothelial binding site for lipoprotein lipase (LPL), the key enzyme for intravascular lipolysis of triglyceride-rich lipoproteins (TGRL). We have identified two new missense mutations of the GPIHBP1 gene, C89F and G175R, by systematic sequencing in a cohort of 376 hyperchylomicronemic patients without mutations on the LPL, APOC2, or APOA5 gene. OBJECTIVE: Phenotypic expression and functional consequences of these two mutations were studied. DESIGN: We performed clinical and genotypic studies of probands and their families. GPIHBP1 functional alterations were studied in CHO pgsA-745 transfected cells. RESULTS: Probands are an adult with a homozygous G175R mutation and a child with a hemizygous C89F neomutation and a deletion of the second allele. C89F mutation was associated with a C14F signal peptide polymorphism on the same haplotype. Both patients had resistant hyperchylomicronemia, low LPL activity, and history of acute pancreatitis. In CHO pgsA-745 cells, both G175R and C14F variants reduce the expression of GPIHBP1 at the cell surface. C89F mutation is responsible for a drastic LPL-binding defect to GPIHBP1. C14F may further potentiate C89F effect. CONCLUSIONS: The emergence of hyperchylomicronemia in the generation after a neomutation further establishes a critical role for GPIHBP1 in TGRL physiopathology in humans. Our results highlight the crucial role of C65-C89 disulfide bond in LPL binding by GPIHBP1 Ly6 domain. Furthermore, we first report a mutation of the hydrophobic C-terminal domain that impairs GPIHBP1 membrane targeting.


Asunto(s)
Proteínas Portadoras/genética , Quilomicrones/sangre , Quilomicrones/genética , Hiperlipoproteinemia Tipo I/sangre , Hiperlipoproteinemia Tipo I/genética , Adulto , Animales , Apolipoproteína A-V , Apolipoproteína C-II/genética , Apolipoproteína C-II/metabolismo , Apolipoproteínas A/genética , Apolipoproteínas A/metabolismo , Células CHO , Estudios de Cohortes , Cricetinae , Cricetulus , ADN/genética , Humanos , Lactante , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Hígado/enzimología , Masculino , Mutación/genética , Mutación/fisiología , Mutación Missense , Pancreatitis/complicaciones , Pancreatitis/genética , Linaje , Receptores de Lipoproteína
17.
J Biol Chem ; 286(37): 32198-207, 2011 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-21795711

RESUMEN

Dietary retinoids (vitamin A and its derivatives) contribute to normal embryonic development. However, the mechanism(s) involved in the transfer of recently ingested vitamin A from mother to embryo is not fully understood. We investigated in vivo whether lipoprotein lipase (LPL) facilitates the placental uptake of dietary retinyl ester incorporated in chylomicrons and their remnants and its transfer to the embryo. We examined the effects of both genetic ablation (MCK-L0 mice) and pharmacological inhibition (P-407) of LPL by maintaining wild type and MCK-L0 mice on diets with different vitamin A content or administering them an oral gavage dose of [(3)H]retinol with or without P-407 treatment. We showed that LPL expressed in placenta facilitates uptake of retinoids by this organ and their transfer to the embryo, mainly through its catalytic activity. In addition, through its "bridging function," LPL can mediate the acquisition of nascent chylomicrons by the placenta, although less efficiently. Quantitative real-time PCR and Western blot analysis showed that placental LPL acts in concert with LDL receptor and LRP1. Finally, by knocking out the retinol-binding protein (RBP) gene in the MCK-L0 background (MCK-L0-RBP(-/-) mice) we demonstrated that the placenta acquires dietary retinoids also via the maternal circulating RBP-retinol complex. RBP expressed in the placenta facilitate the transfer of postprandial retinoids across the placental layers toward the embryo.


Asunto(s)
Suplementos Dietéticos , Lipoproteína Lipasa/biosíntesis , Intercambio Materno-Fetal/fisiología , Placenta/enzimología , Proteínas Gestacionales/biosíntesis , Embarazo/fisiología , Vitamina A/farmacocinética , Vitaminas/farmacocinética , Animales , Quilomicrones/genética , Quilomicrones/metabolismo , Embrión de Mamíferos/enzimología , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/genética , Lipoproteína Lipasa/genética , Ratones , Ratones Noqueados , Receptores de LDL/genética , Receptores de LDL/metabolismo , Proteínas de Unión al Retinol/genética , Proteínas de Unión al Retinol/metabolismo , Vitamina A/farmacología , Vitaminas/farmacología
18.
J Biol Chem ; 286(28): 25201-10, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21610069

RESUMEN

The membrane glycoprotein CD36 binds nanomolar concentrations of long chain fatty acids (LCFA) and is highly expressed on the luminal surface of enterocytes. CD36 deficiency reduces chylomicron production through unknown mechanisms. In this report, we provide novel insights into some of the underlying mechanisms. Our in vivo data demonstrate that CD36 gene deletion in mice does not affect LCFA uptake and subsequent esterification into triglycerides by the intestinal mucosa exposed to the micellar LCFA concentrations prevailing in the intestine. In rodents, the CD36 protein disappears early from the luminal side of intestinal villi during the postprandial period, but only when the diet contains lipids. This drop is significant 1 h after a lipid supply and associates with ubiquitination of CD36. Using CHO cells expressing CD36, it is shown that the digestion products LCFA and diglycerides trigger CD36 ubiquitination. In vivo treatment with the proteasome inhibitor MG132 prevents the lipid-mediated degradation of CD36. In vivo and ex vivo, CD36 is shown to be required for lipid activation of ERK1/2, which associates with an increase of the key chylomicron synthesis proteins, apolipoprotein B48 and microsomal triglyceride transfer protein. Therefore, intestinal CD36, possibly through ERK1/2-mediated signaling, is involved in the adaptation of enterocyte metabolism to the postprandial lipid challenge by promoting the production of large triglyceride-rich lipoproteins that are rapidly cleared in the blood. This suggests that CD36 may be a therapeutic target for reducing the postprandial hypertriglyceridemia and associated cardiovascular risks.


Asunto(s)
Antígenos CD36/metabolismo , Quilomicrones/biosíntesis , Enterocitos/metabolismo , Sistema de Señalización de MAP Quinasas/fisiología , Ubiquitinación/fisiología , Animales , Apolipoproteína B-48/genética , Apolipoproteína B-48/metabolismo , Antígenos CD36/genética , Células CHO , Quilomicrones/genética , Cricetinae , Cricetulus , Enterocitos/citología , Hipertrigliceridemia , Metabolismo de los Lípidos/fisiología , Masculino , Ratones , Ratones Noqueados , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Periodo Posprandial , Ratas , Ratas Wistar
19.
Proc Natl Acad Sci U S A ; 108(19): 7980-4, 2011 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-21518912

RESUMEN

GPIHBP1, a glycosylphosphatidylinositol-anchored protein of capillary endothelial cells, shuttles lipoprotein lipase (LPL) from subendothelial spaces to the capillary lumen. An absence of GPIHBP1 prevents the entry of LPL into capillaries, blocking LPL-mediated triglyceride hydrolysis and leading to markedly elevated triglyceride levels in the plasma (i.e., chylomicronemia). Earlier studies have established that chylomicronemia can be caused by LPL mutations that interfere with catalytic activity. We hypothesized that some cases of chylomicronemia might be caused by LPL mutations that interfere with LPL's ability to bind to GPIHBP1. Any such mutation would provide insights into LPL sequences required for GPIHBP1 binding. Here, we report that two LPL missense mutations initially identified in patients with chylomicronemia, C418Y and E421K, abolish LPL's ability to bind to GPIHBP1 without interfering with LPL catalytic activity or binding to heparin. Both mutations abolish LPL transport across endothelial cells by GPIHBP1. These findings suggest that sequences downstream from LPL's principal heparin-binding domain (amino acids 403-407) are important for GPIHBP1 binding. In support of this idea, a chicken LPL (cLPL)-specific monoclonal antibody, xCAL 1-11 (epitope, cLPL amino acids 416-435), blocks cLPL binding to GPIHBP1 but not to heparin. Also, changing cLPL residues 421 to 425, 426 to 430, and 431 to 435 to alanine blocks cLPL binding to GPIHBP1 without inhibiting catalytic activity. Together, these data define a mechanism by which LPL mutations could elicit disease and provide insights into LPL sequences required for binding to GPIHBP1.


Asunto(s)
Proteínas Portadoras/metabolismo , Células Endoteliales/metabolismo , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Mutación Missense , Sustitución de Aminoácidos , Animales , Sitios de Unión/genética , Células CHO , Quilomicrones/sangre , Quilomicrones/genética , Cricetinae , Cricetulus , Humanos , Hiperlipoproteinemia Tipo IV/sangre , Hiperlipoproteinemia Tipo IV/enzimología , Hiperlipoproteinemia Tipo IV/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Unión Proteica , Receptores de Lipoproteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfección
20.
J Intern Med ; 270(3): 224-8, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21314738

RESUMEN

OBJECTIVES: Deficiency in the catabolism of triglyceride-rich lipoproteins is the main cause of childhood-onset chylomicronaemia syndrome. Missense mutations in lipoprotein lipase (LPL) or in proteins influencing LPL activity or stability have been shown to be critical determinants of chylomicronaemia syndrome. The main objective of this study was to assess the primary deficiency in five cases of childhood-onset chylomicronaemia syndrome. SETTING: Lipid clinic at a university hospital, SUBJECTS: Subjects presenting with severe hypertriglyceridaemia and chylomicronaemia syndrome in which reduced LPL activity and mass were observed. INTERVENTIONS: Analysis of LPL and GPIHBP1 genes. RESULTS: Amongst the five patients, one novel homozygous missense mutation (p.C68Y) in exon 3 of GPIHBP1 was identified. The other four patients were homozygous for the common LPL mutation p.G188E. CONCLUSION: These findings provide further evidence that GPIHBP1 is involved in the catabolism of triglyceride-rich lipoproteins and plays a role in childhood-onset chylomicronaemia.


Asunto(s)
Proteínas Portadoras/genética , Quilomicrones/sangre , Hipertrigliceridemia/sangre , Lipoproteína Lipasa/sangre , Mutación Missense , Edad de Inicio , Niño , Quilomicrones/genética , Exones , Femenino , Homocigoto , Humanos , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Masculino , Receptores de Lipoproteína , Síndrome
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...