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










Publication year range
3.
Nat Commun ; 9(1): 4333, 2018 10 15.
Article in English | MEDLINE | ID: mdl-30323191

ABSTRACT

The original version of this Article incorrectly gave a publication date of 8 October 2018; this should have been 28 September 2018. This has now been corrected in the PDF and HTML versions of the Article.

4.
Nat Commun ; 9(1): 3191, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30266909

ABSTRACT

Huntington's disease is a progressive neurodegenerative disorder caused by polyglutamine-expanded mutant huntingtin (mHTT). Here, we show that the deubiquitinase Usp12 rescues mHTT-mediated neurodegeneration in Huntington's disease rodent and patient-derived human neurons, and in Drosophila. The neuroprotective role of Usp12 may be specific amongst related deubiquitinases, as the closely related homolog Usp46 does not suppress mHTT-mediated toxicity. Mechanistically, we identify Usp12 as a potent inducer of neuronal autophagy. Usp12 overexpression accelerates autophagic flux and induces an approximately sixfold increase in autophagic structures as determined by ultrastructural analyses, while suppression of endogenous Usp12 slows autophagy. Surprisingly, the catalytic activity of Usp12 is not required to protect against neurodegeneration or induce autophagy. These findings identify the deubiquitinase Usp12 as a regulator of neuronal proteostasis and mHTT-mediated neurodegeneration.


Subject(s)
Autophagy/genetics , Neurons/metabolism , Neuroprotection/genetics , Ubiquitin Thiolesterase/genetics , Animals , Cells, Cultured , Drosophila melanogaster , Gene Expression Regulation , HEK293 Cells , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Huntington Disease/metabolism , Mutation , Neurons/cytology , Neurons/ultrastructure , RNA Interference , Rats , Ubiquitin Thiolesterase/metabolism
5.
Cell Metab ; 23(3): 492-504, 2016 Mar 08.
Article in English | MEDLINE | ID: mdl-26833026

ABSTRACT

Phospholipids are important determinants of membrane biophysical properties, but the impact of membrane acyl chain composition on dietary-lipid absorption is unknown. Here we demonstrate that the LXR-responsive phospholipid-remodeling enzyme Lpcat3 modulates intestinal fatty acid and cholesterol absorption and is required for survival on a high-fat diet. Mice lacking Lpcat3 in the intestine thrive on carbohydrate-based chow but lose body weight rapidly and become moribund on a triglyceride-rich diet. Lpcat3-dependent incorporation of polyunsaturated fatty acids into phospholipids is required for the efficient transport of dietary lipids into enterocytes. Furthermore, loss of Lpcat3 amplifies the production of gut hormones, including GLP-1 and oleoylethanolamide, in response to high-fat feeding, contributing to the paradoxical cessation of food intake in the setting of starvation. These results reveal that membrane phospholipid composition is a gating factor in passive lipid absorption and implicate LXR-Lpcat3 signaling in a gut-brain feedback loop that couples absorption to food intake.


Subject(s)
Diet, High-Fat/adverse effects , Intestinal Mucosa/metabolism , Phospholipids/metabolism , 1-Acylglycerophosphocholine O-Acyltransferase/genetics , 1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Animals , Apolipoproteins/metabolism , Cholesterol/blood , Dietary Fats/metabolism , Female , Glucagon-Like Peptide 1/genetics , Glucagon-Like Peptide 1/metabolism , Intestinal Absorption , Intestines/pathology , Lipid Metabolism , Male , Membrane Fluidity , Mice, Inbred C57BL , Mice, Knockout , Obesity/blood , Obesity/etiology , Transcriptional Activation , Triglycerides/blood
6.
Antimicrob Agents Chemother ; 59(11): 7109-12, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26324264

ABSTRACT

GS-5806 is a small-molecule inhibitor of human respiratory syncytial virus fusion protein-mediated viral entry. During viral entry, the fusion protein undergoes major conformational changes, resulting in fusion of the viral envelope with the host cell membrane. This process is reproduced in vitro using a purified, truncated respiratory syncytial virus (RSV) fusion protein. GS-5806 blocked these conformational changes, suggesting a possible mechanism for antiviral activity.


Subject(s)
Antiviral Agents/pharmacology , Pyrazoles/pharmacology , Respiratory Syncytial Virus, Human/drug effects , Respiratory Syncytial Virus, Human/metabolism , Sulfonamides/pharmacology , Viral Proteins/chemistry , Viral Proteins/metabolism , Indazoles , Protein Conformation , Respiratory Syncytial Virus Infections
7.
Elife ; 42015 Mar 25.
Article in English | MEDLINE | ID: mdl-25806685

ABSTRACT

The role of specific phospholipids (PLs) in lipid transport has been difficult to assess due to an inability to selectively manipulate membrane composition in vivo. Here we show that the phospholipid remodeling enzyme lysophosphatidylcholine acyltransferase 3 (Lpcat3) is a critical determinant of triglyceride (TG) secretion due to its unique ability to catalyze the incorporation of arachidonate into membranes. Mice lacking Lpcat3 in the intestine fail to thrive during weaning and exhibit enterocyte lipid accumulation and reduced plasma TGs. Mice lacking Lpcat3 in the liver show reduced plasma TGs, hepatosteatosis, and secrete lipid-poor very low-density lipoprotein (VLDL) lacking arachidonoyl PLs. Mechanistic studies indicate that Lpcat3 activity impacts membrane lipid mobility in living cells, suggesting a biophysical basis for the requirement of arachidonoyl PLs in lipidating lipoprotein particles. These data identify Lpcat3 as a key factor in lipoprotein production and illustrate how manipulation of membrane composition can be used as a regulatory mechanism to control metabolic pathways.


Subject(s)
1-Acylglycerophosphocholine O-Acyltransferase/metabolism , Arachidonic Acid/metabolism , Phospholipids/metabolism , Triglycerides/metabolism , 1-Acylglycerophosphocholine O-Acyltransferase/deficiency , Animals , Apolipoproteins B/metabolism , Breeding , Diet , Golgi Apparatus/metabolism , Golgi Apparatus/ultrastructure , Hepatocytes/metabolism , Inflammation/pathology , Lipid Metabolism , Lipoproteins, VLDL , Liver/metabolism , Liver/pathology , Liver X Receptors , Lysophosphatidylcholines/metabolism , Membrane Lipids/metabolism , Mice, Inbred C57BL , Mice, Knockout , Organ Specificity , Orphan Nuclear Receptors/metabolism , Phenotype
8.
JAMA Neurol ; 71(10): 1228-36, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25111166

ABSTRACT

IMPORTANCE: The identification of a patient with a rare form of severe dysbetalipoproteinemia allowed the study of the consequences of total absence of apolipoprotein E (apoE). OBJECTIVES: To discover the molecular basis of this rare disorder and to determine the effects of complete absence of apoE on neurocognitive and visual function and on lipoprotein metabolism. DESIGN, SETTING, AND PARTICIPANTS: Whole-exome sequencing was performed on the patient's DNA. He underwent detailed neurological and visual function testing and lipoprotein analysis. Lipoprotein analysis was also performed in the Cardiovascular Research Institute, University of California, San Francisco, on blood samples from the proband's mother, wife, 2 daughters, and normolipidemic control participants. MAIN OUTCOME MEASURES: Whole-exome sequencing, lipoprotein analysis, and neurocognitive function. RESULTS: The patient was homozygous for an ablative APOE frameshift mutation (c.291del, p.E97fs). No other mutations likely to contribute to the phenotype were discovered, with the possible exception of two, in ABCC2 (p.I670T) and LIPC (p.G137R). Despite complete absence of apoE, he had normal vision, exhibited normal cognitive, neurological, and retinal function, had normal findings on brain magnetic resonance imaging, and had normal cerebrospinal fluid levels of ß-amyloid and tau proteins. He had no significant symptoms of cardiovascular disease except a suggestion of myocardial ischemia on treadmill testing and mild atherosclerosis noted on carotid ultrasonography. He had exceptionally high cholesterol content (760 mg/dL; to convert to millimoles per liter, multiply by 0.0259) and a high cholesterol to triglycerides ratio (1.52) in very low-density lipoproteins with elevated levels of small-diameter high-density lipoproteins, including high levels of prebeta-1 high-density lipoprotein. Intermediate-density lipoproteins, low-density lipoproteins, and very low-density lipoproteins contained elevated apoA-I and apoA-IV levels. The patient's apoC-III and apoC-IV levels were decreased in very low-density lipoproteins. Electron microscopy revealed large lamellar particles having electron-opaque cores attached to electron-lucent zones in intermediate-density and low-density lipoproteins. Low-density lipoprotein particle diameters were distributed bimodally. CONCLUSIONS AND RELEVANCE: Despite a profound effect on lipoprotein metabolism, detailed neurocognitive and retinal studies failed to demonstrate any defects. This suggests that functions of apoE in the brain and eye are not essential or that redundant mechanisms exist whereby its role can be fulfilled. Targeted knockdown of apoE in the central nervous system might be a therapeutic modality in neurodegenerative disorders.


Subject(s)
Apolipoproteins E/genetics , Hyperlipoproteinemia Type III/genetics , Lipase/genetics , Multidrug Resistance-Associated Proteins/genetics , Adult , Apolipoproteins A/blood , Apolipoproteins C/blood , Apolipoproteins E/deficiency , Carotid Artery Diseases/diagnostic imaging , Exercise Test , Exome , Frameshift Mutation , Genotype , High-Density Lipoproteins, Pre-beta/blood , Humans , Hyperlipoproteinemia Type III/physiopathology , Hyperlipoproteinemia Type III/psychology , Lipid Metabolism/genetics , Lipoproteins, HDL/blood , Lipoproteins, LDL/blood , Lipoproteins, VLDL/blood , Male , Multidrug Resistance-Associated Protein 2 , Phenotype , Retina , Sequence Analysis, DNA , Severity of Illness Index , Skin Diseases/genetics , Triglycerides/blood , Ultrasonography , Xanthomatosis/genetics
9.
Proc Natl Acad Sci U S A ; 107(39): 16982-7, 2010 Sep 28.
Article in English | MEDLINE | ID: mdl-20833817

ABSTRACT

Autophagy is an intracellular turnover pathway. It has special relevance for neurodegenerative proteinopathies, such as Alzheimer disease, Parkinson disease, and Huntington disease (HD), which are characterized by the accumulation of misfolded proteins. Although induction of autophagy enhances clearance of misfolded protein and has therefore been suggested as a therapy for proteinopathies, neurons appear to be less responsive to classic autophagy inducers than nonneuronal cells. Searching for improved inducers of neuronal autophagy, we discovered an N(10)-substituted phenoxazine that, at proper doses, potently and safely up-regulated autophagy in neurons in an Akt- and mTOR-independent fashion. In a neuron model of HD, this compound was neuroprotective and decreased the accumulation of diffuse and aggregated misfolded protein. A structure/activity analysis with structurally similar compounds approved by the US Food and Drug Administration revealed a defined pharmacophore for inducing neuronal autophagy. This pharmacophore should prove useful in studying autophagy in neurons and in developing therapies for neurodegenerative proteinopathies.


Subject(s)
Autophagy , Cytoprotection , Huntington Disease/metabolism , Neurons/drug effects , Neuroprotective Agents/pharmacology , Oxazines/pharmacology , Animals , Cells, Cultured , Drug Approval , Huntington Disease/pathology , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Oxazines/chemistry , Protein Folding , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Rats , TOR Serine-Threonine Kinases
10.
J Clin Invest ; 119(7): 1871-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19603543

ABSTRACT

Maintenance of vascular integrity is critical for homeostasis, and temporally and spatially regulated vascular leak is a central feature of inflammation. Sphingosine-1-phosphate (S1P) can regulate endothelial barrier function, but the sources of the S1P that provide this activity in vivo and its importance in modulating different inflammatory responses are unknown. We report here that mutant mice engineered to selectively lack S1P in plasma displayed increased vascular leak and impaired survival after anaphylaxis, administration of platelet-activating factor (PAF) or histamine, and exposure to related inflammatory challenges. Increased leak was associated with increased interendothelial cell gaps in venules and was reversed by transfusion with wild-type erythrocytes (which restored plasma S1P levels) and by acute treatment with an agonist for the S1P receptor 1 (S1pr1). S1pr1 agonist did not protect wild-type mice from PAF-induced leak, consistent with plasma S1P levels being sufficient for S1pr1 activation in wild-type mice. However, an agonist for another endothelial cell Gi-coupled receptor, Par2, did protect wild-type mice from PAF-induced vascular leak, and systemic treatment with pertussis toxin prevented rescue by Par2 agonist and sensitized wild-type mice to leak-inducing stimuli in a manner that resembled the loss of plasma S1P. Our results suggest that the blood communicates with blood vessels via plasma S1P to maintain vascular integrity and regulate vascular leak. This pathway prevents lethal responses to leak-inducing mediators in mouse models.


Subject(s)
Capillary Permeability , Inflammation/metabolism , Lysophospholipids/physiology , Sphingosine/analogs & derivatives , Animals , Erythrocyte Transfusion , Female , GTP-Binding Protein alpha Subunits, Gi-Go/physiology , Lysophospholipids/blood , Male , Mice , Mice, Inbred C57BL , Oligopeptides/physiology , Platelet Activating Factor/pharmacology , Receptors, Lysosphingolipid/physiology , Sphingosine/blood , Sphingosine/physiology
11.
Neural Dev ; 4: 22, 2009 Jun 23.
Article in English | MEDLINE | ID: mdl-19549326

ABSTRACT

BACKGROUND: Regulated secretion of specialized neuropeptides in the vertebrate neuroendocrine system is critical for ensuring physiological homeostasis. Expression of these cell-specific peptide markers in the differentiating hypothalamus commences prior to birth, often predating the physiological demand for secreted neuropeptides. The conserved function and spatial expression of hypothalamic peptides in vertebrates prompted us to search for critical neuroendocrine genes in newly hatched zebrafish larvae. RESULTS: We screened mutant 5 days post-fertilization zebrafish larvae that fail to undergo visually mediated background adaptation for disruption in hypothalamic pomc expression. To our surprise, the ATPase N-ethylmaleimide sensitive factor (nsf) was identified as an essential gene for maintenance of neuroendocrine transcriptional programs during the embryo-to-larva transition. Despite normal hypothalamic development in nsf(st53) mutants, neuropeptidergic cells exhibited a dramatic loss of cell-specific markers by 5 days post-fertilization that is accompanied by elevated intracellular neuropeptide protein. Consistent with the role of NSF in vesicle-membrane fusion events and intracellular trafficking, cytoplasmic endoplasmic reticulum-like membranes accumulate in nsf(-/-) hypothalamic neurons similar to that observed for SEC18 (nsf ortholog) yeast mutants. Our data support a model in which unspent neuropeptide cargo feedbacks to extinguish transcription in neuropeptidergic cells just as they become functionally required. In support of this model we found that gnrh3 transcripts remained unchanged in pre-migratory, non-functional gonadotropin-releasing hormone (GnRH) neurons in nsf(-/-) zebrafish. Furthermore, oxytocin-like (oxtl, intp) transcripts, which are found in osmoreceptive neurons and persist in mutant zebrafish, drop precipitously after mutant zebrafish are acutely challenged with high salt. CONCLUSION: Our analyses of nsf mutant zebrafish reveal an unexpected role for NSF in hypothalamic development, with mutant 5 days post-fertilization larvae exhibiting a stage-dependent loss of neuroendocrine transcripts and a corresponding accumulation of neuropeptides in the soma. Based on our collective findings, we speculate that neuroendocrine transcriptional programs adapt dynamically to both the supply and demand for neuropeptides to ensure adequate homeostatic responses.


Subject(s)
Gene Expression Regulation, Developmental/genetics , Hypothalamus , Mutation/genetics , N-Ethylmaleimide-Sensitive Proteins/metabolism , Neurons/metabolism , Neuropeptides/metabolism , Zebrafish Proteins/metabolism , Animals , Animals, Newborn , Apoptosis/physiology , Embryo, Nonmammalian , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/cytology , Hypothalamus/embryology , Hypothalamus/growth & development , In Situ Nick-End Labeling/methods , Larva , Mice , Models, Biological , N-Ethylmaleimide-Sensitive Proteins/genetics , Neuropeptides/genetics , Oxytocin/genetics , Oxytocin/metabolism , Pro-Opiomelanocortin/metabolism , Pyrrolidonecarboxylic Acid/analogs & derivatives , Pyrrolidonecarboxylic Acid/metabolism , Zebrafish/genetics , Zebrafish/metabolism , Zebrafish Proteins/genetics
12.
Nat Med ; 15(4): 442-6, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19287392

ABSTRACT

Animals are remarkably efficient in absorbing dietary fat and assimilating this energy-dense nutrient into the white adipose tissue (WAT) for storage. Although this metabolic efficiency may confer an advantage in times of calorie deprivation, it contributes to obesity and associated metabolic disorders when dietary fat is abundant. Here we show that the intestinal lipid synthesis enzyme acyl CoA:monoacylglycerol acyltransferase-2 (MGAT2) has a crucial role in the assimilation of dietary fat and the accretion of body fat in mice. Mice lacking MGAT2 have a normal phenotype on a low-fat diet. However, on a high-fat diet, MGAT2-deficient mice are protected against developing obesity, glucose intolerance, hypercholesterolemia and fatty livers. Caloric intake is normal in MGAT2-deficient mice, and dietary fat is absorbed fully. However, entry of dietary fat into the circulation occurs at a reduced rate. This altered kinetics of fat absorption apparently results in more partitioning of dietary fat toward energy dissipation rather than toward storage in the WAT. Thus, our studies identify MGAT2 as a key determinant of energy metabolism in response to dietary fat and suggest that the inhibition of this enzyme may prove to be a useful strategy for treating obesity and other metabolic diseases associated with excessive fat intake.


Subject(s)
Acyltransferases/deficiency , Dietary Fats/adverse effects , Obesity/genetics , Adipose Tissue/drug effects , Adipose Tissue/physiopathology , Animals , Body Temperature , Body Weight , Energy Intake , Energy Metabolism , Female , Growth , Hypercholesterolemia/prevention & control , Male , Mice , Obesity/prevention & control
13.
Nature ; 453(7195): 657-61, 2008 May 29.
Article in English | MEDLINE | ID: mdl-18408709

ABSTRACT

Eukaryotic cells store neutral lipids in cytoplasmic lipid droplets enclosed in a monolayer of phospholipids and associated proteins. These dynamic organelles serve as the principal reservoirs for storing cellular energy and for the building blocks for membrane lipids. Excessive lipid accumulation in cells is a central feature of obesity, diabetes and atherosclerosis, yet remarkably little is known about lipid-droplet cell biology. Here we show, by means of a genome-wide RNA interference (RNAi) screen in Drosophila S2 cells that about 1.5% of all genes function in lipid-droplet formation and regulation. The phenotypes of the gene knockdowns sorted into five distinct phenotypic classes. Genes encoding enzymes of phospholipid biosynthesis proved to be determinants of lipid-droplet size and number, suggesting that the phospholipid composition of the monolayer profoundly affects droplet morphology and lipid utilization. A subset of the Arf1-COPI vesicular transport proteins also regulated droplet morphology and lipid utilization, thereby identifying a previously unrecognized function for this machinery. These phenotypes are conserved in mammalian cells, suggesting that insights from these studies are likely to be central to our understanding of human diseases involving excessive lipid storage.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/cytology , Drosophila melanogaster/genetics , Genes, Insect/genetics , Genome, Insect/genetics , Genomics , Lipid Metabolism/genetics , ADP-Ribosylation Factors/metabolism , Animals , Cell Line , Coat Protein Complex I/metabolism , Lipolysis , Phenotype , Phosphatidylcholines/metabolism , RNA Interference
14.
Cell Metab ; 5(4): 279-91, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17403372

ABSTRACT

The triglycerides in chylomicrons are hydrolyzed by lipoprotein lipase (LpL) along the luminal surface of the capillaries. However, the endothelial cell molecule that facilitates chylomicron processing by LpL has not yet been defined. Here, we show that glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein 1 (GPIHBP1) plays a critical role in the lipolytic processing of chylomicrons. Gpihbp1-deficient mice exhibit a striking accumulation of chylomicrons in the plasma, even on a low-fat diet, resulting in milky plasma and plasma triglyceride levels as high as 5000 mg/dl. Normally, Gpihbp1 is expressed highly in heart and adipose tissue, the same tissues that express high levels of LpL. In these tissues, GPIHBP1 is located on the luminal face of the capillary endothelium. Expression of GPIHBP1 in cultured cells confers the ability to bind both LpL and chylomicrons. These studies strongly suggest that GPIHBP1 is an important platform for the LpL-mediated processing of chylomicrons in capillaries.


Subject(s)
Chylomicrons/metabolism , Lipolysis/genetics , Receptors, Lipoprotein/physiology , Animals , CHO Cells , Chylomicrons/blood , Cricetinae , Cricetulus , Eating/physiology , Endothelial Cells/metabolism , Gene Expression Regulation , Lipoprotein Lipase/metabolism , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Rabbits , Receptors, Lipoprotein/genetics , Receptors, Lipoprotein/metabolism , Transfection
15.
J Lipid Res ; 47(10): 2297-305, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16877777

ABSTRACT

Increased triglyceride synthesis resulting from enhanced flux of fatty acids into liver is frequently associated with VLDL overproduction. This has led to the common belief that hepatic triglyceride synthesis can directly modulate VLDL production. We used adenoviral vectors containing either murine acyl-coenzyme A:diacylglycerol transferase 1 (DGAT1) or DGAT2 cDNA to determine the effect of a short-term increase in hepatic triglyceride synthesis on VLDL triglyceride and apolipoprotein B (apoB) production in female wild-type mice. Hepatic DGAT1 and DGAT2 overexpression resulted in 2.0-fold and 2.4-fold increases in the triglyceride content of liver, respectively. However, the increase in hepatic triglyceride content had no effect on the production rate of VLDL triglyceride or apoB in either case. Liver subfractionation showed that DGAT1 and DGAT2 overexpression significantly increased the content of triglyceride within the cytoplasmic lipid fraction, with no change in the triglyceride content of the microsomal membrane or microsomal VLDL. The increased cytoplasmic triglyceride content was observed in electron micrographs of liver sections from mice overexpressing DGAT1 or DGAT2. Overexpression of DGAT1 or DGAT2 resulted in enhanced [(3)H]glycerol tracer incorporation into triglyceride within cytoplasmic lipids. These results suggest that increasing the cytoplasmic triglyceride pool in hepatocytes does not directly influence VLDL triglyceride or apoB production. In the presence of adequate cytoplasmic lipid stores, factors other than triglyceride synthesis are rate-limiting for VLDL production.


Subject(s)
Apolipoproteins B/biosynthesis , Diacylglycerol O-Acyltransferase/biosynthesis , Lipoproteins, VLDL/metabolism , Liver/metabolism , Triglycerides/metabolism , Animals , Diacylglycerol O-Acyltransferase/genetics , Female , Mice , Mice, Inbred C57BL , Mice, Transgenic , Subcellular Fractions , Time Factors
16.
J Biol Chem ; 281(7): 4075-86, 2006 Feb 17.
Article in English | MEDLINE | ID: mdl-16354657

ABSTRACT

Microsomal TG transfer protein (MTTP) is required for the assembly and secretion of TG (TG)-rich lipoproteins from both enterocytes and hepatocytes. Liver-specific deletion of Mttp produced a dramatic reduction in plasma very low density lipoprotein-TG and virtually eliminated apolipoprotein B100 (apoB100) secretion yet caused only modest reductions in plasma apoB48 and apoB48 secretion from primary hepatocytes. These observations prompted us to examine the phenotype following intestine-specific Mttp deletion because murine, like human enterocytes, secrete virtually exclusively apoB48. We generated mice with conditional Mttp deletion in villus enterocytes (Mttp-IKO), using a tamoxifen-inducible, intestine-specific Cre transgene. Villus enterocytes from chow-fed Mttp-IKO mice contained large cytoplasmic TG droplets and no chylomicron-sized particles within the secretory pathway. Chow-fed, Mttp-IKO mice manifested steatorrhea, growth arrest, and decreased cholesterol absorption, features that collectively recapitulate the phenotype associated with abetalipoproteinemia. Chylomicron secretion was reduced dramatically in vivo, in conjunction with an approximately 80% decrease in apoB48 secretion from primary enterocytes. Additionally, although plasma and hepatic cholesterol and TG content were decreased, Mttp-IKO mice demonstrated a paradoxical increase in both hepatic lipogenesis and very low density lipoprotein secretion. These findings establish distinctive features for MTTP involvement in intestinal chylomicron assembly and secretion and suggest that hepatic lipogenesis undergoes compensatory induction in the face of defective intestinal TG secretion.


Subject(s)
Carrier Proteins/physiology , Intestinal Mucosa/metabolism , Lipogenesis , Liver/metabolism , Animals , Chylomicrons/metabolism , Hydroxymethylglutaryl CoA Reductases/genetics , Intestinal Absorption , Lipid Metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Triglycerides/metabolism , Weight Gain
17.
Arterioscler Thromb Vasc Biol ; 23(9): 1653-9, 2003 Sep 01.
Article in English | MEDLINE | ID: mdl-12893687

ABSTRACT

OBJECTIVE: We have previously shown that intravenous apolipoprotein A-I/phosphatidylcholine (apoA-I/PC) discs increase plasma pre-beta HDL concentration and stimulate reverse cholesterol transport (RCT) in humans. We have now investigated the associated changes in the following 3 HDL components that play key roles in RCT: lecithin:cholesterol acyltransferase (LCAT), cholesteryl ester transfer protein (CETP), and phospholipid transfer protein (PLTP). METHODS AND RESULTS: apoA-I/PC discs (40 mg/kg over 4 hours) were infused into 8 healthy men. Samples of blood and prenodal peripheral lymph were collected for 24 to 48 hours. At 12 hours, plasma LCAT concentration had increased by 0.40+/-0.90 mg/L (+7.8%; mean+/-SD; P<0.05), plasma cholesterol esterification rate by 29.0+/-9.0 nmol/mL per h (+69.5%; P<0.01), plasma CETP concentration by 0.5+/-0.2 mg/L (+29.7%; P<0.01), and plasma PLTP activity by 1.45+/-0.67 micromol/mL per h (+23.9%; P<0.01). In contrast, plasma PLTP concentration had decreased by 4.4+/-2.7 mg/L (-44.8%; P<0.01). The changes in PLTP were accompanied by alterations in the relative proportions of large lipoproteins containing inactive PLTP and small particles containing PLTP of high specific activity. No changes were detected in peripheral lymph. CONCLUSIONS: Nascent HDL secretion may induce changes in PLTP, LCAT, and CETP that promote RCT by catalyzing pre-beta HDL production, cholesterol esterification in HDLs, and cholesteryl ester transfer from HDLs to other lipoproteins.


Subject(s)
Apolipoprotein A-I/administration & dosage , Apolipoprotein A-I/pharmacology , Blood Proteins/metabolism , Glycoproteins , Lymph/chemistry , Phosphatidylcholines/administration & dosage , Phosphatidylcholines/pharmacology , Phospholipid Transfer Proteins , Adult , Apolipoproteins/blood , Carrier Proteins/blood , Carrier Proteins/metabolism , Cholesterol Ester Transfer Proteins , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacology , Drug Delivery Systems , Humans , Infusions, Intravenous , Lipids/blood , Lipoproteins/ultrastructure , Male , Membrane Proteins/blood , Membrane Proteins/metabolism , Microscopy, Electron , Phosphatidylcholine-Sterol O-Acyltransferase/blood , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism
18.
Circulation ; 107(9): 1315-21, 2003 Mar 11.
Article in English | MEDLINE | ID: mdl-12628954

ABSTRACT

BACKGROUND: LDL receptor-deficient "apolipoprotein (apo)-B100-only" mice (Ldlr-/-Apob100/100 have elevated LDL cholesterol levels on a chow diet and develop severe aortic atherosclerosis. We hypothesized that both the hypercholesterolemia and the susceptibility to atherosclerosis could be eliminated by switching off hepatic lipoprotein production. METHODS AND RESULTS: We bred Ldlr-/-Apob100/100 mice that were homozygous for a conditional allele for Mttp (the gene for microsomal triglyceride transfer protein) and the inducible Mx1-Cre transgene. In these animals, which we called "Reversa mice," the hypercholesterolemia could be reversed, without modifying the diet or initiating a hypolipidemic drug, by the transient induction of Cre expression in the liver. After Cre induction, hepatic Mttp expression was virtually eliminated (as judged by quantitative real-time PCR), hepatic lipoprotein secretion was abolished (as judged by electron microscopy), and LDLs were virtually eliminated from the plasma. Intestinal lipoprotein production was unaffected. In mice fed a chow diet, Cre induction reduced plasma cholesterol levels from 233.9+/-46.0 to 37.2+/-6.5 mg/dL. In mice fed a high-fat diet, cholesterol levels fell from 525.7+/-32.2 to 100.6+/-14.3 mg/dL. The elimination of hepatic lipoprotein production completely prevented both the development of atherosclerosis and the changes in gene expression that accompany atherogenesis. CONCLUSIONS: We developed mice in which hypercholesterolemia can be reversed with a genetic switch. These mice will be useful for understanding gene-expression changes that accompany the reversal of hypercholesterolemia and atherosclerosis.


Subject(s)
Arteriosclerosis/prevention & control , Carrier Proteins/genetics , Lipoproteins/metabolism , Liver/metabolism , Animals , Apolipoprotein B-100 , Apolipoproteins B/genetics , Arteries/metabolism , Arteriosclerosis/etiology , Arteriosclerosis/metabolism , Cholesterol/blood , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Hypercholesterolemia/blood , Hypercholesterolemia/therapy , Integrases/genetics , Integrases/metabolism , Lipids/blood , Lipoproteins/chemistry , Lipoproteins/ultrastructure , Mice , Particle Size , Poly I-C/pharmacology , Receptors, LDL/genetics , Viral Proteins/genetics , Viral Proteins/metabolism
19.
J Biol Chem ; 277(28): 25474-9, 2002 Jul 12.
Article in English | MEDLINE | ID: mdl-11959864

ABSTRACT

Dietary triacylglycerols are a major source of energy for animals. The absorption of dietary triacylglycerols involves their hydrolysis to free fatty acids and monoacylglycerols in the intestinal lumen, the uptake of these products into enterocytes, the resynthesis of triacylgylcerols, and the incorporation of newly synthesized triacylglycerols into nascent chylomicrons for secretion. In enterocytes, the final step in triacylglycerol synthesis is believed to be catalyzed primarily through the actions of acyl-CoA:diacylglycerol acyltransferase (DGAT) enzymes. In this study, we analyzed intestinal triacylglycerol absorption and chylomicron synthesis and secretion in DGAT1-deficient (Dgat1(-/-)) mice. Surprisingly, DGAT1 was not essential for quantitative dietary triacylglycerol absorption, even in mice fed a high fat diet, or for the synthesis of chylomicrons. However, Dgat1(-/-) mice had reduced postabsorptive chylomicronemia (1 h after a high fat challenge) and accumulated neutral-lipid droplets in the cytoplasm of enterocytes when chronically fed a high fat diet. These results suggest a reduced rate of triacylglycerol absorption in Dgat1(-/-) mice. Analysis of intestine from Dgat1(-/-) mice revealed activity for two other enzymes, DGAT2 and diacylglycerol transacylase, that catalyze triacylglycerol synthesis and apparently help to compensate for the absence of DGAT1. Our findings indicate that multiple mechanisms for triacylglycerol synthesis in the intestine facilitate triacylglycerol absorption.


Subject(s)
Acyltransferases/physiology , Chylomicrons/biosynthesis , Intestinal Absorption/physiology , Triglycerides/metabolism , Acyltransferases/genetics , Animals , Diacylglycerol O-Acyltransferase , Gene Expression , Intestine, Small/metabolism , Intestine, Small/physiology , Intestine, Small/ultrastructure , Mice , Mice, Inbred C57BL , Mice, Knockout
20.
J Lipid Res ; 43(4): 544-9, 2002 Apr.
Article in English | MEDLINE | ID: mdl-11907136

ABSTRACT

Scavenger receptor class B type I (SR-BI) mediates the selective uptake of HDL cholesteryl ester into liver and steroidogenic tissues. In steroidogenic cells, juxtaposed microvilli, or microvilli snuggled against the plasma membrane create microvillar channels that fill with HDL. Microvillar membranes contain SR-BI and are believed to be the site of HDL cholesteryl ester uptake. A recent study showed that SR-BI expression in insect cells elicits membrane structures that contain SR-BI, bind HDL, and closely resemble the ultrastructure of microvillar channels. In the present study we compared the ultrastructure of adrenal gland microvillar membranes in Srb1+/+ and Srb1-/- mice to test whether SR-BI is required for the formation of microvillar channels. The results show that SR-BI is absolutely required for microvillar channel formation and that the microvillar membranes of Srb1-/- mice are 17% thinner than in Srb1+/+ mice. We conclude that SR-BI has a major influence on plasma membrane ultrastructure and organization in vivo.


Subject(s)
Adrenal Cortex/metabolism , CD36 Antigens/metabolism , Lipoproteins, HDL/metabolism , Membrane Proteins , Microvilli/metabolism , Receptors, Immunologic , Receptors, Lipoprotein , Adrenal Cortex/cytology , Adrenal Cortex/ultrastructure , Animals , Biological Transport , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Mice , Mice, Inbred C57BL , Microscopy, Electron , Microvilli/ultrastructure , Particle Size , Receptors, Scavenger , Scavenger Receptors, Class B
SELECTION OF CITATIONS
SEARCH DETAIL
...