Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 27
Filtrer
1.
Arterioscler Thromb Vasc Biol ; 44(6): 1407-1418, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38695168

RÉSUMÉ

BACKGROUND: LCAT (lecithin cholesterol acyl transferase) catalyzes the conversion of unesterified, or free cholesterol, to cholesteryl ester, which moves from the surface of HDL (high-density lipoprotein) into the neutral lipid core. As this iterative process continues, nascent lipid-poor HDL is converted to a series of larger, spherical cholesteryl ester-enriched HDL particles that can be cleared by the liver in a process that has been termed reverse cholesterol transport. METHODS: We conducted a randomized, placebocontrolled, crossover study in 5 volunteers with atherosclerotic cardiovascular disease, to examine the effects of an acute increase of recombinant human (rh) LCAT via intravenous administration (300-mg loading dose followed by 150 mg at 48 hours) on the in vivo metabolism of HDL APO (apolipoprotein)A1 and APOA2, and the APOB100-lipoproteins, very low density, intermediate density, and low-density lipoproteins. RESULTS: As expected, recombinant human LCAT treatment significantly increased HDL-cholesterol (34.9 mg/dL; P≤0.001), and this was mostly due to the increase in cholesteryl ester content (33.0 mg/dL; P=0.014). This change did not affect the fractional clearance or production rates of HDL-APOA1 and HDL-APOA2. There were also no significant changes in the metabolism of APOB100-lipoproteins. CONCLUSIONS: Our results suggest that an acute increase in LCAT activity drives greater flux of cholesteryl ester through the reverse cholesterol transport pathway without significantly altering the clearance and production of the main HDL proteins and without affecting the metabolism of APOB100-lipoproteins. Long-term elevations of LCAT might, therefore, have beneficial effects on total body cholesterol balance and atherogenesis.


Sujet(s)
Apolipoprotéine A-II , Apolipoprotéine A-I , Cholestérol HDL , Études croisées , Phosphatidylcholine-Sterol O-Acyltransferase , Protéines recombinantes , Humains , Phosphatidylcholine-Sterol O-Acyltransferase/métabolisme , Mâle , Apolipoprotéine A-I/sang , Adulte d'âge moyen , Cholestérol HDL/sang , Apolipoprotéine A-II/sang , Femelle , Cholestérol ester/sang , Cholestérol ester/métabolisme , Athérosclérose/traitement médicamenteux , Athérosclérose/enzymologie , Athérosclérose/sang , Apolipoprotéine B-100/sang , Sujet âgé , Adulte , Lipoprotéines/sang , Lipoprotéines/métabolisme
2.
JCI Insight ; 9(3)2024 Feb 08.
Article de Anglais | MEDLINE | ID: mdl-38194265

RÉSUMÉ

Depletion of torsinA from hepatocytes leads to reduced liver triglyceride secretion and marked hepatic steatosis. TorsinA is an atypical ATPase that lacks intrinsic activity unless it is bound to its activator, lamina-associated polypeptide 1 (LAP1) or luminal domain-like LAP1 (LULL1). We previously demonstrated that depletion of LAP1 from hepatocytes has more modest effects on liver triglyceride secretion and steatosis development than depletion of torsinA. We now show that depletion of LULL1 alone does not significantly decrease triglyceride secretion or cause steatosis. However, simultaneous depletion of both LAP1 and LULL1 leads to defective triglyceride secretion and marked steatosis similar to that observed with depletion of torsinA. Depletion of both LAP1 and torsinA from hepatocytes generated phenotypes similar to those observed with only torsinA depletion, implying that the 2 proteins act in the same pathway in liver lipid metabolism. Our results demonstrate that torsinA and its activators dynamically regulate hepatic lipid metabolism.


Sujet(s)
Protéines de transport , Métabolisme lipidique , Protéines de transport/génétique , Protéines membranaires/métabolisme , Foie/métabolisme , Triglycéride/métabolisme
3.
bioRxiv ; 2023 Jul 26.
Article de Anglais | MEDLINE | ID: mdl-37547008

RÉSUMÉ

TorsinA is an atypical ATPase that lacks intrinsic activity unless it is bound to its activators lamina-associated polypeptide 1 (LAP1) in the perinuclear space or luminal domain-like LAP1 (LULL1) throughout the endoplasmic reticulum. However, the interaction of torsinA with LAP1 and LULL1 has not yet been shown to modulate a defined physiological process in mammals in vivo . We previously demonstrated that depletion of torsinA from mouse hepatocytes leads to reduced liver triglyceride secretion and marked steatosis, whereas depletion of LAP1 had more modest similar effects. We now show that depletion of LULL1 alone does not significantly decrease liver triglyceride secretion or cause steatosis. However, simultaneous depletion of both LAP1 and LULL1 from hepatocytes leads to defective triglyceride secretion and marked steatosis similar to that observed with depletion of torsinA. Our results demonstrate that torsinA and its activators dynamically regulate a physiological process in mammals in vivo .

4.
J Lipid Res ; 63(10): 100277, 2022 10.
Article de Anglais | MEDLINE | ID: mdl-36100089

RÉSUMÉ

Lipid droplets (LDs) are generally considered to be synthesized in the ER and utilized in the cytoplasm. However, LDs have been observed inside nuclei in some cells, although recent research on nuclear LDs has focused on cultured cell lines. To better understand nuclear LDs that occur in vivo, here we examined LDs in primary hepatocytes from mice following depletion of the nuclear envelope protein lamina-associated polypeptide 1 (LAP1). Microscopic image analysis showed that LAP1-depleted hepatocytes contain frequent nuclear LDs, which differ from cytoplasmic LDs in their associated proteins. We found type 1 nucleoplasmic reticula, which are invaginations of the inner nuclear membrane, are often associated with nuclear LDs in these hepatocytes. Furthermore, in vivo depletion of the nuclear envelope proteins lamin A and C from mouse hepatocytes led to severely abnormal nuclear morphology, but significantly fewer nuclear LDs than were observed upon depletion of LAP1. In addition, we show both high-fat diet feeding and fasting of mice increased cytoplasmic lipids in LAP1-depleted hepatocytes but reduced nuclear LDs, demonstrating a relationship of LD formation with nutritional state. Finally, depletion of microsomal triglyceride transfer protein did not change the frequency of nuclear LDs in LAP1-depleted hepatocytes, suggesting that it is not required for the biogenesis of nuclear LDs in these cells. Together, these data show that LAP1-depleted hepatocytes represent an ideal mammalian system to investigate the biogenesis of nuclear LDs and their partitioning between the nucleus and cytoplasm in response to changes in nutritional state and cellular metabolism in vivo.


Sujet(s)
Gouttelettes lipidiques , Enveloppe nucléaire , Souris , Animaux , Gouttelettes lipidiques/métabolisme , Enveloppe nucléaire/métabolisme , Lamine A/métabolisme , Hépatocytes/métabolisme , Protéines membranaires/métabolisme , Peptides/métabolisme , Lipides , Mammifères/métabolisme
5.
Mol Metab ; 56: 101412, 2022 02.
Article de Anglais | MEDLINE | ID: mdl-34890852

RÉSUMÉ

OBJECTIVE: Multiple genome-wide association studies (GWAS) have identified SNPs in the 8q24 locus near TRIB1 that are significantly associated with plasma lipids and other markers of cardiometabolic health, and prior studies have revealed the roles of hepatic and myeloid Trib1 in plasma lipid regulation and atherosclerosis. The same 8q24 SNPs are additionally associated with plasma adiponectin levels in humans, implicating TRIB1 in adipocyte biology. Here, we hypothesize that TRIB1 in adipose tissue regulates plasma adiponectin, lipids, and metabolic health. METHODS: We investigate the metabolic phenotype of adipocyte-specific Trib1 knockout mice (Trib1_ASKO) fed on chow and high-fat diet (HFD). Through secretomics of adipose tissue explants and RNA-seq of adipocytes and livers from these mice, we further investigate the mechanism of TRIB1 in adipose tissue. RESULTS: Trib1_ASKO mice have an improved metabolic phenotype with increased plasma adiponectin levels, improved glucose tolerance, and decreased plasma lipids. Trib1_ASKO adipocytes have increased adiponectin production and secretion independent of the known TRIB1 function of regulating proteasomal degradation. RNA-seq analysis of adipocytes and livers from Trib1_ASKO mice indicates that alterations in adipocyte function underlie the observed plasma lipid changes. Adipose tissue explant secretomics further reveals that Trib1_ASKO adipose tissue has decreased ANGPTL4 production, and we demonstrate an accompanying increase in the lipoprotein lipase (LPL) activity that likely underlies the triglyceride phenotype. CONCLUSIONS: This study shows that adipocyte Trib1 regulates multiple aspects of metabolic health, confirming previously observed genetic associations in humans and shedding light on the further mechanisms by which TRIB1 regulates plasma lipids and metabolic health.


Sujet(s)
Adiponectine , Étude d'association pangénomique , Adipocytes/métabolisme , Adiponectine/génétique , Adiponectine/métabolisme , Animaux , Protéines et peptides de signalisation intracellulaire , Souris , Souris knockout , Protein-Serine-Threonine Kinases/antagonistes et inhibiteurs , Triglycéride/métabolisme
6.
Biology (Basel) ; 9(10)2020 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-33076344

RÉSUMÉ

Nonalcoholic fatty liver disease (NAFLD) is a burgeoning public health problem worldwide. Despite its tremendous significance for public health, we lack a comprehensive understanding of the pathogenic mechanisms of NAFLD and its more advanced stage, nonalcoholic steatohepatitis (NASH). Identification of novel pathways or cellular mechanisms that regulate liver lipid metabolism has profound implications for the understanding of the pathology of NAFLD and NASH. The nuclear envelope is topologically connected to the ER, where protein synthesis and lipid synthesis occurs. Emerging evidence points toward that the nuclear lamins and nuclear membrane-associated proteins are involved in lipid metabolism and homeostasis. We review published reports that link these nuclear envelope proteins to lipid metabolism. In particular, we focus on the recent work demonstrating the essential roles for the nuclear envelope-localized torsinA/lamina-associated polypeptide (LAP1) complex in hepatic steatosis, lipid secretion, and NASH development. We also discuss plausible pathogenic mechanisms by which the loss of either protein in hepatocytes leads to hepatic dyslipidemia and NASH development.

7.
J Clin Invest ; 129(11): 4885-4900, 2019 08 13.
Article de Anglais | MEDLINE | ID: mdl-31408437

RÉSUMÉ

Deciphering novel pathways regulating liver lipid content has profound implications for understanding the pathophysiology of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. Recent evidence suggests that the nuclear envelope is a site of regulation of lipid metabolism but there is limited appreciation of the responsible mechanisms and molecular components within this organelle. We showed that conditional hepatocyte deletion of the inner nuclear membrane protein lamina-associated polypeptide 1 (LAP1) caused defective VLDL secretion and steatosis, including intranuclear lipid accumulation. LAP1 binds to and activates torsinA, an AAA+ ATPase that resides in the perinuclear space and continuous main ER. Deletion of torsinA from mouse hepatocytes caused even greater reductions in VLDL secretion and profound steatosis. Both of these mutant mouse lines developed hepatic steatosis and subsequent steatohepatitis on a regular chow diet in the absence of whole-body insulin resistance or obesity. Our results establish an essential role for the nuclear envelope-localized torsinA-LAP1 complex in hepatic VLDL secretion and suggest that the torsinA pathway participates in the pathophysiology of nonalcoholic fatty liver disease.


Sujet(s)
Protéines de transport/métabolisme , Hépatocytes/métabolisme , Lipoprotéines VLDL/métabolisme , Protéines membranaires/métabolisme , Chaperons moléculaires/métabolisme , Stéatose hépatique non alcoolique/métabolisme , Enveloppe nucléaire/métabolisme , Animaux , Protéines de transport/génétique , Hépatocytes/anatomopathologie , Métabolisme lipidique , Lipoprotéines VLDL/génétique , Protéines membranaires/génétique , Souris , Souris knockout , Chaperons moléculaires/génétique , Stéatose hépatique non alcoolique/génétique , Stéatose hépatique non alcoolique/anatomopathologie , Enveloppe nucléaire/génétique , Enveloppe nucléaire/anatomopathologie
8.
J Biol Chem ; 292(11): 4395-4410, 2017 03 17.
Article de Anglais | MEDLINE | ID: mdl-28031458

RÉSUMÉ

Niemann-Pick type C (NP-C) disease is a fatal genetic lipidosis for which there is no Food and Drug Administration (FDA)-approved therapy. Vorinostat, an FDA-approved inhibitor of histone deacetylases, ameliorates lysosomal lipid accumulation in cultured NP-C patient fibroblasts. To assess the therapeutic potential of histone deacetylase inhibition, we pursued these in vitro observations in two murine models of NP-C disease. Npc1nmf164 mice, which express a missense mutation in the Npc1 gene, were treated intraperitoneally, from weaning, with the maximum tolerated dose of vorinostat (150 mg/kg, 5 days/week). Disease progression was measured via gene expression, liver function and pathology, serum and tissue lipid levels, body weight, and life span. Transcriptome analyses of treated livers indicated multiple changes consistent with reversal of liver dysfunction that typifies NP-C disease. Significant improvements in liver pathology and function were achieved by this treatment regimen; however, NPC1 protein maturation and levels, disease progression, weight loss, and animal morbidity were not detectably altered. Vorinostat concentrations were >200 µm in the plasma compartment of treated animals but were almost 100-fold lower in brain tissue. Apolipoprotein B metabolism and the expression of key components of lipid homeostasis in primary hepatocytes from null (Npc1-/-) and missense (Npc1nmf164 ) mutant mice were altered by vorinostat treatment, consistent with a response by these cells independent of the status of the Npc1 locus. These results suggest that HDAC inhibitors have utility to treat visceral NP-C disease. However, it is clear that improved blood-brain barrier penetration will be required to alleviate the neurological symptoms of human NP-C disease.


Sujet(s)
Inhibiteurs de désacétylase d'histone/usage thérapeutique , Acides hydroxamiques/usage thérapeutique , Foie/effets des médicaments et des substances chimiques , Foie/physiopathologie , Maladie de Niemann-Pick de type C/traitement médicamenteux , Maladie de Niemann-Pick de type C/génétique , Protéines/génétique , Animaux , Apolipoprotéines B/métabolisme , Cellules cultivées , Cholestérol/génétique , Cholestérol/métabolisme , Modèles animaux de maladie humaine , Inhibiteurs de désacétylase d'histone/pharmacocinétique , Homéostasie/effets des médicaments et des substances chimiques , Humains , Acides hydroxamiques/pharmacocinétique , Protéines et peptides de signalisation intracellulaire , Foie/métabolisme , Foie/anatomopathologie , Souris , Souris de lignée C57BL , Mutation faux-sens , Protéine NPC1 , Maladie de Niemann-Pick de type C/anatomopathologie , Maladie de Niemann-Pick de type C/physiopathologie , Protéines/métabolisme , Transcriptome/effets des médicaments et des substances chimiques , Vorinostat
9.
J Clin Invest ; 126(10): 3852-3867, 2016 10 03.
Article de Anglais | MEDLINE | ID: mdl-27599291

RÉSUMÉ

Inhibition of VLDL secretion reduces plasma levels of atherogenic apolipoprotein B (apoB) lipoproteins but can also cause hepatic steatosis. Approaches targeting apoB synthesis, which lies upstream of VLDL secretion, have potential to effectively reduce dyslipidemia but can also lead to hepatic accumulation of unsecreted triglycerides (TG). Here, we found that treating mice with apoB antisense oligonucleotides (ASOs) for 6 weeks decreased VLDL secretion and plasma cholesterol without causing steatosis. The absence of steatosis was linked to an increase in ER stress in the first 3 weeks of ASO treatment, followed by development of ER autophagy at the end of 6 weeks of treatment. The latter resulted in increased fatty acid (FA) oxidation that was inhibited by both chloroquine and 3-methyl adenine, consistent with trafficking of ER TG through the autophagic pathway before oxidation. These findings support the concept that inhibition of apoB synthesis traps lipids that have been transferred to the ER by microsomal TG transfer protein (MTP), inducing ER stress. ER stress then triggers ER autophagy and subsequent lysosomal lipolysis of TG, followed by mitochondrial oxidation of released FA, leading to prevention of steatosis. The identification of this pathway indicates that inhibition of VLDL secretion remains a viable target for therapies aiming to reduce circulating levels of atherogenic apoB lipoproteins.


Sujet(s)
Apolipoprotéines B/biosynthèse , Autophagie , Réticulum endoplasmique/métabolisme , Stéatose hépatique/thérapie , Animaux , Apolipoprotéines B/génétique , Apolipoprotéines B/métabolisme , Athérosclérose/étiologie , Athérosclérose/anatomopathologie , Protéines de transport/biosynthèse , Protéines de transport/génétique , Cellules cultivées , Dyslipidémies/complications , Dyslipidémies/anatomopathologie , Stress du réticulum endoplasmique , Acides gras/métabolisme , Stéatose hépatique/métabolisme , Stéatose hépatique/anatomopathologie , Techniques de knock-down de gènes , Lipogenèse , Foie/métabolisme , Foie/anatomopathologie , Souris de lignée C57BL , Souris knockout , Oligonucléotides antisens/génétique , Oxydoréduction , Biosynthèse des protéines , Triglycéride/métabolisme
10.
Arterioscler Thromb Vasc Biol ; 36(7): 1356-66, 2016 07.
Article de Anglais | MEDLINE | ID: mdl-27150392

RÉSUMÉ

OBJECTIVE: Plasma levels of high-density lipoprotein cholesterol (HDL-C) and apolipoprotein A-I (ApoA-I) are reduced in individuals with defective insulin signaling. Initial studies using liver-specific insulin receptor (InsR) knockout mice identified reduced expression of type 1 deiodinase (Dio1) as a potentially novel link between defective hepatic insulin signaling and reduced expression of the ApoA-I gene. Our objective was to examine the regulation of ApoA-I expression by Dio1. APPROACH AND RESULTS: Acute inactivation of InsR by adenoviral delivery of Cre recombinase to InsR floxed mice reduced HDL-C and expression of both ApoA-I and Dio1. Overexpression of Dio1 in InsR knockout mice restored HDL-C and ApoA-I levels and increased the expression of ApoA-I. Dio1 knockout mice had low expression of ApoA-I and reduced serum levels of HDL-C and ApoA-I. Treatment of C57BL/6J mice with antisense to Dio1 reduced ApoA-I mRNA, HDL-C, and serum ApoA-I. Hepatic 3,5,3'-triiodothyronine content was normal or elevated in InsR knockout mice or Dio1 knockout mice. Knockdown of either InsR or Dio1 by siRNA in HepG2 cells decreased the expression of ApoA-I and ApoA-I synthesis and secretion. siRNA knockdown of InsR or Dio1 decreased activity of a region of the ApoA-I promoter lacking thyroid hormone response elements (region B). Electrophoretic mobility shift assay demonstrated that reduced Dio1 expression decreased the binding of nuclear proteins to region B. CONCLUSIONS: Reductions in Dio1 expression reduce the expression of ApoA-I in a 3,5,3'-triiodothyronine-/thyroid hormone response element-independent manner.


Sujet(s)
Apolipoprotéine A-I/métabolisme , Iodide peroxidase/métabolisme , Foie/enzymologie , Transduction du signal , Tri-iodothyronine/métabolisme , Animaux , Apolipoprotéine A-I/sang , Apolipoprotéine A-I/déficit , Apolipoprotéine A-I/génétique , Cholestérol HDL/sang , Régulation de l'expression des gènes , Génotype , Cellules HepG2 , Humains , Iodide peroxidase/déficit , Iodide peroxidase/génétique , Mâle , Souris de lignée C57BL , Souris knockout , Oligonucléotides antisens/génétique , Oligonucléotides antisens/métabolisme , Phénotype , Régions promotrices (génétique) , Liaison aux protéines , Interférence par ARN , ARN messager/génétique , ARN messager/métabolisme , Récepteur à l'insuline/déficit , Récepteur à l'insuline/génétique , Éléments de réponse , Transfection
11.
Sci Transl Med ; 8(323): 323ra12, 2016 Jan 27.
Article de Anglais | MEDLINE | ID: mdl-26819195

RÉSUMÉ

Mipomersen is a 20mer antisense oligonucleotide (ASO) that inhibits apolipoprotein B (apoB) synthesis; its low-density lipoprotein (LDL)-lowering effects should therefore result from reduced secretion of very-low-density lipoprotein (VLDL). We enrolled 17 healthy volunteers who received placebo injections weekly for 3 weeks followed by mipomersen weekly for 7 to 9 weeks. Stable isotopes were used after each treatment to determine fractional catabolic rates and production rates of apoB in VLDL, IDL (intermediate-density lipoprotein), and LDL, and of triglycerides in VLDL. Mipomersen significantly reduced apoB in VLDL, IDL, and LDL, which was associated with increases in fractional catabolic rates of VLDL and LDL apoB and reductions in production rates of IDL and LDL apoB. Unexpectedly, the production rates of VLDL apoB and VLDL triglycerides were unaffected. Small interfering RNA-mediated knockdown of apoB expression in human liver cells demonstrated preservation of apoB secretion across a range of apoB synthesis. Titrated ASO knockdown of apoB mRNA in chow-fed mice preserved both apoB and triglyceride secretion. In contrast, titrated ASO knockdown of apoB mRNA in high-fat-fed mice resulted in stepwise reductions in both apoB and triglyceride secretion. Mipomersen lowered all apoB lipoproteins without reducing the production rate of either VLDL apoB or triglyceride. Our human data are consistent with long-standing models of posttranscriptional and posttranslational regulation of apoB secretion and are supported by in vitro and in vivo experiments. Targeting apoB synthesis may lower levels of apoB lipoproteins without necessarily reducing VLDL secretion, thereby lowering the risk of steatosis associated with this therapeutic strategy.


Sujet(s)
Apolipoprotéine B-100/antagonistes et inhibiteurs , Foie/métabolisme , Adolescent , Adulte , Sujet âgé , Animaux , Apolipoprotéines B/génétique , Femelle , Volontaires sains , Cellules HepG2 , Humains , Lipoprotéines IDL/sang , Lipoprotéines LDL/sang , Lipoprotéines VLDL/sang , Mâle , Souris , Adulte d'âge moyen , Oligonucléotides/composition chimique , Oligonucléotides antisens/composition chimique , Petit ARN interférent/métabolisme , Triglycéride/sang , Triglycéride/métabolisme , Jeune adulte
12.
Cell Metab ; 19(2): 209-20, 2014 Feb 04.
Article de Anglais | MEDLINE | ID: mdl-24506864

RÉSUMÉ

The underlying molecular genetic basis of combined hyperlipidemia, the most common atherogenic lipid disorder, is poorly characterized. Rare, nonconservative mutations in the Wnt coreceptor, LRP6, underlie autosomal dominant atherosclerosis, combined hyperlipidemia, and fatty liver disease. Mice with LRP6(R611C) mutation similarly developed elevated plasma LDL and TG levels and fatty liver. Further investigation showed that LRP6(R611C) mutation triggers hepatic de novo lipogenesis, lipid and cholesterol biosynthesis, and apoB secretion by an Sp1-dependent activation of IGF1, AKT, and both mTORC1 and mTORC2. These pathways were normalized after in vitro treatment of primary hepatocytes from LRP6(R611C) mice with either the IGF1R antagonist PPP, rapamycin, or rmWnt3a. Strikingly, in vivo administration of rmWnt3a to LRP6(R611C) mice normalized the altered expression of enzymes of DNL and cholesterol biosynthesis, and restored plasma TG and LDL levels to normal. These findings identify Wnt signaling as a regulator of plasma lipids and a target for treatment of hyperlipidemia.


Sujet(s)
Athérosclérose/métabolisme , Hyperlipidémies/métabolisme , Protéine-6 apparentée au récepteur des LDL/métabolisme , Animaux , Cellules cultivées , Stéatose hépatique/métabolisme , Hépatocytes/métabolisme , Complexe-1 cible mécanistique de la rapamycine , Complexe-2 cible mécanistique de la rapamycine , Souris , Modèles biologiques , Complexes multiprotéiques/métabolisme , Mutation , Stéatose hépatique non alcoolique , Sérine-thréonine kinases TOR/métabolisme , Protéine Wnt3A/métabolisme
13.
J Lipid Res ; 53(11): 2364-79, 2012 Nov.
Article de Anglais | MEDLINE | ID: mdl-22911105

RÉSUMÉ

Acyl CoA:diacylglycerol acyltransferase (DGAT) 1 catalyzes the final step of triglyceride (TG) synthesis. We show that acute administration of a DGAT1 inhibitor (DGAT1i) by oral gavage or genetic deletion of intestinal Dgat1 (intestine-Dgat1(-/-)) markedly reduced postprandial plasma TG and retinyl ester excursions by inhibiting chylomicron secretion in mice. Loss of DGAT1 activity did not affect the efficiency of retinol esterification, but it did reduce TG and retinoid accumulation in the small intestine. In contrast, inhibition of microsomal triglyceride transfer protein (MTP) reduced chylomicron secretion after oral fat/retinol loads, but with accumulation of dietary TG and retinoids in the small intestine. Lack of intestinal accumulation of TG and retinoids in DGAT1i-treated or intestine-Dgat1(-/-) mice resulted, in part, from delayed gastric emptying associated with increased plasma levels of glucagon-like peptide (GLP)-1. However, neither bypassing the stomach through duodenal oil injection nor inhibiting the receptor for GLP-1 normalized postprandial TG or retinyl esters excursions in the absence of DGAT1 activity. In summary, intestinal DGAT1 inhibition or deficiency acutely delayed gastric emptying and inhibited chylomicron secretion; however, the latter occurred when gastric emptying was normal or when lipid was administered directly into the small intestine. Long-term hepatic retinoid metabolism was not impacted by DGAT1 inhibition.


Sujet(s)
Chylomicron/métabolisme , Diacylglycerol O-acyltransferase/déficit , Diacylglycerol O-acyltransferase/métabolisme , Vidange gastrique/génétique , Période post-prandiale/physiologie , Triglycéride/métabolisme , Animaux , Carbamates/pharmacologie , Protéines de transport/antagonistes et inhibiteurs , Protéines de transport/métabolisme , Diacylglycerol O-acyltransferase/antagonistes et inhibiteurs , Diacylglycerol O-acyltransferase/génétique , Vidange gastrique/effets des médicaments et des substances chimiques , Glucagon-like peptide 1/sang , Récepteur du peptide-1 similaire au glucagon , Indoles/pharmacologie , Mâle , Souris , Souches mutantes de souris , Fragments peptidiques/pharmacologie , Période post-prandiale/génétique , Récepteurs au glucagon/antagonistes et inhibiteurs , Rétinoïdes/métabolisme , Triglycéride/sang
14.
J Clin Invest ; 122(5): 1677-87, 2012 May.
Article de Anglais | MEDLINE | ID: mdl-22466652

RÉSUMÉ

Recent GWAS have identified SNPs at a human chromosom1 locus associated with coronary artery disease risk and LDL cholesterol levels. The SNPs are also associated with altered expression of hepatic sortilin-1 (SORT1), which encodes a protein thought to be involved in apoB trafficking and degradation. Here, we investigated the regulation of Sort1 expression in mouse models of obesity. Sort1 expression was markedly repressed in both genetic (ob/ob) and high-fat diet models of obesity; restoration of hepatic sortilin-1 levels resulted in reduced triglyceride and apoB secretion. Mouse models of obesity also exhibit increased hepatic activity of mammalian target of rapamycin complex 1 (mTORC1) and ER stress, and we found that administration of the mTOR inhibitor rapamycin to ob/ob mice reduced ER stress and increased hepatic sortilin-1 levels. Conversely, genetically increased hepatic mTORC1 activity was associated with repressed Sort1 and increased apoB secretion. Treating WT mice with the ER stressor tunicamycin led to marked repression of hepatic sortilin-1 expression, while administration of the chemical chaperone PBA to ob/ob mice led to amelioration of ER stress, increased sortilin-1 expression, and reduced apoB and triglyceride secretion. Moreover, the ER stress target Atf3 acted at the SORT1 promoter region as a transcriptional repressor, whereas knockdown of Atf3 mRNA in ob/ob mice led to increased hepatic sortilin-1 levels and decreased apoB and triglyceride secretion. Thus, in mouse models of obesity, induction of mTORC1 and ER stress led to repression of hepatic Sort1 and increased VLDL secretion via Atf3. This pathway may contribute to dyslipidemia in metabolic disease.


Sujet(s)
Protéines adaptatrices du transport vésiculaire/métabolisme , Stress du réticulum endoplasmique , Foie/métabolisme , Protéines/métabolisme , Facteur de transcription ATF-3/génétique , Facteur de transcription ATF-3/métabolisme , Facteur de transcription ATF-3/physiologie , Protéines adaptatrices du transport vésiculaire/génétique , Animaux , Apolipoprotéines B/sang , Apolipoprotéines B/métabolisme , Séquence nucléotidique , Sites de fixation , Alimentation riche en graisse , Régulation négative , Régulation de l'expression des gènes , Humains , Métabolisme lipidique , Lipoprotéines VLDL/sang , Lipoprotéines VLDL/métabolisme , Complexe-1 cible mécanistique de la rapamycine , Souris , Souris de lignée C57BL , Souris obèse , Complexes multiprotéiques , Obésité/métabolisme , Régions promotrices (génétique) , Protéines/antagonistes et inhibiteurs , Protéines/génétique , Sirolimus/pharmacologie , Protéine-1 de liaison à l'élément de régulation des stérols/génétique , Protéine-1 de liaison à l'élément de régulation des stérols/métabolisme , Sérine-thréonine kinases TOR , Transcription génétique , Triglycéride/sang , Triglycéride/métabolisme , Antigènes CD95/génétique , Antigènes CD95/métabolisme
15.
Metabolism ; 61(6): 823-35, 2012 Jun.
Article de Anglais | MEDLINE | ID: mdl-22209663

RÉSUMÉ

We recently reported that inhibition of 11ß-hydroxysteroid dehydrogenase 1 (11ß-HSD1) by antisense oligonucleotide (ASO) improved hepatic lipid metabolism independent of food intake. In that study, 11ß-HSD1 ASO-treated mice lost weight compared with food-matched control ASO-treated mice, suggesting treatment-mediated increased energy expenditure. We have now examined the effects of 11ß-HSD1 ASO treatment on adipose tissue metabolism, insulin sensitivity, and whole-body energy expenditure. We used an ASO to knock down 11ß-HSD1 in C57BL/6J mice consuming a Western-type diet (WTD). The 11ß-HSD1 ASO-treated mice consumed less food, so food-matched control ASO-treated mice were also evaluated. We characterized body composition, gene expression of individual adipose depots, and measures of energy metabolism. We also investigated glucose/insulin tolerance as well as acute insulin signaling in several tissues. Knockdown of 11ß-HSD1 protected against WTD-induced obesity by reducing epididymal, mesenteric, and subcutaneous white adipose tissue while activating thermogenesis in brown adipose tissue. The latter was confirmed by demonstrating increased energy expenditure in 11ß-HSD1 ASO-treated mice. The 11ß-HSD1 ASO treatment also protected against WTD-induced glucose intolerance and insulin resistance; this protection was associated with smaller cells and fewer macrophages in epididymal white adipose tissue as well as enhanced in vivo insulin signaling. Our results indicate that ASO-mediated inhibition of 11ß-HSD1 can protect against several WTD-induced metabolic abnormalities. These effects are, at least in part, mediated by increases in the oxidative capacity of brown adipose tissue.


Sujet(s)
11-beta-Hydroxysteroid dehydrogenase type 1/génétique , Tissu adipeux/métabolisme , Consommation alimentaire/physiologie , Métabolisme énergétique/physiologie , Insulinorésistance/physiologie , 11-beta-Hydroxysteroid dehydrogenase type 1/antagonistes et inhibiteurs , 11-beta-Hydroxysteroid dehydrogenase type 1/métabolisme , Animaux , Composition corporelle/génétique , Composition corporelle/physiologie , Consommation alimentaire/génétique , Métabolisme énergétique/génétique , Hyperglycémie provoquée , Insuline/métabolisme , Insulinorésistance/génétique , Mâle , Souris , Souris de lignée C57BL , Transduction du signal/génétique , Transduction du signal/physiologie
16.
Arterioscler Thromb Vasc Biol ; 32(2): 236-46, 2012 Feb.
Article de Anglais | MEDLINE | ID: mdl-22155452

RÉSUMÉ

OBJECTIVE: States of insulin resistance, hyperinsulinemia, and hepatic steatosis are associated with increased secretion of triglycerides (TG) and apolipoprotein B (apoB), even though insulin targets apoB for degradation. We used hepatic-specific "phosphatase and tensin homologue deleted on chromosome 10" (Pten) knockout (hPten-ko) mice, with increased hepatic insulin signaling, to determine the relative roles of insulin signaling and hepatic TG in regulating apoB secretion. METHODS AND RESULTS: TG and apoB secretion was elevated in hPten-ko mice. When hepatic TG was reduced by inhibition of diacylglycerol acyltransferase 1/diacylglycerol acyltransferase 2 or sterol regulatory element-binding protein-1c, both TG secretion and apoB secretion fell without changes in hepatic insulin signaling. Acute reconstitution of hPten reduced hepatic TG content, and both TG and apoB secretion fell within 4 days despite decreased hepatic insulin signaling. Acute depletion of hepatic Pten by adenoviral introduction of Cre into Pten floxed mice caused steatosis within 4 days, and secretion of both TG and apoB increased despite increased hepatic insulin signaling. Even when steatosis after acute Pten depletion was prevented by pretreatment with SREBP-1c antisense oligonucleotides, apoB secretion was not reduced after 4 days. Ex vivo results were in primary hepatocytes were similar. CONCLUSIONS: Either hepatic TG is the dominant regulator of apoB secretion or any inhibitory effects of hepatic insulin signaling on apoB secretion is very short-lived.


Sujet(s)
Apolipoprotéines B/métabolisme , Insuline/métabolisme , Foie/métabolisme , Transduction du signal/physiologie , Triglycéride/métabolisme , Animaux , Diacylglycerol O-acyltransferase/déficit , Diacylglycerol O-acyltransferase/génétique , Stéatose hépatique/métabolisme , Lipogenèse/physiologie , Mâle , Souris , Souris knockout , Modèles animaux , Phosphohydrolase PTEN/déficit , Phosphohydrolase PTEN/génétique
17.
J Lipid Res ; 52(9): 1636-51, 2011 Sep.
Article de Anglais | MEDLINE | ID: mdl-21719579

RÉSUMÉ

Although short-term incubation of hepatocytes with oleic acid (OA) stimulates secretion of apolipoprotein B100 (apoB100), exposure to higher doses of OA for longer periods inhibits secretion in association with induction of endoplasmic reticulum (ER) stress. Palmitic acid (PA) induces ER stress, but its effects on apoB100 secretion are unclear. Docosahexaenoic acid (DHA) inhibits apoB100 secretion, but its effects on ER stress have not been studied. We compared the effects of each of these fatty acids on ER stress and apoB100 secretion in McArdle RH7777 (McA) cells: OA and PA induced ER stress and inhibited apoB100 secretion at higher doses; PA was more potent because it also increased the synthesis of ceramide. DHA did not induce ER stress but was the most potent inhibitor of apoB100 secretion, acting via stimulation of autophagy. These unique effects of each fatty acid were confirmed when they were infused into C57BL6J mice. Our results suggest that when both increased hepatic secretion of VLDL apoB100 and hepatic steatosis coexist, reducing ER stress might alleviate hepatic steatosis but at the expense of increased VLDL secretion. In contrast, increasing autophagy might reduce VLDL secretion without causing steatosis.


Sujet(s)
Apolipoprotéine B-100/métabolisme , Autophagie/effets des médicaments et des substances chimiques , Céramides/métabolisme , Réticulum endoplasmique/effets des médicaments et des substances chimiques , Acides gras/pharmacologie , Stress oxydatif/effets des médicaments et des substances chimiques , Animaux , Lignée cellulaire , Réticulum endoplasmique/métabolisme , Acides gras/composition chimique , Acides gras monoinsaturés/pharmacologie , Foie/cytologie , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Mâle , Souris , Souris de lignée C57BL , Phénylbutyrates/pharmacologie , Acyl-(acyl-carrier-protein)desaturase/génétique , Acyl-(acyl-carrier-protein)desaturase/métabolisme
18.
J Lipid Res ; 52(5): 971-81, 2011 May.
Article de Anglais | MEDLINE | ID: mdl-21364201

RÉSUMÉ

11ß-hydroxysteroid dehydrogenase 1 (11ß-HSD1) converts inactive 11-keto derivatives to active glucocorticoids within tissues and may play a role in the metabolic syndrome (MS). We used an antisense oligonucleotide (ASO) to knock down 11ß-HSD1 in livers of C57BL/6J mice consuming a Western-type diet (WTD). 11ß-HSD1 ASO-treated mice consumed less food, so we compared them to ad libitum-fed mice and to food-matched mice receiving control ASO. Knockdown of 11ß-HSD1 directly protected mice from WTD-induced steatosis and dyslipidemia by reducing synthesis and secretion of triglyceride (TG) and increasing hepatic fatty acid oxidation. These changes in hepatic and plasma lipids were not associated with reductions in genes involved in de novo lipogenesis. However, protein levels of both sterol regulatory element-binding protein (SREBP) 1 and fatty acid synthase were significantly reduced in mice treated with 11ß-HSD1 ASO. There was no change in hepatic secretion of apolipoprotein (apo)B, indicating assembly and secretion of smaller apoB-containing lipoproteins by the liver in the 11ß-HSD1-treated mice. Our results indicate that inhibition of 11ß-HSD1 by ASO treatment of WTD-fed mice resulted in improved plasma and hepatic lipid levels, reduced lipogenesis by posttranslational regulation, and secretion of similar numbers of apoB-containing lipoproteins containing less TG per particle.


Sujet(s)
11-beta-Hydroxysteroid dehydrogenase type 1/métabolisme , Oligonucléotides antisens/pharmacologie , 11-beta-Hydroxysteroid dehydrogenase type 1/antagonistes et inhibiteurs , Animaux , Apolipoprotéines B/métabolisme , Poids/effets des médicaments et des substances chimiques , Cellules cultivées , Chromatographie en phase liquide , Matières grasses alimentaires/effets indésirables , Dyslipidémies/induit chimiquement , Dyslipidémies/prévention et contrôle , Consommation alimentaire/effets des médicaments et des substances chimiques , Stéatose hépatique/induit chimiquement , Stéatose hépatique/prévention et contrôle , Hépatocytes/effets des médicaments et des substances chimiques , Hépatocytes/métabolisme , Immunotransfert , Métabolisme lipidique/effets des médicaments et des substances chimiques , Lipoprotéines VLDL/métabolisme , Foie/effets des médicaments et des substances chimiques , Foie/métabolisme , Mâle , Souris , Souris de lignée C57BL , Réaction de polymérisation en chaîne , Triglycéride/métabolisme
19.
Cell Metab ; 7(2): 125-34, 2008 Feb.
Article de Anglais | MEDLINE | ID: mdl-18249172

RÉSUMÉ

Insulin resistance plays a central role in the development of the metabolic syndrome, but how it relates to cardiovascular disease remains controversial. Liver insulin receptor knockout (LIRKO) mice have pure hepatic insulin resistance. On a standard chow diet, LIRKO mice have a proatherogenic lipoprotein profile with reduced high-density lipoprotein (HDL) cholesterol and very low-density lipoprotein (VLDL) particles that are markedly enriched in cholesterol. This is due to increased secretion and decreased clearance of apolipoprotein B-containing lipoproteins, coupled with decreased triglyceride secretion secondary to increased expression of Pgc-1 beta (Ppargc-1b), which promotes VLDL secretion, but decreased expression of Srebp-1c (Srebf1), Srebp-2 (Srebf2), and their targets, the lipogenic enzymes and the LDL receptor. Within 12 weeks on an atherogenic diet, LIRKO mice show marked hypercholesterolemia, and 100% of LIRKO mice, but 0% of controls, develop severe atherosclerosis. Thus, insulin resistance at the level of the liver is sufficient to produce the dyslipidemia and increased risk of atherosclerosis associated with the metabolic syndrome.


Sujet(s)
Athérosclérose/étiologie , Dyslipidémies/étiologie , Insulinorésistance , Animaux , Prédisposition aux maladies , Hypercholestérolémie/étiologie , Lipoprotéines/sang , Maladies du foie , Souris , Souris knockout , Récepteur à l'insuline/déficit
20.
Metabolism ; 56(11): 1566-75, 2007 Nov.
Article de Anglais | MEDLINE | ID: mdl-17950109

RÉSUMÉ

To investigate the effect of dietary 1,3-diacylglycerol (DAG) on the development of insulin resistance (IR) and obesity, brown adipose tissue-deficient mice, a model of high-fat diet-induced IR and obesity, were fed Western-type diets (WTD) containing either DAG oil (n = 8) or standard triacylglycerol (TAG) oil (n = 9) for 15 weeks, beginning at 8 weeks of age. Although brown adipose tissue-deficient mice became obese on both TAG- and DAG-enriched WTD (TAG-WTD and DAG-WTD), the mice eating DAG-WTD gained less weight and had less body fat accumulation. The results of glucose tolerance tests conducted after 5 weeks of each WTD were not different. However, after 10 weeks of each WTD, impaired glucose tolerance developed in the TAG-WTD group but was prevented by DAG-WTD. Exploratory analyses of gene expression suggested that consumption of DAG-WTD was associated with reduced phosphoenolpyruvate carboxykinase gene expression in liver and increased expression of the genes for peroxisome proliferator-activated receptor alpha, lipoprotein lipase, and uncoupling proteins 2 and 3 in skeletal muscle. There were no effects of the DAG-WTD on fasting and postprandial plasma triglyceride (TG) levels, hepatic TG content, or the rate of secretion of TG from the liver. These findings suggest that diets enriched in 1,3-DAG oil may reduce WTD-induced IR and body fat accumulation by suppressing gluconeogenesis in liver and stimulating fat oxidation in skeletal muscle.


Sujet(s)
Régime alimentaire , Diglycéride/pharmacologie , Insulinorésistance , Obésité/prévention et contrôle , Animaux , Séquence nucléotidique , Amorces ADN , Diglycéride/administration et posologie , Hyperglycémie provoquée , Souris , Réaction de polymérisation en chaîne
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...