Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Front Physiol ; 15: 1371096, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38694206

RESUMO

Introduction: The Aster-C protein (encoded by the Gramd1c gene) is an endoplasmic reticulum (ER) resident protein that has been reported to transport cholesterol from the plasma membrane to the ER. Although there is a clear role for the closely-related Aster-B protein in cholesterol transport and downstream esterification in the adrenal gland, the specific role for Aster-C in cholesterol homeostasis is not well understood. Here, we have examined whole body cholesterol balance in mice globally lacking Aster-C under low or high dietary cholesterol conditions. Method: Age-matched Gramd1c +/+ and Gramd1c -/- mice were fed either low (0.02%, wt/wt) or high (0.2%, wt/wt) dietarycholesterol and levels of sterol-derived metabolites were assessed in the feces, liver, and plasma. Results: Compared to wild type controls (Gramd1c +/+) mice, mice lackingGramd1c (Gramd1c -/-) have no significant alterations in fecal, liver, or plasma cholesterol. Given the potential role for Aster C in modulating cholesterol metabolism in diverse tissues, we quantified levels of cholesterol metabolites such as bile acids, oxysterols, and steroid hormones. Compared to Gramd1c +/+ controls, Gramd1c -/- mice had modestly reduced levels of select bile acid species and elevated cortisol levels, only under low dietary cholesterol conditions. However, the vast majority of bile acids, oxysterols, and steroid hormones were unaltered in Gramd1c -/- mice. Bulk RNA sequencing in the liver showed that Gramd1c -/- mice did not exhibit alterations in sterol-sensitive genes, but instead showed altered expression of genes in major urinary protein and cytochrome P450 (CYP) families only under low dietary cholesterol conditions. Discussion: Collectively, these data indicate nominal effects of Aster-C on whole body cholesterol transport and metabolism under divergent dietary cholesterol conditions. These results strongly suggest that Aster-C alone is not sufficient to control whole body cholesterol balance, but can modestly impact circulating cortisol and bile acid levels when dietary cholesterol is limited.

2.
J Lipid Res ; 63(2): 100166, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35016907

RESUMO

Apolipoprotein F (ApoF) modulates lipoprotein metabolism by selectively inhibiting cholesteryl ester transfer protein activity on LDL. This ApoF activity requires that it is bound to LDL. How hyperlipidemia alters total plasma ApoF and its binding to LDL are poorly understood. In this study, total plasma ApoF and LDL-bound ApoF were quantified by ELISA (n = 200). Plasma ApoF was increased 31% in hypercholesterolemic plasma but decreased 20% in hypertriglyceridemia. However, in donors with combined hypercholesterolemia and hypertriglyceridemia, the elevated triglyceride ameliorated the rise in ApoF caused by hypercholesterolemia alone. Compared with normolipidemic LDL, hypercholesterolemic LDL contained ∼2-fold more ApoF per LDL particle, whereas ApoF bound to LDL in hypertriglyceridemia plasma was <20% of control. To understand the basis for altered association of ApoF with hyperlipidemic LDL, the physiochemical properties of LDL were modified in vitro by cholesteryl ester transfer protein ± LCAT activities. The time-dependent change in LDL lipid composition, proteome, core and surface lipid packing, LDL surface charge, and LDL size caused by these factors were compared with the ApoF binding capacity of these LDLs. Only LDL particle size correlated with ApoF binding capacity. This positive association between LDL size and ApoF content was confirmed in hyperlipidemic plasmas. Similarly, when in vitro produced and enlarged LDLs with elevated ApoF binding capacity were incubated with LPL to reduce their size, ApoF binding was reduced by 90%. Thus, plasma ApoF levels and the activation status of this ApoF are differentially altered by hypercholesterolemia and hypertriglyceridemia. LDL size is a key determinate of ApoF binding and activation.


Assuntos
Apolipoproteínas
3.
Lipids ; 57(1): 69-79, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34866179

RESUMO

We previously reported that overexpression of full-length cholesteryl ester transfer protein (FL-CETP), but not its exon 9-deleted variant (∆E9-CETP), in an adipose cell line reduces their triacylglycerol (TAG) content. This provided mechanistic insight into several in vivo studies where FL-CETP levels are inversely correlated with adiposity. However, increased FL-CETP is also associated with elevated hepatic lipids, suggesting that the effect of CETP on cellular lipid metabolism may be tissue-specific. Here, we directly investigated the role of FL-CETP and ∆E9-CETP in hepatic lipid metabolism. FL- or ∆E9-CETP was overexpressed in HepG2-C3A by adenovirus transduction. Overexpression of either FL or ∆E9-CETP in hepatocytes increased cellular TAG mass by 25% but reduced TAG secretion. This cellular TAG was contained in larger and more numerous lipid droplets. Analysis of TAG synthetic and catabolic pathways showed that this elevated TAG content was due to increased incorporation of fatty acid into TAG (24%), and higher de novo synthesis of fatty acid (50%) and TAG from acetate (40%). siRNA knockdown of CETP had the opposite effect on TAG synthesis and lipogenesis, and decreased cellular TAG. This novel increase in cellular TAG by FL-CETP overexpression was reproduced in Caco-2 intestinal epithelial cells. We conclude that, unlike that seen in adipocyte cells, overexpression of either CETP isoform in lipoprotein-secreting cells promotes the accumulation of TAG. These data suggest that the in vivo correlation between CETP levels and hepatic steatosis can be explained, in part, by a direct effect of CETP on hepatocyte cellular metabolism.


Assuntos
Proteínas de Transferência de Ésteres de Colesterol , Hepatócitos , Células CACO-2 , Proteínas de Transferência de Ésteres de Colesterol/genética , Ésteres do Colesterol , Éxons , Células Hep G2 , Humanos , Triglicerídeos
4.
J Lipid Res ; 62: 100027, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33515552

RESUMO

Cholesteryl ester transfer protein (CETP) modulates lipoprotein metabolism by transferring cholesteryl ester (CE) and triglyceride (TG) between lipoproteins. However, differences in the way CETP functions exist across species. Unlike human CETP, hamster CETP prefers TG over CE as a substrate, raising questions regarding how substrate preference may impact lipoprotein metabolism. To understand how altering the CE versus TG substrate specificity of CETP might impact lipoprotein metabolism in humans, we modified CETP expression in fat/cholesterol-fed hamsters, which have a human-like lipoprotein profile. Hamsters received adenoviruses expressing no CETP, hamster CETP, or human CETP. Total plasma CETP mass increased up to 70% in the hamster and human CETP groups. Hamsters expressing human CETP exhibited decreased endogenous hamster CETP, resulting in an overall CE:TG preference of plasma CETP that was similar to that in humans. Hamster CETP overexpression had little impact on lipoproteins, whereas human CETP expression reduced HDL by 60% without affecting LDL. HDLs were TG enriched and CE depleted and much smaller, causing the HDL3:HDL2 ratio to increase threefold. HDL from hamsters expressing human CETP supported higher LCAT activity and greater cholesterol efflux. The fecal excretion of HDL-associated CE in human CETP animals was unchanged. However, much of this cholesterol accumulated in the liver and was associated with a 1.8-fold increase in hepatic cholesterol mass. Overall, these data show in a human-like lipoprotein model that modification of CETP's lipid substrate preference selectively alters HDL concentration and function. This provides a powerful tool for modulating HDL metabolism and impacting sterol balance in vivo.


Assuntos
Proteínas de Transferência de Ésteres de Colesterol
5.
J Lipid Res ; 61(8): 1168-1179, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32591337

RESUMO

Cholesteryl ester transfer protein (CETP) facilitates the net transfer of cholesteryl esters (CEs) and TGs between lipoproteins, impacting the metabolic fate of these lipoproteins. Previous studies have shown that a CETP antibody can alter CETP's preference for CE versus TG as transfer substrate, suggesting that CETP substrate preference can be manipulated in vivo. Hamster and human CETPs have very different preferences for CE and TG. To assess the effect of altering CETP's substrate preference on lipoproteins in vivo, here, we expressed human CETP in hamsters. Chow-fed hamsters received adenoviruses expressing no CETP, hamster CETP, or human CETP. Plasma CETP mass increased 2-fold in both the hamster and human CETP groups. Although the animals expressing human CETP still had low levels of hamster CETP, the CE versus TG preference of their plasma CETP was similar to that of the human ortholog. Hamster CETP overexpression had little impact on lipoproteins. However, expression of human CETP reduced HDL up to 50% and increased VLDL cholesterol 2.5-fold. LDL contained 20% more CE, whereas HDL CE was reduced 40%, and TG increased 6-fold. The HDL3:HDL2 ratio increased from 0.32 to 0.60. Hepatic expression of three cholesterol-related genes (LDLR, SCARB1, and CYP7A1) was reduced up to 40%. However, HDL-associated CE excretion into feces was unchanged. We conclude that expression of human CETP in hamsters humanizes their lipoprotein profile with respect to the relative concentrations of VLDL, LDL, HDL, and the HDL3:HDL2 ratio. Altering the lipid substrate preference of CETP provides a novel approach for modifying plasma lipoproteins.


Assuntos
Proteínas de Transferência de Ésteres de Colesterol/metabolismo , Lipoproteínas/sangue , Lipoproteínas/química , Animais , Proteínas de Transferência de Ésteres de Colesterol/genética , Cricetinae , Humanos , Fígado/metabolismo
6.
Curr Opin Lipidol ; 31(4): 194-199, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32520778

RESUMO

PURPOSE OF REVIEW: The aim of this study is to highlight recent studies that have advanced our understanding of apolipoprotein F (ApoF) and its role in lipid metabolism. RECENT FINDINGS: Previous studies showed that ApoF hepatic mRNA levels are suppressed by fat-enriched diets. Recent studies show this downregulation is mediated by agonist-induced binding of liver X receptor (LXR) and PPARalpha to a regulatory element in the ApoF promoter. First-of-kind in-vivo studies show ApoF lowers low-density lipoprotein levels and enhances reverse cholesterol transport in fat-fed hamsters. SUMMARY: Diverse studies collectively provide compelling evidence that cholesteryl ester transfer protein (CETP) plays an important role in regulating lipid metabolism. Inhibiting CETP raises HDL cholesterol. However, considering the recent failures of pharmacological inhibitors of CETP in clinical trials, it does not seem likely that global inhibition of CETP will be beneficial. ApoF is a minor apolipoprotein that functions as a natural inhibitor of CETP. However, ApoF is not a general inhibitor of CETP, but rather it preferentially inhibits CETP activity with LDL. Therefore, ApoF tailors CETP activity so that less tissue-derived cholesterol traffics from HDL into the LDL compartment. Lower LDL cholesterol levels have recognized clinical benefit for reduced cardiovascular disease.


Assuntos
Apolipoproteínas/metabolismo , Proteínas de Transferência de Ésteres de Colesterol/antagonistas & inibidores , Animais , Doenças Cardiovasculares/metabolismo , Proteínas de Transferência de Ésteres de Colesterol/metabolismo , Humanos
7.
J Lipid Res ; 61(3): 422-431, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31988147

RESUMO

Cholesteryl ester transfer protein (CETP) exists as full-length (FL) and exon 9 (E9)-deleted isoforms. The function of E9-deleted CETP is poorly understood. Here, we investigated the role of E9-deleted CETP in regulating the secretion of FL-CETP by cells and explored its possible role in intracellular lipid metabolism. CETP overexpression in cells that naturally express CETP confirmed that E9-deleted CETP is not secreted, and showed that cellular FL- and E9-deleted CETP form an isolatable complex. Coexpression of CETP isoforms lowered cellular levels of both proteins and impaired FL-CETP secretion. These effects were due to reduced synthesis of both isoforms; however, the predominate consequence of FL- and E9-deleted CETP coexpression is impaired FL-CETP synthesis. We reported previously that reducing both CETP isoforms or overexpressing FL-CETP impairs cellular triglyceride (TG) storage. To investigate this further, E9-deleted CETP was expressed in SW872 cells that naturally synthesize CETP and in mouse 3T3-L1 cells that do not. E9-deleted CETP overexpression stimulated SW872 triglyceride synthesis and increased stored TG 2-fold. Expression of E9-deleted CETP in mouse 3T3-L1 cells produced a similar lipid phenotype. In vitro, FL-CETP promotes the transfer of TG from ER-enriched membranes to lipid droplets. E9-deleted CETP also promoted this transfer, although less effectively, and it inhibited the transfer driven by FL-CETP. We conclude that FL- and E9-deleted CETP isoforms interact to mutually decrease their intracellular levels and impair FL-CETP secretion by reducing CETP biosynthesis. E9-deleted CETP, like FL-CETP, alters cellular TG metabolism and storage but in a contrary manner.


Assuntos
Proteínas de Transferência de Ésteres de Colesterol/biossíntese , Triglicerídeos/metabolismo , Células 3T3-L1 , Animais , Células Cultivadas , Proteínas de Transferência de Ésteres de Colesterol/genética , Éxons , Humanos , Camundongos
8.
Artigo em Inglês | MEDLINE | ID: mdl-31812787

RESUMO

Apolipoprotein F (ApoF) regulates cholesteryl ester transfer protein activity. We previously observed that hepatic APOF mRNA levels are decreased by high fat, cholesterol-enriched diets. Here we show in human liver C3A cells that APOF mRNA levels are reduced by agonists of LXR and PPARα nuclear receptors. This negative regulation requires co-incubation with the RXR agonist, retinoic acid. Bioinformatic analysis of the ~2 kb sequence upstream of the APOF promoter identified one potential LXR and 4 potential PPARα binding sites clustered between nucleotides -2007 and -1961. ChIP analysis confirmed agonist-dependent binding of LXRα, PPARα, and RXRα to this hormone response element complex (HREc). A luciferase reporter containing the 2 kb 5' APOF sequence was negatively regulated by LXR and PPARα ligands as seen in cells. This regulation was maintained in constructs lacking the ~1700 nucleotides between the HREc and the APOF proximal promoter. Mutations of the HREc that disrupted LXRα and PPARα binding led to the loss of reporter construct inhibition by agonists of these nuclear receptors. siRNA knockdown studies showed that APOF gene regulation by LXRα or PPARα agonists did not require an interaction between these two nuclear receptors. Thus, APOF is subject to negative regulation by agonist-activated LXR or PPARα nuclear receptors binding to a regulatory element ~1900 bases 5' to the APOF promoter. High fat, cholesterol-enriched diets likely reduce APOF gene expression via these receptors interacting at this regulatory site.


Assuntos
Apolipoproteínas/genética , Receptores X do Fígado/metabolismo , PPAR alfa/metabolismo , Elementos de Resposta , Apolipoproteínas/metabolismo , Colesterol/farmacologia , Células HEK293 , Células Hep G2 , Humanos , Receptores X do Fígado/agonistas , PPAR alfa/agonistas , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Tretinoína/farmacologia
9.
J Lipid Res ; 60(11): 1868-1879, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31511396

RESUMO

Cholesteryl ester transfer protein (CETP) regulates intravascular lipoprotein metabolism. In vitro studies indicate that ApoF alters CETP function by inhibiting its activity with LDL. To explore in vivo the complexities driving ApoF's effects on CETP, we developed a siRNA-based hamster model of ApoF knockdown. In both male and female hamsters on chow- or fat-fed diets, we measured lipoprotein levels and composition, determined CETP-mediated transfer of cholesteryl esters (CEs) between lipoproteins, and quantified reverse cholesterol transport (RCT). We found that apoF knockdown in chow-fed hamsters had no effect on lipoprotein levels or composition, but these ApoF-deficient lipoproteins supported 50-100% higher LDL CETP activity in vitro. ApoF knockdown in fat-fed male hamsters created a phenotype in which endogenous CETP-mediated CE transfer from HDL to LDL increased up to 2-fold, LDL cholesterol increased 40%, HDL declined 25%, LDL and HDL lipid compositions were altered, and hepatic LDLR gene expression was decreased. Diet-induced hypercholesterolemia obscured this phenotype on occasion. In fat-fed female hamsters, ApoF knockdown caused similar but smaller changes in plasma CETP activity and LDL cholesterol. Notably, ApoF knockdown impaired HDL RCT in fat-fed hamsters but increased sterol excretion in chow-fed animals. These in vivo data validate in vitro findings that ApoF regulates lipid transfer to LDL. The consequences of ApoF knockdown on lipoproteins and sterol excretion depend on the underlying lipid status. By minimizing the transfer of HDL-derived CE to LDL, ApoF helps control LDL cholesterol levels when LDL clearance mechanisms are limiting.


Assuntos
Apoproteínas/deficiência , Apoproteínas/genética , Ésteres do Colesterol/metabolismo , LDL-Colesterol/metabolismo , Dieta , Técnicas de Silenciamento de Genes , Animais , Proteínas de Transferência de Ésteres de Colesterol/metabolismo , Cricetinae , Feminino , Regulação da Expressão Gênica/genética , Células HEK293 , Humanos , Fígado/metabolismo , Masculino , RNA Interferente Pequeno/genética
10.
Arterioscler Thromb Vasc Biol ; 38(1): 218-231, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29074585

RESUMO

OBJECTIVE: Human genetic variants near the FADS (fatty acid desaturase) gene cluster (FADS1-2-3) are strongly associated with cardiometabolic traits including dyslipidemia, fatty liver, type 2 diabetes mellitus, and coronary artery disease. However, mechanisms underlying these genetic associations are unclear. APPROACH AND RESULTS: Here, we specifically investigated the physiological role of the Δ-5 desaturase FADS1 in regulating diet-induced cardiometabolic phenotypes by treating hyperlipidemic LDLR (low-density lipoprotein receptor)-null mice with antisense oligonucleotides targeting the selective knockdown of Fads1. Fads1 knockdown resulted in striking reorganization of both ω-6 and ω-3 polyunsaturated fatty acid levels and their associated proinflammatory and proresolving lipid mediators in a highly diet-specific manner. Loss of Fads1 activity promoted hepatic inflammation and atherosclerosis, yet was associated with suppression of hepatic lipogenesis. Fads1 knockdown in isolated macrophages promoted classic M1 activation, whereas suppressing alternative M2 activation programs, and also altered systemic and tissue inflammatory responses in vivo. Finally, the ability of Fads1 to reciprocally regulate lipogenesis and inflammation may rely in part on its role as an effector of liver X receptor signaling. CONCLUSIONS: These results position Fads1 as an underappreciated regulator of inflammation initiation and resolution, and suggest that endogenously synthesized arachidonic acid and eicosapentaenoic acid are key determinates of inflammatory disease progression and liver X receptor signaling.


Assuntos
Aorta/enzimologia , Doenças da Aorta/enzimologia , Aterosclerose/enzimologia , Dislipidemias/enzimologia , Ácidos Graxos Dessaturases/metabolismo , Mediadores da Inflamação/metabolismo , Inflamação/enzimologia , Lipogênese , Animais , Aorta/patologia , Doenças da Aorta/genética , Doenças da Aorta/patologia , Ácido Araquidônico/metabolismo , Aterosclerose/genética , Aterosclerose/patologia , Células Cultivadas , Dessaturase de Ácido Graxo Delta-5 , Modelos Animais de Doenças , Dislipidemias/genética , Dislipidemias/patologia , Ácido Eicosapentaenoico/metabolismo , Ácidos Graxos Dessaturases/genética , Inflamação/genética , Inflamação/patologia , Fígado/metabolismo , Receptores X do Fígado/metabolismo , Ativação de Macrófagos , Macrófagos Peritoneais/enzimologia , Macrófagos Peritoneais/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/metabolismo , Placa Aterosclerótica , Receptores de LDL/deficiência , Receptores de LDL/genética
12.
Cell Rep ; 19(12): 2451-2461, 2017 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-28636934

RESUMO

Emerging evidence suggests that microbes resident in the human intestine represent a key environmental factor contributing to obesity-associated disorders. Here, we demonstrate that the gut microbiota-initiated trimethylamine N-oxide (TMAO)-generating pathway is linked to obesity and energy metabolism. In multiple clinical cohorts, systemic levels of TMAO were observed to strongly associate with type 2 diabetes. In addition, circulating TMAO levels were associated with obesity traits in the different inbred strains represented in the Hybrid Mouse Diversity Panel. Further, antisense oligonucleotide-mediated knockdown or genetic deletion of the TMAO-producing enzyme flavin-containing monooxygenase 3 (FMO3) conferred protection against obesity in mice. Complimentary mouse and human studies indicate a negative regulatory role for FMO3 in the beiging of white adipose tissue. Collectively, our studies reveal a link between the TMAO-producing enzyme FMO3 and obesity and the beiging of white adipose tissue.


Assuntos
Metilaminas/sangue , Obesidade/enzimologia , Oxigenases/fisiologia , Gordura Subcutânea/enzimologia , Adipócitos Bege/enzimologia , Animais , Diabetes Mellitus Tipo 2/sangue , Feminino , Expressão Gênica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Obesidade/sangue , Obesidade/patologia , Gordura Subcutânea/patologia , Gordura Subcutânea/fisiopatologia
13.
J Lipid Res ; 56(9): 1669-78, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26203075

RESUMO

We previously reported that reducing the expression of cholesteryl ester transfer protein (CETP) disrupts cholesterol homeostasis in SW872 cells and causes an ∼50% reduction in TG. The causes of this reduced TG content, investigated here, could not be attributed to changes in the differentiation status of CETP-deficient cells, nor was there evidence of endoplasmic reticulum (ER) stress. In short-term studies, the total flux of oleate through the TG biosynthetic pathway was not altered in CETP-deficient cells, although mRNA levels of some pathway enzymes were different. However, the conversion of diglyceride (DG) to TG was impaired. In longer-term studies, newly synthesized TG was not effectively transported to lipid droplets, yet this lipid did not accumulate in the ER, apparently due to elevated lipase activity in this organelle. DG, shown to be a novel CETP substrate, was also inefficiently transferred to lipid droplets. This may reduce TG synthesis on droplets by resident diacylglycerol acyltransferase. Overall, these data suggest that the decreased TG content of CETP-deficient cells arises from the reduced conversion of DG to TG in the ER and/or on the lipid droplet surface, and enhanced TG degradation in the ER due to its ineffective transport from this organelle.


Assuntos
Proteínas de Transferência de Ésteres de Colesterol/metabolismo , Colesterol/metabolismo , Retículo Endoplasmático/metabolismo , Triglicerídeos/biossíntese , Linhagem Celular , Colesterol/biossíntese , Proteínas de Transferência de Ésteres de Colesterol/deficiência , Proteínas de Transferência de Ésteres de Colesterol/genética , Diglicerídeos/metabolismo , Estresse do Retículo Endoplasmático/genética , Humanos , Gotículas Lipídicas/metabolismo , Erros Inatos do Metabolismo Lipídico/genética , Erros Inatos do Metabolismo Lipídico/metabolismo , RNA Mensageiro/biossíntese , Triglicerídeos/metabolismo
14.
PLoS One ; 10(5): e0125126, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25938460

RESUMO

There is strong epidemiological association between periodontal disease and cardiovascular disease but underlying mechanisms remain ill-defined. Because the human periodontal disease pathogen, Porphyromonas gingivalis (Pg), interacts with innate immune receptors Toll-like Receptor (TLR) 2 and CD36/scavenger receptor-B2 (SR-B2), we studied how CD36/SR-B2 and TLR pathways promote Pg-mediated atherosclerosis. Western diet fed low density lipoprotein receptor knockout (Ldlr°) mice infected orally with Pg had a significant increase in lesion burden compared with uninfected controls.This increase was entirely CD36/SR-B2-dependent, as there was no significant change in lesion burden between infected and uninfected Cd36o/Ldlro mice [corrected]. Western diet feeding promoted enhanced CD36/SR-B2-dependent IL1ß generation and foam cell formation as a result of Pg lipopolysaccharide (PgLPS) exposure. CD36/SR-B2 and TLR2 were necessary for inflammasome activation and optimal IL1ß generation, but also resulted in LPS induced lethality (pyroptosis). Modified forms of LDL inhibited Pg-mediated IL1ß generation in a CD36/SR-B2-dependent manner and prevented pyroptosis, but promoted foam cell formation. Our data show that Pg infection in the oral cavity can lead to significant TLR2-CD36/SR-B2 dependent IL1ß release. In the vessel wall, macrophages encountering systemic release of IL1ß, PgLPS and modified LDL have increased lipid uptake, foam cell formation, and release of IL1ß, but because pyroptosis is inhibited, this enables macrophage survival and promotes increased plaque development. These studies may explain increased lesion burden as a result of periodontal disease, and suggest strategies for development of therapeutics.


Assuntos
Aterosclerose/complicações , Aterosclerose/microbiologia , Antígenos CD36/metabolismo , Porphyromonas gingivalis/fisiologia , Receptores de LDL/deficiência , Receptor 2 Toll-Like/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Apoptose/efeitos dos fármacos , Aterosclerose/sangue , Infecções por Bacteroidaceae/sangue , Infecções por Bacteroidaceae/complicações , Infecções por Bacteroidaceae/metabolismo , Infecções por Bacteroidaceae/patologia , Peso Corporal/efeitos dos fármacos , Proteínas de Transporte/metabolismo , Modelos Animais de Doenças , Comportamento Alimentar , Feminino , Células Espumosas/metabolismo , Inflamassomos/metabolismo , Interferon gama/sangue , Interleucina-1beta/metabolismo , Interleucina-6/sangue , Lipopolissacarídeos/farmacologia , Lipoproteínas LDL/farmacologia , Masculino , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR , Porphyromonas gingivalis/efeitos dos fármacos , Receptores de LDL/metabolismo , Fatores de Risco , Seio Aórtico/efeitos dos fármacos , Seio Aórtico/microbiologia , Seio Aórtico/patologia
15.
J Lipid Res ; 56(3): 515-525, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25593327

RESUMO

Cells produce two cholesteryl ester transfer protein (CETP) isoforms, full-length and a shorter variant produced by alternative splicing. Blocking synthesis of both isoforms disrupts lipid metabolism and storage. To further define the role of CETP in cellular lipid metabolism, we stably overexpressed full-length CETP in SW872 cells. These CETP(+) cells had several-fold higher intracellular CETP and accumulated 50% less TG due to a 26% decrease in TG synthesis and 2.5-fold higher TG turnover rate. Reduced TG synthesis was due to decreased fatty acid uptake and impaired conversion of diglyceride to TG even though diacylglycerol acyltransferase activity was normal. Sterol-regulatory element binding protein 1 mRNA levels were normal, and although PPARγ expression was reduced, the expression of several of its target genes including adipocyte triglyceride lipase, FASN, and APOE was normal. CETP(+) cells contained smaller lipid droplets, consistent with their higher levels of perilipin protein family (PLIN) 3 compared with PLIN1 and PLIN2. Intracellular CETP was mostly associated with the endoplasmic reticulum, although CETP near lipid droplets poorly colocalized with this membrane. A small pool of CETP resided in the cytoplasm, and a subfraction coisolated with lipid droplets. These data show that overexpression of full-length CETP disrupts lipid homeostasis resulting in the formation of smaller, more metabolically active lipid droplets.


Assuntos
Proteínas de Transferência de Ésteres de Colesterol/metabolismo , Citoplasma/metabolismo , Metabolismo dos Lipídeos/fisiologia , Triglicerídeos/metabolismo , Apolipoproteínas E/biossíntese , Apolipoproteínas E/genética , Linhagem Celular Tumoral , Proteínas de Transferência de Ésteres de Colesterol/genética , Citoplasma/genética , Ácido Graxo Sintase Tipo I/genética , Ácido Graxo Sintase Tipo I/metabolismo , Regulação da Expressão Gênica/fisiologia , Humanos , PPAR gama/biossíntese , PPAR gama/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Triglicerídeos/genética
16.
J Lipid Res ; 56(3): 612-619, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25616437

RESUMO

We previously determined that hamster cholesteryl ester transfer protein (CETP), unlike human CETP, promotes a novel one-way transfer of TG from VLDL to HDL, causing HDL to gain lipid. We hypothesize that this nonreciprocal lipid transfer activity arises from the usually high TG/cholesteryl ester (CE) substrate preference of hamster CETP. Consistent with this, we report here that ∼25% of the total lipid transfer promoted by the human Q199A CETP mutant, which prefers TG as substrate, is nonreciprocal transfer. Other human CETP mutants with TG/CE substrate preferences higher or lower than wild-type also possess nonreciprocal lipid transfer activity. Mutants with high TG/CE substrate preference promote the nonreciprocal lipid transfer of TG from VLDL to HDL, but mutants with low TG/CE substrate preference promote the nonreciprocal lipid transfer of CE, not TG, and this lipid flow is in the reverse direction (from HDL to VLDL). Anti-CETP TP2 antibody alters the TG/CE substrate preference of CETP and also changes the extent of nonreciprocal lipid transfer, showing the potential for externally acting agents to modify the transfer properties of CETP. Overall, these data show that the lipid transfer properties of CETP can be manipulated. Function-altering pharmaceuticals may offer a novel approach to modify CETP activity and achieve specific modifications in lipoprotein metabolism.


Assuntos
Proteínas de Transferência de Ésteres de Colesterol/química , Ésteres do Colesterol/química , Desenho de Fármacos , Triglicerídeos/química , Substituição de Aminoácidos , Animais , Proteínas de Transferência de Ésteres de Colesterol/antagonistas & inibidores , Proteínas de Transferência de Ésteres de Colesterol/metabolismo , Ésteres do Colesterol/genética , Ésteres do Colesterol/metabolismo , Cricetinae , Humanos , Lipoproteínas HDL/química , Lipoproteínas HDL/genética , Lipoproteínas HDL/metabolismo , Lipoproteínas VLDL/química , Lipoproteínas VLDL/genética , Lipoproteínas VLDL/metabolismo , Mutação de Sentido Incorreto , Relação Estrutura-Atividade , Especificidade por Substrato , Triglicerídeos/genética , Triglicerídeos/metabolismo
17.
J Exp Med ; 211(5): 887-907, 2014 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-24752299

RESUMO

Low-grade systemic inflammation is often associated with metabolic syndrome, which plays a critical role in the development of the obesity-associated inflammatory diseases, including insulin resistance and atherosclerosis. Here, we investigate how Toll-like receptor-MyD88 signaling in myeloid and endothelial cells coordinately participates in the initiation and progression of high fat diet-induced systemic inflammation and metabolic inflammatory diseases. MyD88 deficiency in myeloid cells inhibits macrophage recruitment to adipose tissue and their switch to an M1-like phenotype. This is accompanied by substantially reduced diet-induced systemic inflammation, insulin resistance, and atherosclerosis. MyD88 deficiency in endothelial cells results in a moderate reduction in diet-induced adipose macrophage infiltration and M1 polarization, selective insulin sensitivity in adipose tissue, and amelioration of spontaneous atherosclerosis. Both in vivo and ex vivo studies suggest that MyD88-dependent GM-CSF production from the endothelial cells might play a critical role in the initiation of obesity-associated inflammation and development of atherosclerosis by priming the monocytes in the adipose and arterial tissues to differentiate into M1-like inflammatory macrophages. Collectively, these results implicate a critical MyD88-dependent interplay between myeloid and endothelial cells in the initiation and progression of obesity-associated inflammatory diseases.


Assuntos
Células Endoteliais/metabolismo , Inflamação/metabolismo , Inflamação/fisiopatologia , Insulina/metabolismo , Células Mieloides/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Obesidade/complicações , Análise de Variância , Animais , Antígeno CD11b/metabolismo , Citometria de Fluxo , Imuno-Histoquímica , Inflamação/etiologia , Camundongos , Reação em Cadeia da Polimerase em Tempo Real
18.
J Lipid Res ; 55(2): 258-65, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24293641

RESUMO

Site-specific changes in the amino acid composition of human cholesteryl ester transfer protein (CETP) modify its preference for triglyceride (TG) versus cholesteryl ester (CE) as substrate. CETP homologs are found in many species but little is known about their activity. Here, we examined the lipid transfer properties of CETP species with 80-96% amino acid identity to human CETP. TG/CE transfer ratios for recombinant rabbit, monkey, and hamster CETPs were 1.40-, 1.44-, and 6.08-fold higher than human CETP, respectively. In transfer assays between VLDL and HDL, net transfers of CE into VLDL by human and monkey CETPs were offset by equimolar net transfers of TG toward HDL. For hamster CETP this process was not equimolar but resulted in a net flow of lipid (TG) into HDL. When assayed for the ability to transfer lipid to an acceptor particle lacking CE and TG, monkey and hamster CETPs were most effective, although all CETP species were able to promote this one-way movement of neutral lipid. We conclude that CETPs from human, monkey, rabbit, and hamster are not functionally equivalent. Most unique was hamster CETP, which strongly prefers TG as a substrate and promotes the net flow of lipid from VLDL to HDL.


Assuntos
Proteínas de Transferência de Ésteres de Colesterol/metabolismo , Animais , Transporte Biológico , Membrana Celular/metabolismo , Ésteres do Colesterol/metabolismo , Cricetinae , Células HEK293 , Haplorrinos , Humanos , Lipoproteínas/metabolismo , Coelhos , Especificidade da Espécie , Especificidade por Substrato , Triglicerídeos/metabolismo
19.
Mol Biol Cell ; 24(21): 3309-25, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24025716

RESUMO

Niemann-Pick disease type C (NPC) is caused by mutations in NPC1 or NPC2, which coordinate egress of low-density-lipoprotein (LDL)-cholesterol from late endosomes. We previously reported that the adenovirus-encoded protein RIDα rescues the cholesterol storage phenotype in NPC1-mutant fibroblasts. We show here that RIDα reconstitutes deficient endosome-to-endoplasmic reticulum (ER) transport, allowing excess LDL-cholesterol to be esterified by acyl-CoA:cholesterol acyltransferase and stored in lipid droplets (LDs) in NPC1-deficient cells. Furthermore, the RIDα pathway is regulated by the oxysterol-binding protein ORP1L. Studies have classified ORP1L as a sterol sensor involved in LE positioning downstream of GTP-Rab7. Our data, however, suggest that ORP1L may play a role in transport of LDL-cholesterol to a specific ER pool designated for LD formation. In contrast to NPC1, which is dispensable, the RIDα/ORP1L-dependent route requires functional NPC2. Although NPC1/NPC2 constitutes the major pathway, therapies that amplify minor egress routes for LDL-cholesterol could significantly improve clinical management of patients with loss-of-function NPC1 mutations. The molecular identity of putative alternative pathways, however, is poorly characterized. We propose RIDα as a model system for understanding physiological egress routes that use ORP1L to activate ER feedback responses involved in LD formation.


Assuntos
Proteínas E3 de Adenovirus/metabolismo , Proteínas de Transporte/metabolismo , Grânulos Citoplasmáticos/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Receptores de Esteroides/metabolismo , Proteínas E3 de Adenovirus/genética , Animais , Transporte Biológico/genética , Células CHO , Proteínas de Transporte/genética , Células Cultivadas , LDL-Colesterol/metabolismo , Cricetinae , Cricetulus , Retículo Endoplasmático/metabolismo , Endossomos/metabolismo , Esterificação , Fibroblastos/metabolismo , Fibroblastos/patologia , Glicoproteínas/genética , Glicoproteínas/metabolismo , Células HEK293 , Humanos , Immunoblotting , Peptídeos e Proteínas de Sinalização Intracelular , Metabolismo dos Lipídeos , Glicoproteínas de Membrana/genética , Proteínas de Membrana/genética , Microscopia Confocal , Mutação , Proteína C1 de Niemann-Pick , Doenças de Niemann-Pick/genética , Doenças de Niemann-Pick/metabolismo , Doenças de Niemann-Pick/patologia , Interferência de RNA , Receptores de Esteroides/genética , Transdução de Sinais , Proteínas de Transporte Vesicular
20.
Cell Metab ; 15(6): 861-72, 2012 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-22632897

RESUMO

Akt, a serine-threonine protein kinase, exists as three isoforms. The Akt signaling pathway controls multiple cellular functions in the cardiovascular system, and the atheroprotective endothelial cell-dependent role of Akt1 has been recently demonstrated. The role of Akt3 isoform in cardiovascular pathophysiology is not known. We explored the role of Akt3 in atherosclerosis using mice with a genetic ablation of the Akt3 gene. Using hyperlipidemic ApoE(-/-) mice, we demonstrated a macrophage-dependent, atheroprotective role for Akt3. In vitro experiments demonstrated differential subcellular localization of Akt1 and Akt3 in macrophages and showed that Akt3 specifically inhibits macrophage cholesteryl ester accumulation and foam cell formation, a critical early event in atherogenesis. Mechanistically, Akt3 suppresses foam cell formation by reducing lipoprotein uptake and promoting ACAT-1 degradation via the ubiquitin-proteasome pathway. These studies demonstrate the nonredundant atheroprotective role for Akt3 exerted via the previously unknown link between the Akt signaling pathway and cholesterol metabolism.


Assuntos
Aterosclerose/enzimologia , Células Espumosas/enzimologia , Macrófagos Peritoneais/enzimologia , Proteínas Proto-Oncogênicas c-akt/deficiência , Acetil-CoA C-Acetiltransferase/metabolismo , Animais , Aorta/patologia , Apolipoproteínas E/deficiência , Apolipoproteínas E/genética , Aterosclerose/etiologia , Células da Medula Óssea/enzimologia , Sobrevivência Celular , Células Cultivadas , Colesterol/biossíntese , Colesterol/sangue , Colesterol/metabolismo , Ésteres do Colesterol/metabolismo , Feminino , Hiperlipidemias/complicações , Hiperlipidemias/enzimologia , Lipoproteínas/sangue , Lipoproteínas/metabolismo , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Seio Aórtico/patologia , Triglicerídeos/sangue
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...