Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros











Intervalo de año de publicación
1.
ACS Chem Biol ; 19(8): 1820-1835, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39099090

RESUMEN

Neuropilin-1 acts as a coreceptor with vascular endothelial growth factor receptors to facilitate binding of its ligand, vascular endothelial growth factor. Neuropilin-1 also binds to heparan sulfate, but the functional significance of this interaction has not been established. A combinatorial library screening using heparin oligosaccharides followed by molecular dynamics simulations of a heparin tetradecasaccharide suggested a highly conserved binding site composed of amino acid residues extending across the b1 and b2 domains of murine neuropilin-1. Mutagenesis studies established the importance of arginine513 and lysine514 for binding of heparin to a recombinant form of Nrp1 composed of the a1, a2, b1, and b2 domains. Recombinant Nrp1 protein bearing R513A,K514A mutations showed a significant loss of heparin-binding, heparin-induced dimerization, and heparin-dependent thermal stabilization. Isothermal calorimetry experiments suggested a 1:2 complex of heparin tetradecasaccharide:Nrp1. To study the impact of altered heparin binding in vivo, a mutant allele of Nrp1 bearing the R513A,K514A mutations was created in mice (Nrp1D) and crossbred to Nrp1+/- mice to examine the impact of altered heparan sulfate binding. Analysis of tumor formation showed variable effects on tumor growth in Nrp1D/D mice, resulting in a frank reduction in tumor growth in Nrp1D/- mice. Expression of mutant Nrp1D protein was normal in tissues, suggesting that the reduction in tumor growth was due to the altered binding of heparin/heparan sulfate to neuropilin-1. These findings suggest that the interaction of neuropilin-1 with heparan sulfate modulates its stability and its role in tumor formation and growth.


Asunto(s)
Heparitina Sulfato , Neuropilina-1 , Neuropilina-1/metabolismo , Neuropilina-1/genética , Neuropilina-1/química , Animales , Heparitina Sulfato/metabolismo , Ratones , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Unión Proteica , Sitios de Unión , Ratones Endogámicos C57BL , Heparina/metabolismo , Heparina/química , Simulación de Dinámica Molecular , Mutación
2.
J Biol Chem ; 298(8): 102159, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35750212

RESUMEN

Lysosomal storage diseases result in various developmental and physiological complications, including cachexia. To study the causes for the negative energy balance associated with cachexia, we assessed the impact of sulfamidase deficiency and heparan sulfate storage on energy homeostasis and metabolism in a mouse model of type IIIa mucopolysaccharidosis (MPS IIIa, Sanfilippo A syndrome). At 12-weeks of age, MPS IIIa mice exhibited fasting and postprandial hypertriglyceridemia compared with wildtype mice, with a reduction of white and brown adipose tissues. Partitioning of dietary [3H]triolein showed a marked increase in intestinal uptake and secretion, whereas hepatic production and clearance of triglyceride-rich lipoproteins did not differ from wildtype controls. Uptake of dietary triolein was also elevated in brown adipose tissue (BAT), and notable increases in beige adipose tissue occurred, resulting in hyperthermia, hyperphagia, hyperdipsia, and increased energy expenditure. Furthermore, fasted MPS IIIa mice remained hyperthermic when subjected to low temperature but became cachexic and profoundly hypothermic when treated with a lipolytic inhibitor. We demonstrated that the reliance on increased lipid fueling of BAT was driven by a reduced ability to generate energy from stored lipids within the depot. These alterations arose from impaired autophagosome-lysosome fusion, resulting in increased mitochondria content in beige and BAT. Finally, we show that increased mitochondria content in BAT and postprandial dyslipidemia was partially reversed upon 5-week treatment with recombinant sulfamidase. We hypothesize that increased BAT activity and persistent increases in energy demand in MPS IIIa mice contribute to the negative energy balance observed in patients with MPS IIIa.


Asunto(s)
Hipertrigliceridemia , Mucopolisacaridosis III , Tejido Adiposo Pardo/metabolismo , Animales , Caquexia , Ratones , Mitofagia , Mucopolisacaridosis III/metabolismo , Mucopolisacaridosis III/terapia , Trioleína
3.
Nat Chem Biol ; 17(6): 684-692, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33846619

RESUMEN

Heparan sulfate (HS) proteoglycans bind extracellular proteins that participate in cell signaling, attachment and endocytosis. These interactions depend on the arrangement of sulfated sugars in the HS chains generated by well-characterized biosynthetic enzymes; however, the regulation of these enzymes is largely unknown. We conducted genome-wide CRISPR-Cas9 screens with a small-molecule ligand that binds to HS. Screening of A375 melanoma cells uncovered additional genes and pathways impacting HS formation. The top hit was the epigenetic factor KDM2B, a histone demethylase. KDM2B inactivation suppressed multiple HS sulfotransferases and upregulated the sulfatase SULF1. These changes differentially affected the interaction of HS-binding proteins. KDM2B-deficient cells displayed decreased growth rates, which was rescued by SULF1 inactivation. In addition, KDM2B deficiency altered the expression of many extracellular matrix genes. Thus, KDM2B controls proliferation of A375 cells through the regulation of HS structure and serves as a master regulator of the extracellular matrix.


Asunto(s)
Proteínas F-Box/antagonistas & inhibidores , Estudio de Asociación del Genoma Completo , Heparitina Sulfato/metabolismo , Histona Demetilasas con Dominio de Jumonji/antagonistas & inhibidores , Algoritmos , Sistemas CRISPR-Cas , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Descubrimiento de Drogas , Matriz Extracelular/genética , Ensayos Analíticos de Alto Rendimiento , Humanos , Unión Proteica/genética , RNA-Seq , Sulfotransferasas/antagonistas & inhibidores
4.
bioRxiv ; 2021 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-33791697

RESUMEN

We identify the prolyl-tRNA synthetase (PRS) inhibitor halofuginone 1 , a compound in clinical trials for anti-fibrotic and anti-inflammatory applications 2 , as a potent inhibitor of SARS-CoV-2 infection and replication. The interaction of SARS-CoV-2 spike protein with cell surface heparan sulfate (HS) promotes viral entry 3 . We find that halofuginone reduces HS biosynthesis, thereby reducing spike protein binding, SARS-CoV-2 pseudotyped virus, and authentic SARS-CoV-2 infection. Halofuginone also potently suppresses SARS-CoV-2 replication post-entry and is 1,000-fold more potent than Remdesivir 4 . Inhibition of HS biosynthesis and SARS-CoV-2 infection depends on specific inhibition of PRS, possibly due to translational suppression of proline-rich proteins. We find that pp1a and pp1ab polyproteins of SARS-CoV-2, as well as several HS proteoglycans, are proline-rich, which may make them particularly vulnerable to halofuginone's translational suppression. Halofuginone is orally bioavailable, has been evaluated in a phase I clinical trial in humans and distributes to SARS-CoV-2 target organs, including the lung, making it a near-term clinical trial candidate for the treatment of COVID-19.

7.
Proc Natl Acad Sci U S A ; 116(32): 16036-16045, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31332008

RESUMEN

Cardiovascular disease (CVD) events due to atherosclerosis cause one-third of worldwide deaths and risk factors include physical inactivity, age, dyslipidemia, hypertension, diabetes, obesity, smoking, and red meat consumption. However, ∼15% of first-time events occur without such factors. In contrast, coronary events are extremely rare even in closely related chimpanzees in captivity, despite human-like CVD-risk-prone blood lipid profiles, hypertension, and mild atherosclerosis. Similarly, red meat-associated enhancement of CVD event risk does not seem to occur in other carnivorous mammals. Thus, heightened CVD risk may be intrinsic to humans, and genetic changes during our evolution need consideration. Humans exhibit a species-specific deficiency of the sialic acid N-glycolylneuraminic acid (Neu5Gc), due to pseudogenization of cytidine monophosphate-N-acetylneuraminic acid (Neu5Ac) hydroxylase (CMAH), which occurred in hominin ancestors ∼2 to 3 Mya. Ldlr-/- mice with human-like Cmah deficiency fed a sialic acids (Sias)-free high-fat diet (HFD) showed ∼1.9-fold increased atherogenesis over Cmah wild-type Ldlr-/- mice, associated with elevated macrophage cytokine expression and enhanced hyperglycemia. Human consumption of Neu5Gc (from red meat) acts as a "xeno-autoantigen" via metabolic incorporation into endogenous glycoconjugates, as interactions with circulating anti-Neu5Gc "xeno-autoantibodies" potentiate chronic inflammation ("xenosialitis"). Cmah-/-Ldlr-/- mice immunized with Neu5Gc-bearing antigens to generate human-like anti-Neu5Gc antibodies suffered a ∼2.4-fold increased atherosclerosis on a Neu5Gc-rich HFD, compared with Neu5Ac-rich or Sias-free HFD. Lesions in Neu5Gc-immunized and Neu5Gc-rich HFD-fed Cmah-/-Ldlr-/- mice were more advanced but unexplained by lipoprotein or glucose changes. Human evolutionary loss of CMAH likely contributes to atherosclerosis predisposition via multiple intrinsic and extrinsic mechanisms, and future studies could consider this more human-like model.


Asunto(s)
Aterosclerosis/enzimología , Oxigenasas de Función Mixta/deficiencia , Animales , Bovinos , Citocinas/metabolismo , Dieta Alta en Grasa , Femenino , Humanos , Hiperglucemia/patología , Inflamación/patología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Oxigenasas de Función Mixta/metabolismo , Modelos Biológicos , Fenotipo , Receptores de LDL/deficiencia , Receptores de LDL/metabolismo , Ácidos Siálicos/metabolismo , Especificidad de la Especie
8.
J Biol Chem ; 294(36): 13292-13303, 2019 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-31315930

RESUMEN

Hepcidin is a liver-derived peptide hormone that controls systemic iron homeostasis. Its expression is regulated by the bone morphogenetic protein 6 (BMP6)/SMAD1/5/8 pathway and by the proinflammatory cytokine interleukin 6 (IL6). Proteoglycans that function as receptors of these signaling proteins in the liver are commonly decorated by heparan sulfate, but the potential role of hepatic heparan sulfate in hepcidin expression and iron homeostasis is unclear. Here, we show that modulation of hepatic heparan sulfate significantly alters hepcidin expression and iron metabolism both in vitro and in vivo Specifically, enzymatic removal of heparan sulfate from primary human hepatocytes, CRISPR/Cas9 manipulation of heparan sulfate biosynthesis in human hepatoma cells, or pharmacological manipulation of heparan sulfate-protein interactions using sodium chlorate or surfen dramatically reduced baseline and BMP6/SMAD1/5/8-dependent hepcidin expression. Moreover inactivation of the heparan sulfate biosynthetic gene N-deacetylase and N-sulfotransferase 1 (Ndst1) in murine hepatocytes (Ndst1f/fAlbCre+) reduced hepatic hepcidin expression and caused a redistribution of systemic iron, leading to iron accumulation in the liver and serum of mice. Manipulation of heparan sulfate had a similar effect on IL6-dependent hepcidin expression in vitro and suppressed IL6-mediated iron redistribution induced by lipopolysaccharide in vivo These results provide compelling evidence that hepatocyte heparan sulfate plays a key role in regulating hepcidin expression and iron homeostasis in mice and in human hepatocytes.


Asunto(s)
Heparitina Sulfato/metabolismo , Hepatocitos/metabolismo , Hepcidinas/genética , Homeostasis , Hierro/metabolismo , Animales , Línea Celular , Perfilación de la Expresión Génica , Hepcidinas/metabolismo , Humanos , Ratones , Ratones Endogámicos , Regiones Promotoras Genéticas/genética
9.
Glycobiology ; 29(8): 582-592, 2019 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-31094413

RESUMEN

Binding and uptake of triglyceride-rich lipoproteins (TRLs) in mice depend on heparan sulfate and the hepatic proteoglycan, syndecan-1 (SDC1). Alteration of glucosamine N-sulfation by deletion of glucosamine N-deacetylase-N-sulfotransferase 1 (Ndst1) and 2-O-sulfation of uronic acids by deletion of uronyl 2-O-sulfotransferase (Hs2st) led to diminished lipoprotein metabolism, whereas inactivation of glucosaminyl 6-O-sulfotransferase 1 (Hs6st1), which encodes one of the three 6-O-sulfotransferases, had little effect on lipoprotein binding. However, other studies have suggested that 6-O-sulfation may be important for TRL binding and uptake. In order to explain these discrepant findings, we used CRISPR/Cas9 gene editing to create a library of mutants in the human hepatoma cell line, Hep3B. Inactivation of EXT1 encoding the heparan sulfate copolymerase, NDST1 and HS2ST dramatically reduced binding of TRLs. Inactivation of HS6ST1 had no effect, but deletion of HS6ST2 reduced TRL binding. Compounding mutations in HS6ST1 and HS6ST2 did not exacerbate this effect indicating that HS6ST2 is the dominant 6-O-sulfotransferase and that binding of TRLs indeed depends on 6-O-sulfation of glucosamine residues. Uptake studies showed that TRL internalization was also affected in 6-O-sulfation deficient cells. Interestingly, genetic deletion of SDC1 only marginally impacted binding of TRLs but reduced TRL uptake to the same extent as treating the cells with heparin lyases. These findings confirm that SDC1 is the dominant endocytic proteoglycan receptor for TRLs in human Hep3B cells and that binding and uptake of TRLs depend on SDC1 and N- and 2-O-sulfation as well as 6-O-sulfation of heparan sulfate chains catalyzed by HS6ST2.


Asunto(s)
Lipoproteínas/metabolismo , Mutación con Pérdida de Función , N-Acetilglucosaminiltransferasas/metabolismo , Sulfotransferasas/metabolismo , Sistemas CRISPR-Cas , Línea Celular Tumoral , Células Cultivadas , Hepatocitos/metabolismo , Humanos , Lipoproteínas/química , N-Acetilglucosaminiltransferasas/genética , Unión Proteica , Sulfotransferasas/genética , Sindecano-1/genética , Sindecano-1/metabolismo , Triglicéridos/química , Triglicéridos/metabolismo
10.
Mol Metab ; 16: 88-99, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30100244

RESUMEN

OBJECTIVE: Insulin resistance is associated with impaired receptor dependent hepatic uptake of triglyceride-rich lipoproteins (TRL), promoting hypertriglyceridemia and atherosclerosis. Next to low-density lipoprotein (LDL) receptor (LDLR) and syndecan-1, the LDLR-related protein 1 (LRP1) stimulated by insulin action contributes to the rapid clearance of TRL in the postprandial state. Here, we investigated the hypothesis that the adaptor protein phosphotyrosine interacting domain-containing protein 1 (PID1) regulates LRP1 function, thereby controlling hepatic endocytosis of postprandial lipoproteins. METHODS: Localization and interaction of PID1 and LRP1 in cultured hepatocytes was studied by confocal microscopy of fluorescent tagged proteins, by indirect immunohistochemistry of endogenous proteins, by GST-based pull down and by immunoprecipitation experiments. The in vivo relevance of PID1 was assessed using whole body as well as liver-specific Pid1-deficient mice on a wild type or Ldlr-deficient (Ldlr-/-) background. Intravital microscopy was used to study LRP1 translocation in the liver. Lipoprotein metabolism was investigated by lipoprotein profiling, gene and protein expression as well as organ-specific uptake of radiolabelled TRL. RESULTS: PID1 co-localized in perinuclear endosomes and was found associated with LRP1 under fasting conditions. We identified the distal NPxY motif of the intracellular C-terminal domain (ICD) of LRP1 as the site critical for the interaction with PID1. Insulin-mediated NPxY-phosphorylation caused the dissociation of PID1 from the ICD, causing LRP1 translocation to the plasma membrane. PID1 deletion resulted in higher LRP1 abundance at the cell surface, higher LDLR protein levels and, paradoxically, reduced total LRP1. The latter can be explained by higher receptor shedding, which we observed in cultured Pid1-deficient hepatocytes. Consistently, PID1 deficiency alone led to increased LDLR-dependent endocytosis of postprandial lipoproteins and lower plasma triglycerides. In contrast, hepatic PID1 deletion on an Ldlr-/- background reduced lipoprotein uptake into liver and caused plasma TRL accumulation. CONCLUSIONS: By acting as an insulin-dependent retention adaptor, PID1 serves as a regulator of LRP1 function controlling the disposal of postprandial lipoproteins. PID1 inhibition provides a novel approach to lower plasma levels of pro-atherogenic TRL remnants by stimulating endocytic function of both LRP1 and LDLR in the liver.


Asunto(s)
Proteínas Portadoras/metabolismo , Hipertrigliceridemia/metabolismo , Lipoproteínas/metabolismo , Triglicéridos/metabolismo , Animales , Carcinoma Hepatocelular , Proteínas Portadoras/genética , Línea Celular Tumoral , Endocitosis/fisiología , Hepatocitos/metabolismo , Humanos , Hipertrigliceridemia/genética , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Lipoproteínas/fisiología , Hígado/metabolismo , Neoplasias Hepáticas , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Periodo Posprandial , Receptores de LDL/metabolismo , Sinapsinas/metabolismo , Sinapsinas/fisiología , Triglicéridos/fisiología , Proteínas Supresoras de Tumor/metabolismo
11.
J Lipid Res ; 59(5): 854-863, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29559522

RESUMEN

Apolipoprotein A-I binding protein (AIBP) has been shown to augment cholesterol efflux from endothelial cells and macrophages. In zebrafish and mice, AIBP-mediated regulation of cholesterol levels in the plasma membrane of endothelial cells controls angiogenesis. The goal of this work was to evaluate metabolic changes and atherosclerosis in AIBP loss-of-function and gain-of-function animal studies. Here, we show that Apoa1bp-/-Ldlr-/- mice fed a high-cholesterol, high-fat diet had exacerbated weight gain, liver steatosis, glucose intolerance, hypercholesterolemia, hypertriglyceridemia, and larger atherosclerotic lesions compared with Ldlr-/- mice. Feeding Apoa1bp-/-Ldlr-/- mice a high-cholesterol, normal-fat diet did not result in significant differences in lipid levels or size of atherosclerotic lesions from Ldlr-/- mice. Conversely, adeno-associated virus-mediated overexpression of AIBP reduced hyperlipidemia and atherosclerosis in high-cholesterol, high-fat diet-fed Ldlr-/- mice. Injections of recombinant AIBP reduced aortic inflammation in Ldlr-/- mice fed a short high-cholesterol, high-fat diet. Conditional overexpression of AIBP in zebrafish also reduced diet-induced vascular lipid accumulation. In experiments with isolated macrophages, AIBP facilitated cholesterol efflux to HDL, reduced lipid rafts content, and inhibited inflammatory responses to lipopolysaccharide.jlr Our data demonstrate that AIBP confers protection against diet-induced metabolic abnormalities and atherosclerosis.


Asunto(s)
Aterosclerosis/metabolismo , Proteínas Portadoras/metabolismo , Síndrome Metabólico/metabolismo , Fosfoproteínas/metabolismo , Animales , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfoproteínas/deficiencia , Racemasas y Epimerasas , Receptores de LDL/deficiencia , Receptores de LDL/metabolismo
12.
J Clin Invest ; 126(8): 2855-66, 2016 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-27400128

RESUMEN

Hypertriglyceridemia is an independent risk factor for cardiovascular disease, and plasma triglycerides (TGs) correlate strongly with plasma apolipoprotein C-III (ApoC-III) levels. Antisense oligonucleotides (ASOs) for ApoC-III reduce plasma TGs in primates and mice, but the underlying mechanism of action remains controversial. We determined that a murine-specific ApoC-III-targeting ASO reduces fasting TG levels through a mechanism that is dependent on low-density lipoprotein receptors (LDLRs) and LDLR-related protein 1 (LRP1). ApoC-III ASO treatment lowered plasma TGs in mice lacking lipoprotein lipase (LPL), hepatic heparan sulfate proteoglycan (HSPG) receptors, LDLR, or LRP1 and in animals with combined deletion of the genes encoding HSPG receptors and LDLRs or LRP1. However, the ApoC-III ASO did not lower TG levels in mice lacking both LDLR and LRP1. LDLR and LRP1 were also required for ApoC-III ASO-induced reduction of plasma TGs in mice fed a high-fat diet, in postprandial clearance studies, and when ApoC-III-rich or ApoC-III-depleted lipoproteins were injected into mice. ASO reduction of ApoC-III had no effect on VLDL secretion, heparin-induced TG reduction, or uptake of lipids into heart and skeletal muscle. Our data indicate that ApoC-III inhibits turnover of TG-rich lipoproteins primarily through a hepatic clearance mechanism mediated by the LDLR/LRP1 axis.


Asunto(s)
Apolipoproteína C-III/sangre , Lipoproteínas/sangre , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad/metabolismo , Receptores de LDL/metabolismo , Triglicéridos/sangre , Proteínas Supresoras de Tumor/metabolismo , Animales , Femenino , Genotipo , Heparina/farmacología , Hepatocitos/metabolismo , Cetonas/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Miocardio/metabolismo , Factores de Riesgo
13.
Cytokine ; 72(1): 118-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25573804

RESUMEN

Macrophages are important mediators of diseases associated with metabolic inflammation such as obesity and atherosclerosis. In this Stimulus we discuss recent findings showing that heparan sulfate proteoglycans on macrophages serve as an important inflammatory rheostat. This observation has significant implications as the degree of macrophage proteoglycan sulfation can determine and possibly predict disease outcomes of metabolic inflammatory disorders.


Asunto(s)
Proteoglicanos de Heparán Sulfato/inmunología , Inflamación , Interferón beta/metabolismo , Macrófagos/inmunología , Animales , Células Cultivadas , Humanos , Inflamación/metabolismo , Interferón beta/genética , Macrófagos/fisiología , Ratones
14.
Cell Metab ; 20(5): 813-826, 2014 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-25440058

RESUMEN

Heparan sulfate proteoglycans (HSPGs) are an important constituent of the macrophage glycocalyx and extracellular microenvironment. To examine their role in atherogenesis, we inactivated the biosynthetic gene N-acetylglucosamine N-deacetylase-N-sulfotransferase 1 (Ndst1) in macrophages and crossbred the strain to Ldlr(-/-) mice. When placed on an atherogenic diet, Ldlr(-/-)Ndst1(f/f)LysMCre(+) mice had increased atherosclerotic plaque area and volume compared to Ldlr(-/-) mice. Diminished sulfation of heparan sulfate resulted in enhanced chemokine expression; increased macrophages in plaques; increased expression of ACAT2, a key enzyme in cholesterol ester storage; and increased foam cell conversion. Motif analysis of promoters of upregulated genes suggested increased type I interferon signaling, which was confirmed by elevation of STAT1 phosphorylation induced by IFN-ß. The proinflammatory macrophages derived from Ndst1(f/f)LysMCre(+) mice also sensitized the animals to diet-induced obesity. We propose that macrophage HSPGs control basal activation of macrophages by maintaining type I interferon reception in a quiescent state through sequestration of IFN-ß.


Asunto(s)
Aterosclerosis/inmunología , Proteoglicanos de Heparán Sulfato/inmunología , Interferón Tipo I/inmunología , Macrófagos/inmunología , Obesidad/inmunología , Transducción de Señal , Animales , Aterosclerosis/genética , Aterosclerosis/patología , Dieta Aterogénica/efectos adversos , Femenino , Regulación de la Expresión Génica , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Modelos Moleculares , Obesidad/genética , Obesidad/patología , Sulfotransferasas/genética
15.
Arterioscler Thromb Vasc Biol ; 33(9): 2065-74, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23846497

RESUMEN

OBJECTIVE: Chylomicron and very low-density lipoprotein remnants are cleared from the circulation in the liver by heparan sulfate proteoglycan (HSPG) receptors (syndecan-1), the low-density lipoprotein receptor (LDLR), and LDLR-related protein-1 (LRP1), but the relative contribution of each class of receptors under different dietary conditions remains unclear. APPROACH AND RESULTS: Triglyceride-rich lipoprotein clearance was measured in AlbCre(+)Ndst1(f/f), Ldlr(-/-), and AlbCre(+)Lrp1(f/f) mice and mice containing combinations of these mutations. Triglyceride measurements in single and double mutant mice showed that HSPGs and LDLR dominate clearance under fasting conditions and postprandial conditions, but LRP1 contributes significantly when LDLR is absent. Mice lacking hepatic expression of all 3 receptors (AlbCre(+)Ndst1(f/f) Lrp1(f/f) Ldlr(-/-)) displayed dramatic hyperlipidemia (870 ± 270 mg triglyceride/dL; 1300 ± 350 mg of total cholesterol/dL) and exhibited persistent elevated postprandial triglyceride levels because of reduced hepatic clearance. Analysis of the particles accumulating in mutants showed that HSPGs preferentially clear a subset of small triglyceride-rich lipoproteins (≈ 20-40 nm diameter), whereas LDLR and LRP1 clear larger particles (≈ 40-60 nm diameter). Finally, we show that HSPGs play a major role in clearance of triglyceride-rich lipoproteins in mice fed normal chow or under postprandial conditions but seem to play a less significant role on a high-fat diet. CONCLUSIONS: These data show that HSPGs, LDLR, and LRP1 clear distinct subsets of particles, that HSPGs work independently of LDLR and LRP1, and that HSPGs, LDLR, and LRP1 are the 3 major hepatic triglyceride-rich lipoprotein clearance receptors in mice.


Asunto(s)
Colesterol en la Dieta/metabolismo , Remanentes de Quilomicrones/metabolismo , Dieta Alta en Grasa , Sacarosa en la Dieta/metabolismo , Proteoglicanos de Heparán Sulfato/metabolismo , Hepatocitos/metabolismo , Hígado/metabolismo , Receptores de LDL/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Células Cultivadas , Colesterol en la Dieta/administración & dosificación , Remanentes de Quilomicrones/sangre , Sacarosa en la Dieta/administración & dosificación , Proteoglicanos de Heparán Sulfato/sangre , Hiperlipidemias/genética , Hiperlipidemias/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Tamaño de la Partícula , Periodo Posprandial , Receptores de LDL/deficiencia , Receptores de LDL/genética , Sulfotransferasas/genética , Sulfotransferasas/metabolismo , Factores de Tiempo , Triglicéridos/metabolismo , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética
16.
J Clin Invest ; 123(6): 2742-51, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23676495

RESUMEN

The heparan sulfate proteoglycan (HSPG) syndecan-1 (SDC1) acts as a major receptor for triglyceride-rich lipoprotein (TRL) clearance in the liver. We sought to identify the relevant apolipoproteins on TRLs that mediate binding to SDC1 and determine their clinical relevance. Evidence supporting ApoE as a major determinant arose from its enrichment in TRLs from mice defective in hepatic heparan sulfate (Ndst1f/fAlbCre⁺ mice), decreased binding of ApoE-deficient TRLs to HSPGs on human hepatoma cells, and decreased clearance of ApoE-deficient [³H]TRLs in vivo. Evidence for a second ligand was suggested by the faster clearance of ApoE-deficient TRLs after injection into WT Ndst1f/fAlbCre⁻ versus mutant Ndst1f/fAlbCre⁺ mice and elevated fasting and postprandial plasma triglycerides in compound Apoe⁻/⁻Ndst1f/fAlbCre⁺ mice compared with either single mutant. ApoAV emerged as a candidate based on 6-fold enrichment of ApoAV in TRLs accumulating in Ndst1f/fAlbCre⁺ mice, decreased binding of TRLs to proteoglycans after depletion of ApoAV or addition of anti-ApoAV mAb, and decreased heparan sulfate-dependent binding of ApoAV-deficient particles to hepatocytes. Importantly, disruption of hepatic heparan sulfate-mediated clearance increased atherosclerosis. We conclude that clearance of TRLs by hepatic HSPGs is atheroprotective and mediated by multivalent binding to ApoE and ApoAV.


Asunto(s)
Apolipoproteínas E/metabolismo , Apolipoproteínas/metabolismo , Sindecano-1/metabolismo , Animales , Aorta Torácica/metabolismo , Aorta Torácica/patología , Apolipoproteína A-V , Apolipoproteínas B/metabolismo , Aterosclerosis/sangre , Aterosclerosis/patología , Línea Celular Tumoral , Femenino , Humanos , Lipoproteínas/metabolismo , Hígado/metabolismo , Masculino , Ratones , Ratones Transgénicos , Unión Proteica , Triglicéridos/sangre
17.
Nat Med ; 17(2): 200-5, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21258337

RESUMEN

Brown adipose tissue (BAT) burns fatty acids for heat production to defend the body against cold and has recently been shown to be present in humans. Triglyceride-rich lipoproteins (TRLs) transport lipids in the bloodstream, where the fatty acid moieties are liberated by the action of lipoprotein lipase (LPL). Peripheral organs such as muscle and adipose tissue take up the fatty acids, whereas the remaining cholesterol-rich remnant particles are cleared by the liver. Elevated plasma triglyceride concentrations and prolonged circulation of cholesterol-rich remnants, especially in diabetic dyslipidemia, are risk factors for cardiovascular disease. However, the precise biological role of BAT for TRL clearance remains unclear. Here we show that increased BAT activity induced by short-term cold exposure controls TRL metabolism in mice. Cold exposure drastically accelerated plasma clearance of triglycerides as a result of increased uptake into BAT, a process crucially dependent on local LPL activity and transmembrane receptor CD36. In pathophysiological settings, cold exposure corrected hyperlipidemia and improved deleterious effects of insulin resistance. In conclusion, BAT activity controls vascular lipoprotein homeostasis by inducing a metabolic program that boosts TRL turnover and channels lipids into BAT. Activation of BAT might be a therapeutic approach to reduce elevated triglyceride concentrations and combat obesity in humans.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Triglicéridos/metabolismo , Tejido Adiposo Pardo/fisiología , Animales , Regulación de la Temperatura Corporal/fisiología , Antígenos CD36/metabolismo , Colesterol/metabolismo , Colesterol/fisiología , Frío , Humanos , Hiperlipidemias/metabolismo , Hiperlipidemias/fisiopatología , Resistencia a la Insulina/fisiología , Lipoproteína Lipasa/metabolismo , Lipoproteínas/metabolismo , Lipoproteínas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/metabolismo , Obesidad/fisiopatología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/fisiología
18.
Cell Mol Life Sci ; 67(1): 135-45, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19856143

RESUMEN

The proximal NPXY and distal NPXYXXL motifs in the intracellular domain of LRP1 play an important role in regulation of the function of the receptor. The impact of single and double inactivating knock-in mutations of these motifs on receptor maturation, cell surface expression, and ligand internalization was analyzed in mutant and control wild-type mice and MEFs. Single inactivation of the proximal NPXY or in combination with inactivation of the distal NPXYXXL motif are both shown to be associated with an impaired maturation and premature proteasomal degradation of full-length LRP1. Therefore, only a small mature LRP1 pool is able to reach the cell surface resulting indirectly in severe impairment of ligand internalization. Single inactivation of the NPXYXXL motif revealed normal maturation, but direct impairment of ligand internalization. In conclusion, the proximal NPXY motif proves to be essential for early steps in the LRP1 biosynthesis, whereas NPXYXXL appears rather relevant for internalization.


Asunto(s)
Receptores de LDL/biosíntesis , Proteínas Supresoras de Tumor/biosíntesis , Secuencias de Aminoácidos , Animales , Desarrollo Embrionario , Endocitosis , Técnicas de Sustitución del Gen , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Ratones , Mutación , Complejo de la Endopetidasa Proteasomal/metabolismo , Dominios y Motivos de Interacción de Proteínas , Estructura Terciaria de Proteína , Receptores de LDL/genética , Receptores de LDL/metabolismo , Análisis de Supervivencia , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
19.
Arterioscler Thromb Vasc Biol ; 29(9): 1258-64, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19667105

RESUMEN

OBJECTIVE: The purpose of this study was to determine the significance of the intracellular NPxYxxL motif of LRP1 for the atheroprotective role of this multifunctional receptor. METHODS AND RESULTS: LRP1 knock-in mice carrying an inactivating mutation in the NPxYxxL motif were crossed with LDLR-deficient mice, a model for atherosclerosis. In this LDLR(-/-) background the mutated mice showed a more atherogenic lipoprotein profile, which was associated with a decreased clearance of postprandial lipids because of a compromised endocytosis rate and reduced lipase activity. On an atherogenic diet LRP1 mutant mice revealed a 50% increased development of atherosclerosis. This aggravation was accompanied by an increase in smooth muscle cell (SMC) and collagen content and apoptotic cells in the lesions. The mutation showed, however, a limited impact on basal PDGFR-beta expression and signaling and the antimigratory property of apoE on PDGF-BB-stimulated SMCs. Additionally, levels of LRP1 atherogenic ligands, like MMP2, t-PA, FVIII, and the inflammatory ligand TNF-alpha showed to be significantly elevated. CONCLUSIONS: These findings demonstrate that the NPxYxxL motif is essential for the atheroprotective role of LRP1. This motif is relevant for normal control of lipid metabolism and of atherogenic and inflammatory ligands, but has no pronounced effect on regulating PDGF-BB/PDGFR-beta signaling in SMCs.


Asunto(s)
Aterosclerosis/metabolismo , Dislipidemias/metabolismo , Metabolismo de los Lípidos , Periodo Posprandial , Receptores de LDL/deficiencia , Proteínas Supresoras de Tumor/metabolismo , Secuencias de Aminoácidos , Animales , Apoptosis , Aterosclerosis/genética , Aterosclerosis/patología , Becaplermina , Colágeno/metabolismo , Modelos Animales de Enfermedad , Dislipidemias/genética , Dislipidemias/patología , Endocitosis , Técnicas de Sustitución del Gen , Metabolismo de los Lípidos/genética , Lipoproteína Lipasa/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Macrófagos/metabolismo , Ratones , Ratones Noqueados , Músculo Liso Vascular/metabolismo , Músculo Liso Vascular/patología , Mutación , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-sis , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores de LDL/química , Receptores de LDL/genética , Receptores de LDL/metabolismo , Transducción de Señal , Factores de Tiempo , Factor de Necrosis Tumoral alfa/metabolismo , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA