Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Lipid Res ; 58(1): 216-225, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27811232

RESUMEN

In mice lacking glycosylphosphatidylinositol-anchored high density lipoprotein binding protein 1 (GPIHBP1), the LPL secreted by adipocytes and myocytes remains bound to heparan sulfate proteoglycans (HSPGs) on all cells within tissues. That observation raises a perplexing issue: Why isn't the freshly secreted LPL in wild-type mice captured by the same HSPGs, thereby preventing LPL from reaching GPIHBP1 on capillaries? We hypothesized that LPL-HSPG interactions are transient, allowing the LPL to detach and move to GPIHBP1 on capillaries. Indeed, we found that LPL detaches from HSPGs on cultured cells and moves to: 1) soluble GPIHBP1 in the cell culture medium; 2) GPIHBP1-coated agarose beads; and 3) nearby GPIHBP1-expressing cells. Movement of HSPG-bound LPL to GPIHBP1 did not occur when GPIHBP1 contained a Ly6 domain missense mutation (W109S), but was almost normal when GPIHBP1's acidic domain was mutated. To test the mobility of HSPG-bound LPL in vivo, we injected GPIHBP1-coated agarose beads into the brown adipose tissue of GPIHBP1-deficient mice. LPL moved quickly from HSPGs on adipocytes to GPIHBP1-coated beads, thereby depleting LPL stores on the surface of adipocytes. We conclude that HSPG-bound LPL in the interstitial spaces of tissues is mobile, allowing the LPL to move to GPIHBP1 on endothelial cells.


Asunto(s)
Adipocitos/metabolismo , Proteoglicanos de Heparán Sulfato/metabolismo , Lipoproteína Lipasa/genética , Receptores de Lipoproteína/genética , Animales , Capilares/enzimología , Capilares/metabolismo , Línea Celular , Quilomicrones/metabolismo , Medios de Cultivo/química , Células Hep G2 , Humanos , Lipólisis/genética , Lipoproteína Lipasa/metabolismo , Ratones
2.
Circ Res ; 116(4): 624-32, 2015 Feb 13.
Artículo en Inglés | MEDLINE | ID: mdl-25387803

RESUMEN

RATIONALE: GPIHBP1, a GPI-anchored protein of capillary endothelial cells, binds lipoprotein lipase (LPL) in the subendothelial spaces and shuttles it to the capillary lumen. GPIHBP1 missense mutations that interfere with LPL binding cause familial chylomicronemia. OBJECTIVE: We sought to understand mechanisms by which GPIHBP1 mutations prevent LPL binding and lead to chylomicronemia. METHODS AND RESULTS: We expressed mutant forms of GPIHBP1 in Chinese hamster ovary cells, rat and human endothelial cells, and Drosophila S2 cells. In each expression system, mutation of cysteines in GPIHBP1's Ly6 domain (including mutants identified in patients with chylomicronemia) led to the formation of disulfide-linked dimers and multimers. GPIHBP1 dimerization/multimerization was not unique to cysteine mutations; mutations in other amino acid residues, including several associated with chylomicronemia, also led to protein dimerization/multimerization. The loss of GPIHBP1 monomers is relevant to the pathogenesis of chylomicronemia because only GPIHBP1 monomers-and not dimers or multimers-are capable of binding LPL. One GPIHBP1 mutant, GPIHBP1-W109S, had distinctive properties. GPIHBP1-W109S lacked the ability to bind LPL but had a reduced propensity for forming dimers or multimers, suggesting that W109 might play a more direct role in binding LPL. In support of that idea, replacing W109 with any of 8 other amino acids abolished LPL binding-and often did so without promoting the formation of dimers and multimers. CONCLUSIONS: Many amino acid substitutions in GPIHBP1's Ly6 domain that abolish LPL binding lead to protein dimerization/multimerization. Dimerization/multimerization is relevant to disease pathogenesis, given that only GPIHBP1 monomers are capable of binding LPL.


Asunto(s)
Hiperlipoproteinemia Tipo I/enzimología , Lipoproteína Lipasa/metabolismo , Mutación Missense , Receptores de Lipoproteína/metabolismo , Animales , Sitios de Unión , Células CHO , Cricetulus , Cisteína , Drosophila/citología , Drosophila/metabolismo , Células Endoteliales de la Vena Umbilical Humana/enzimología , Humanos , Hiperlipoproteinemia Tipo I/genética , Modelos Moleculares , Fosfoinositido Fosfolipasa C/metabolismo , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Ratas , Receptores de Lipoproteína/química , Receptores de Lipoproteína/genética , Transfección
3.
J Lipid Res ; 57(9): 1670-83, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27034464

RESUMEN

LPL hydrolyzes triglycerides in triglyceride-rich lipoproteins along the capillaries of heart, skeletal muscle, and adipose tissue. The activity of LPL is repressed by angiopoietin-like 4 (ANGPTL4) but the underlying mechanisms have not been fully elucidated. Our objective was to study the cellular location and mechanism for LPL inhibition by ANGPTL4. We performed studies in transfected cells, ex vivo studies, and in vivo studies with Angptl4(-/-) mice. Cotransfection of CHO pgsA-745 cells with ANGPTL4 and LPL reduced intracellular LPL protein levels, suggesting that ANGPTL4 promotes LPL degradation. This conclusion was supported by studies of primary adipocytes and adipose tissue explants from wild-type and Angptl4(-/-) mice. Absence of ANGPTL4 resulted in accumulation of the mature-glycosylated form of LPL and increased secretion of LPL. Blocking endoplasmic reticulum (ER)-Golgi transport abolished differences in LPL abundance between wild-type and Angptl4(-/-) adipocytes, suggesting that ANGPTL4 acts upon LPL after LPL processing in the ER. Finally, physiological changes in adipose tissue ANGPTL4 expression during fasting and cold resulted in inverse changes in the amount of mature-glycosylated LPL in wild-type mice, but not Angptl4(-/-) mice. We conclude that ANGPTL4 promotes loss of intracellular LPL by stimulating LPL degradation after LPL processing in the ER.


Asunto(s)
Adipocitos/metabolismo , Angiopoyetinas/metabolismo , Lipoproteína Lipasa/metabolismo , Lipoproteínas/genética , Triglicéridos/genética , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/genética , Animales , Citoplasma/metabolismo , Aparato de Golgi/metabolismo , Humanos , Lipoproteína Lipasa/genética , Ratones , Ratones Noqueados , Proteolisis , Triglicéridos/metabolismo
4.
J Lipid Res ; 57(10): 1889-1898, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27494936

RESUMEN

LPL contains two principal domains: an amino-terminal catalytic domain (residues 1-297) and a carboxyl-terminal domain (residues 298-448) that is important for binding lipids and binding glycosylphosphatidylinositol-anchored high density lipoprotein binding protein 1 (GPIHBP1) (an endothelial cell protein that shuttles LPL to the capillary lumen). The LPL sequences required for GPIHBP1 binding have not been examined in detail, but one study suggested that sequences near LPL's carboxyl terminus (residues ∼403-438) were crucial. Here, we tested the ability of LPL-specific monoclonal antibodies (mAbs) to block the binding of LPL to GPIHBP1. One antibody, 88B8, abolished LPL binding to GPIHBP1. Consistent with those results, antibody 88B8 could not bind to GPIHBP1-bound LPL on cultured cells. Antibody 88B8 bound poorly to LPL proteins with amino acid substitutions that interfered with GPIHBP1 binding (e.g., C418Y, E421K). However, the sequences near LPL's carboxyl terminus (residues ∼403-438) were not sufficient for 88B8 binding; upstream sequences (residues 298-400) were also required. Additional studies showed that these same sequences are required for LPL binding to GPIHBP1. In conclusion, we identified an LPL mAb that binds to LPL's GPIHBP1-binding domain. The binding of both antibody 88B8 and GPIHBP1 to LPL depends on large segments of LPL's carboxyl-terminal domain.


Asunto(s)
Anticuerpos Monoclonales de Origen Murino/química , Lipoproteína Lipasa/química , Receptores de Lipoproteína/química , Sustitución de Aminoácidos , Animales , Línea Celular , Drosophila melanogaster , Humanos , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Mutación Missense , Unión Proteica , Dominios Proteicos , Receptores de Lipoproteína/genética , Receptores de Lipoproteína/metabolismo
5.
J Biol Chem ; 289(28): 19491-9, 2014 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-24847059

RESUMEN

GPIHBP1, a glycosylphosphatidylinositol-anchored glycoprotein of microvascular endothelial cells, binds lipoprotein lipase (LPL) within the interstitial spaces and transports it across endothelial cells to the capillary lumen. The ability of GPIHBP1 to bind LPL depends on the Ly6 domain, a three-fingered structure containing 10 cysteines and a conserved pattern of disulfide bond formation. Here, we report a patient with severe hypertriglyceridemia who was homozygous for a GPIHBP1 point mutation that converted a serine in the GPIHBP1 Ly6 domain (Ser-107) to a cysteine. Two hypertriglyceridemic siblings were homozygous for the same mutation. All three homozygotes had very low levels of LPL in the preheparin plasma. We suspected that the extra cysteine in GPIHBP1-S107C might prevent the trafficking of the protein to the cell surface, but this was not the case. However, nearly all of the GPIHBP1-S107C on the cell surface was in the form of disulfide-linked dimers and multimers, whereas wild-type GPIHBP1 was predominantly monomeric. An insect cell GPIHBP1 expression system confirmed the propensity of GPIHBP1-S107C to form disulfide-linked dimers and to form multimers. Functional studies showed that only GPIHBP1 monomers bind LPL. In keeping with that finding, there was no binding of LPL to GPIHBP1-S107C in either cell-based or cell-free binding assays. We conclude that an extra cysteine in the GPIHBP1 Ly6 motif results in multimerization of GPIHBP1, defective LPL binding, and severe hypertriglyceridemia.


Asunto(s)
Homocigoto , Hiperlipoproteinemia Tipo I , Lipoproteína Lipasa/metabolismo , Mutación Missense , Multimerización de Proteína/genética , Receptores de Lipoproteína , Adulto , Sustitución de Aminoácidos , Línea Celular , Humanos , Hiperlipoproteinemia Tipo I/genética , Hiperlipoproteinemia Tipo I/metabolismo , Hiperlipoproteinemia Tipo I/patología , Lipoproteína Lipasa/genética , Masculino , Unión Proteica/genética , Estructura Terciaria de Proteína , Transporte de Proteínas/genética , Receptores de Lipoproteína/genética , Receptores de Lipoproteína/metabolismo
6.
Biochim Biophys Acta ; 1841(7): 970-6, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24681165

RESUMEN

Lipoprotein lipase (LPL) has been highly conserved through vertebrate evolution, making it challenging to generate useful antibodies. Some polyclonal antibodies against LPL have turned out to be nonspecific, and the available monoclonal antibodies (Mabs) against LPL, all of which bind to LPL's carboxyl terminus, have drawbacks for some purposes. We report a new LPL-specific monoclonal antibody, Mab 4-1a, which binds to the amino terminus of LPL (residues 5-25). Mab 4-1a binds human and bovine LPL avidly; it does not inhibit LPL catalytic activity nor does it interfere with the binding of LPL to heparin. Mab 4-1a does not bind to human hepatic lipase. Mab 4-1a binds to GPIHBP1-bound LPL and does not interfere with the ability of the LPL-GPIHBP1 complex to bind triglyceride-rich lipoproteins. Mab 4-1a will be a useful reagent for both biochemists and clinical laboratories.


Asunto(s)
Anticuerpos Monoclonales/metabolismo , Lipoproteína Lipasa/metabolismo , Receptores de Lipoproteína/metabolismo , Triglicéridos/metabolismo , Secuencia de Aminoácidos , Animales , Anticuerpos Monoclonales/biosíntesis , Anticuerpos Monoclonales/aislamiento & purificación , Afinidad de Anticuerpos , Especificidad de Anticuerpos , Células CHO , Bovinos , Cricetulus , Expresión Génica , Heparina/metabolismo , Humanos , Lipasa/metabolismo , Lipoproteína Lipasa/genética , Ratones , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Receptores de Lipoproteína/genética , Transfección
7.
Biochim Biophys Acta ; 1841(7): 963-9, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24704550

RESUMEN

The S447X polymorphism in lipoprotein lipase (LPL), which shortens LPL by two amino acids, is associated with low plasma triglyceride levels and reduced risk for coronary heart disease. S447X carriers have higher LPL levels in the pre- and post-heparin plasma, raising the possibility that the S447X polymorphism leads to higher LPL levels within capillaries. One potential explanation for increased amounts of LPL in capillaries would be more avid binding of S447X-LPL to GPIHBP1 (the protein that binds LPL dimers and shuttles them to the capillary lumen). This explanation seems plausible because sequences within the carboxyl terminus of LPL are known to mediate LPL binding to GPIHBP1. To assess the impact of the S447X polymorphism on LPL binding to GPIHBP1, we compared the ability of internally tagged versions of wild-type LPL (WT-LPL) and S447X-LPL to bind to GPIHBP1 in both cell-based and cell-free binding assays. In the cell-based assay, we compared the binding of WT-LPL and S447X-LPL to GPIHBP1 on the surface of cultured cells. This assay revealed no differences in the binding of WT-LPL and S447X-LPL to GPIHBP1. In the cell-free assay, we compared the binding of internally tagged WT-LPL and S447X-LPL to soluble GPIHBP1 immobilized on agarose beads. Again, no differences in the binding of WT-LPL and S447X-LPL to GPIHBP1 were observed. We conclude that increased binding of S447X-LPL to GPIHBP1 is unlikely to be the explanation for more efficient lipolysis and lower plasma triglyceride levels in S447X carriers.


Asunto(s)
Proteínas Inmovilizadas/metabolismo , Lipoproteína Lipasa/metabolismo , Polimorfismo de Nucleótido Simple , Receptores de Lipoproteína/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Triglicéridos/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Bioensayo , Células CHO , Cricetulus , Expresión Génica , Humanos , Proteínas Inmovilizadas/genética , Metabolismo de los Lípidos , Lipoproteína Lipasa/genética , Datos de Secuencia Molecular , Unión Proteica , Transporte de Proteínas , Receptores de Lipoproteína/genética , Proteínas Recombinantes de Fusión/genética
8.
Hum Mol Genet ; 21(13): 2961-72, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22493000

RESUMEN

Lipoprotein lipase (LPL) is a 448-amino-acid head-to-tail dimeric enzyme that hydrolyzes triglycerides within capillaries. LPL is secreted by parenchymal cells into the interstitial spaces; it then binds to GPIHBP1 (glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1) on the basolateral face of endothelial cells and is transported to the capillary lumen. A pair of amino acid substitutions, C418Y and E421K, abolish LPL binding to GPIHBP1, suggesting that the C-terminal portion of LPL is important for GPIHBP1 binding. However, a role for LPL's N terminus has not been excluded, and published evidence has suggested that only full-length homodimers are capable of binding GPIHBP1. Here, we show that LPL's C-terminal domain is sufficient for GPIHBP1 binding. We found, serendipitously, that two LPL missense mutations, G409R and E410V, render LPL susceptible to cleavage at residue 297 (a known furin cleavage site). The C terminus of these mutants (residues 298-448), bound to GPIHBP1 avidly, independent of the N-terminal fragment. We also generated an LPL construct with an in-frame deletion of the N-terminal catalytic domain (residues 50-289); this mutant was secreted but also was cleaved at residue 297. Once again, the C-terminal domain (residues 298-448) bound GPIHBP1 avidly. The binding of the C-terminal fragment to GPIHBP1 was eliminated by C418Y or E421K mutations. After exposure to denaturing conditions, the C-terminal fragment of LPL refolds and binds GPIHBP1 avidly. Thus, the binding of LPL to GPIHBP1 requires only the C-terminal portion of LPL and does not depend on full-length LPL homodimers.


Asunto(s)
Proteínas Portadoras/metabolismo , Lipoproteína Lipasa/metabolismo , Sustitución de Aminoácidos , Animales , Células CHO , Proteínas Portadoras/genética , Quilomicrones/sangre , Cricetinae , Células Endoteliales/metabolismo , Furina/metabolismo , Lipoproteína Lipasa/química , Lipoproteína Lipasa/genética , Mutación Missense , Péptidos/genética , Péptidos/metabolismo , Unión Proteica
9.
Arterioscler Thromb Vasc Biol ; 33(7): 1706-13, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23661675

RESUMEN

OBJECTIVE: Angiopoietin-like 3 (Angptl3) is a regulator of lipoprotein metabolism at least by inhibiting lipoprotein lipase activity. Loss-of-function mutations in ANGPTL3 cause familial combined hypolipidemia through an unknown mechanism. APPROACH AND RESULTS: We compared lipolytic activities, lipoprotein composition, and other lipid-related enzyme/lipid transfer proteins in carriers of the S17X loss-of-function mutation in ANGPTL3 and in age- and sex-matched noncarrier controls. Gel filtration analysis revealed a severely disturbed lipoprotein profile and a reduction in size and triglyceride content of very low density lipoprotein in homozygotes as compared with heterozygotes and noncarriers. S17X homozygotes had significantly higher lipoprotein lipase activity and mass in postheparin plasma, whereas heterozygotes showed no difference in these parameters when compared with noncarriers. No changes in hepatic lipase, endothelial lipase, paraoxonase 1, phospholipid transfer protein, and cholesterol ester transfer protein activities were associated with the S17X mutation. Plasma free fatty acid, insulin, glucose, and homeostatic model assessment of insulin resistance were significantly lower in homozygous subjects compared with heterozygotes and noncarriers subjects. CONCLUSIONS: These results indicate that, although partial Angptl3 deficiency did not affect the activities of lipolytic enzymes, the complete absence of Angptl3 results in an increased lipoprotein lipase activity and mass and low circulating free fatty acid levels. This latter effect is probably because of decreased mobilization of free fatty acid from fat stores in human adipose tissue and may result in reduced hepatic very low density lipoprotein synthesis and secretion via attenuated hepatic free fatty acid supply. Altogether, Angptl3 may affect insulin sensitivity and play a role in modulating both lipid and glucose metabolism.


Asunto(s)
Angiopoyetinas/deficiencia , Ácidos Grasos no Esterificados/sangre , Hipobetalipoproteinemias/enzimología , Resistencia a la Insulina , Lipoproteína Lipasa/sangre , Adulto , Anciano , Análisis de Varianza , Proteína 3 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Angiopoyetinas/genética , Biomarcadores/sangre , Glucemia/análisis , Estudios de Casos y Controles , Distribución de Chi-Cuadrado , Regulación hacia Abajo , Femenino , Heterocigoto , Homocigoto , Humanos , Hipobetalipoproteinemias/sangre , Hipobetalipoproteinemias/genética , Hipobetalipoproteinemias/fisiopatología , Insulina/sangre , Italia , Modelos Lineales , Lipasa/sangre , Lipoproteínas LDL/sangre , Masculino , Persona de Mediana Edad , Mutación , Triglicéridos/sangre , Regulación hacia Arriba
10.
Proc Natl Acad Sci U S A ; 108(19): 7980-4, 2011 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-21518912

RESUMEN

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


Asunto(s)
Proteínas Portadoras/metabolismo , Células Endoteliales/metabolismo , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Mutación Missense , Sustitución de Aminoácidos , Animales , Sitios de Unión/genética , Células CHO , Quilomicrones/sangre , Quilomicrones/genética , Cricetinae , Cricetulus , Humanos , Hiperlipoproteinemia Tipo IV/sangre , Hiperlipoproteinemia Tipo IV/enzimología , Hiperlipoproteinemia Tipo IV/genética , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Unión Proteica , Receptores de Lipoproteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transfección
11.
Arterioscler Thromb Vasc Biol ; 32(5): 1204-10, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22345169

RESUMEN

OBJECTIVE: Lipoprotein lipase (LPL) is a principal enzyme in lipoprotein metabolism, tissue lipid utilization, and energy metabolism. LPL is synthesized by parenchymal cells in adipose, heart, and muscle tissues followed by secretion to extracellular sites, where lipolyic function is exerted. The catalytic activity of LPL is attained during posttranslational maturation, which involves glycosylation, folding, and subunit assembly within the endoplasmic reticulum. A lipase-chaperone, lipase maturation factor 1 (Lmf1), has recently emerged as a critical factor in this process. Previous studies demonstrated that loss-of-function mutations of Lmf1 result in diminished lipase activity and severe hypertriglyceridemia in mice and human subjects. The objective of this study is to investigate whether, beyond its role as a required factor in lipase maturation, variation in Lmf1 expression is sufficient to modulate LPL activity in vivo. METHODS AND RESULTS: To assess the effects of Lmf1 overexpression in adipose and muscle tissues, we generated aP2-Lmf1 and Mck-Lmf1 transgenic mice. Characterization of relevant tissues revealed increased LPL activity in both mouse strains. In the omental and subcutaneous adipose depots, Lmf1 overexpression was associated with increased LPL specific activity without changes in LPL mass. In contrast, increased LPL activity was due to elevated LPL protein level in heart and gonadal adipose tissue. To extend these studies to humans, we detected association between LMF1 gene variants and postheparin LPL activity in a dyslipidemic cohort. CONCLUSIONS: Our results suggest that variation in Lmf1 expression is a posttranslational determinant of LPL activity.


Asunto(s)
ADN/genética , Metabolismo Energético/fisiología , Regulación de la Expresión Génica , Variación Genética , Hipertrigliceridemia/genética , Lipoproteína Lipasa/genética , Proteínas de la Membrana/genética , Tejido Adiposo/metabolismo , Animales , Humanos , Hipertrigliceridemia/metabolismo , Lipoproteína Lipasa/biosíntesis , Proteínas de la Membrana/biosíntesis , Ratones , Ratones Transgénicos , Músculo Esquelético/metabolismo , Miocardio/metabolismo
12.
Cell Metab ; 5(4): 279-91, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17403372

RESUMEN

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


Asunto(s)
Quilomicrones/metabolismo , Lipólisis/genética , Receptores de Lipoproteína/fisiología , Animales , Células CHO , Quilomicrones/sangre , Cricetinae , Cricetulus , Ingestión de Alimentos/fisiología , Células Endoteliales/metabolismo , Regulación de la Expresión Génica , Lipoproteína Lipasa/metabolismo , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Conejos , Receptores de Lipoproteína/genética , Receptores de Lipoproteína/metabolismo , Transfección
13.
J Lipid Res ; 53(12): 2690-7, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23008484

RESUMEN

Lipoprotein lipase (LPL) is secreted into the interstitial spaces by adipocytes and myocytes but then must be transported to the capillary lumen by GPIHBP1, a glycosylphosphatidylinositol-anchored protein of capillary endothelial cells. The mechanism by which GPIHBP1 and LPL move across endothelial cells remains unclear. We asked whether the transport of GPIHBP1 and LPL across endothelial cells was uni- or bidirectional. We also asked whether GPIHBP1 and LPL are transported across cells in vesicles and whether this transport process requires caveolin-1. The movement of GPIHBP1 and LPL across cultured endothelial cells was bidirectional. Also, GPIHBP1 moved bidirectionally across capillary endothelial cells in live mice. The transport of LPL across endothelial cells was inhibited by dynasore and genistein, consistent with a vesicular transport process. Also, transmission electron microscopy (EM) and dual-axis EM tomography revealed GPIHBP1 and LPL in invaginations of the plasma membrane and in vesicles. The movement of GPIHBP1 across capillary endothelial cells was efficient in the absence of caveolin-1, and there was no defect in the internalization of LPL by caveolin-1-deficient endothelial cells in culture. Our studies show that GPIHBP1 and LPL move bidirectionally across endothelial cells in vesicles and that transport is efficient even when caveolin-1 is absent.


Asunto(s)
Células Endoteliales/metabolismo , Lipoproteína Lipasa/metabolismo , Receptores de Lipoproteína/metabolismo , Animales , Células CHO , Cricetinae , Células Endoteliales/química , Células Endoteliales/enzimología , Genisteína/farmacología , Humanos , Hidrazonas/farmacología , Lipoproteína Lipasa/antagonistas & inhibidores , Ratones , Ratones Noqueados , Ratas , Receptores de Lipoproteína/deficiencia , Relación Estructura-Actividad
14.
J Biol Chem ; 286(22): 19735-43, 2011 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-21478160

RESUMEN

Glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1 (GPIHBP1) is an endothelial cell protein that transports lipoprotein lipase (LPL) from the subendothelial spaces to the capillary lumen. GPIHBP1 contains two main structural motifs, an amino-terminal acidic domain enriched in aspartates and glutamates and a lymphocyte antigen 6 (Ly6) motif containing 10 cysteines. All of the cysteines in the Ly6 domain are disulfide-bonded, causing the protein to assume a three-fingered structure. The acidic domain of GPIHBP1 is known to be important for LPL binding, but the involvement of the Ly6 domain in LPL binding requires further study. To assess the importance of the Ly6 domain, we created a series of GPIHBP1 mutants in which each residue of the Ly6 domain was changed to alanine. The mutant proteins were expressed in Chinese hamster ovary (CHO) cells, and their expression level on the cell surface and their ability to bind LPL were assessed with an immunofluorescence microscopy assay and a Western blot assay. We identified 12 amino acids within GPIHBP1, aside from the conserved cysteines, that are important for LPL binding; nine of those were clustered in finger 2 of the GPIHBP1 three-fingered motif. The defective GPIHBP1 proteins also lacked the ability to transport LPL from the basolateral to the apical surface of endothelial cells. Our studies demonstrate that the Ly6 domain of GPIHBP1 is important for the ability of GPIHBP1 to bind and transport LPL.


Asunto(s)
Proteínas Portadoras/metabolismo , Células Endoteliales/metabolismo , Lipoproteína Lipasa/metabolismo , Sustitución de Aminoácidos , Animales , Células CHO , Proteínas Portadoras/genética , Cricetinae , Cricetulus , Humanos , Lipoproteína Lipasa/genética , Mutación Missense , Mapeo Peptídico/métodos , Estructura Terciaria de Proteína , Transporte de Proteínas/fisiología , Receptores de Lipoproteína
15.
J Inherit Metab Dis ; 35(3): 531-40, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22008945

RESUMEN

Lipoprotein lipase (LPL) is a hydrolase that cleaves circulating triglycerides to release fatty acids to the surrounding tissues. The enzyme is synthesized in parenchymal cells and is transported to its site of action on the capillary endothelium by glycophosphatidylinositol (GPI)-anchored high-density lipoprotein-binding protein 1 (GPIHBP1). Inactivating mutations in LPL; in its cofactor, apolipoprotein (Apo) C2; or in GPIHBP1 cause severe hypertriglyceridemia. Here we describe an individual with complete deficiency of GPIHBP1. The proband was an Asian Indian boy who had severe chylomicronemia at 2 months of age. Array-based copy-number analysis of his genomic DNA revealed homozygosity for a 17.5-kb deletion that included GPIHBP1. A 44-year-old aunt with a history of hypertriglyceridemia and pancreatitis was also homozygous for the deletion. A bolus of intravenously administered heparin caused a rapid increase in circulating LPL and decreased plasma triglyceride levels in control individuals but not in two GPIHBP1-deficient patients. Thus, short-term treatment with heparin failed to attenuate the hypertriglyceridemia in patients with GPIHBP1 deficiency. The increasing resolution of copy number microarrays and their widespread adoption for routine cytogenetic analysis is likely to reveal a greater role for submicroscopic deletions in Mendelian conditions. We describe the first neonate with complete GPIHBP1 deficiency due to homozygosity for a deletion of GPIHBP1.


Asunto(s)
Proteínas Portadoras/genética , Quilomicrones/química , Eliminación de Gen , Hipertrigliceridemia/genética , Adulto , Apolipoproteína C-II/metabolismo , Niño , Preescolar , Citogenética , Femenino , Dosificación de Gen , Heparina/metabolismo , Homocigoto , Humanos , Hipertrigliceridemia/etiología , Lactante , Lipoproteína Lipasa/química , Masculino , Persona de Mediana Edad , Mutación , Receptores de Lipoproteína
16.
Arterioscler Thromb Vasc Biol ; 31(1): 176-82, 2011 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20966398

RESUMEN

OBJECTIVE: To define the ability of GPIHBP1 to bind other lipase family members and other apolipoproteins (apos) and lipoproteins. METHODS AND RESULTS: GPIHBP1, a GPI-anchored lymphocyte antigen (Ly)6 protein of capillary endothelial cells, binds lipoprotein lipase (LPL) avidly, but its ability to bind related lipase family members has never been evaluated. As judged by cell-based and cell-free binding assays, LPL binds to GPIHBP1, but other members of the lipase family do not. We also examined the binding of apoAV-phospholipid disks to GPIHBP1. ApoAV binds avidly to GPIHBP1-transfected cells; this binding requires GPIHBP1's amino-terminal acidic domain and is independent of its cysteine-rich Ly6 domain (the latter domain is essential for LPL binding). GPIHBP1-transfected cells did not bind high-density lipoprotein. Chylomicrons bind avidly to GPIHBP1-transfected Chinese hamster ovary cells, but this binding is dependent on GPIHBP1's ability to bind LPL within the cell culture medium. CONCLUSIONS: GPIHBP1 binds LPL but does not bind other lipase family members. GPIHBP1 binds apoAV but does not bind apoAI or high-density lipoprotein. The ability of GPIHBP1-transfected Chinese hamster ovary cells to bind chylomicrons is mediated by LPL; chylomicron binding does not occur unless GPIHBP1 first captures LPL from the cell culture medium.


Asunto(s)
Capilares/metabolismo , Proteínas Portadoras/metabolismo , Células Endoteliales/metabolismo , Animales , Apolipoproteínas A/metabolismo , Células CHO , Capilares/citología , Proteínas Portadoras/genética , Quilomicrones/metabolismo , Cricetinae , Cricetulus , Humanos , Lipoproteína Lipasa/metabolismo , Lipoproteínas HDL/metabolismo , Mutación , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Mapeo de Interacción de Proteínas , Receptores de Lipoproteína , Proteínas Recombinantes de Fusión/metabolismo , Transfección
17.
BMC Physiol ; 12: 14, 2012 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-23186339

RESUMEN

BACKGROUND: Lipoprotein lipase (LPL) hydrolyzes triglycerides in plasma lipoproteins and enables uptake of lipolysis products for energy production or storage in tissues. Our aim was to study the localization of LPL and its endothelial anchoring protein glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1 (GPIHBP1) in mouse pancreas, and effects of diet and leptin deficiency on their expression patterns. For this, immunofluorescence microscopy was used on pancreatic tissue from C57BL/6 mouse embryos (E18), adult mice on normal or high-fat diet, and adult ob/ob-mice treated or not with leptin. The distribution of LPL and GPIHBP1 was compared to insulin, glucagon and CD31. Heparin injections were used to discriminate between intracellular and extracellular LPL. RESULTS: In the exocrine pancreas LPL was found in capillaries, and was mostly co-localized with GPIHBP1. LPL was releasable by heparin, indicating localization on cell surfaces. Within the islets, most of the LPL was associated with beta cells and could not be released by heparin, indicating that the enzyme remained mostly within cells. Staining for LPL was found also in the glucagon-producing alpha cells, both in embryos (E18) and in adult mice. Only small amounts of LPL were found together with GPIHBP1 within the capillaries of islets. Neither a high fat diet nor fasting/re-feeding markedly altered the distribution pattern of LPL or GPIHBP1 in mouse pancreas. Islets from ob/ob mice appeared completely deficient of LPL in the beta cells, while LPL-staining was normal in alpha cells and in the exocrine pancreas. Leptin treatment of ob/ob mice for 12 days reversed this pattern, so that most of the islets expressed LPL in beta cells. CONCLUSIONS: We conclude that both LPL and GPIHBP1 are present in mouse pancreas, and that LPL expression in beta cells is dependent on leptin.


Asunto(s)
Leptina/deficiencia , Leptina/metabolismo , Lipoproteína Lipasa/metabolismo , Páncreas/metabolismo , Receptores de Lipoproteína/metabolismo , Animales , Capilares/metabolismo , Dieta Alta en Grasa/métodos , Células Endoteliales/metabolismo , Glucagón/metabolismo , Células Secretoras de Glucagón/metabolismo , Glicosilfosfatidilinositoles/metabolismo , Heparina/metabolismo , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo
18.
J Lipid Res ; 52(11): 1869-84, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21844202

RESUMEN

Interest in lipolysis and the metabolism of triglyceride-rich lipoproteins was recently reignited by the discovery of severe hypertriglyceridemia (chylomicronemia) in glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1 (GPIHBP1)-deficient mice. GPIHBP1 is expressed exclusively in capillary endothelial cells and binds lipoprotein lipase (LPL) avidly. These findings prompted speculation that GPIHBP1 serves as a binding site for LPL in the capillary lumen, creating "a platform for lipolysis." More recent studies have identified a second and more intriguing role for GPIHBP1-picking up LPL in the subendothelial spaces and transporting it across endothelial cells to the capillary lumen. Here, we review the studies that revealed that GPIHBP1 is the LPL transporter and discuss which amino acid sequences are required for GPIHBP1-LPL interactions. We also discuss the human genetics of LPL transport, focusing on cases of chylomicronemia caused by GPIHBP1 mutations that abolish GPIHBP1's ability to bind LPL, and LPL mutations that prevent LPL binding to GPIHBP1.


Asunto(s)
Proteínas Portadoras/metabolismo , Células Endoteliales/metabolismo , Lipoproteína Lipasa/metabolismo , Secuencia de Aminoácidos , Animales , Células CHO , Proteínas Portadoras/química , Proteínas Portadoras/genética , Cricetinae , Cricetulus , Humanos , Datos de Secuencia Molecular , Transporte de Proteínas , Receptores de Lipoproteína
19.
J Biol Chem ; 285(50): 39239-48, 2010 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-20889497

RESUMEN

Glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1 (GPIHBP1), a GPI-anchored endothelial cell protein, binds lipoprotein lipase (LPL) and transports it into the lumen of capillaries where it hydrolyzes triglycerides in lipoproteins. GPIHBP1 is assumed to be expressed mainly within the heart, skeletal muscle, and adipose tissue, the sites where most lipolysis occurs, but the tissue pattern of GPIHBP1 expression has never been evaluated systematically. Because GPIHBP1 is found on the luminal face of capillaries, we predicted that it would be possible to define GPIHBP1 expression patterns with radiolabeled GPIHBP1-specific antibodies and positron emission tomography (PET) scanning. In Gpihbp1(-/-) mice, GPIHBP1-specific antibodies were cleared slowly from the blood, and PET imaging showed retention of the antibodies in the blood pools (heart and great vessels). In Gpihbp1(+/+) mice, the antibodies were cleared extremely rapidly from the blood and, to our surprise, were taken up mainly by lung and liver. Immunofluorescence microscopy confirmed the presence of GPIHBP1 in the capillary endothelium of both lung and liver. In most tissues with high levels of Gpihbp1 expression, Lpl expression was also high, but the lung was an exception (very high Gpihbp1 expression and extremely low Lpl expression). Despite low Lpl transcript levels, however, LPL protein was readily detectable in the lung, suggesting that some of that LPL originates elsewhere and then is captured by GPIHBP1 in the lung. In support of this concept, lung LPL levels were significantly lower in Gpihbp1(-/-) mice than in Gpihbp1(+/+) mice. In addition, Lpl(-/-) mice expressing human LPL exclusively in muscle contained high levels of human LPL in the lung.


Asunto(s)
Regulación de la Expresión Génica , Glicosilfosfatidilinositoles/metabolismo , Receptores de Lipoproteína/química , Animales , Sitios de Unión , Capilares/metabolismo , Membrana Celular/metabolismo , Endotelio/metabolismo , Cinética , Pulmón/metabolismo , Ratones , Ratones Transgénicos , Modelos Biológicos , Tomografía de Emisión de Positrones/métodos
20.
Arterioscler Thromb Vasc Biol ; 30(11): 2106-13, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20814015

RESUMEN

OBJECTIVE: To determine whether plasma triglyceride levels in adult Glycosylphosphatidylinositol HDL-binding protein 1 (GPIHBP1)-deficient (Gpihbp1(-/-)) mice would be sensitive to cholesterol intake. METHODS AND RESULTS: Gpihbp1(-/-) mice were fed a Western diet containing 0.15% cholesterol. After 4 to 8 weeks, their plasma triglyceride levels were 113 to 135 mmol/L. When 0.005% ezetimibe was added to the diet to block cholesterol absorption, Lpl expression in the liver was reduced significantly, and the plasma triglyceride levels were significantly higher (>170 mmol/L). We also assessed plasma triglyceride levels in Gpihbp1(-/-) mice fed Western diets containing either high (1.3%) or low (0.05%) amounts of cholesterol. The high-cholesterol diet significantly increased Lpl expression in the liver and lowered plasma triglyceride levels. CONCLUSIONS: Treatment of Gpihbp1(-/-) mice with ezetimibe lowers Lpl expression in the liver and increases plasma triglyceride levels. A high-cholesterol diet had the opposite effects. Thus, cholesterol intake modulates plasma triglyceride levels in Gpihbp1(-/-) mice.


Asunto(s)
Anticolesterolemiantes/farmacología , Azetidinas/farmacología , Colesterol/metabolismo , Grasas de la Dieta/metabolismo , Receptores de Lipoproteína/deficiencia , Triglicéridos/metabolismo , Animales , Modelos Animales de Enfermedad , Ezetimiba , Hígado/efectos de los fármacos , Hígado/metabolismo , Ratones , Triglicéridos/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA