Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 45
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
N Engl J Med ; 376(17): 1647-1658, 2017 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-28402248

RESUMEN

BACKGROUND: A protein that is expressed on capillary endothelial cells, called GPIHBP1 (glycosylphosphatidylinositol-anchored high-density lipoprotein binding protein 1), binds lipoprotein lipase and shuttles it to its site of action in the capillary lumen. A deficiency in GPIHBP1 prevents lipoprotein lipase from reaching the capillary lumen. Patients with GPIHBP1 deficiency have low plasma levels of lipoprotein lipase, impaired intravascular hydrolysis of triglycerides, and severe hypertriglyceridemia (chylomicronemia). During the characterization of a monoclonal antibody-based immunoassay for GPIHBP1, we encountered two plasma samples (both from patients with chylomicronemia) that contained an interfering substance that made it impossible to measure GPIHBP1. That finding raised the possibility that those samples might contain GPIHBP1 autoantibodies. METHODS: Using a combination of immunoassays, Western blot analyses, and immunocytochemical studies, we tested the two plasma samples (as well as samples from other patients with chylomicronemia) for the presence of GPIHBP1 autoantibodies. We also tested the ability of GPIHBP1 autoantibodies to block the binding of lipoprotein lipase to GPIHBP1. RESULTS: We identified GPIHBP1 autoantibodies in six patients with chylomicronemia and found that these autoantibodies blocked the binding of lipoprotein lipase to GPIHBP1. As in patients with GPIHBP1 deficiency, those with GPIHBP1 autoantibodies had low plasma levels of lipoprotein lipase. Three of the six patients had systemic lupus erythematosus. One of these patients who had GPIHBP1 autoantibodies delivered a baby with plasma containing maternal GPIHBP1 autoantibodies; the infant had severe but transient chylomicronemia. Two of the patients with chylomicronemia and GPIHBP1 autoantibodies had a response to treatment with immunosuppressive agents. CONCLUSIONS: In six patients with chylomicronemia, GPIHBP1 autoantibodies blocked the ability of GPIHBP1 to bind and transport lipoprotein lipase, thereby interfering with lipoprotein lipase-mediated processing of triglyceride-rich lipoproteins and causing severe hypertriglyceridemia. (Funded by the National Heart, Lung, and Blood Institute and the Leducq Foundation.).


Asunto(s)
Autoanticuerpos/sangre , Hiperlipoproteinemia Tipo I/inmunología , Lipoproteína Lipasa/metabolismo , Receptores de Lipoproteína/inmunología , Adulto , Autoanticuerpos/fisiología , Femenino , Humanos , Hiperlipoproteinemia Tipo I/sangre , Inmunoensayo , Lipólisis , Lipoproteína Lipasa/sangre , Masculino , Persona de Mediana Edad , Unión Proteica , Transporte de Proteínas , Receptores de Lipoproteína/metabolismo
2.
J Lipid Res ; 59(4): 706-713, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29449313

RESUMEN

Glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1 (GPIHBP1), an endothelial cell protein, binds LPL in the subendothelial spaces and transports it to the capillary lumen. In Gpihbp1-/- mice, LPL remains stranded in the subendothelial spaces, causing hypertriglyceridemia, but how Gpihbp1-/- mice respond to metabolic stress (e.g., cold exposure) has never been studied. In wild-type mice, cold exposure increases LPL-mediated processing of triglyceride-rich lipoproteins (TRLs) in brown adipose tissue (BAT), providing fuel for thermogenesis and leading to lower plasma triglyceride levels. We suspected that defective TRL processing in Gpihbp1-/- mice might impair thermogenesis and blunt the fall in plasma triglyceride levels. Indeed, Gpihbp1-/- mice exhibited cold intolerance, but the effects on plasma triglyceride levels were paradoxical. Rather than falling, the plasma triglyceride levels increased sharply (from ∼4,000 to ∼15,000 mg/dl), likely because fatty acid release by peripheral tissues drives hepatic production of TRLs that cannot be processed. We predicted that the sharp increase in plasma triglyceride levels would not occur in Gpihbp1-/-Angptl4-/- mice, where LPL activity is higher and baseline plasma triglyceride levels are lower. Indeed, the plasma triglyceride levels in Gpihbp1-/-Angptl4-/- mice fell during cold exposure. Metabolic studies revealed increased levels of TRL processing in the BAT of Gpihbp1-/-Angptl4-/- mice.


Asunto(s)
Frío , Receptores de Lipoproteína/sangre , Receptores de Lipoproteína/deficiencia , Termogénesis , Triglicéridos/sangre , Animales , Apolipoproteínas B/sangre , Ratones , Ratones Noqueados
3.
J Lipid Res ; 58(5): 840-852, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28159869

RESUMEN

Plasma apoC-III levels correlate with triglyceride (TG) levels and are a strong predictor of CVD outcomes. ApoC-III elevates TG in part by inhibiting LPL. ApoC-III likely inhibits LPL by competing for lipid binding. To probe this, we used oil-drop tensiometry to characterize binding of six apoC-III variants to lipid/water interfaces. This technique monitors the dependence of lipid binding on surface pressure, which increases during TG hydrolysis by LPL. ApoC-III adsorption increased surface pressure by upward of 18 mN/m at phospholipid/TG/water interfaces. ApoC-III was retained to high pressures at these interfaces, desorbing at 21-25 mN/m. Point mutants, which substituted alanine for aromatic residues, impaired the lipid binding of apoC-III. Adsorption and retention pressures decreased by 1-6 mN/m in point mutants, with the magnitude determined by the location of alanine substitutions. Trp42 was most critical to mediating lipid binding. These results strongly correlate with our previous results, linking apoC-III point mutants to increased LPL binding and activity at lipid surfaces. We propose that aromatic residues in the C-terminal half of apoC-III mediate binding to TG-rich lipoproteins. Increased apoC-III expression in the hypertriglyceridemic state allows apoC-III to accumulate on lipoproteins and inhibit LPL by preventing binding and/or access to substrate.


Asunto(s)
Apolipoproteína C-II/química , Apolipoproteína C-II/metabolismo , Metabolismo de los Lípidos , Lipoproteína Lipasa/antagonistas & inhibidores , Adsorción , Secuencia de Aminoácidos , Apolipoproteína C-II/genética , Humanos , Mutación , Relación Estructura-Actividad , Triglicéridos/metabolismo
4.
J Lipid Res ; 58(9): 1893-1902, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28694296

RESUMEN

apoC-III is often assumed to retard the intravascular processing of triglyceride-rich lipoproteins (TRLs) by inhibiting LPL, but that view is based largely on studies of free LPL. We now recognize that intravascular LPL is neither free nor loosely bound, but instead is tightly bound to glycosylphosphatidylinositol-anchored HDL-binding protein 1 (GPIHBP1) on endothelial cells. Here, we revisited the effects of apoC-III on LPL, focusing on apoC-III's capacity to affect the activity of GPIHBP1-bound LPL. We found that TRLs from APOC3 transgenic mice bound normally to GPIHBP1-bound LPL on cultured cells in vitro and to heart capillaries in vivo. However, the triglycerides in apoC-III-enriched TRLs were hydrolyzed more slowly by free LPL, and the inhibitory effect of apoC-III on triglyceride lipolysis was exaggerated when LPL was bound to GPIHBP1 on the surface of agarose beads. Also, recombinant apoC-III reduced triglyceride hydrolysis by free LPL only modestly, but the inhibitory effect was greater when the LPL was bound to GPIHBP1. A mutant apoC-III associated with low plasma triglyceride levels (p.A23T) displayed a reduced capacity to inhibit free and GPIHBP1-bound LPL. Our results show that apoC-III potently inhibits triglyceride hydrolysis when LPL is bound to GPIHBP1.


Asunto(s)
Apolipoproteína C-III/metabolismo , Lipoproteína Lipasa/metabolismo , Receptores de Lipoproteína/metabolismo , Triglicéridos/metabolismo , Animales , Células CHO , Cricetulus , Humanos , Hidrólisis , Ratones , Unión Proteica
5.
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
6.
J Lipid Res ; 58(7): 1453-1461, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28476858

RESUMEN

Mutation of conserved cysteines in proteins of the Ly6 family cause human disease-chylomicronemia in the case of glycosylphosphatidylinositol-anchored HDL binding protein 1 (GPIHBP1) and paroxysmal nocturnal hemoglobinuria in the case of CD59. A mutation in a conserved cysteine in CD59 prevented the protein from reaching the surface of blood cells. In contrast, mutation of conserved cysteines in human GPIHBP1 had little effect on GPIHBP1 trafficking to the surface of cultured CHO cells. The latter findings were somewhat surprising and raised questions about whether CHO cell studies accurately model the fate of mutant GPIHBP1 proteins in vivo. To explore this concern, we created mice harboring a GPIHBP1 cysteine mutation (p.C63Y). The p.C63Y mutation abolished the ability of mouse GPIHBP1 to bind LPL, resulting in severe chylomicronemia. The mutant GPIHBP1 was detectable by immunohistochemistry on the surface of endothelial cells, but the level of expression was ∼70% lower than in WT mice. The mutant GPIHBP1 protein in mouse tissues was predominantly monomeric. We conclude that mutation of a conserved cysteine in GPIHBP1 abolishes the ability of GPIHBP1 to bind LPL, resulting in mislocalization of LPL and severe chylomicronemia. The mutation reduced but did not eliminate GPIHBP1 on the surface of endothelial cells in vivo.


Asunto(s)
Secuencia Conservada , Cisteína , Lipoproteína Lipasa/metabolismo , Mutación , Receptores de Lipoproteína/química , Receptores de Lipoproteína/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Femenino , Humanos , Lipoproteína Lipasa/genética , Ratones , Unión Proteica/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Lipoproteína/genética , Triglicéridos/sangre
7.
J Lipid Res ; 58(1): 208-215, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27875259

RESUMEN

GPIHBP1, an endothelial cell protein, binds LPL in the interstitial spaces and shuttles it to its site of action inside blood vessels. For years, studies of human GPIHBP1 have been hampered by an absence of useful antibodies. We reasoned that monoclonal antibodies (mAbs) against human GPIHBP1 would be useful for 1) defining the functional relevance of GPIHBP1's Ly6 and acidic domains to the binding of LPL; 2) ascertaining whether human GPIHBP1 is expressed exclusively in capillary endothelial cells; and 3) testing whether GPIHBP1 is detectable in human plasma. Here, we report the development of a panel of human GPIHBP1-specific mAbs. Two mAbs against GPIHBP1's Ly6 domain, RE3 and RG3, abolished LPL binding, whereas an antibody against the acidic domain, RF4, did not. Also, mAbs RE3 and RG3 bound with reduced affinity to a mutant GPIHBP1 containing an Ly6 domain mutation (W109S) that abolishes LPL binding. Immunohistochemistry studies with the GPIHBP1 mAbs revealed that human GPIHBP1 is expressed only in capillary endothelial cells. Finally, we created an ELISA that detects GPIHBP1 in human plasma. That ELISA should make it possible for clinical lipidologists to determine whether plasma GPIHBP1 levels are a useful biomarker of metabolic or vascular disease.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Lipoproteína Lipasa/inmunología , Receptores de Lipoproteína/inmunología , Triglicéridos/metabolismo , Animales , Sitios de Unión/inmunología , Línea Celular , Drosophila , Células Endoteliales/enzimología , Células Endoteliales/inmunología , Humanos , Lipoproteína Lipasa/antagonistas & inhibidores , Lipoproteína Lipasa/aislamiento & purificación , Ratones , Receptores de Lipoproteína/genética , Triglicéridos/inmunología
8.
Biofouling ; 33(2): 184-194, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28198663

RESUMEN

Shortly after a surface is submerged in the sea, a conditioning film is generally formed by adsorption of organic molecules, such as polysaccharides. This could affect transport of molecules and ions between the seawater and the surface. An artificial seawater model system was developed to understand how adsorbed polysaccharides impact copper binding by glutaraldehyde-crosslinked polyethyleneimine coatings. Coating performance was also determined when competed against copper-chelating EDTA. Polysaccharide adsorption and copper binding and distribution were investigated using advanced analytical techniques, including depth-resolved time-of-flight secondary ion mass spectroscopy, grazing incidence X-ray absorption near-edge spectroscopy, quartz crystal microbalance with dissipation monitoring and X-ray photoelectron spectroscopy. In artificial seawater, the polysaccharides adsorbed in a swollen state that copper readily penetrated and the glutaraldehyde-polyethyleneimine coatings outcompeted EDTA for copper binding. Furthermore, the depth distribution of copper species was determined with nanometre precision. The results are highly relevant for copper-binding and copper-releasing materials in seawater.


Asunto(s)
Cobre/análisis , Ácido Edético/química , Glutaral/química , Polietileneimina/química , Polisacáridos/química , Agua de Mar/química , Adsorción , Incrustaciones Biológicas/prevención & control , Reactivos de Enlaces Cruzados/química , Iones , Ligandos , Modelos Químicos , Propiedades de Superficie , Contaminación Química del Agua/prevención & control
9.
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
10.
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
11.
J Biol Chem ; 290(29): 18029-18044, 2015 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-26026161

RESUMEN

Apolipoprotein C-II (apoC-II) is the co-factor for lipoprotein lipase (LPL) at the surface of triacylglycerol-rich lipoproteins. LPL hydrolyzes triacylglycerol, which increases local surface pressure as surface area decreases and amphipathic products transiently accumulate at the lipoprotein surface. To understand how apoC-II adapts to these pressure changes, we characterized the behavior of apoC-II at multiple lipid/water interfaces. ApoC-II adsorption to a triacylglycerol/water interface resulted in large increases in surface pressure. ApoC-II was exchangeable at this interface and desorbed on interfacial compressions. These compressions increase surface pressure and mimic the action of LPL. Analysis of gradual compressions showed that apoC-II undergoes a two-step desorption, which indicates that lipid-bound apoC-II can exhibit at least two conformations. We characterized apoC-II at phospholipid/triacylglycerol/water interfaces, which more closely mimic lipoprotein surfaces. ApoC-II had a large exclusion pressure, similar to that of apoC-I and apoC-III. However, apoC-II desorbed at retention pressures higher than those seen with the other apoCs. This suggests that it is unlikely that apoC-I and apoC-III inhibit LPL via displacement of apoC-II from the lipoprotein surface. Upon rapid compressions and re-expansions, re-adsorption of apoC-II increased pressure by lower amounts than its initial adsorption. This indicates that apoC-II removed phospholipid from the interface upon desorption. These results suggest that apoC-II regulates the activity of LPL in a pressure-dependent manner. ApoC-II is provided as a component of triacylglycerol-rich lipoproteins and is the co-factor for LPL as pressure increases. Above its retention pressure, apoC-II desorbs and removes phospholipid. This triggers release of LPL from lipoproteins.


Asunto(s)
Apolipoproteína C-II/metabolismo , Lipoproteína Lipasa/metabolismo , Adsorción , Secuencia de Aminoácidos , Apolipoproteína C-II/química , Humanos , Metabolismo de los Lípidos , Datos de Secuencia Molecular , Fosfolípidos/metabolismo , Presión , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Propiedades de Superficie , Agua/metabolismo
12.
J Biol Chem ; 290(22): 13919-34, 2015 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-25873395

RESUMEN

GPIHBP1 is an endothelial membrane protein that transports lipoprotein lipase (LPL) from the subendothelial space to the luminal side of the capillary endothelium. Here, we provide evidence that two regions of GPIHBP1, the acidic N-terminal domain and the central Ly6 domain, interact with LPL as two distinct binding sites. This conclusion is based on comparative binding studies performed with a peptide corresponding to the N-terminal domain of GPIHBP1, the Ly6 domain of GPIHBP1, wild type GPIHBP1, and the Ly6 domain mutant GPIHBP1 Q114P. Although LPL and the N-terminal domain formed a tight but short lived complex, characterized by fast on- and off-rates, the complex between LPL and the Ly6 domain formed more slowly and persisted for a longer time. Unlike the interaction of LPL with the Ly6 domain, the interaction of LPL with the N-terminal domain was significantly weakened by salt. The Q114P mutant bound LPL similarly to the N-terminal domain of GPIHBP1. Heparin dissociated LPL from the N-terminal domain, and partially from wild type GPIHBP1, but was unable to elute the enzyme from the Ly6 domain. When LPL was in complex with the acidic peptide corresponding to the N-terminal domain of GPIHBP1, the enzyme retained its affinity for the Ly6 domain. Furthermore, LPL that was bound to the N-terminal domain interacted with lipoproteins, whereas LPL bound to the Ly6 domain did not. In summary, our data suggest that the two domains of GPIHBP1 interact independently with LPL and that the functionality of LPL depends on its localization on GPIHBP1.


Asunto(s)
Glicosilfosfatidilinositoles/química , Lipoproteína Lipasa/química , Lipoproteínas/química , Receptores de Lipoproteína/química , Animales , Anisotropía , Sitios de Unión , Bovinos , Reactivos de Enlaces Cruzados/química , Endotelio Vascular/metabolismo , Epítopos/química , Colorantes Fluorescentes/química , Heparina/química , Humanos , Concentración de Iones de Hidrógeno , Espectrometría de Masas , Ratones , Mutación , Péptidos/química , Unión Proteica , Mapeo de Interacción de Proteínas , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Ratas , Resonancia por Plasmón de Superficie
13.
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
14.
Mol Pharm ; 12(4): 1242-9, 2015 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-25760774

RESUMEN

Targeting controlled release core-shell nanocarriers with the potential to overcome multidrug resistant (MDR) lung cancer were prepared based on demethoxycurcumin (DMC) loaded amphiphilic chitosan nanoparticles coated with an anti-EGFR antibody layer. The nanocarriers were characterized with regard to size with dynamic light scattering, SEM, and TEM. The characterization confirmed the nanocarriers to have a surface coating of the anti-EGFR antibody and a final size excellently suited for circulating targeting nanocarriers, i.e., <200 nm in diameter. In vitro drug release revealed extended quasi-Fickian release from the nanocarriers, with the anti-EGFR layer further reducing the release rate. Cell culture experiments using normoxic and MDR hypoxic cells overexpressing EGFR confirmed improved DMC delivery for anti-EGFR coated particles and revealed that the DMC was delivered to the cytoplasmic region of the cells, forming nanoprecipitates in lysosomes and endosomes. The effective endocytosis and targeting of the core-shell nanoparticles resulted in the nanocarriers achieving high cytotoxicity also against MDR cells. The therapeutic potential was further confirmed in an A549 xenograft lung tumor mouse model, where DMC loaded core-shell nanocarriers achieved about 8-fold reduction in tumor volume compared with control group over the 8 weeks of the investigation. Both in vitro and in vivo data suggest the anti-EGFR coated core-shell nanocarriers as highly promising for treatment of hypoxic MDR cancers, especially for non-small cell lung cancer.


Asunto(s)
Quitosano/química , Curcumina/análogos & derivados , Portadores de Fármacos , Nanopartículas/química , Animales , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Línea Celular Tumoral , Curcumina/química , Citoplasma/metabolismo , Diarilheptanoides , Sistemas de Liberación de Medicamentos , Receptores ErbB/metabolismo , Humanos , Concentración 50 Inhibidora , Luz , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Nanomedicina , Trasplante de Neoplasias , Dispersión de Radiación
15.
J Biol Chem ; 288(47): 33997-34008, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24121499

RESUMEN

Apolipoproteins (apo) C-I and C-III are known to inhibit lipoprotein lipase (LPL) activity, but the molecular mechanisms for this remain obscure. We present evidence that either apoC-I or apoC-III, when bound to triglyceride-rich lipoproteins, prevent binding of LPL to the lipid/water interface. This results in decreased lipolytic activity of the enzyme. Site-directed mutagenesis revealed that hydrophobic amino acid residues centrally located in the apoC-III molecule are critical for attachment to lipid emulsion particles and consequently inhibition of LPL activity. Triglyceride-rich lipoproteins stabilize LPL and protect the enzyme from inactivating factors such as angiopoietin-like protein 4 (angptl4). The addition of either apoC-I or apoC-III to triglyceride-rich particles severely diminished their protective effect on LPL and rendered the enzyme more susceptible to inactivation by angptl4. These observations were seen using chylomicrons as well as the synthetic lipid emulsion Intralipid. In the presence of the LPL activator protein apoC-II, more of apoC-I or apoC-III was needed for displacement of LPL from the lipid/water interface. In conclusion, we show that apoC-I and apoC-III inhibit lipolysis by displacing LPL from lipid emulsion particles. We also propose a role for these apolipoproteins in the irreversible inactivation of LPL by factors such as angptl4.


Asunto(s)
Apolipoproteína C-III/química , Apolipoproteína C-I/química , Lipoproteína Lipasa/química , Triglicéridos/química , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/química , Angiopoyetinas/genética , Angiopoyetinas/metabolismo , Animales , Apolipoproteína C-I/genética , Apolipoproteína C-I/metabolismo , Apolipoproteína C-III/genética , Apolipoproteína C-III/metabolismo , Bovinos , Emulsiones , Humanos , Lipólisis/fisiología , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Mutagénesis Sitio-Dirigida , Triglicéridos/genética , Triglicéridos/metabolismo
16.
Biochem Biophys Res Commun ; 450(2): 1063-9, 2014 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-24984153

RESUMEN

Patients at increased cardiovascular risk commonly display high levels of plasma triglycerides (TGs), elevated LDL cholesterol, small dense LDL particles and low levels of HDL-cholesterol. Many remain at high risk even after successful statin therapy, presumably because TG levels remain high. Lipoprotein lipase (LPL) maintains TG homeostasis in blood by hydrolysis of TG-rich lipoproteins. Efficient clearance of TGs is accompanied by increased levels of HDL-cholesterol and decreased levels of small dense LDL. Given the central role of LPL in lipid metabolism we sought to find small molecules that could increase LPL activity and serve as starting points for drug development efforts against cardiovascular disease. Using a small molecule screening approach we have identified small molecules that can protect LPL from inactivation by the controller protein angiopoietin-like protein 4 during incubations in vitro. One of the selected compounds, 50F10, was directly shown to preserve the active homodimer structure of LPL, as demonstrated by heparin-Sepharose chromatography. On injection to hypertriglyceridemic apolipoprotein A-V deficient mice the compound ameliorated the postprandial response after an olive oil gavage. This is a potential lead compound for the development of drugs that could reduce the residual risk associated with elevated plasma TGs in dyslipidemia.


Asunto(s)
Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Hipolipemiantes/farmacología , Lipoproteína Lipasa/metabolismo , Triglicéridos/sangre , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/metabolismo , Animales , Apolipoproteína A-V , Apolipoproteínas/genética , Estabilidad de Enzimas , Compuestos Heterocíclicos de 4 o más Anillos/química , Hipertrigliceridemia/sangre , Hipertrigliceridemia/tratamiento farmacológico , Lipoproteína Lipasa/química , Ratones Endogámicos C57BL , Ratones Noqueados , Periodo Posprandial , Unión Proteica , Multimerización de Proteína , Piridinas/química , Piridinas/farmacología , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad
17.
J Biol Chem ; 287(35): 29739-52, 2012 Aug 24.
Artículo en Inglés | MEDLINE | ID: mdl-22773878

RESUMEN

Angiopoietin-like protein 4 (Angptl4), a potent regulator of plasma triglyceride metabolism, binds to lipoprotein lipase (LPL) through its N-terminal coiled-coil domain (ccd-Angptl4) inducing dissociation of the dimeric enzyme to inactive monomers. In this study, we demonstrate that fatty acids reduce the inactivation of LPL by Angptl4. This was the case both with ccd-Angptl4 and full-length Angptl4, and the effect was seen in human plasma or in the presence of albumin. The effect decreased in the sequence oleic acid > palmitic acid > myristic acid > linoleic acid > linolenic acid. Surface plasmon resonance, isothermal titration calorimetry, fluorescence, and chromatography measurements revealed that fatty acids bind with high affinity to ccd-Angptl4. The interactions were characterized by fast association and slow dissociation rates, indicating formation of stable complexes. The highest affinity for ccd-Angptl4 was detected for oleic acid with a subnanomolar equilibrium dissociation constant (K(d)). The K(d) values for palmitic and myristic acid were in the nanomolar range. Linoleic and linolenic acid bound with much lower affinity. On binding of fatty acids, ccd-Angptl4 underwent conformational changes resulting in a decreased helical content, weakened structural stability, dissociation of oligomers, and altered fluorescence properties of the Trp-38 residue that is located close to the putative LPL-binding region. Based on these results, we propose that fatty acids play an important role in modulating the effects of Angptl4.


Asunto(s)
Angiopoyetinas/metabolismo , Ácidos Grasos/metabolismo , Lipoproteína Lipasa/metabolismo , Multimerización de Proteína , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/química , Angiopoyetinas/genética , Animales , Sitios de Unión , Bovinos , Ácidos Grasos/química , Humanos , Lipoproteína Lipasa/química , Lipoproteína Lipasa/genética , Ratones , Plasma/química , Plasma/metabolismo , Estabilidad Proteica , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Albúmina Sérica/química , Albúmina Sérica/genética , Albúmina Sérica/metabolismo , Especificidad por Sustrato , Resonancia por Plasmón de Superficie
18.
Biochim Biophys Acta ; 1821(10): 1370-8, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22732211

RESUMEN

Lipoprotein lipase (LPL) is important for clearance of triacylglycerols (TG) from plasma both as an enzyme and as a bridging factor between lipoproteins and receptors for endocytosis. The amount of LPL at the luminal side of the capillary endothelium determines to what extent lipids are taken up. Mechanisms to control both the activity of LPL and its transport to the endothelial sites are regulated, but poorly understood. Angiopoietin-like proteins (ANGPTLs) 3 and 4 are potential control proteins for LPL, but plasma concentrations of ANGPTLs do not correlate with plasma TG levels. We investigated the effects of recombinant human N-terminal (NT) ANGPTLs3 and 4 on LPL-mediated bridging of TG-rich lipoproteins to primary mouse hepatocytes and found that the NT-ANGPTLs, in concentrations sufficient to cause inactivation of LPL in vitro, were unable to prevent LPL-mediated lipoprotein uptake. We therefore investigated the effects of lipoproteins (chylomicrons, VLDL and LDL) on the inactivation of LPL in vitro by NT-ANGPTLs3 and 4 and found that LPL activity was protected by TG-rich lipoproteins. In vivo, postprandial TG protected LPL from inactivation by recombinant NT-ANGPTL4 injected to mice. We conclude that lipoprotein-bound LPL is stabilized against inactivation by ANGPTLs. The levels of ANGPTLs found in blood may not be sufficient to overcome this stabilization. Therefore it is likely that the prime site of action of ANGPTLs on LPL is in subendothelial compartments where TG-rich lipoprotein concentration is lower than in blood. This could explain why the plasma levels of TG and ANGPTLs do not correlate.


Asunto(s)
Angiopoyetinas/farmacología , Lipoproteína Lipasa/metabolismo , Lipoproteínas/fisiología , Triglicéridos/fisiología , Proteína 3 Similar a la Angiopoyetina , Proteína 4 Similar a la Angiopoyetina , Proteínas Similares a la Angiopoyetina , Animales , Quilomicrones/fisiología , Activación Enzimática , Hepatocitos/metabolismo , Humanos , Lipoproteínas LDL/fisiología , Lipoproteínas VLDL/fisiología , Ratones
19.
PLoS One ; 18(1): e0281010, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36701280

RESUMEN

Maritime archaeological investigations of the wreck of the medieval warship Gribshunden (1495), flagship of King Hans of Denmark and Norway, have revealed diverse artifacts including exotic spices imported from far distant origins: saffron, ginger, clove, peppercorns, and almond. The special circumstances of the vessel's last voyage add unique context to the assemblage. Gribshunden and an accompanying squadron conveyed the king, courtiers, noblemen, and soldiers from Copenhagen to a political summit in Kalmar, Sweden. At that conference, Hans expected the Swedish Council to elect him king of Sweden, and thereby fulfill his ambition to reunify the Nordic region under a single crown. To achieve this, Hans assembled in his fleet and particularly aboard his flagship the people and elite cultural signifiers that would convince the Swedish delegation to accept his rule. Along the way, the ships anchored near Ronneby, Blekinge. Written sources record that an explosion and fire caused Gribshunden to sink off Stora Ekön (Great Oak Island). Exotic spices were status markers among the aristocracy in Scandinavia and around the Baltic Sea during the Middle Ages (1050-1550 CE). Until the Gribshunden finds, these extravagances have rarely or never been represented archaeologically. Evidence of their use and consumption in medieval Scandinavia has been limited to sparse written references. We present here the botanical remains from the Gribshunden shipwreck and compare them to previous archaeobotanical finds from the medieval Baltic region. These opulent status symbols traveled with a medieval king en route to a major historical event. The combination of textual and archaeological evidence allows a novel analytical view of the social environment in which these luxurious foods were consumed.


Asunto(s)
Especias , Viaje , Humanos , Masculino , Suecia , Países Escandinavos y Nórdicos , Noruega
20.
Cardiovasc Res ; 118(2): 489-502, 2022 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-33693480

RESUMEN

AIMS: Fibroblast growth factor (FGF) 21, a key regulator of energy metabolism, is currently evaluated in humans for treatment of type 2 diabetes and non-alcoholic steatohepatitis. However, the effects of FGF21 on cardiovascular benefit, particularly on lipoprotein metabolism in relation to atherogenesis, remain elusive. METHODS AND RESULTS: Here, the role of FGF21 in lipoprotein metabolism in relation to atherosclerosis development was investigated by pharmacological administration of a half-life extended recombinant FGF21 protein to hypercholesterolaemic APOE*3-Leiden.CETP mice, a well-established model mimicking atherosclerosis initiation and development in humans. FGF21 reduced plasma total cholesterol, explained by a reduction in non-HDL-cholesterol. Mechanistically, FGF21 promoted brown adipose tissue (BAT) activation and white adipose tissue (WAT) browning, thereby enhancing the selective uptake of fatty acids from triglyceride-rich lipoproteins into BAT and into browned WAT, consequently accelerating the clearance of the cholesterol-enriched remnants by the liver. In addition, FGF21 reduced body fat, ameliorated glucose tolerance and markedly reduced hepatic steatosis, related to up-regulated hepatic expression of genes involved in fatty acid oxidation and increased hepatic VLDL-triglyceride secretion. Ultimately, FGF21 largely decreased atherosclerotic lesion area, which was mainly explained by the reduction in non-HDL-cholesterol as shown by linear regression analysis, decreased lesion severity, and increased atherosclerotic plaque stability index. CONCLUSION: FGF21 improves hypercholesterolaemia by accelerating triglyceride-rich lipoprotein turnover as a result of activating BAT and browning of WAT, thereby reducing atherosclerotic lesion severity and increasing atherosclerotic lesion stability index. We have thus provided additional support for the clinical use of FGF21 in the treatment of atherosclerotic cardiovascular disease.


Asunto(s)
Anticolesterolemiantes/farmacología , Aterosclerosis/prevención & control , Colesterol/sangre , Factores de Crecimiento de Fibroblastos/farmacología , Hipercolesterolemia/tratamiento farmacológico , Placa Aterosclerótica , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/patología , Tejido Adiposo Blanco/efectos de los fármacos , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/patología , Adiposidad/efectos de los fármacos , Animales , Apolipoproteína E3/genética , Apolipoproteína E3/metabolismo , Aterosclerosis/sangre , Aterosclerosis/genética , Aterosclerosis/patología , Biomarcadores/sangre , Modelos Animales de Enfermedad , Metabolismo Energético/efectos de los fármacos , Hipercolesterolemia/sangre , Hipercolesterolemia/genética , Hipercolesterolemia/patología , Metabolismo de los Lípidos/efectos de los fármacos , Lipoproteínas VLDL/sangre , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Ratones Transgénicos , Proteínas Recombinantes/farmacología , Triglicéridos/sangre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA