Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 200
Filtrar
1.
Subcell Biochem ; 104: 139-179, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38963487

RESUMEN

Lipoprotein lipase (LPL) is a critical enzyme in humans that provides fuel to peripheral tissues. LPL hydrolyzes triglycerides from the cores of lipoproteins that are circulating in plasma and interacts with receptors to mediate lipoprotein uptake, thus directing lipid distribution via catalytic and non-catalytic functions. Functional losses in LPL or any of its myriad of regulators alter lipid homeostasis and potentially affect the risk of developing cardiovascular disease-either increasing or decreasing the risk depending on the mutated protein. The extensive LPL regulatory network tunes LPL activity to allocate fatty acids according to the energetic needs of the organism and thus is nutritionally responsive and tissue dependent. Multiple pharmaceuticals in development manipulate or mimic these regulators, demonstrating their translational importance. Another facet of LPL biology is that the oligomeric state of the enzyme is also central to its regulation. Recent structural studies have solidified the idea that LPL is regulated not only by interactions with other binding partners but also by self-associations. Here, we review the complexities of the protein-protein and protein-lipid interactions that govern LPL structure and function.


Asunto(s)
Lipoproteína Lipasa , Lipoproteína Lipasa/metabolismo , Lipoproteína Lipasa/química , Lipoproteína Lipasa/genética , Humanos , Animales , Unión Proteica , Triglicéridos/metabolismo , Metabolismo de los Lípidos
2.
Proc Natl Acad Sci U S A ; 118(12)2021 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-33723082

RESUMEN

The complex between lipoprotein lipase (LPL) and its endothelial receptor (GPIHBP1) is responsible for the lipolytic processing of triglyceride-rich lipoproteins (TRLs) along the capillary lumen, a physiologic process that releases lipid nutrients for vital organs such as heart and skeletal muscle. LPL activity is regulated in a tissue-specific manner by endogenous inhibitors (angiopoietin-like [ANGPTL] proteins 3, 4, and 8), but the molecular mechanisms are incompletely understood. ANGPTL4 catalyzes the inactivation of LPL monomers by triggering the irreversible unfolding of LPL's α/ß-hydrolase domain. Here, we show that this unfolding is initiated by the binding of ANGPTL4 to sequences near LPL's catalytic site, including ß2, ß3-α3, and the lid. Using pulse-labeling hydrogen‒deuterium exchange mass spectrometry, we found that ANGPTL4 binding initiates conformational changes that are nucleated on ß3-α3 and progress to ß5 and ß4-α4, ultimately leading to the irreversible unfolding of regions that form LPL's catalytic pocket. LPL unfolding is context dependent and varies with the thermal stability of LPL's α/ß-hydrolase domain (Tm of 34.8 °C). GPIHBP1 binding dramatically increases LPL stability (Tm of 57.6 °C), while ANGPTL4 lowers the onset of LPL unfolding by ∼20 °C, both for LPL and LPL•GPIHBP1 complexes. These observations explain why the binding of GPIHBP1 to LPL retards the kinetics of ANGPTL4-mediated LPL inactivation at 37 °C but does not fully suppress inactivation. The allosteric mechanism by which ANGPTL4 catalyzes the irreversible unfolding and inactivation of LPL is an unprecedented pathway for regulating intravascular lipid metabolism.


Asunto(s)
Proteína 4 Similar a la Angiopoyetina/química , Proteína 4 Similar a la Angiopoyetina/metabolismo , Hidrolasas/química , Hidrolasas/metabolismo , Lipoproteína Lipasa/química , Lipoproteína Lipasa/metabolismo , Dominios Proteicos , Secuencia de Aminoácidos , Sitios de Unión , Catálisis , Dominio Catalítico , Susceptibilidad a Enfermedades , Humanos , Cinética , Lipólisis , Espectrometría de Masas , Unión Proteica , Estabilidad Proteica , Desplegamiento Proteico , Temperatura
3.
Proc Natl Acad Sci U S A ; 117(19): 10254-10264, 2020 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-32332168

RESUMEN

Lipases are enzymes necessary for the proper distribution and utilization of lipids in the human body. Lipoprotein lipase (LPL) is active in capillaries, where it plays a crucial role in preventing dyslipidemia by hydrolyzing triglycerides from packaged lipoproteins. Thirty years ago, the existence of a condensed and inactive LPL oligomer was proposed. Although recent work has shed light on the structure of the LPL monomer, the inactive oligomer remained opaque. Here we present a cryo-EM reconstruction of a helical LPL oligomer at 3.8-Å resolution. Helix formation is concentration-dependent, and helices are composed of inactive dihedral LPL dimers. Heparin binding stabilizes LPL helices, and the presence of substrate triggers helix disassembly. Superresolution fluorescent microscopy of endogenous LPL revealed that LPL adopts a filament-like distribution in vesicles. Mutation of one of the helical LPL interaction interfaces causes loss of the filament-like distribution. Taken together, this suggests that LPL is condensed into its inactive helical form for storage in intracellular vesicles.


Asunto(s)
Lipoproteína Lipasa/química , Lipoproteína Lipasa/metabolismo , Triglicéridos/metabolismo , Animales , Bovinos , Microscopía por Crioelectrón , Células HEK293 , Humanos , Hidrólisis , Lipoproteína Lipasa/genética , Ratones , Modelos Moleculares , Mutación , Células 3T3 NIH , Conformación Proteica , Especificidad por Sustrato
4.
Proc Natl Acad Sci U S A ; 117(8): 4337-4346, 2020 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-32034094

RESUMEN

The binding of lipoprotein lipase (LPL) to GPIHBP1 focuses the intravascular hydrolysis of triglyceride-rich lipoproteins on the surface of capillary endothelial cells. This process provides essential lipid nutrients for vital tissues (e.g., heart, skeletal muscle, and adipose tissue). Deficiencies in either LPL or GPIHBP1 impair triglyceride hydrolysis, resulting in severe hypertriglyceridemia. The activity of LPL in tissues is regulated by angiopoietin-like proteins 3, 4, and 8 (ANGPTL). Dogma has held that these ANGPTLs inactivate LPL by converting LPL homodimers into monomers, rendering them highly susceptible to spontaneous unfolding and loss of enzymatic activity. Here, we show that binding of an LPL-specific monoclonal antibody (5D2) to the tryptophan-rich lipid-binding loop in the carboxyl terminus of LPL prevents homodimer formation and forces LPL into a monomeric state. Of note, 5D2-bound LPL monomers are as stable as LPL homodimers (i.e., they are not more prone to unfolding), but they remain highly susceptible to ANGPTL4-catalyzed unfolding and inactivation. Binding of GPIHBP1 to LPL alone or to 5D2-bound LPL counteracts ANGPTL4-mediated unfolding of LPL. In conclusion, ANGPTL4-mediated inactivation of LPL, accomplished by catalyzing the unfolding of LPL, does not require the conversion of LPL homodimers into monomers. Thus, our findings necessitate changes to long-standing dogma on mechanisms for LPL inactivation by ANGPTL proteins. At the same time, our findings align well with insights into LPL function from the recent crystal structure of the LPL•GPIHBP1 complex.


Asunto(s)
Proteína 4 Similar a la Angiopoyetina/metabolismo , Lipoproteína Lipasa/química , Triglicéridos/sangre , Secuencias de Aminoácidos , Proteína 4 Similar a la Angiopoyetina/genética , Animales , Anticuerpos Monoclonales/metabolismo , Dimerización , Humanos , Hipertrigliceridemia/enzimología , Hipertrigliceridemia/genética , Hipertrigliceridemia/metabolismo , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Desplegamiento Proteico , Receptores de Lipoproteína/química , Receptores de Lipoproteína/genética , Receptores de Lipoproteína/metabolismo
5.
J Biol Chem ; 296: 100312, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33482195

RESUMEN

Elevated plasma triglycerides are a risk factor for coronary artery disease, which is the leading cause of death worldwide. Lipoprotein lipase (LPL) reduces triglycerides in the blood by hydrolyzing them from triglyceride-rich lipoproteins to release free fatty acids. LPL activity is regulated in a nutritionally responsive manner by macromolecular inhibitors including angiopoietin-like proteins 3 and 4 (ANGPTL3 and ANGPTL4). However, the mechanism by which ANGPTL3 inhibits LPL is unclear, in part due to challenges in obtaining pure protein for study. We used a new purification protocol for the N-terminal domain of ANGPTL3, removing a DNA contaminant, and found DNA-free ANGPTL3 showed enhanced inhibition of LPL. Structural analysis showed that ANGPTL3 formed elongated, flexible trimers and hexamers that did not interconvert. ANGPTL4 formed only elongated flexible trimers. We compared the inhibition of ANGPTL3 and ANGPTL4 using human very-low-density lipoproteins as a substrate and found both were noncompetitive inhibitors. The inhibition constants for the trimeric ANGPTL3 (7.5 ± 0.7 nM) and ANGPTL4 (3.6 ± 1.0 nM) were only 2-fold different. Heparin has previously been reported to interfere with ANGPTL3 binding to LPL, so we questioned if the negatively charged heparin was acting in a similar fashion to the DNA contaminant. We found that ANGPTL3 inhibition is abolished by binding to low-molecular-weight heparin, whereas ANGPTL4 inhibition is not. Our data show new similarities and differences in how ANGPTL3 and ANGPTL4 regulate LPL and opens new avenues of investigating the effect of heparin on LPL inhibition by ANGPTL3.


Asunto(s)
Proteína 4 Similar a la Angiopoyetina/química , Proteínas Similares a la Angiopoyetina/química , Enfermedad de la Arteria Coronaria/genética , Lipoproteína Lipasa/química , Conformación Proteica , Proteína 3 Similar a la Angiopoyetina , Proteína 4 Similar a la Angiopoyetina/genética , Proteína 4 Similar a la Angiopoyetina/ultraestructura , Proteínas Similares a la Angiopoyetina/genética , Proteínas Similares a la Angiopoyetina/ultraestructura , Enfermedad de la Arteria Coronaria/sangre , Enfermedad de la Arteria Coronaria/patología , Heparina/farmacología , Humanos , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/ultraestructura , Lipoproteínas VLDL/química , Lipoproteínas VLDL/genética , Unión Proteica/efectos de los fármacos , Especificidad por Sustrato , Triglicéridos/sangre
6.
Proc Natl Acad Sci U S A ; 116(21): 10360-10365, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31072929

RESUMEN

Lipoprotein lipase (LPL) plays a central role in triglyceride (TG) metabolism. By catalyzing the hydrolysis of TGs present in TG-rich lipoproteins (TRLs), LPL facilitates TG utilization and regulates circulating TG and TRL concentrations. Until very recently, structural information for LPL was limited to homology models, presumably due to the propensity of LPL to unfold and aggregate. By coexpressing LPL with a soluble variant of its accessory protein glycosylphosphatidylinositol-anchored high-density lipoprotein binding protein 1 (GPIHBP1) and with its chaperone protein lipase maturation factor 1 (LMF1), we obtained a stable and homogenous LPL/GPIHBP1 complex that was suitable for structure determination. We report here X-ray crystal structures of human LPL in complex with human GPIHBP1 at 2.5-3.0 Å resolution, including a structure with a novel inhibitor bound to LPL. Binding of the inhibitor resulted in ordering of the LPL lid and lipid-binding regions and thus enabled determination of the first crystal structure of LPL that includes these important regions of the protein. It was assumed for many years that LPL was only active as a homodimer. The structures and additional biochemical data reported here are consistent with a new report that LPL, in complex with GPIHBP1, can be active as a monomeric 1:1 complex. The crystal structures illuminate the structural basis for LPL-mediated TRL lipolysis as well as LPL stabilization and transport by GPIHBP1.


Asunto(s)
Lipoproteína Lipasa/química , Lipoproteína Lipasa/metabolismo , Receptores de Lipoproteína/química , Receptores de Lipoproteína/metabolismo , Células HEK293 , Humanos , Hidrólisis , Metabolismo de los Lípidos/fisiología , Lipólisis/fisiología , Lipoproteínas/metabolismo , Triglicéridos/metabolismo
7.
Proc Natl Acad Sci U S A ; 116(13): 6319-6328, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30850549

RESUMEN

Lipoprotein lipase (LPL), the enzyme that hydrolyzes triglycerides in plasma lipoproteins, is assumed to be active only as a homodimer. In support of this idea, several groups have reported that the size of LPL, as measured by density gradient ultracentrifugation, is ∼110 kDa, twice the size of LPL monomers (∼55 kDa). Of note, however, in those studies the LPL had been incubated with heparin, a polyanionic substance that binds and stabilizes LPL. Here we revisited the assumption that LPL is active only as a homodimer. When freshly secreted human LPL (or purified preparations of LPL) was subjected to density gradient ultracentrifugation (in the absence of heparin), LPL mass and activity peaks exhibited the size expected of monomers (near the 66-kDa albumin standard). GPIHBP1-bound LPL also exhibited the size expected for a monomer. In the presence of heparin, LPL size increased, overlapping with a 97.2-kDa standard. We also used density gradient ultracentrifugation to characterize the LPL within the high-salt and low-salt peaks from a heparin-Sepharose column. The catalytically active LPL within the high-salt peak exhibited the size of monomers, whereas most of the inactive LPL in the low-salt peak was at the bottom of the tube (in aggregates). Consistent with those findings, the LPL in the low-salt peak, but not that in the high-salt peak, was easily detectable with single mAb sandwich ELISAs, in which LPL is captured and detected with the same antibody. We conclude that catalytically active LPL can exist in a monomeric state.


Asunto(s)
Lipoproteína Lipasa/química , Lipoproteína Lipasa/aislamiento & purificación , Animales , Células CHO , Bovinos , Centrifugación por Gradiente de Densidad/métodos , Cromatografía de Afinidad , Cromatografía en Agarosa , Cricetulus , Epítopos , Heparina , Humanos , Lipoproteína Lipasa/sangre , Receptores de Lipoproteína/sangre , Receptores de Lipoproteína/química , Receptores de Lipoproteína/aislamiento & purificación , Sefarosa/análogos & derivados , Triglicéridos/metabolismo , Ultracentrifugación
8.
J Biol Chem ; 295(10): 2900-2912, 2020 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-31645434

RESUMEN

Lipoprotein lipase (LPL) is central to triglyceride metabolism. Severely compromised LPL activity causes familial chylomicronemia syndrome (FCS), which is associated with very high plasma triglyceride levels and increased risk of life-threatening pancreatitis. Currently, no approved pharmacological intervention can acutely lower plasma triglycerides in FCS. Low yield, high aggregation, and poor stability of recombinant LPL have thus far prevented development of enzyme replacement therapy. Recently, we showed that LPL monomers form 1:1 complexes with the LPL transporter glycosylphosphatidylinositol-anchored high-density lipoprotein-binding protein 1 (GPIHBP1) and solved the structure of the complex. In the present work, we further characterized the monomeric LPL/GPIHBP1 complex and its derivative, the LPL-GPIHBP1 fusion protein, with the goal of contributing to the development of an LPL enzyme replacement therapy. Fusion of LPL to GPIHBP1 increased yields of recombinant LPL, prevented LPL aggregation, stabilized LPL against spontaneous inactivation, and made it resistant to inactivation by the LPL antagonists angiopoietin-like protein 3 (ANGPTL3) or ANGPTL4. The high stability of the fusion protein enabled us to identify LPL amino acids that interact with ANGPTL4. Additionally, the LPL-GPIHBP1 fusion protein exhibited high enzyme activity in in vitro assays. Importantly, both intravenous and subcutaneous administrations of the fusion protein lowered triglycerides in several mouse strains without causing adverse effects. These results indicate that the LPL-GPIHBP1 fusion protein has potential for use as a therapeutic for managing FCS.


Asunto(s)
Lipoproteína Lipasa/metabolismo , Receptores de Lipoproteína/metabolismo , Triglicéridos/sangre , Secuencia de Aminoácidos , Proteína 3 Similar a la Angiopoyetina , Proteína 4 Similar a la Angiopoyetina/química , Proteína 4 Similar a la Angiopoyetina/metabolismo , Proteínas Similares a la Angiopoyetina/química , Proteínas Similares a la Angiopoyetina/metabolismo , Animales , Sitios de Unión , Modelos Animales de Enfermedad , Terapia de Reemplazo Enzimático , Humanos , Hiperlipoproteinemia Tipo I/tratamiento farmacológico , Hiperlipoproteinemia Tipo I/patología , Infusiones Subcutáneas , Lipoproteína Lipasa/química , Lipoproteína Lipasa/genética , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Agregado de Proteínas/efectos de los fármacos , Estabilidad Proteica , Receptores de Lipoproteína/química , Receptores de Lipoproteína/genética , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/uso terapéutico
9.
Proc Natl Acad Sci U S A ; 115(26): E6020-E6029, 2018 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-29899144

RESUMEN

The intravascular processing of triglyceride-rich lipoproteins depends on lipoprotein lipase (LPL) and GPIHBP1, a membrane protein of endothelial cells that binds LPL within the subendothelial spaces and shuttles it to the capillary lumen. In the absence of GPIHBP1, LPL remains mislocalized within the subendothelial spaces, causing severe hypertriglyceridemia (chylomicronemia). The N-terminal domain of GPIHBP1, an intrinsically disordered region (IDR) rich in acidic residues, is important for stabilizing LPL's catalytic domain against spontaneous and ANGPTL4-catalyzed unfolding. Here, we define several important properties of GPIHBP1's IDR. First, a conserved tyrosine in the middle of the IDR is posttranslationally modified by O-sulfation; this modification increases both the affinity of GPIHBP1-LPL interactions and the ability of GPIHBP1 to protect LPL against ANGPTL4-catalyzed unfolding. Second, the acidic IDR of GPIHBP1 increases the probability of a GPIHBP1-LPL encounter via electrostatic steering, increasing the association rate constant (kon) for LPL binding by >250-fold. Third, we show that LPL accumulates near capillary endothelial cells even in the absence of GPIHBP1. In wild-type mice, we expect that the accumulation of LPL in close proximity to capillaries would increase interactions with GPIHBP1. Fourth, we found that GPIHBP1's IDR is not a key factor in the pathogenicity of chylomicronemia in patients with the GPIHBP1 autoimmune syndrome. Finally, based on biophysical studies, we propose that the negatively charged IDR of GPIHBP1 traverses a vast space, facilitating capture of LPL by capillary endothelial cells and simultaneously contributing to GPIHBP1's ability to preserve LPL structure and activity.


Asunto(s)
Células Endoteliales/metabolismo , Lipoproteína Lipasa/metabolismo , Receptores de Lipoproteína/metabolismo , Proteína 4 Similar a la Angiopoyetina/química , Proteína 4 Similar a la Angiopoyetina/genética , Proteína 4 Similar a la Angiopoyetina/metabolismo , Animales , Células Endoteliales/patología , Humanos , Hiperlipoproteinemia Tipo I/genética , Hiperlipoproteinemia Tipo I/metabolismo , Hiperlipoproteinemia Tipo I/patología , Lipoproteína Lipasa/química , Lipoproteína Lipasa/genética , Ratones , Unión Proteica , Dominios Proteicos , Receptores de Lipoproteína/química , Receptores de Lipoproteína/genética , Tirosina/química , Tirosina/genética , Tirosina/metabolismo
10.
J Lipid Res ; 61(10): 1347-1359, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32690595

RESUMEN

For three decades, the LPL-specific monoclonal antibody 5D2 has been used to investigate LPL structure/function and intravascular lipolysis. 5D2 has been used to measure LPL levels, block the triglyceride hydrolase activity of LPL, and prevent the propensity of concentrated LPL preparations to form homodimers. Two early studies on the location of the 5D2 epitope reached conflicting conclusions, but the more convincing report suggested that 5D2 binds to a tryptophan (Trp)-rich loop in the carboxyl terminus of LPL. The same loop had been implicated in lipoprotein binding. Using surface plasmon resonance, we showed that 5D2 binds with high affinity to a synthetic LPL peptide containing the Trp-rich loop of human (but not mouse) LPL. We also showed, by both fluorescence and UV resonance Raman spectroscopy, that the Trp-rich loop binds lipids. Finally, we used X-ray crystallography to solve the structure of the Trp-rich peptide bound to a 5D2 Fab fragment. The Trp-rich peptide contains a short α-helix, with two Trps projecting into the antigen recognition site. A proline substitution in the α-helix, found in mouse LPL, is expected to interfere with several hydrogen bonds, explaining why 5D2 cannot bind to mouse LPL.


Asunto(s)
Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Lipoproteína Lipasa/química , Lipoproteína Lipasa/inmunología , Animales , Sitios de Unión , Humanos , Ratones , Triptófano
11.
Biochemistry ; 57(2): 241-254, 2018 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-29303250

RESUMEN

Lipoprotein lipase (LPL) is a dimeric enzyme that is responsible for clearing triglyceride-rich lipoproteins from the blood. Although LPL plays a key role in cardiovascular health, an experimentally derived three-dimensional structure has not been determined. Such a structure would aid in understanding mutations in LPL that cause familial LPL deficiency in patients and help in the development of therapeutic strategies to target LPL. A major obstacle to structural studies of LPL is that LPL is an unstable protein that is difficult to produce in the quantities needed for nuclear magnetic resonance or crystallography. We present updated LPL structural models generated by combining disulfide mapping, computational modeling, and data derived from single-molecule Förster resonance energy transfer (smFRET). We pioneer the technique of smFRET for use with LPL by developing conditions for imaging active LPL and identifying positions in LPL for the attachment of fluorophores. Using this approach, we measure LPL-LPL intermolecular interactions to generate experimental constraints that inform new computational models of the LPL dimer structure. These models suggest that LPL may dimerize using an interface that is different from the dimerization interface suggested by crystal packing contacts seen in structures of pancreatic lipase.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia/métodos , Imagen Individual de Molécula/métodos , Biotinilación , Biología Computacional , Cisteína/química , Dimerización , Células HEK293 , Humanos , Lipoproteína Lipasa/química , Lipoproteína Lipasa/genética , Lipoproteínas/metabolismo , Modelos Moleculares , Simulación del Acoplamiento Molecular , Conformación Proteica , Proteínas Recombinantes/química , Triglicéridos/metabolismo
12.
Biochem Biophys Res Commun ; 504(1): 54-60, 2018 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-30170729

RESUMEN

The ß-isoform of diacylglycerol kinase (DGK) localizes predominantly to neurons and induces neurite outgrowth and spine formation. However, the detailed molecular mechanisms underlying the functions of DGKß remain elusive. During the course of studies on other DGK isozymes, we unexpectedly found that the overexpression of wild-type DGKß in COS-7 cells markedly induced filopodium formation. Because filopodium formation is closely related to neurite outgrowth and spine formation, we constructed various DGKß mutants and compared their abilities to induce filopodium formation in order to elucidate the structure-function relationships of DGKß. We found that the C-terminal, C1 and catalytic domains and catalytic activity were indispensable for filopodium formation, but the recoverin homology domain and EF-hand motifs were not. Moreover, the extent of plasma membrane localization and F-actin colocalization were positively correlated with filopodium formation. Intriguingly, DGKß selectively interacted and colocalized at the plasma membrane with a Rac1-GTPase-activating protein, ß2-chimaerin, which is an inducer of filopodia; it also interacted, to lesser extent, with α2-chimaerin, but not with α1- or ß1-chimaerin. Moreover, DGKß enhanced the plasma membrane localization of ß2-chimaerin. These results suggest that DGKß plays an important role in neurite outgrowth and spine formation in neurons via its ability to induce filopodium formation.


Asunto(s)
Proteínas Activadoras de GTPasa/metabolismo , Lipoproteína Lipasa/metabolismo , Proteínas de Neoplasias/metabolismo , Seudópodos/fisiología , Animales , Células COS , Dominio Catalítico , Chlorocebus aethiops , Lipoproteína Lipasa/química , Lipoproteína Lipasa/genética , Mutación , Dominios Proteicos , Seudópodos/ultraestructura
13.
Org Biomol Chem ; 16(29): 5250-5253, 2018 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-30004552

RESUMEN

Diacylglycerol lipases (DAGL) produce the endocannabinoid 2-arachidonoylglycerol, a key modulator of neurotransmitter release. Chemical tools that visualize endogenous DAGL activity are desired. Here, we report the design, synthesis and application of a triazole urea probe for DAGL equipped with a norbornene as a biorthogonal handle. The activity and selectivity of the probe was assessed with activity-based protein profiling. This probe was potent against endogenous DAGLα (IC50 = 5 nM) and it was successfully applied as a two-step activity-based probe for labeling of DAGLα using an inverse electron-demand Diels-Alder ligation in living cells.


Asunto(s)
Lipoproteína Lipasa/química , Lipoproteína Lipasa/metabolismo , Animales , Encéfalo/metabolismo , Línea Celular Tumoral , Reacción de Cicloadición , Teoría Funcional de la Densidad , Endocannabinoides/química , Humanos , Lipoproteína Lipasa/antagonistas & inhibidores , Ratones , Sondas Moleculares/química , Sondas Moleculares/toxicidad , Norbornanos/química , Proteoma , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Triazoles/química , Urea/química
14.
Bioorg Chem ; 80: 347-360, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29986183

RESUMEN

Studies on the lipid-regulating effects of alisol compounds are reported that include alisol B, alisol A 24-acetate (24A), alisol A and an alisol B - 24A - alisol A mixture (content ratio = 1:1:1). The effects on the activity of lipoprotein lipase (LPL), a key lipid-modulating enzyme, were studied to investigate the molecular mechanism of lipid-regulating activity of alisols. The effects of alisols on regulating blood lipids and the activities of LPL were determined using a reagent kit method. The structure of LPL was obtained by homology modeling and the interactive mechanism of alisol monomers and the mixture with LPL was investigated by molecular simulation. The alisol monomer and mixture were shown to regulate blood lipids, suggesting that alisols may decrease the level of triglyceride (TG) by improving the activity of LPL. The order of intensity was: mixture > alisol A > alisol B > 24A, indicating that alisols of alismatis rhizoma feature a synergistic effect on LPL. The N- and C-terminus of LPL both represented the catalytic active domains of this lipid-regulating effect. Cys306, Gln129 and Ser166 were the key amino acid residues resulting in the lipid-regulating effect of the alisol monomer while Ser166 and Arg18 were found to be responsible for the lipid-regulating effect of the mixture. The C-terminus of LPL was indirectly involved in the enzymatic process. A folded side chain of alisols or the parent ring was found to bind somewhat weaker to LPL than an open side chain or parent ring. The hydroxyl groups on the C14-, C22-, C28-, C30- and C31-terminus in the side chain, the ring ether structure in C23-position, and the acetyl group in C29-position represented the key sites for the lipid-regulating action of alisols. Meanwhile, the C30-site hydroxyl group played an important role in the synergistic effect of the alisol mixture.


Asunto(s)
Colestenonas/metabolismo , Lipoproteína Lipasa/metabolismo , Animales , Sitios de Unión , Colestenonas/química , Colestenonas/uso terapéutico , Enlace de Hidrógeno , Interacciones Hidrofóbicas e Hidrofílicas , Hiperlipidemias/tratamiento farmacológico , Hiperlipidemias/metabolismo , Hiperlipidemias/veterinaria , Lípidos/sangre , Lipoproteína Lipasa/química , Masculino , Ratones , Ratones Endogámicos ICR , Simulación de Dinámica Molecular , Electricidad Estática
15.
Biochemistry ; 56(3): 525-533, 2017 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-27984852

RESUMEN

Lipoprotein lipase (LPL) is responsible for the hydrolysis of triglycerides from circulating lipoproteins. Whereas most identified mutations in the LPL gene are deleterious, one mutation, LPLS447X, causes a gain of function. This mutation truncates two amino acids from LPL's C-terminus. Carriers of LPLS447X have decreased VLDL levels and increased HDL levels, a cardioprotective phenotype. LPLS447X is used in Alipogene tiparvovec, the gene therapy product for individuals with familial LPL deficiency. It is unclear why LPLS447X results in a serum lipid profile more favorable than that of LPL. In vitro reports vary as to whether LPLS447X is more active than LPL. We report a comprehensive, biochemical comparison of purified LPLS447X and LPL dimers. We found no difference in specific activity on synthetic and natural substrates. We also did not observe a difference in the Ki for ANGPTL4 inhibition of LPLS447X relative to that of LPL. Finally, we analyzed LPL-mediated uptake of fluorescently labeled lipoprotein particles and found that LPLS447X enhanced lipoprotein uptake to a greater degree than LPL did. An LPL structural model suggests that the LPLS447X truncation exposes residues implicated in LPL binding to uptake receptors.


Asunto(s)
HDL-Colesterol/química , LDL-Colesterol/química , Lipoproteína Lipasa/química , Mutación , Receptores de Lipoproteína/química , Triglicéridos/química , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/química , Angiopoyetinas/genética , Angiopoyetinas/metabolismo , Animales , Transporte Biológico , HDL-Colesterol/metabolismo , LDL-Colesterol/metabolismo , VLDL-Colesterol/química , VLDL-Colesterol/metabolismo , Expresión Génica , Humanos , Hiperlipidemias/sangre , Hiperlipidemias/genética , Hiperlipidemias/patología , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Ratones , Modelos Moleculares , Unión Proteica , Dominios Proteicos , Multimerización de Proteína , Estructura Secundaria de Proteína , Receptores de Lipoproteína/genética , Receptores de Lipoproteína/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Serina/química , Serina/metabolismo , Especificidad por Sustrato , Triglicéridos/metabolismo
16.
Biotechnol Appl Biochem ; 64(4): 464-470, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27097985

RESUMEN

The human lipoprotein lipase (LPL) is a therapeutic target for obesity, and inhibition of LPL with the approved small molecule agent orlistat has been widely used in clinic to treat obesity-related health problems such as diabetes and cardiovascular diseases. However, a variety of missense mutations in LPL protein have been observed, which may cause resistance or sensitization for orlistat, largely limiting the clinical applications of orlistat in obesity therapy. Here, we integrated molecular dynamics simulations and enzyme inhibition to investigate orlistat response to 16 disorder-associated missense mutations in LPL catalytic domain. It was found that most mutations have a modest effect on orlistat binding, and only few can exert strong impact to the binding. Three unfavorable (Trp86Arg, Ile194Thr, and Glu242Lys) and two favorable (His136Arg and Gly188Glu) mutations were identified, which can alter the binding affinity and inhibitory activity of orlistat considerably. Structural and energetic analysis revealed that these potent mutations induce orlistat resistance and sensitization by directly influencing the intermolecular interaction between LPL and orlistat or by indirectly addressing allosteric effect on LPL structure.


Asunto(s)
Lactonas/farmacología , Lipoproteína Lipasa/antagonistas & inhibidores , Lipoproteína Lipasa/química , Mutación Missense/genética , Regulación Alostérica/efectos de los fármacos , Humanos , Lactonas/química , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/metabolismo , Simulación de Dinámica Molecular , Estructura Molecular , Orlistat , Relación Estructura-Actividad
17.
Curr Opin Lipidol ; 27(3): 233-41, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27031275

RESUMEN

PURPOSE OF REVIEW: A major step in energy metabolism is hydrolysis of triacylglycerol-rich lipoproteins (TRLs) to release fatty acids that can be used or stored. This is accomplished by lipoprotein lipase (LPL) at 'binding lipolysis sites' at the vascular endothelium. A multitude of interactions are involved in this seemingly simple reaction. Recent advances in the understanding of some of these factors will be discussed in an attempt to build a comprehensive picture. RECENT FINDINGS: The first event in catabolism of TRLs is that they dock at the vascular endothelium. This requires LPL and GPIHBP1, the endothelial transporter of LPL.Kinetic studies in rats with labeled chylomicrons showed that once a chylomicron has docked in the heart it stays for minutes and a large number of triacylglycerol molecules are split. The distribution of binding between tissues reflects the amount of LPL, as evident from studies with mutant mice.Clearance of TRLs is often slowed down in metabolic disease, as was demonstrated both in mice and men. In mice, this was directly connected to decreased amounts of endothelial LPL. SUMMARY: The LPL system is central in energy metabolism and results from interplay between several factors. Rapid and exciting progress is being made.


Asunto(s)
Metabolismo de los Lípidos , Lipoproteína Lipasa/metabolismo , Animales , Endotelio/metabolismo , Humanos , Lipoproteína Lipasa/química , Síndrome Metabólico/enzimología , Síndrome Metabólico/metabolismo , Pliegue de Proteína
18.
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
19.
Biochemistry ; 55(19): 2713-21, 2016 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-27115711

RESUMEN

The diacylglycerol lipases (DAGLα and DAGLß) hydrolyze DAG to generate 2-arachidonoylglycerol (2-AG), the principal endocannabinoid and main precursor of arachidonic acid (AA). The DAGLs make distinct tissue specific contributions toward 2-AG and AA levels, and therefore, selective modulators for these enzymes could play crucial roles toward harnessing their therapeutic potential. Relatively high-throughput assays have recently been reported for DAGLα and have proven useful toward the characterization of inhibitors of this enzyme. Similar assays are also warranted for DAGLß which was the aim of this study. We first adapted previously reported DAGLα membrane assays (using PNPB and DiFMUO as substrates) to measure recombinant DAGLß activity in membranes. In contrast to results with DAGLα, both substrates provided a relatively limited signal window for measuring DAGLß activity, however, an improved window was obtained when employing a third commercially available substrate, EnzChek. In order to further improve on the assay parameters, we successfully purified the glutathione S-transferase (GST) tagged catalytic domain of DAGLß. Activity of the enzyme was confirmed using EnzChek as well as two DAGL inhibitors (THL and OMDM-188). The purified DAGLß catalytic domain assay described here provides the basis for a relatively clean and convenient assay with the potential to be adapted for high-throughput drug discovery efforts.


Asunto(s)
Descubrimiento de Drogas/métodos , Inhibidores Enzimáticos/química , Isoleucina/análogos & derivados , Lactonas/química , Lipoproteína Lipasa/antagonistas & inhibidores , Lipoproteína Lipasa/química , Membranas Artificiales , Humanos , Isoleucina/química , Lipoproteína Lipasa/genética , Lipoproteína Lipasa/aislamiento & purificación , Dominios Proteicos , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo
20.
J Lipid Res ; 57(6): 1074-85, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27040450

RESUMEN

Reduced white adipose tissue (WAT) LPL activity delays plasma clearance of TG-rich lipoproteins (TRLs). We reported the secretion of apoC-I, an LPL inhibitor, from WAT ex vivo in women. Therefore we hypothesized that WAT-secreted apoC-I associates with reduced WAT LPL activity and TRL clearance. WAT apoC-I secretion averaged 86.9 ± 31.4 pmol/g/4 h and 74.1 ± 36.6 pmol/g/4 h in 28 women and 11 men with BMI ≥27 kg/m(2), respectively, with no sex differences. Following the ingestion of a (13)C-triolein-labeled high-fat meal, subjects with high WAT apoC-I secretion (above median) had delayed postprandial plasma clearance of dietary TRLs, assessed from plasma (13)C-triolein-labeled TGs and apoB48. They also had reduced hydrolysis and storage of synthetic (3)H-triolein-labeled ((3)H)-TRLs in WAT ex vivo (i.e., in situ LPL activity). Adjusting for WAT in situ LPL activity eliminated group differences in chylomicron clearance; while adjusting for plasma apoC-I, (3)H-NEFA uptake by WAT, or body composition did not. apoC-I inhibited in situ LPL activity in adipocytes in both a concentration- and time-dependent manner. There was no change in postprandial WAT apoC-I secretion. WAT apoC-I secretion may inhibit WAT LPL activity and promote delayed chylomicron clearance in overweight and obese subjects. We propose that reducing WAT apoC-I secretion ameliorates postprandial TRL clearance in humans.


Asunto(s)
Tejido Adiposo Blanco/enzimología , Apolipoproteína C-I/sangre , Lipoproteína Lipasa/sangre , Obesidad/sangre , Tejido Adiposo Blanco/química , Anciano , Animales , Apolipoproteína B-48/química , Apolipoproteína B-48/metabolismo , Apolipoproteínas E/química , Apolipoproteínas E/metabolismo , Índice de Masa Corporal , Isótopos de Carbono/química , Quilomicrones/sangre , Dieta Alta en Grasa , Femenino , Humanos , Lipoproteína Lipasa/química , Lipoproteína Lipasa/genética , Lipoproteínas HDL/sangre , Masculino , Ratones , Persona de Mediana Edad , Obesidad/genética , Obesidad/patología , Periodo Posprandial , Triglicéridos/sangre , Trioleína/química , Trioleína/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA