Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters










Publication year range
1.
JCI Insight ; 2(20)2017 10 19.
Article in English | MEDLINE | ID: mdl-29046479

ABSTRACT

In mammals, GPIHBP1 is absolutely essential for transporting lipoprotein lipase (LPL) to the lumen of capillaries, where it hydrolyzes the triglycerides in triglyceride-rich lipoproteins. In all lower vertebrate species (e.g., birds, amphibians, reptiles, fish), a gene for LPL can be found easily, but a gene for GPIHBP1 has never been found. The obvious question is whether the LPL in lower vertebrates is able to reach the capillary lumen. Using purified antibodies against chicken LPL, we showed that LPL is present on capillary endothelial cells of chicken heart and adipose tissue, colocalizing with von Willebrand factor. When the antibodies against chicken LPL were injected intravenously into chickens, they bound to LPL on the luminal surface of capillaries in heart and adipose tissue. LPL was released rapidly from chicken hearts with an infusion of heparin, consistent with LPL being located inside blood vessels. Remarkably, chicken LPL bound in a specific fashion to mammalian GPIHBP1. However, we could not identify a gene for GPIHBP1 in the chicken genome, nor could we identify a transcript for GPIHBP1 in a large chicken RNA-seq data set. We conclude that LPL reaches the capillary lumen in chickens - as it does in mammals - despite an apparent absence of GPIHBP1.


Subject(s)
Capillaries/metabolism , Chickens/metabolism , Lipoprotein Lipase/metabolism , Receptors, Lipoprotein/metabolism , Adipose Tissue/blood supply , Adipose Tissue/metabolism , Animals , Antibodies , Endothelial Cells/metabolism , Female , Goats , Heart , Heparin , Humans , Immunoglobulin G , Lipid Metabolism , Lipoprotein Lipase/genetics , Lipoproteins/metabolism , Male , Mice , Receptors, Lipoprotein/analysis , Receptors, Lipoprotein/genetics , Triglycerides/metabolism
2.
J Lipid Res ; 58(1): 216-225, 2017 01.
Article in English | MEDLINE | ID: mdl-27811232

ABSTRACT

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.


Subject(s)
Adipocytes/metabolism , Heparan Sulfate Proteoglycans/metabolism , Lipoprotein Lipase/genetics , Receptors, Lipoprotein/genetics , Animals , Capillaries/enzymology , Capillaries/metabolism , Cell Line , Chylomicrons/metabolism , Culture Media/chemistry , Hep G2 Cells , Humans , Lipolysis/genetics , Lipoprotein Lipase/metabolism , Mice
3.
J Lipid Res ; 57(10): 1889-1898, 2016 10.
Article in English | MEDLINE | ID: mdl-27494936

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/chemistry , Lipoprotein Lipase/chemistry , Receptors, Lipoprotein/chemistry , Amino Acid Substitution , Animals , Cell Line , Drosophila melanogaster , Humans , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Mutation, Missense , Protein Binding , Protein Domains , Receptors, Lipoprotein/genetics , Receptors, Lipoprotein/metabolism
4.
Trends Endocrinol Metab ; 27(7): 455-469, 2016 07.
Article in English | MEDLINE | ID: mdl-27185325

ABSTRACT

GPIHBP1, a GPI-anchored protein in capillary endothelial cells, is crucial for the lipolytic processing of triglyceride-rich lipoproteins (TRLs). GPIHBP1 shuttles lipoprotein lipase (LPL) to its site of action in the capillary lumen and is essential for the margination of TRLs along capillaries - such that lipolytic processing can proceed. GPIHBP1 also reduces the unfolding of the LPL catalytic domain, thereby stabilizing LPL catalytic activity. Many different GPIHBP1 mutations have been identified in patients with severe hypertriglyceridemia (chylomicronemia), the majority of which interfere with folding of the protein and abolish its capacity to bind and transport LPL. The discovery of GPIHBP1 has substantially revised our understanding of intravascular triglyceride metabolism but has also raised many new questions for future research.


Subject(s)
Receptors, Lipoprotein/metabolism , Triglycerides/blood , Animals , Humans , Hypertriglyceridemia/blood , Hypertriglyceridemia/metabolism , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Receptors, Lipoprotein/genetics , Triglycerides/metabolism
5.
J Lipid Res ; 57(9): 1670-83, 2016 09.
Article in English | MEDLINE | ID: mdl-27034464

ABSTRACT

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.


Subject(s)
Adipocytes/metabolism , Angiopoietins/metabolism , Lipoprotein Lipase/metabolism , Lipoproteins/genetics , Triglycerides/genetics , Angiopoietin-Like Protein 4 , Angiopoietins/genetics , Animals , Cytoplasm/metabolism , Golgi Apparatus/metabolism , Humans , Lipoprotein Lipase/genetics , Mice , Mice, Knockout , Proteolysis , Triglycerides/metabolism
6.
Circ Res ; 116(4): 624-32, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25387803

ABSTRACT

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.


Subject(s)
Hyperlipoproteinemia Type I/enzymology , Lipoprotein Lipase/metabolism , Mutation, Missense , Receptors, Lipoprotein/metabolism , Animals , Binding Sites , CHO Cells , Cricetulus , Cysteine , Drosophila/cytology , Drosophila/metabolism , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Hyperlipoproteinemia Type I/genetics , Models, Molecular , Phosphoinositide Phospholipase C/metabolism , Protein Binding , Protein Conformation , Protein Multimerization , Rats , Receptors, Lipoprotein/chemistry , Receptors, Lipoprotein/genetics , Transfection
7.
Cell Metab ; 20(3): 458-70, 2014 Sep 02.
Article in English | MEDLINE | ID: mdl-25066055

ABSTRACT

Sel1L is an essential adaptor protein for the E3 ligase Hrd1 in the endoplasmic reticulum (ER)-associated degradation (ERAD), a universal quality-control system in the cell; but its physiological role remains unclear. Here we show that mice with adipocyte-specific Sel1L deficiency are resistant to diet-induced obesity and exhibit postprandial hypertriglyceridemia. Further analyses reveal that Sel1L is indispensable for the secretion of lipoprotein lipase (LPL), independent of its role in Hrd1-mediated ERAD and ER homeostasis. Sel1L physically interacts with and stabilizes the LPL maturation complex consisting of LPL and lipase maturation factor 1 (LMF1). In the absence of Sel1L, LPL is retained in the ER and forms protein aggregates, which are degraded primarily by autophagy. The Sel1L-mediated control of LPL secretion is also seen in other LPL-expressing cell types including cardiac myocytes and macrophages. Thus, our study reports a role of Sel1L in LPL secretion and systemic lipid metabolism.


Subject(s)
Lipid Metabolism , Lipoprotein Lipase/metabolism , Proteins/metabolism , Adipocytes/metabolism , Animals , Cells, Cultured , Diet, High-Fat/adverse effects , Endoplasmic Reticulum-Associated Degradation , Female , Gene Deletion , Hyperglycemia/genetics , Hyperglycemia/metabolism , Intracellular Signaling Peptides and Proteins , Lipoprotein Lipase/chemistry , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/genetics , Obesity/metabolism , Protein Aggregates , Protein Multimerization , Proteins/genetics
8.
J Biol Chem ; 289(28): 19491-9, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24847059

ABSTRACT

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.


Subject(s)
Homozygote , Hyperlipoproteinemia Type I , Lipoprotein Lipase/metabolism , Mutation, Missense , Protein Multimerization/genetics , Receptors, Lipoprotein , Adult , Amino Acid Substitution , Cell Line , Humans , Hyperlipoproteinemia Type I/genetics , Hyperlipoproteinemia Type I/metabolism , Hyperlipoproteinemia Type I/pathology , Lipoprotein Lipase/genetics , Male , Protein Binding/genetics , Protein Structure, Tertiary , Protein Transport/genetics , Receptors, Lipoprotein/genetics , Receptors, Lipoprotein/metabolism
9.
Biochim Biophys Acta ; 1841(7): 970-6, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24681165

ABSTRACT

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.


Subject(s)
Antibodies, Monoclonal/metabolism , Lipoprotein Lipase/metabolism , Receptors, Lipoprotein/metabolism , Triglycerides/metabolism , Amino Acid Sequence , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/isolation & purification , Antibody Affinity , Antibody Specificity , CHO Cells , Cattle , Cricetulus , Gene Expression , Heparin/metabolism , Humans , Lipase/metabolism , Lipoprotein Lipase/genetics , Mice , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Protein Transport , Receptors, Lipoprotein/genetics , Transfection
10.
Biochim Biophys Acta ; 1841(7): 963-9, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24704550

ABSTRACT

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.


Subject(s)
Immobilized Proteins/metabolism , Lipoprotein Lipase/metabolism , Polymorphism, Single Nucleotide , Receptors, Lipoprotein/metabolism , Recombinant Fusion Proteins/metabolism , Triglycerides/metabolism , Amino Acid Sequence , Animals , Binding Sites , Biological Assay , CHO Cells , Cricetulus , Gene Expression , Humans , Immobilized Proteins/genetics , Lipid Metabolism , Lipoprotein Lipase/genetics , Molecular Sequence Data , Protein Binding , Protein Transport , Receptors, Lipoprotein/genetics , Recombinant Fusion Proteins/genetics
11.
Cell Metab ; 19(5): 849-60, 2014 May 06.
Article in English | MEDLINE | ID: mdl-24726386

ABSTRACT

Triglyceride-rich lipoproteins (TRLs) undergo lipolysis by lipoprotein lipase (LPL), an enzyme that is transported to the capillary lumen by an endothelial cell protein, GPIHBP1. For LPL-mediated lipolysis to occur, TRLs must bind to the lumen of capillaries. This process is often assumed to involve heparan sulfate proteoglycans (HSPGs), but we suspected that TRL margination might instead require GPIHBP1. Indeed, TRLs marginate along the heart capillaries of wild-type but not Gpihbp1⁻/⁻ mice, as judged by fluorescence microscopy, quantitative assays with infrared-dye-labeled lipoproteins, and EM tomography. Both cell-culture and in vivo studies showed that TRL margination depends on LPL bound to GPIHBP1. Notably, the expression of LPL by endothelial cells in Gpihbp1⁻/⁻ mice did not restore defective TRL margination, implying that the binding of LPL to HSPGs is ineffective in promoting TRL margination. Our studies show that GPIHBP1-bound LPL is the main determinant of TRL margination.


Subject(s)
Capillaries/metabolism , Lipoprotein Lipase/metabolism , Lipoproteins/metabolism , Receptors, Lipoprotein/metabolism , Triglycerides/metabolism , Animals , Cell Line , Endothelial Cells/metabolism , Heart/physiology , Mice
12.
Arterioscler Thromb Vasc Biol ; 33(7): 1706-13, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23661675

ABSTRACT

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.


Subject(s)
Angiopoietins/deficiency , Fatty Acids, Nonesterified/blood , Hypobetalipoproteinemias/enzymology , Insulin Resistance , Lipoprotein Lipase/blood , Adult , Aged , Analysis of Variance , Angiopoietin-Like Protein 3 , Angiopoietin-like Proteins , Angiopoietins/genetics , Biomarkers/blood , Blood Glucose/analysis , Case-Control Studies , Chi-Square Distribution , Down-Regulation , Female , Heterozygote , Homozygote , Humans , Hypobetalipoproteinemias/blood , Hypobetalipoproteinemias/genetics , Hypobetalipoproteinemias/physiopathology , Insulin/blood , Italy , Linear Models , Lipase/blood , Lipoproteins, LDL/blood , Male , Middle Aged , Mutation , Triglycerides/blood , Up-Regulation
13.
BMC Physiol ; 12: 14, 2012 Nov 27.
Article in English | MEDLINE | ID: mdl-23186339

ABSTRACT

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.


Subject(s)
Leptin/deficiency , Leptin/metabolism , Lipoprotein Lipase/metabolism , Pancreas/metabolism , Receptors, Lipoprotein/metabolism , Animals , Capillaries/metabolism , Diet, High-Fat/methods , Endothelial Cells/metabolism , Glucagon/metabolism , Glucagon-Secreting Cells/metabolism , Glycosylphosphatidylinositols/metabolism , Heparin/metabolism , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Platelet Endothelial Cell Adhesion Molecule-1/metabolism
14.
J Lipid Res ; 53(12): 2690-7, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23008484

ABSTRACT

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.


Subject(s)
Endothelial Cells/metabolism , Lipoprotein Lipase/metabolism , Receptors, Lipoprotein/metabolism , Animals , CHO Cells , Cricetinae , Endothelial Cells/chemistry , Endothelial Cells/enzymology , Genistein/pharmacology , Humans , Hydrazones/pharmacology , Lipoprotein Lipase/antagonists & inhibitors , Mice , Mice, Knockout , Rats , Receptors, Lipoprotein/deficiency , Structure-Activity Relationship
15.
Hum Mol Genet ; 21(13): 2961-72, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22493000

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Lipoprotein Lipase/metabolism , Amino Acid Substitution , Animals , CHO Cells , Carrier Proteins/genetics , Chylomicrons/blood , Cricetinae , Endothelial Cells/metabolism , Furin/metabolism , Lipoprotein Lipase/chemistry , Lipoprotein Lipase/genetics , Mutation, Missense , Peptides/genetics , Peptides/metabolism , Protein Binding
16.
Arterioscler Thromb Vasc Biol ; 32(5): 1204-10, 2012 May.
Article in English | MEDLINE | ID: mdl-22345169

ABSTRACT

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.


Subject(s)
DNA/genetics , Energy Metabolism/physiology , Gene Expression Regulation , Genetic Variation , Hypertriglyceridemia/genetics , Lipoprotein Lipase/genetics , Membrane Proteins/genetics , Adipose Tissue/metabolism , Animals , Humans , Hypertriglyceridemia/metabolism , Lipoprotein Lipase/biosynthesis , Membrane Proteins/biosynthesis , Mice , Mice, Transgenic , Muscle, Skeletal/metabolism , Myocardium/metabolism
17.
J Inherit Metab Dis ; 35(3): 531-40, 2012 May.
Article in English | MEDLINE | ID: mdl-22008945

ABSTRACT

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.


Subject(s)
Carrier Proteins/genetics , Chylomicrons/chemistry , Gene Deletion , Hypertriglyceridemia/genetics , Adult , Apolipoprotein C-II/metabolism , Child , Child, Preschool , Cytogenetics , Female , Gene Dosage , Heparin/metabolism , Homozygote , Humans , Hypertriglyceridemia/etiology , Infant , Lipoprotein Lipase/chemistry , Male , Middle Aged , Mutation , Receptors, Lipoprotein
19.
J Lipid Res ; 52(11): 1869-84, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21844202

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Endothelial Cells/metabolism , Lipoprotein Lipase/metabolism , Amino Acid Sequence , Animals , CHO Cells , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cricetinae , Cricetulus , Humans , Molecular Sequence Data , Protein Transport , Receptors, Lipoprotein
20.
Proc Natl Acad Sci U S A ; 108(19): 7980-4, 2011 May 10.
Article in English | MEDLINE | ID: mdl-21518912

ABSTRACT

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.


Subject(s)
Carrier Proteins/metabolism , Endothelial Cells/metabolism , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Mutation, Missense , Amino Acid Substitution , Animals , Binding Sites/genetics , CHO Cells , Chylomicrons/blood , Chylomicrons/genetics , Cricetinae , Cricetulus , Humans , Hyperlipoproteinemia Type IV/blood , Hyperlipoproteinemia Type IV/enzymology , Hyperlipoproteinemia Type IV/genetics , Mutant Proteins/genetics , Mutant Proteins/metabolism , Protein Binding , Receptors, Lipoprotein , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...