Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Type of study
Publication year range
1.
J Biol Chem ; 290(26): 16077-87, 2015 Jun 26.
Article in English | MEDLINE | ID: mdl-25918162

ABSTRACT

Apolipoprotein D (apoD), a member of the lipocalin family, is a 29-kDa secreted glycoprotein that binds and transports small lipophilic molecules. Expressed in several tissues, apoD is up-regulated under different stress stimuli and in a variety of pathologies. Numerous studies have revealed that overexpression of apoD led to neuroprotection in various mouse models of acute stress and neurodegeneration. This multifunctional protein is internalized in several cells types, but the specific internalization mechanism remains unknown. In this study, we demonstrate that the internalization of apoD involves a specific cell surface receptor in 293T cells, identified as the transmembrane glycoprotein basigin (BSG, CD147); more particularly, its low glycosylated form. Our results show that internalized apoD colocalizes with BSG into vesicular compartments. Down-regulation of BSG disrupted the internalization of apoD in cells. In contrast, overexpression of basigin in SH-5YSY cells, which poorly express BSG, restored the uptake of apoD. Cyclophilin A, a known ligand of BSG, competitively reduced apoD internalization, confirming that BSG is a key player in the apoD internalization process. In summary, our results demonstrate that basigin is very likely the apoD receptor and provide additional clues on the mechanisms involved in apoD-mediated functions, including neuroprotection.


Subject(s)
Apolipoproteins D/metabolism , Basigin/metabolism , Apolipoproteins D/genetics , Basigin/genetics , Cell Line , Cell Membrane/genetics , Cell Membrane/metabolism , Humans , Protein Transport
2.
Physiol Rep ; 2(10)2014 Oct 01.
Article in English | MEDLINE | ID: mdl-25281615

ABSTRACT

Scavenger receptor class B type I (SR-BI), the Scarb1 gene product, is a high-density lipoprotein (HDL) receptor which was shown to influence bone metabolism. Its absence in mice is associated with alterations of the glucocorticoid/adrenocorticotropic hormone axis, and translated in high bone mass and enhanced bone formation. Since the cellular alterations underlying the enhanced bone formation remain unknown, we investigated Scarb1-deficient marrow stromal cells (MSC) behavior in vitro. No difference in HDL3, cholesteryl ester (CE) or estradiol (E) association/binding was measured between Scarb1-null and wild-type (WT) cells. Scarb1 genic expression was down-regulated twofold following osteogenic treatment. Neither WT nor null cell proliferation was influenced by HDL3 exposure whereas this condition decreased genic expression of osteoblastic marker osterix (Sp7), and osteocyte markers sclerostin (Sost) and dentin matrix protein 1 (Dmp1) independently of genotype. Sost and Dmp1 basal expression in null cells was 40% and 50% that of WT cells; accordingly, osteocyte density was 20% lower in vertebrae from Scarb1-null mice. Genic expression of co-receptors for Wnt signaling, namely LDL-related protein (Lrp) 5 and Lrp8, was increased, respectively, by two- and threefold, and of transcription target-genes axis inhibition protein 2 (Axin2) and lymphoid enhancer-binding factor 1 (Lef1) over threefold. Gene expression of Wnt signaling agonist Wnt5a and of the antagonist dickkopfs-related protein 1 (Dkk1) were found to be increased 10- to 20-fold in null MSC. These data suggest alterations of Wnt pathways in Scarb1-deficient MSC potentially explaining their enhanced function, hence contributing to the high bone mass observed in these mice.

3.
J Endocrinol ; 222(2): 277-88, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24928939

ABSTRACT

A positive correlation between plasma levels of HDL and bone mass has been reported by epidemiological studies. As scavenger receptor class B, type I (SR-BI), the gene product of Scarb1, is known to regulate HDL metabolism, we recently characterized bone metabolism in Scarb1-null mice. These mice display high femoral bone mass associated with enhanced bone formation. As gender differences have been reported in HDL metabolism and SR-BI function, we investigated gender-specific bone alterations in Scarb1-null mice by microtomography and histology. We found 16% greater relative bone volume and 39% higher bone formation rate in the vertebrae from 2-month-old Scarb1-null females. No such alteration was seen in males, indicating gender- and region-specific differences in skeletal phenotype. Total and HDL-associated cholesterol levels, as well as ACTH plasma levels, were increased in both Scarb1-null genders, the latter being concurrent to impaired corticosterone response to fasting. Plasma levels of estradiol did not differ between null and WT females, suggesting that the estrogen metabolism alteration is not relevant to the higher vertebral bone mass in female Scarb1-null mice. Constitutively, high plasma levels of leptin along with 2.5-fold increase in its expression in white adipose tissue were measured in female Scarb1-null mice only. In vitro exposure of bone marrow stromal cells to ACTH and leptin promoted osteoblast differentiation as evidenced by increased gene expression of osterix and collagen type I alpha. Our results suggest that hyperleptinemia may account for the gender-specific high bone mass seen in the vertebrae of female Scarb1-null mice.


Subject(s)
Bone Density , Femur/metabolism , Leptin/blood , Osteogenesis , Scavenger Receptors, Class B/deficiency , Spine/metabolism , Adrenocorticotropic Hormone/blood , Adrenocorticotropic Hormone/pharmacology , Animals , Cell Differentiation/drug effects , Cholesterol, HDL/metabolism , Collagen Type I/biosynthesis , Collagen Type I, alpha 1 Chain , Female , Leptin/pharmacology , Male , Mesenchymal Stem Cells/drug effects , Mice , Mice, Knockout , Sex Factors , Sp7 Transcription Factor , Transcription Factors/biosynthesis , X-Ray Microtomography
4.
Am J Physiol Endocrinol Metab ; 306(1): E48-57, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24253048

ABSTRACT

Scavenger receptor class B, type I (SR-BI), the Scarb1 gene product, is a receptor associated with cholesteryl ester uptake from high-density lipoproteins (HDL), which drives cholesterol movement from peripheral tissues toward the liver for excretion, and, consequently, Scarb1 null mice are prone to atherosclerosis. Because studies have linked atherosclerosis incidence with osteoporosis, we characterized the bone metabolism in these mice. Bone morphometry was assessed through microcomputed tomography and histology. Marrow stromal cells (MSCs) were used to characterize influence of endogenous SR-BI in cell functions. Total and HDL-associated cholesterol in null mice were increased by 32-60%, correlating with its role in lipoprotein metabolism. Distal metaphyses from 2- and 4-mo-old null mice showed correspondingly 46 and 37% higher bone volume fraction associated with a higher number of trabeculae. Histomorphometric analyses in 2-mo-old null male mice revealed 1.42-fold greater osteoblast surface, 1.37-fold higher percent mineralizing surface, and 1.69-fold enhanced bone formation rate. In vitro assays for MSCs from null mice revealed 37% higher proliferation rate, 48% more alkaline phosphatase activity, 70% greater mineralization potential and a 2-fold osterix (Sp7) expression, yet a 0.5-fold decrease in caveolin-1 (Cav1) expression. Selective uptake levels of HDL-associated cholesteryl oleate and estradiol were similar between MSC from wild-type and Scarb1 null mice, suggesting that its contribution to this process is not its main role in these cells. However, Scarb1 knockout stunted the HDL-dependent regulation of Cav1 genic expression. Scarb1 null mice are not prone to osteoporosis but show higher bone mass associated with enhanced bone formation.


Subject(s)
Atherosclerosis/complications , Bone and Bones/metabolism , Osteoporosis/etiology , Scavenger Receptors, Class B/deficiency , Scavenger Receptors, Class B/physiology , Animals , Atherosclerosis/pathology , Bone Marrow Cells/pathology , Bone and Bones/pathology , Calcification, Physiologic , Caveolin 1/genetics , Cell Proliferation , Cholesterol/blood , Cholesterol, HDL/blood , Disease Models, Animal , Female , Gene Expression , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/pathology , Osteogenesis , Stromal Cells/pathology
5.
PLoS One ; 8(10): e77701, 2013.
Article in English | MEDLINE | ID: mdl-24204923

ABSTRACT

Bone tissue is continuously remodeled by bone cells and maintenance of its mass relies on the balance between the processes of resorption and formation. We have reported the expression of numerous scavenger receptors, namely scavenger receptor (SR) class B type I and II (SR-BI and SR-BII), and CD36, in bone-forming osteoblasts but their physiological roles in bone metabolism are still unknown. To unravel the role of CD36 in bone metabolism, we determined the bone phenotype of CD36 knockout (CD36KO) mice and characterized the cell functions of osteoblasts lacking CD36. Weights of CD36KO mice were significantly lower than corresponding wild-type (WT) mice, yet no significant difference was found in femoral nor tibial length between CD36KO and WT mice. Analysis of bone architecture by micro-computed tomography revealed a low bone mass phenotype in CD36KO mice of both genders. Femoral trabecular bone from 1 to 6 month-old CD36KO mice showed lower bone volume, higher trabecular separation and reduced trabeculae number compared to WT mice; similar alterations were noticed for lumbar vertebrae. Plasma levels of osteocalcin (OCN) and N-terminal propeptide of type I procollagen (PINP), two known markers of bone formation, were significantly lower in CD36KO mice than in WT mice, whereas plasma levels of bone resorption markers were similar. Accordingly, histology highlighted lower osteoblast perimeter and reduced bone formation rate. In vitro functional characterization of bone marrow stromal cells and osteoblasts isolated from CD36KO mice showed reduced cell culture expansion and survival, lower gene expression of osteoblastic Runt-related transcription factor 2 (Runx2) and osterix (Osx), as well as bone sialoprotein (BSP) and osteocalcin (OCN). Our results indicate that CD36 is mandatory for adequate bone metabolism, playing a role in osteoblast functions ensuring adequate bone formation.


Subject(s)
Blood Platelet Disorders/complications , Blood Platelet Disorders/pathology , Bone and Bones/pathology , CD36 Antigens/deficiency , Genetic Diseases, Inborn/complications , Genetic Diseases, Inborn/pathology , Animals , Blood Platelet Disorders/genetics , Blood Platelet Disorders/metabolism , Bone Density/genetics , Bone Density/physiology , Bone and Bones/metabolism , CD36 Antigens/genetics , CD36 Antigens/metabolism , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line , Femur/metabolism , Femur/pathology , Gene Expression/genetics , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoblasts/metabolism , Osteoblasts/pathology , Osteocalcin/blood , Osteocalcin/genetics , Osteocalcin/metabolism , Osteogenesis/genetics , Osteogenesis/physiology , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phenotype , Procollagen/genetics , Procollagen/metabolism , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism , Tibia/metabolism , Tibia/pathology
6.
Atherosclerosis ; 229(2): 408-14, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23880196

ABSTRACT

AIMS: The CD36 selective ligand, EP 80317, features potent anti-atherosclerotic and hypocholesterolemic effects that are associated with an increase in macrophage cholesterol efflux through the activation of the peroxisome proliferator-activated receptor γ-liver X receptor α (LXRα)-ATP-binding cassette (ABC) transporter pathway. Cholesterol efflux is the first step of reverse cholesterol transport (RCT). However, whether EP 80317 exerts its hypocholesterolemic and anti-atherosclerotic activity through RCT in vivo has yet to be determined. In the present study, we investigated the effects of EP 80317 on RCT, in particular on macrophage-to-feces RCT and the expression of selected genes associated with hepatic cholesterol metabolism and intestinal cholesterol transport. METHODS AND RESULTS: Reverse cholesterol transport was assessed following the intraperitoneal injection of [(3)H]-cholesterol-labelled J774 macrophages to hypercholesterolemic apoE- and apoE/CD36 double-deficient mice that had been treated for 12 weeks with EP 80317. Forty-eight hours after the administration of [(3)H]-cholesterol-labelled cells, blood, liver, intestines and feces were harvested. The radioactivity recovered in the feces (cholesterol and bile acid combined) was significantly increased by 311% (P = 0.0259) in EP 80317-treated mice compared with that found in vehicle-treated mice despite no significant change in [(3)H]-tracer recovery in plasma between groups. Whereas the mRNA levels of LXRα in the gut were significantly upregulated, mRNA and protein levels of the Niemann-Pick C1-like 1 protein (NPC1L1) transporter, a LXRα target which regulates intestinal cholesterol absorption, were downregulated in EP 80317-treated mice. In contrast, neither mRNA nor protein levels of investigated transporters and receptors were modulated in the small intestine of double-deficient mice, nor was the fecal recovery of radioactivity. No change was observed in targeted genes in liver of either apoE- or apoE/CD36 double-deficient mice after a chronic treatment with EP 80317. CONCLUSION: This study shows that EP 80317 elicits macrophage-to-feces reverse cholesterol transport in a manner dependent on CD36 expression. This effect is associated with the upregulation of LXRα and the downregulation of NPC1L1 expression.


Subject(s)
Apolipoproteins E/genetics , Atherosclerosis/drug therapy , Atherosclerosis/metabolism , CD36 Antigens/metabolism , Cholesterol/metabolism , Intestinal Absorption/drug effects , Oligopeptides/pharmacology , ATP Binding Cassette Transporter, Subfamily G, Member 5 , ATP Binding Cassette Transporter, Subfamily G, Member 8 , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Female , Intestinal Absorption/physiology , Lipoproteins/genetics , Lipoproteins/metabolism , Liver/metabolism , Liver X Receptors , Macrophages/cytology , Macrophages/drug effects , Macrophages/metabolism , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Naphthols , Orphan Nuclear Receptors/genetics , Orphan Nuclear Receptors/metabolism , RNA, Messenger/metabolism , Triazines
7.
Cell Biochem Funct ; 28(6): 480-9, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20629037

ABSTRACT

In non-hepatic cells, scavenger receptor class B type I (SR-BI), cluster of differentiation 36 (CD36), and caveolin-1 were described as mediators of cholesterol efflux, the first step of reverse cholesterol transport (RCT). Stable transformants of HepG2 cells overexpressing SR-BI, CD36, or caveolin-1 were generated, as well as cells overexpressing both caveolin-1 and SR-BI or caveolin-1 and CD36 in order to address the effect of caveolin-1 on both receptor activities. These cells were analyzed for their ability to efflux cholesterol to HDL(3). Our results show that overexpressing SR-BI, CD36, or caveolin-1 increases cholesterol efflux by 106, 92, and 48%, respectively. Moreover, the dual overexpressions of caveolin-1 and SR-BI or caveolin-1 and CD36 lead to a more prominent increase in cholesterol efflux. Studies were also conducted with primary cultures of SR-BI knockout (KO), CD36 KO, and SR-BI/CD36 double-KO (dKO) mice. SR-BI KO and SR-BI/CD36 dKO hepatic cells show 41 and 56% less cholesterol efflux, respectively, than normal hepatic cells. No significant difference was observed between the efflux of normal and CD36 KO cells. The difference between the role of human and murine CD36 correlated with the absence of CD36 dimers in mouse caveolae/rafts. Overall, our results show that SR-BI is clearly involved in cholesterol efflux in mouse and human hepatic cells, while CD36 plays a significant role in human cells.


Subject(s)
CD36 Antigens/metabolism , Caveolin 1/metabolism , Cholesterol/metabolism , Hepatocytes/metabolism , Scavenger Receptors, Class B/metabolism , Animals , CD36 Antigens/genetics , Caveolin 1/genetics , Cells, Cultured , Female , Hep G2 Cells , Humans , Male , Mice , Mice, Knockout , Scavenger Receptors, Class B/genetics
8.
Biochim Biophys Acta ; 1801(1): 42-8, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19761869

ABSTRACT

Plasma cholesterol from low- and high-density lipoproteins (LDL and HDL) are cleared from the circulation by specific receptors that either totally degrade lipoproteins as the LDL receptor or selectively take up their cholesteryl esters (CE) like the scavenger receptor class B type I (SR-BI). The aim of the present study was to define the effect of apoC-I on the uptake of LDL and HDL(3) by HepG2 cells. In experiments conducted with exogenously added purified apoC-I, no significant effect was observed on lipoprotein-protein association and degradation; however, LDL- and HDL(3)-CE selective uptake was significantly reduced in a dose-dependent manner. This study also shows that apoC-I has the ability to associate with HepG2 cells and with LDL and HDL(3). Moreover, pre-incubation of HepG2 cells with apoC-I reduces HDL(3)-CE selective uptake and pre-incubation of LDL and HDL(3) with apoC-I decreases their CE selective uptake by HepG2 cells. Thus, apoC-I can accomplish its inhibitory effect on SR-BI activity by either binding to SR-BI or lipoproteins. We conclude that by reducing hepatic lipoprotein-CE selective uptake, apoC-I has an atherogenic character.


Subject(s)
Apolipoprotein C-I/metabolism , Cholesterol Esters/metabolism , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Lipoproteins/metabolism , Binding Sites , Cells, Cultured , Humans , Lipoproteins/blood , Lipoproteins, HDL/blood , Lipoproteins, LDL/blood
9.
J Cell Biochem ; 108(4): 906-15, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19718657

ABSTRACT

Oxidized LDL (OxLDL) that are positively associated with the risk of developing cardiovascular diseases are ligands of scavenger receptor-class B type I (SR-BI) and cluster of differentiation-36 (CD36) which can be found in caveolae. The contribution of these receptors in human hepatic cell is however unknown. The HepG2 cell, a human hepatic parenchymal cell model, expresses these receptors and is characterized by a very low level of caveolin-1. Our aim was to define the contribution of human CD36, SR-BI, and caveolin-1 in the metabolism of OxLDL in HepG2 cells and conversely the effects of OxLDL on the levels/localization of these receptors. By comparing mildly (M)- and heavily (H)-OxLDL metabolism between control HepG2 cells and HepG2 cells overexpressing CD36, SR-BI, or caveolin-1, we found that (1) CD36 increases M- and H-OxLDL-protein uptake; (2) SR-BI drives M-OxLDL through a degradation pathway at the expense of the cholesterol ester (CE) selective uptake pathway; (3) caveolin-1 increases M- and H-OxLDL-protein uptake and decreases CE selective uptake from M-OxLDL. Also, incubation with M- or H-OxLDL decreases the levels of SR-BI and LDL-receptor in control HepG2 cells which can be overcome by caveolin-1 expression. In addition, OxLDL move CD36 from low to high buoyant density membrane fractions, as well as caveolin-1 in cells overexpressing this protein. Thus, hepatic caveolin-1 expression has significant effects on OxLDL metabolism and on lipoprotein receptor levels.


Subject(s)
Caveolin 1/biosynthesis , Gene Expression Regulation , Lipoproteins, LDL/metabolism , Lipoproteins/chemistry , Liver/cytology , Liver/metabolism , Receptors, LDL/metabolism , Biological Transport , CD36 Antigens/biosynthesis , CD36 Antigens/metabolism , Cell Line , Cell Survival , Hep G2 Cells , Humans , Models, Biological
10.
J Cell Biochem ; 105(6): 1374-85, 2008 Dec 15.
Article in English | MEDLINE | ID: mdl-18980242

ABSTRACT

Oxidized low density lipoproteins (OxLDL) are known to promote atherosclerosis, but it is only recently that OxLDL have been associated with alterations of the functions of bone-forming osteoblasts and osteoporosis. Although high density lipoproteins (HDL) are recognized for their anti-atherogenic action, there is less information about their ability to protect against osteoporosis. Therefore, we investigated the capacity of HDL3 to prevent the cell death induced by OxLDL in human osteoblastic cells. Simultaneous exposure of the cells to HDL3 and OxLDL abolished the reduction of cell viability monitored by MTT activity measurement and the induction of apoptosis determined by annexin V staining indicating that HDL3 prevent the apoptosis of osteoblasts induced by OxLDL. This protection correlated with the displacement by HDL3 of OxLDL association to osteoblasts, signifying that OxLDL binding and/or internalization are/is necessary for their cytotoxic effects. We also found that exposition of osteoblastic cells to HDL3 prior to incubation with OxLDL reduced cell death and preserved the lysosomal integrity. This protection was correlated with an increase of SR-BI expression, a modification of OxLDL metabolism with less global uptake of OxLDL and greater selective uptake of cholesterol from OxLDL. These results strongly suggest that, as for atherosclerosis, HDL may exert beneficial actions on bone metabolism.


Subject(s)
Lipoproteins, HDL3/metabolism , Lipoproteins, LDL/metabolism , Osteoblasts/metabolism , Cell Death/physiology , Cells, Cultured , Humans , Lipoproteins, LDL/pharmacology
11.
Arterioscler Thromb Vasc Biol ; 28(7): 1290-5, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18436808

ABSTRACT

OBJECTIVE: The cluster of differentiation-36 (CD36) is a multifunctional protein which is recognized for its in vitro ability to take up oxidized low-density lipoproteins (oxLDL) in macrophages and is therefore considered atherogenic. It also binds LDL. Our objective was to define the physiological role of CD36 in both native LDL and oxLDL metabolism in mice. METHODS AND RESULTS: Clearance studies of labeled LDL and oxLDL were conducted in wild-type, CD36 knockout (KO), scavenger receptor class B, type I (SR-BI) KO, and SR-BI/CD36 double KO mice. We found that CD36 impedes the disappearance of native LDL and favors that of oxLDL. This was confirmed by association and degradation assays with primary cultures of hepatic cells from wild-type and CD36 KO mice. In addition, our in vivo work indicates that neither SR-BI nor CD36 plays a significant role in cholesteryl esters (CE) selective uptake (SU) from oxLDL, whereas CD36, in absence of SR-BI, can selectively take CE from LDL. CONCLUSIONS: Our investigation showed for the first time that CD36 plays a significant role in oxLDL uptake in vivo in the mouse. As CD36 also retards LDL clearance, its atherogenic character may also relate to its negative effect on LDL catabolism.


Subject(s)
CD36 Antigens/metabolism , Hepatocytes/metabolism , Lipoproteins, LDL/metabolism , Animals , CD36 Antigens/genetics , Cells, Cultured , Cholesterol Esters/metabolism , Hepatocytes/immunology , Kinetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Binding , Scavenger Receptors, Class B/genetics , Scavenger Receptors, Class B/metabolism
12.
Free Radic Biol Med ; 44(4): 506-17, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-18241787

ABSTRACT

Cardiovascular diseases have recently been noted as potential risk factors for osteoporosis development. Although it is poorly understood how these two pathologies are related, it is a known fact that oxidized low-density lipoproteins (OxLDL) constitute potential determinants for both of them. The current study investigated the metabolism of OxLDL by osteoblasts and its effect on osteoblastic viability. The results obtained show that OxLDL are internalized but not degraded by osteoblasts while they can selectively transfer their CE to these cells. It is also demonstrated that OxLDL induce proliferation at low concentrations but cell death at high concentrations. This reduction of osteoblast viability was associated with lysosomal membrane damage caused by OxLDL as demonstrated by acridine orange relocalization. Accordingly, chloroquine, an inhibitor of lysosomal activity, accentuated cell death induced by OxLDL. Finally, we demonstrate that osteoblasts have the capacity to oxidize LDL and thereby potentially increase the local concentration of OxLDL. Overall, the current study confirms the potential role of OxLDL in the development of osteoporosis given its influence on osteoblastic viability.


Subject(s)
Lipoproteins, LDL/toxicity , Osteoblasts/drug effects , Osteoporosis/etiology , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Humans , Lipoproteins, LDL/metabolism , Lipoproteins, LDL/pharmacology , Lysosomes/drug effects , Mice , Osteoblasts/metabolism , Oxidation-Reduction , Tetrazolium Salts/metabolism , Thiazoles/metabolism
13.
J Bone Miner Res ; 23(3): 326-37, 2008 Mar.
Article in English | MEDLINE | ID: mdl-17967141

ABSTRACT

UNLABELLED: Lipoproteins transport many vitamins and hormones that have been shown to be necessary for bone formation. However, the metabolism of LDL and HDL3 by bone-forming osteoblastic cells remains unknown. Here we report that osteoblastic cells express scavenger receptors of class B that are implicated in the uptake of cholesterol and estradiol from LDL and HDL3. INTRODUCTION: The bone tissue is continuously remodeled, and its integrity requires a balance between osteoclastic bone resorption and osteoblastic bone formation. Recent studies have reported the importance of triglyceride-rich lipoproteins for the delivery of lipophilic vitamins necessary for normal bone metabolism. However, the ability of osteoblastic cells to process low- and high-density lipoproteins (LDL and HDL3) and the receptors involved remain unknown. MATERIALS AND METHODS: Binding, competition, degradation, and selective uptake assays with LDL and HDL3 radiolabeled in their protein and lipid moieties or with [3H]estradiol were conducted on human osteoblasts (MG-63 cell line and primary cultures of human osteoblasts [hOB cells]) and on mouse osteoblasts (MC3T3-E1 cell line and primary cultures of murine osteoblasts [mOB cells]). The expression of scavenger receptors (SRs) by osteoblastic cells was determined by RT-PCR and Western immunoblotting, and cellular localization was assessed by sucrose gradient fractionation. RESULTS: Osteoblastic cells were able to bind, internalize, and degrade HDL3 and LDL and are capable of selectively taking up cholesteryl esters (CEs) from these lipoproteins. Also, we provide evidence that osteoblastic cells express SR-BI, SR-BII, and CD36 (SR-Bs receptors) and that these receptors are localized in membrane lipid rafts or caveolin-rich membranes. The selective uptake of CE from LDL and HDL3 by osteoblastic cells was strongly inhibited by the known SR-B ligand oxidized LDL, indicating that SR-B receptors are responsible for the selective uptake. Finally, estradiol carried by LDL and HDL3 was selectively transferred to the osteoblastic cells also through SR-B receptors. CONCLUSIONS: Overall, our results suggest a novel mechanism for the routing of cholesterol and estradiol to osteoblasts involving the metabolism of LDL and HDL3 by SR-B receptors.


Subject(s)
Bone Remodeling/physiology , Cholesterol Esters/metabolism , Estradiol/metabolism , Lipoproteins, HDL3/metabolism , Lipoproteins, LDL/metabolism , Osteoblasts/metabolism , Scavenger Receptors, Class B/biosynthesis , Animals , Bone Remodeling/drug effects , Cell Line , Cholesterol Esters/pharmacology , Estradiol/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Lipoproteins, HDL3/pharmacology , Lipoproteins, LDL/pharmacology , Mice
14.
Biochem Cell Biol ; 84(2): 250-6, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16609706

ABSTRACT

The aim of this study was to quantify the abilities of mouse liver parenchymal and nonparenchymal cells with respect to (i) cholesteryl ester (CE) selective uptake from low-density lipoproteins (LDL), oxidized LDL (OxLDL), and high-density lipoprotein (HDL); and (ii) their free cholesterol efflux to HDL. The preparations of cells were incubated with lipoproteins labelled either in protein with iodine-125 or in CE with 3H-cholesterol oleate, and lipoprotein-protein and lipoprotein-CE associations were measured. The associations of LDL-protein and LDL-CE with nonparenchymal cells were 5- and 2-fold greater, respectively, than with parenchymal cells. However, in terms of CE-selective uptake (CE association minus protein association) both types of cell were equivalent. Similar results were obtained with OxLDL, but both types of cell showed higher abilities in OxLDL-CE than in LDL-CE selective uptake (on average by 3.4-fold). The association of HDL-protein with nonparenchymal cells was 3x that with parenchymal cells; however, nonparenchymal cells associated 45% less HDL-CE. Contrary to parenchymal cells, nonparenchymal cells did not show HDL-CE selective uptake activity. Thus parenchymal cells selectively take CE from the 3 types of lipoproteins, whereas nonparenchymal cells exert this function only on LDL and OxLDL. Efflux was 3.5-fold more important in nonparenchymal than in parenchymal cells.


Subject(s)
Cholesterol/metabolism , Hepatocytes/metabolism , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Animals , Biological Transport, Active , Cell Separation , Cells, Cultured , Flow Cytometry , Hepatocytes/classification , Hepatocytes/cytology , In Vitro Techniques , Male , Mice , Oxidation-Reduction
15.
Biochim Biophys Acta ; 1761(1): 24-36, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16443388

ABSTRACT

Receptors of the scavenger class B family were reported to be localized in caveolae, the cell surface microdomains rich in free cholesterol and glycosphyngolipids, which are characterized by the presence of caveolin-1. Parenchymal hepatic and hepatoma HepG2 cells express very low levels of caveolin-1. In the present study, stable transformants of HepG2 cells expressing caveolin-1 were generated to address the effect of caveolin-1 on receptor activity. Compared to normal cells, these cells show higher (125)I-bovine serum albumin (BSA) uptake and cholesterol efflux, two indicators of functional caveolae. By immunoprecipitation, cell fractionation and confocal analyses, we found that caveolin-1 is well colocalized with the cluster of differentiation-36 (CD36) and the low-density lipoprotein (LDL) receptor (LDLr) but to a lesser extent with the scavenger receptor class B type I (SR-BI) in HepG2 cells expressing caveolin-1. However, caveolin-1 expression favors the dimerization of SR-BI. Two clones of cells expressing caveolin-1 were investigated for their lipoprotein metabolism activity. Compared to normal cells, these cells show a 71-144% increase in (125)I-LDL degradation. The analysis of the cholesteryl esters (CE)-selective uptake (CE association minus protein association) revealed that the expression of caveolin-1 in HepG2 cells decreases by 59%-73% LDL-CE selective uptake and increases high-density lipoprotein (HDL)-CE selective uptake by 44%-66%. We conclude that the expression of caveolin-1 in HepG2 cells moves the balance of LDL degradation/CE selective uptake towards degradation and favors HDL-CE selective uptake. Thus, in the normal hepatic parenchymal situation where caveolin-1 is poorly expressed, LDL-CE selective uptake is the preferred pathway.


Subject(s)
Caveolin 1/physiology , Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Biological Transport , Caveolin 1/genetics , Cell Line , Cell Line, Tumor , Humans , Kinetics , Liver , Liver Neoplasms , RNA, Messenger/genetics , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Serum Albumin, Bovine/metabolism , Transfection
16.
Int J Biochem Cell Biol ; 38(7): 1160-70, 2006.
Article in English | MEDLINE | ID: mdl-16427800

ABSTRACT

In blood circulation, low density lipoproteins (LDL) can undergo modification, such as oxidation, and become key factors in the development of atherosclerosis. Although the liver is the major organ involved in the elimination of oxidized LDL (oxLDL), the identity of the receptor(s) involved remains to be defined. Our work aims to clarify the role of the scavenger receptor class B type I (SR-BI) in the hepatic metabolism of mildly and standardly oxLDL as well as the relative contribution of parenchymal (hepatocytes) and nonparenchymal liver cells with a special emphasis on CE-selective uptake. The association of native LDL and mildly or standardly oxLDL labeled either in proteins or in cholesteryl esters (CE) was measured on primary cultures of mouse hepatocytes from normal and SR-BI knock-out (KO) mice. These in vitro assays demonstrated that hepatocytes are able to mediate CE-selective uptake from both LDL and oxLDL and that SR-BI KO hepatocytes have a 60% reduced ability to selectively take CE from LDL but not towards mildly or standardly oxLDL. When lipoproteins were injected in the mouse inferior vena cava, parenchymal and nonparenchymal liver cells accumulated more CE than proteins from native, mildly and standardly oxLDL, indicating that selective uptake of CE from these lipoproteins occurs in vivo in these two cell types. The parenchymal cells contribute near 90% of the LDL-CE selective uptake and SR-BI for 60% of this pathway. Nonparenchymal cells capture mainly standardly oxLDL while parenchymal and nonparenchymal cells equally take up mildly oxLDL. An 82% reduction of standardly oxLDL-CE selective uptake by the nonparenchymal cells of SR-BI KO mice allowed emphasizing the contribution of SR-BI in hepatic metabolism of standardly oxLDL. However, SR-BI is not responsible for mildly oxLDL metabolism. Thus, SR-BI is involved in LDL- and standardly oxLDL-CE selective uptake in parenchymal and nonparenchymal cells, respectively.


Subject(s)
Cholesterol Esters/metabolism , Cholesterol Esters/pharmacokinetics , Cholesterol, LDL/pharmacokinetics , Hepatocytes/metabolism , Lipoproteins, LDL/pharmacokinetics , Scavenger Receptors, Class B/metabolism , Animals , Lipoproteins, LDL/metabolism , Mice , Mice, Inbred Strains/metabolism , Mice, Knockout , Oxidation-Reduction
17.
Int J Biochem Cell Biol ; 37(6): 1308-18, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15778093

ABSTRACT

Plasma low- and high-density lipoproteins (LDL and HDL) are cleared from the circulation by specific receptors and are either totally degraded or their cholesteryl esters (CE) are selectively delivered to cells by receptors such as the scavenger receptor class B type I (SR-BI). The aim of the present study was to define the effect of apoC-II and apoC-III on the uptake of LDL and HDL by HepG2 cells. Stable transformants were obtained with sense or antisense strategies that secrete 47-294% the normal level of apoC-II or 60-200% that of apoC-III. Different levels of secreted apoC-II or apoC-III had little effect on LDL and HDL protein degradation by HepG2 cells. However, compared to controls, cells under-expressing apoC-II showed a 160% higher capacity to selectively take up HDL-CE, while cells under-expressing apoC-III demonstrated 70 and 160% higher capacity to take up CE from LDL and HDL, respectively. In experiments conducted with exogenously added apoC-II or apoC-III, no significant effect was observed on lipoprotein-protein association/degradation; however, LDL-CE and HDL-CE selective uptake was significantly reduced in a dose-dependent manner. These results indicate that apoC-II and apoC-III inhibit CE-selective uptake.


Subject(s)
Apolipoproteins C/physiology , Cholesterol, HDL/antagonists & inhibitors , Cholesterol, HDL/metabolism , Cholesterol, LDL/antagonists & inhibitors , Cholesterol, LDL/metabolism , Apolipoprotein C-II , Apolipoprotein C-III , Apolipoproteins C/metabolism , CD36 Antigens , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Humans , Receptors, Immunologic/metabolism , Receptors, Scavenger , Scavenger Receptors, Class B
18.
J Lipid Res ; 46(4): 687-96, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15654132

ABSTRACT

The physiological role of murine scavenger receptor class B type I (SR-BI) was evaluated by in vivo clearances of human HDL3 and LDL in normal and SR-BI knockout (KO) mice. In normal mice, cholesteryl esters (CEs) were removed faster than proteins, indicating a selective uptake process from both HDL3 and LDL. SR-BI KO mice showed 80% losses of HDL-CE selective uptake and the complete loss of LDL-CE selective uptake in the first phase of clearance. However, the second phase was characterized by an acceleration of CE disappearance in SR-BI KO mice. Thus, SR-BI is the only murine receptor mediating HDL-CE selective uptake, whereas a SR-BI-independent pathway specific to LDL can rescue SR-BI deficiency. The analysis of LDL recovered 3 h after injection in mice from different genotypes revealed that LDLs are significantly depleted in CE (reduction from 19% to 50% of the CE/protein ratios). A smaller LDL size in comparison with that of noninjected LDL was also detectable but was more evident for LDL recovered from normal mice. All LDL preparations migrate faster than noninjected LDL on agarose-barbital gels. Thus, both SR-BI-dependent and -independent pathways lead to substantial changes in LDL.


Subject(s)
Lipoproteins, HDL/metabolism , Lipoproteins, LDL/metabolism , Membrane Proteins/metabolism , Receptors, Lipoprotein/metabolism , Animals , Blotting, Western , Cholesterol Esters/metabolism , Female , Gene Deletion , Gene Expression Regulation , Genotype , Humans , Liver/metabolism , Male , Membrane Proteins/deficiency , Membrane Proteins/genetics , Mice , Mice, Knockout , Receptors, LDL/metabolism , Receptors, Lipoprotein/deficiency , Receptors, Lipoprotein/genetics , Scavenger Receptors, Class B , Sex Characteristics
19.
J Biol Chem ; 279(47): 48865-75, 2004 Nov 19.
Article in English | MEDLINE | ID: mdl-15358785

ABSTRACT

The discovery of autosomal dominant hypercholesterolemic patients with mutations in the PCSK9 gene, encoding the proprotein convertase NARC-1, resulting in the missense mutations suggested a role in low density lipoprotein (LDL) metabolism. We show that the endoplasmic reticulum-localized proNARC-1 to NARC-1 zymogen conversion is Ca2+-independent and that within the zymogen autocatalytic processing site SSVFAQ [downward arrow]SIP Val at P4 and Pro at P3' are critical. The S127R and D374Y mutations result in approximately 50-60% and > or =98% decrease in zymogen processing, respectively. In contrast, the double [D374Y + N157K], F216L, and R218S natural mutants resulted in normal zymogen processing. The cell surface LDL receptor (LDLR) levels are reduced by 35% in lymphoblasts of S127R patients. The LDLR levels are also reduced in stable HepG2 cells overexpressing NARC-1 or its natural mutant S127R, and this reduction is abrogated in the presence of 5 mm ammonium chloride, suggesting that overexpression of NARC-1 increases the turnover rate of the LDLR. Adenoviral expression of wild type human NARC-1 in mice resulted in a maximal approximately 9-fold increase in circulating LDL cholesterol, while in LDLR-/- mice a delayed approximately 2-fold increase in LDL cholesterol was observed. In conclusion, NARC-1 seems to affect both the level of LDLR and that of circulating apoB-containing lipoproteins in an LDLR-dependent and -independent fashion.


Subject(s)
Cholesterol, LDL/metabolism , Enzyme Precursors/metabolism , Mutation , Receptors, LDL/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/physiology , Adenoviridae/genetics , Ammonium Chloride/pharmacology , Animals , Apolipoproteins B/chemistry , Binding Sites , Blotting, Western , Calcium/chemistry , Catalysis , Cell Line , Cell Membrane/metabolism , Cell Separation , Cholesterol/metabolism , DNA, Complementary/metabolism , Endoplasmic Reticulum/metabolism , Female , Flow Cytometry , Gene Deletion , Gene Silencing , Heterozygote , Humans , Hypercholesterolemia/genetics , Lymphocytes/metabolism , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation, Missense , Proprotein Convertase 9 , Proprotein Convertases , Protein Structure, Tertiary , RNA, Messenger/metabolism , Time Factors , Transfection
20.
J Cell Sci ; 117(Pt 15): 3095-105, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15226391

ABSTRACT

The scavenger receptor class B, type I (SR-BI) mediates cholesteryl esters (CE) selective uptake from low density lipoprotein (LDL) and high-density lipoprotein (HDL) particles. In a number of tissues expressing caveolin, SR-BI is localized in caveolae. We show using detergent-free sucrose gradients that SR-BI is found in membrane rafts devoid of caveolin-1 in the human hepatoma HepG2 cell. Perturbation of the structure of HepG2 cell membrane rafts with cholesterol oxidase or sphingomyelinase decreased LDL-CE association due to selective uptake by 60%, while HDL3-CE selective uptake was increased 2.3-fold by cholesterol oxidase but was not affected by sphingomyelinase. Sequestration of membrane cholesterol with filipin III decreased LDL-CE selective uptake by 25%, while it had no effect on HDL3-CE selective uptake. Extraction of cell membrane cholesterol with beta-cyclodextrin increased LDL- and HDL3-CE selective uptake by 1.6-fold and 3-fold, respectively. We found that CE-selective uptake from both HDL and LDL occurs by a pathway involving retro-endocytosis in HepG2 cells. An analysis of the effect of SR-BI level on the expression of critical lipid sensor and lipid binding proteins was conducted with stable transformants of HepG2 cell overexpressing SR-BI. We found that liver-type fatty acid binding protein expression level is higher in SR-BI-overexpressing cells and that caveolin-1 and sterol response element binding protein-2 levels are reduced. Thus, in this hepatic cell model, SR-BI is associated with membrane rafts devoid of caveolin and its expression affects intracellular lipid binding and lipid sensor proteins. SR-BI-dependent LDL- and HDL-CE selective uptake are affected differently by the integrity of membrane rafts, but both occur by a retroendocytic pathway in HepG2 cells.


Subject(s)
Receptors, Immunologic/biosynthesis , CD36 Antigens , Carrier Proteins/metabolism , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Centrifugation, Density Gradient , Cholesterol/metabolism , Cholesterol Esters/metabolism , Detergents/pharmacology , Endocytosis , Fatty Acid-Binding Proteins , Humans , Hydrolysis , Immunoblotting , Lipid Metabolism , Lipoproteins/metabolism , Lipoproteins, LDL/metabolism , Liver/metabolism , Membrane Microdomains/metabolism , Receptors, Scavenger , Scavenger Receptors, Class B , Sphingomyelin Phosphodiesterase/metabolism , Sucrose/pharmacology , beta-Cyclodextrins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...