Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Mol Cell Biol ; 33(19): 3879-92, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23897431

ABSTRACT

Rad23a and Rad23b proteins are linked to nucleotide excision DNA repair (NER) via association with the DNA damage recognition protein xeroderma pigmentosum group C (XPC) are and known to be implicated in protein turnover by the 26S proteasome. Rad23b-null mice are NER proficient, likely due to the redundant function of the Rad23b paralogue, Rad23a. However, Rad23b-null midgestation embryos are anemic, and most embryos die before birth. Using an unbiased proteomics approach, we found that the majority of Rad23b-interacting partners are associated with the ubiquitin-proteasome system (UPS). We tested the requirement for Rad23b-dependent UPS activity in cellular proliferation and more specifically in the process of erythropoiesis. In cultured fibroblasts derived from embryos lacking Rad23b, proliferation rates were reduced. In fetal livers of Rad23b-null embryos, we observed reduced proliferation, accumulation of early erythroid progenitors, and a block during erythroid maturation. In primary wild-type (WT) erythroid cells, knockdown of Rad23b or chemical inhibition of the proteasome reduced survival and differentiation capability. Finally, the defects linked to Rad23b loss specifically affected fetal definitive erythropoiesis and stress erythropoiesis in adult mice. Together, these data indicate a previously unappreciated requirement for Rad23b and the UPS in regulation of proliferation in different cell types.


Subject(s)
Cell Proliferation , DNA-Binding Proteins/genetics , Erythropoiesis/genetics , Proteasome Endopeptidase Complex/genetics , Animals , Blotting, Western , Cell Differentiation/genetics , Cells, Cultured , DNA-Binding Proteins/metabolism , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Erythrocytes/cytology , Erythrocytes/metabolism , Erythroid Precursor Cells/cytology , Erythroid Precursor Cells/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Flow Cytometry , Liver/cytology , Liver/embryology , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteasome Endopeptidase Complex/metabolism , Protein Binding , RNA Interference
2.
Blood ; 121(13): 2553-62, 2013 Mar 28.
Article in English | MEDLINE | ID: mdl-23361909

ABSTRACT

B-cell lymphoma 11A (BCL11A) downregulation in human primary adult erythroid progenitors results in elevated expression of fetal γ-globin. Recent reports showed that BCL11A expression is activated by KLF1, leading to γ-globin repression. To study regulation of erythropoiesis and globin expression by KLF1 and BCL11A in an in vivo model, we used mice carrying a human ß-globin locus transgene with combinations of Klf1 knockout, Bcl11a floxed, and EpoR(Cre) knockin alleles. We found a higher percentage of reticulocytes in adult Klf1(wt/ko) mice and a mild compensated anemia in Bcl11a(cko/cko) mice. These phenotypes were more pronounced in compound Klf1(wt/ko)::Bcl11a(cko/cko) mice. Analysis of Klf1(wt/ko), Bcl11a(cko/cko), and Klf1(wt/ko)::Bcl11a(cko/cko) mutant embryos demonstrated increased expression of mouse embryonic globins during fetal development. Expression of human γ-globin remained high in Bcl11a(cko/cko) embryos during fetal development, and this was further augmented in Klf1(wt/ko)::Bcl11a(cko/cko) embryos. After birth, expression of human γ-globin and mouse embryonic globins decreased in Bcl11a(cko/cko) and Klf1(wt/ko)::Bcl11a(cko/cko) mice, but the levels remained much higher than those observed in control animals. Collectively, our data support an important role for the KLF1-BCL11A axis in erythroid maturation and developmental regulation of globin expression.


Subject(s)
Carrier Proteins/genetics , Erythropoiesis/genetics , Genes, Switch/genetics , Globins/genetics , Kruppel-Like Transcription Factors/genetics , Nuclear Proteins/genetics , Animals , DNA-Binding Proteins , Embryo, Mammalian , Erythropoiesis/physiology , Fetal Development/genetics , Gene Expression Regulation, Developmental , Gene Rearrangement/genetics , Gene Rearrangement/physiology , Genes, Switch/physiology , Humans , Mice , Mice, Mutant Strains , Mice, Transgenic , Repressor Proteins , Reticulocytosis/genetics , Reticulocytosis/physiology , Spleen/cytology , Spleen/embryology , Spleen/metabolism
3.
Atherosclerosis ; 227(1): 37-42, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23313246

ABSTRACT

OBJECTIVE: Plasma phospholipid transfer protein (PLTP) plays a key role in lipoprotein metabolism. Its exact function in the development of atherosclerosis is still under debate however. We studied the effect of elevated PLTP expression in one of the most commonly used models of atherosclerosis, the ApoE deficient mouse. METHODS: Experiment 1: Plasma PLTP activity, total cholesterol, HDL cholesterol and atherosclerosis development was measured in ApoE deficient mice with or without elevated expression of PLTP. Experiment 2: The same parameters were measured in ApoE deficient mice after bone marrow transplantation from wild type mice or mice with elevated PLTP expression. Experiment 3: Similar to experiment 2, but using donor mice with an ApoE deficient background. RESULTS: Experiment 1: ApoE deficient mice have more than two times more atherosclerosis when overexpressing PLTP and a strongly decreased plasma level of HDL. Experiment 2: Bone marrow transplantation with ApoE proficient cells results in a strong reduction of plasma cholesterol in ApoE deficient acceptor mice. Still, elevated PLTP in bone marrow derived cells evoke a reduction of HDL cholesterol and increased atherosclerosis. Experiment 3: Bone marrow transplantation with ApoE deficient cells results in much higher cholesterol levels, but here too HDL cholesterol levels are reduced and atherosclerosis increased. CONCLUSION: In all the models with ApoE deficiency, elevated PLTP expression causes higher levels of diet-induced atherosclerosis coinciding with decreased plasma levels of HDL cholesterol.


Subject(s)
Apolipoproteins E/deficiency , Atherosclerosis/etiology , Phospholipid Transfer Proteins/biosynthesis , Animals , Cholesterol/blood , Cholesterol, HDL/blood , Diet, High-Fat , Humans , Male , Mice , Mice, Inbred C57BL
4.
Epigenetics Chromatin ; 5(1): 9, 2012 Jun 22.
Article in English | MEDLINE | ID: mdl-22726460

ABSTRACT

BACKGROUND: Non-small cell lung carcinoma (NSCLC) is a complex malignancy that owing to its heterogeneity and poor prognosis poses many challenges to diagnosis, prognosis and patient treatment. DNA methylation is an important mechanism of epigenetic regulation involved in normal development and cancer. It is a very stable and specific modification and therefore in principle a very suitable marker for epigenetic phenotyping of tumors. Here we present a genome-wide DNA methylation analysis of NSCLC samples and paired lung tissues, where we combine MethylCap and next generation sequencing (MethylCap-seq) to provide comprehensive DNA methylation maps of the tumor and paired lung samples. The MethylCap-seq data were validated by bisulfite sequencing and methyl-specific polymerase chain reaction of selected regions. RESULTS: Analysis of the MethylCap-seq data revealed a strong positive correlation between replicate experiments and between paired tumor/lung samples. We identified 57 differentially methylated regions (DMRs) present in all NSCLC tumors analyzed by MethylCap-seq. While hypomethylated DMRs did not correlate to any particular functional category of genes, the hypermethylated DMRs were strongly associated with genes encoding transcriptional regulators. Furthermore, subtelomeric regions and satellite repeats were hypomethylated in the NSCLC samples. We also identified DMRs that were specific to two of the major subtypes of NSCLC, adenocarcinomas and squamous cell carcinomas. CONCLUSIONS: Collectively, we provide a resource containing genome-wide DNA methylation maps of NSCLC and their paired lung tissues, and comprehensive lists of known and novel DMRs and associated genes in NSCLC.

5.
Biochim Biophys Acta ; 1791(10): 1031-6, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19524061

ABSTRACT

OBJECTIVE: Elevated plasma phospholipid transfer protein (PLTP) expression may increase atherosclerosis in mice by reducing plasma HDL and increasing hepatic VLDL secretion. Hepatic lipase (HL) is a lipolytic enzyme involved in several aspects of the same pathways of lipoprotein metabolism. We investigated whether the effects of elevated PLTP activity are compromised by HL deficiency. METHODS AND RESULTS: HL deficient mice were crossbred with PLTP transgenic (PLTPtg) mice and studied in the fasted state. Plasma triglycerides were decreased in HL deficiency, explained by reduced hepatic triglyceride secretion. In PLTPtg mice, a redistribution of HL activity between plasma and tissue was evident and plasma triglycerides were also decreased. HL deficiency mitigated or even abolished the stimulatory effect of elevated PLTP activity on hepatic triglyceride secretion. HL deficiency had a modest incremental effect on plasma HDL, which remained present in PLTP transgenic/HL(-/-) mice, thereby partially compensating the decrease in HDL caused by elevation of PLTP activity. HDL decay experiments showed that the fractional turnover rate of HDL cholesteryl esters was delayed in HL deficient mice, increased in PLTPtg mice and intermediate in PLTPtg mice in an HL(-/-) background. CONCLUSIONS: HL affects hepatic VLDL. Elevated PLTP activity lowers plasma HDL-cholesterol by stimulating the plasma turnover and hepatic uptake of HDL cholesteryl esters. HL is not required for the increase in hepatic triglyceride secretion or for the lowering of HDL-cholesterol induced by PLTP overexpression.


Subject(s)
Cholesterol, HDL/metabolism , Cholesterol, VLDL/metabolism , Lipase/metabolism , Phospholipid Transfer Proteins/metabolism , Animals , Cholesterol, HDL/blood , Cholesterol, VLDL/blood , Lipase/blood , Lipoprotein Lipase/blood , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Phospholipid Transfer Proteins/blood
6.
Biochim Biophys Acta ; 1791(8): 790-6, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19422933

ABSTRACT

Phospholipid transfer protein (PLTP) is associated with HDL particles in plasma, where it transfers phospholipids between lipoproteins and remodels HDL particles. Tangier disease patients, with a mutated ABCA1 transporter, have extremely low plasma HDL concentration and reduced PLTP activity levels, a phenotype that is also observed in mice lacking ABCA1. We investigated whether low HDL levels and low PLTP activity are mechanistically related. Firstly, we studied PLTP expression and distribution among lipoproteins in mice lacking ABCA1 (ABCA1(-/-)). Parallel to the strong reduction in PLTP activity in plasma of ABCA1(-/-) mice, decreased PLTP protein levels were observed. Neither PLTP synthesis in liver or macrophages nor the ability of the macrophages to secrete PLTP were impaired in ABCA1(-/-) mice. However, the PLTP activity level in the medium of cultured macrophages was determined by HDL levels in the medium. PLTP was associated with HDL particles in wild type mice, whereas in ABCA1(-/-) mice, PLTP was associated with VLDL and LDL particles. Secondly, we treated different mouse models with varying plasma HDL and PLTP levels (wild type, ABCA1(-/-), apoE(-/-) and PLTPtg mice, overexpressing human PLTP) with a synthetic LXR ligand, and investigated the relationship between LXR-mediated PLTP induction and HDL levels in plasma. Plasma PLTP activity in wild type mice was induced 5.6-fold after LXR activation, whereas in ABCA1(-/-), apoE(-/-) and PLTPtg mice, all having reduced HDL levels, induction of PLTP activity was 2.4- , 3.2- and 2.0-fold, respectively. The less pronounced PLTP induction in these mice compared to wild type mice was not caused by a decreased PLTP gene expression in the liver or macrophages. Our findings indicate that the extent of LXR-mediated PLTP induction depends on plasma HDL levels. In conclusion, we demonstrate that ABCA1 deficiency in mice affects plasma PLTP level and distribution through an indirect effect on HDL metabolism. In addition, we show that the extent of LXR-mediated PLTP induction is HDL-dependent. These findings indicate that plasma HDL level is an important regulator of plasma PLTP and might play a role in the stabilization of PLTP in plasma.


Subject(s)
Lipoproteins, HDL/blood , Phospholipid Transfer Proteins/blood , ATP Binding Cassette Transporter 1 , ATP-Binding Cassette Transporters/metabolism , Animals , DNA-Binding Proteins/metabolism , Humans , Liver X Receptors , Macrophages/metabolism , Mice , Orphan Nuclear Receptors , Receptors, Cytoplasmic and Nuclear/metabolism
7.
Atherosclerosis ; 204(2): 429-34, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19100548

ABSTRACT

Phospholipid transfer protein (PLTP) is a multifunctional protein synthesized by various cell types and secreted into the plasma. Plasma PLTP is able to transfer phospholipids between lipoproteins and modulate HDL particles. Mice with overexpression of human PLTP have an increased ability to generate pre beta-HDL, reduced total HDL levels and an increased susceptibility to atherosclerosis. As the macrophage is a key component of the atherosclerotic lesion and an important site of PLTP expression, we investigated the role of systemic and peripheral PLTP in macrophage cholesterol efflux and reverse cholesterol transport (RCT) in vivo. We used an assay in which (3)H-labelled cholesterol-loaded macrophages were injected intraperitoneally into recipient mice, and radioactivity was quantified in plasma, liver and faeces. Firstly, wild type macrophages were injected into wild type, PLTP transgenic (PLTPtg) and apoAI transgenic (apoAItg) mice. While plasma (3)H-tracer levels in apoAItg mice were increased compared with wild type mice, they were reduced in PLTPtg mice. Moreover, overexpression of PLTP significantly decreased faecal (3)H-tracer levels compared with wild type and apoAItg mice. Secondly, wild type mice were injected with peritoneal macrophages derived from PLTPtg or wild type mice. No significant difference in the amount of (3)H-tracer in plasma, liver or faeces was found between the two groups of mice. Our findings demonstrate that macrophage cholesterol efflux and RCT to faeces is impaired in PLTP transgenic mice, and that elevation of macrophage-PLTP does not affect RCT, indicating that higher systemic PLTP levels may promote atherosclerosis development by decreasing the rate of macrophage RCT.


Subject(s)
Atherosclerosis/enzymology , Cholesterol/metabolism , Macrophages, Peritoneal/enzymology , Phospholipid Transfer Proteins/metabolism , Animals , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Atherosclerosis/blood , Biological Transport , Cells, Cultured , Feces/chemistry , High-Density Lipoproteins, Pre-beta/metabolism , Humans , Liver/metabolism , Macrophages, Peritoneal/transplantation , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phospholipid Transfer Proteins/blood , Phospholipid Transfer Proteins/genetics
8.
J Lipid Res ; 49(12): 2504-12, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18711210

ABSTRACT

Plasma phospholipid transfer protein (PLTP) interacts with HDL particles and facilitates the transfer of phospholipids from triglyceride (TG)-rich lipoproteins to HDL. Overexpressing human PLTP in mice increases the susceptibility to atherosclerosis. In human plasma, high-active and low-active forms of PLTP exist. To elucidate the contribution of phospholipid transfer activity to changes in lipoprotein metabolism and atherogenesis, we developed mice expressing mutant PLTP, still able to associate with HDL but lacking phospholipid transfer activity. In mice heterozygous for the LDL receptor, effects of the mutant and normal human PLTP transgene (mutPLTP tg and PLTP tg, respectively) were compared. In PLTP tg mice, plasma PLTP activity was increased 2.9-fold, resulting in markedly reduced HDL lipid levels. In contrast, in mutPLTP tg mice, lipid levels were not different from controls. Furthermore, hepatic VLDL-TG secretion was stimulated in PLTP tg mice, but not in mutPLTP tg mice. When mice were fed a cholesterol-enriched diet, atherosclerotic lesion size in PLTP tg mice was increased more than 2-fold compared with control mice, whereas in mutPLTP tg mice, there was no change. Our findings demonstrate that PLTP transfer activity is essential for the development of atherosclerosis in PLTP transgenic mice, identifying PLTP activity as a possible target to prevent atherogenesis, independent of plasma PLTP concentration.


Subject(s)
Atherosclerosis/genetics , Mutation , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism , Animals , Atherosclerosis/metabolism , Disease Models, Animal , Lipoproteins, VLDL/metabolism , Mice , Mice, Transgenic , RNA, Messenger/metabolism
9.
PLoS One ; 3(5): e2255, 2008 May 28.
Article in English | MEDLINE | ID: mdl-18509527

ABSTRACT

BACKGROUND: Phospholipid transfer protein (PLTP) is expressed by various cell types. In plasma, it is associated with high density lipoproteins (HDL). Elevated levels of PLTP in transgenic mice result in decreased HDL and increased atherosclerosis. PLTP is present in human atherosclerotic lesions, where it seems to be macrophage derived. The aim of the present study is to evaluate the atherogenic potential of macrophage derived PLTP. METHODS AND FINDINGS: Here we show that macrophages from human PLTP transgenic mice secrete active PLTP. Subsequently, we performed bone marrow transplantations using either wild type mice (PLTPwt/wt), hemizygous PLTP transgenic mice (huPLTPtg/wt) or homozygous PLTP transgenic mice (huPLTPtg/tg) as donors and low density lipoprotein receptor deficient mice (LDLR-/-) as acceptors, in order to establish the role of PLTP expressed by bone marrow derived cells in diet-induced atherogenesis. Atherosclerosis was increased in the huPLTPtg/wt-->LDLR-/- mice (2.3-fold) and even further in the huPLTPtg/tg-->LDLR-/- mice (4.5-fold) compared with the control PLTPwt/wt-->LDLR-/- mice (both P<0.001). Plasma PLTP activity levels and non-HDL cholesterol were increased and HDL cholesterol decreased compared with controls (all P<0.01). PLTP was present in atherosclerotic plaques in the mice as demonstrated by immunohistochemistry and appears to co-localize with macrophages. Isolated macrophages from PLTP transgenic mice do not show differences in cholesterol efflux or in cytokine production. Lipopolysaccharide activation of macrophages results in increased production of PLTP. This effect was strongly amplified in PLTP transgenic macrophages. CONCLUSIONS: We conclude that PLTP expression by bone marrow derived cells results in atherogenic effects on plasma lipids, increased PLTP activity, high local PLTP protein levels in the atherosclerotic lesions and increased atherosclerotic lesion size.


Subject(s)
Atherosclerosis/metabolism , Bone Marrow Cells/metabolism , Phospholipid Transfer Proteins/metabolism , Animals , Bone Marrow Transplantation , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phospholipid Transfer Proteins/biosynthesis , Phospholipid Transfer Proteins/blood , Receptors, LDL/genetics , Receptors, LDL/physiology
10.
Arterioscler Thromb Vasc Biol ; 28(7): 1277-82, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18421000

ABSTRACT

OBJECTIVE: A transgenic mouse model was generated that allows conditional expression of human PLTP, based on the tetracycline-responsive gene system, to study the effects of an acute increase in plasma PLTP activity as may occur in inflammation. METHODS AND RESULTS: The effects of an acute elevation of plasma PLTP activity on the metabolism of apolipoprotein B-containing lipoproteins and on diet-induced pre-existing atherosclerosis were determined in mice displaying a humanized lipoprotein profile (low-density lipoprotein receptor knockout background). Induced expression of PLTP strongly increases plasma VLDL levels in LDL receptor knockout mice, whereas VLDL secretion is not affected. The elevation in plasma triglyceride levels is explained by a PLTP-dependent inhibition of VLDL catabolism, which is caused, at least partly, by a decreased lipoprotein lipase activity. Together with the decreased plasma HDL levels, the acutely increased PLTP expression results in a highly atherogenic lipoprotein profile. Induction of PLTP expression leads to a further increase in size of pre-existing atherosclerotic lesions, even on a chow diet. In addition, the lesions contain more macrophages and less collagen relative to controls, suggesting a less stable lesion phenotype. CONCLUSIONS: In conclusion, acute elevation of PLTP activity destabilizes atherosclerotic lesions and aggravates pre-existing atherosclerosis.


Subject(s)
Atherosclerosis/metabolism , Phospholipid Transfer Proteins/metabolism , Receptors, LDL/metabolism , Animals , Apolipoproteins B/blood , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Cholesterol, Dietary/administration & dosage , Cholesterol, Dietary/blood , Collagen/metabolism , Disease Models, Animal , Disease Progression , Humans , Lipase/blood , Lipoprotein Lipase/blood , Lipoproteins, VLDL/blood , Macrophages/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Phospholipid Transfer Proteins/blood , Phospholipid Transfer Proteins/genetics , Receptors, LDL/deficiency , Receptors, LDL/genetics , Time Factors , Triglycerides/blood , Up-Regulation
11.
J Lipid Res ; 48(12): 2622-31, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17761633

ABSTRACT

In low density lipoprotein receptor (LDLR)-deficient mice, overexpression of human plasma phospholipid transfer protein (PLTP) results in increased atherosclerosis. PLTP strongly decreases HDL levels and might alter the antiatherogenic properties of HDL particles. To study the potential interaction between human PLTP and apolipoprotein A-I (apoA-I), double transgenic animals (hPLTPtg/hApoAItg) were compared with hApoAItg mice. PLTP activity was increased 4.5-fold. Plasma total cholesterol and phospholipid were decreased. Average HDL size (analyzed by gel filtration) increased strongly, hPLTPtg/hApoAItg mice having very large, LDL-sized, HDL particles. Also, after density gradient ultracentrifugation, a substantial part of the apoA-I-containing lipoproteins in hPLTPtg/hApoAItg mice was found in the LDL density range. In cholesterol efflux studies from macrophages, HDL isolated from hPLTPtg/hApoAItg mice was less efficient than HDL isolated from hApoAItg mice. Furthermore, it was found that the largest subfraction of the HDL particles present in hPLTPtg/hApoAItg mice was markedly inferior as a cholesterol acceptor, as no labeled cholesterol was transferred to this fraction. In an LDLR-deficient background, the human PLTP-expressing mouse line showed a 2.2-fold increased atherosclerotic lesion area. These data demonstrate that the action of human PLTP in the presence of human apoA-I results in the formation of a dysfunctional HDL subfraction, which is less efficient in the uptake of cholesterol from cholesterol-laden macrophages.


Subject(s)
Apolipoprotein A-I/genetics , Atherosclerosis/metabolism , Lipoproteins, HDL/metabolism , Phospholipid Transfer Proteins/genetics , Animals , Apolipoprotein A-I/metabolism , Diet, Atherogenic , Humans , Lipoproteins, HDL/chemistry , Mice , Mice, Transgenic , Phospholipid Transfer Proteins/metabolism
12.
Transgenic Res ; 16(4): 503-13, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17437182

ABSTRACT

One main determinant in high-density lipoprotein (HDL) metabolism is phospholipid transfer protein (PLTP), a plasma protein that is associated with HDL. In transgenic mice overexpressing human PLTP we found that elevated plasma PLTP levels dose-dependently increased the susceptibility to diet-induced atherosclerosis. This could be mainly due to the fact that most functions of PLTP are potentially atherogenic, such as decreasing plasma HDL levels. To further elucidate the role of PLTP in lipoprotein metabolism and atherosclerosis we generated a novel transgenic mouse model that allows conditional expression of human PLTP. In this mouse model a human PLTP encoding sequence is controlled by a Tet-On system. Upon induction of PLTP expression, our mouse model showed a strongly increased PLTP activity (from 3.0 +/- 0.6 to 11.4 +/- 2.8 AU, p < 0.001). The increase in PLTP activity resulted in an acute decrease in plasma cholesterol of 33% and a comparable decrease in phospholipids. The decrease in total plasma cholesterol and phospholipids was caused by a 35% decrease in HDL-cholesterol level and a 41% decrease in HDL-phospholipid level. These results demonstrate the feasibility of our mouse model to induce an acute elevation of PLTP activity, which is easily reversible. As a direct consequence of an increase in PLTP activity, HDL-cholesterol and HDL-phospholipid levels strongly decrease. Using this mouse model, it will be possible to study the effects of acute elevation of PLTP activity on lipoprotein metabolism and pre-existing atherosclerosis.


Subject(s)
Mice, Transgenic/physiology , Phospholipid Transfer Proteins/metabolism , Phospholipid Transfer Proteins/physiology , Animals , Female , Humans , Lipids/blood , Male , Mice , Mice, Inbred C57BL , Phospholipid Transfer Proteins/drug effects , Phospholipid Transfer Proteins/genetics , Protein Synthesis Inhibitors/pharmacology , RNA, Messenger/analysis , Tetracycline/pharmacology
13.
Biochim Biophys Acta ; 1761(9): 1070-7, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16935026

ABSTRACT

Plasma phospholipid transfer protein (PLTP) has atherogenic properties in genetically modified mice. PLTP stimulates hepatic triglyceride secretion and reduces plasma levels of high density lipoproteins (HDL). The present study was performed to relate the increased atherosclerosis in PLTP transgenic mice to one of these atherogenic effects. A humanized mouse model was used which had decreased LDL receptor expression and was transgenic for human cholesterylester transfer protein (CETP) in order to obtain a better resemblance to the plasma lipoprotein profile present in humans. It is well known that female mice are more susceptible to atherosclerosis than male mice. Therefore, we compared male and female mice expressing human PLTP. The animals were fed an atherogenic diet and the effects on plasma lipids and lipoproteins, triglyceride secretion and the development of atherosclerosis were measured. The development of atherosclerosis was sex-dependent. This effect was stronger in PLTP transgenic mice, while PLTP activity levels were virtually identical. Also, the rates of hepatic secretion of triglycerides were similar. In contrast, plasma levels of HDL were about 2-fold lower in female mice than in male mice after feeding an atherogenic diet. We conclude that increased atherosclerosis caused by overexpression of PLTP is related to a decrease in HDL, rather than to elevated hepatic secretion of triglycerides.


Subject(s)
Atherosclerosis/metabolism , Lipoproteins, HDL/blood , Phospholipid Transfer Proteins/metabolism , Triglycerides/biosynthesis , Animals , Aortic Valve/pathology , Atherosclerosis/pathology , Cholesterol Ester Transfer Proteins/biosynthesis , Cholesterol Ester Transfer Proteins/blood , Cholesterol Ester Transfer Proteins/genetics , Diet, Atherogenic , Female , Humans , Lipoproteins, VLDL/biosynthesis , Male , Mice , Mice, Transgenic , Phospholipid Transfer Proteins/blood , Phospholipid Transfer Proteins/genetics , Receptors, LDL/genetics , Receptors, LDL/metabolism , Sex Factors , Triglycerides/blood
14.
Biochim Biophys Acta ; 1738(1-3): 48-53, 2005 Dec 30.
Article in English | MEDLINE | ID: mdl-16298159

ABSTRACT

In humans, fibrates are used to treat dyslipidemia, because these drugs lower plasma triglycerides and raise HDL cholesterol. Treatment with fibrates lowers plasma phospholipid transfer protein (PLTP) activity in humans, but increases PLTP activity in mice, without a consistent effect on HDL-cholesterol concentration. Earlier, we found that PLTP overexpression in transgenic mice results in decreased plasma HDL levels and increased diet-induced atherosclerosis. So it seems that the interplay between fibrates, PLTP and HDL is different in mice and man, which may be important for atherosclerosis development. In the present study, we measured the effects of fibrates on PLTP expression in cultured human hepatocytes and effects of fibrate treatment on human PLTP expression, plasma PLTP activity and HDL levels in human PLTP transgenic mice. Fibrate treatment did not influence PLTP mRNA levels in human hepatocytes. Hepatic human PLTP mRNA levels and PLTP activity were both moderately elevated by fenofibrate treatment in human PLTP transgenic mice. In wild-type mice, however, feeding fenofibrate resulted in a strong induction of PLTP mRNA in the liver and a more than 4-fold increase of plasma PLTP activity. Plasma triglycerides were reduced in all mice by 48% or more by fenofibrate treatment. HDL-cholesterol concentrations were substantially increased by fenofibrate in PLTP overexpressing mice (+72%), but unaffected in wild-type mice. We conclude that fenofibrate treatment reverses the HDL-lowering effect of PLTP overexpression in human PLTP transgenic mice.


Subject(s)
Cholesterol, HDL/blood , Fenofibrate/pharmacology , Hypolipidemic Agents/pharmacology , Phospholipid Transfer Proteins/drug effects , Animals , Cells, Cultured , Cholesterol, HDL/drug effects , Gene Expression Regulation/drug effects , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphatidylcholine-Sterol O-Acyltransferase/drug effects , Phosphatidylcholine-Sterol O-Acyltransferase/genetics , Phosphatidylcholine-Sterol O-Acyltransferase/metabolism , Phospholipid Transfer Proteins/genetics , Phospholipid Transfer Proteins/metabolism , RNA, Messenger/drug effects , Triglycerides/blood
15.
J Lipid Res ; 45(5): 805-11, 2004 May.
Article in English | MEDLINE | ID: mdl-14993244

ABSTRACT

Plasma phospholipid transfer protein (PLTP) transfers phospholipids between lipoproteins and mediates HDL conversion. PLTP-overexpressing mice have increased atherosclerosis. However, mice do not express cholesteryl ester transfer protein (CETP), which is involved in the same metabolic pathways as PLTP. Therefore, we studied atherosclerosis in heterozygous LDL receptor-deficient (LDLR(+/-)) mice expressing both human CETP and human PLTP. We used two transgenic lines with moderately and highly elevated plasma PLTP activity. In LDLR(+/-)/huCETPtg mice, cholesterol is present in both LDL and HDL. Both are decreased in LDLR(+/-)/huCETPtg/huPLTPtg mice (>50%). An atherogenic diet resulted in high levels of VLDL+LDL cholesterol. PLTP expression caused a strong PLTP dose-dependent decrease in VLDL and LDL cholesterol (-26% and -69%) and a decrease in HDL cholesterol (-70%). Surprisingly, atherosclerosis was increased in the two transgenic lines with moderately and highly elevated plasma PLTP activity (1.9-fold and 4.4-fold, respectively), indicating that the adverse effect of the reduction in plasma HDL outweighs the beneficial effect of the reduction in apolipoprotein B (apoB)-containing lipoproteins. The activities of the antiatherogenic enzymes paraoxonase and platelet-activating factor acetyl hydrolase were both PLTP dose-dependently reduced ( approximately -33% and -65%, respectively). We conclude that expression of PLTP in this animal model results in increased atherosclerosis in spite of reduced apoB-containing lipoproteins, by reduction of HDL and of HDL-associated antioxidant enzyme activities.


Subject(s)
Apolipoproteins B/metabolism , Arteriosclerosis/blood , Disease Susceptibility , Membrane Proteins/blood , Phospholipid Transfer Proteins/blood , Up-Regulation , Animals , Carrier Proteins/metabolism , Cholesterol/blood , Cholesterol Ester Transfer Proteins , Diet , Glycoproteins/metabolism , Humans , Hypercholesterolemia/blood , Male , Mice , Mice, Transgenic , Models, Animal
16.
Am J Pathol ; 163(4): 1677-86, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14507674

ABSTRACT

The activity of endothelial nitric oxide synthase (eNOS) is subject to complex transcriptional and post-translational regulation including the association with several proteins and variations in subcellular distribution. In the present study we describe a transgenic mouse model expressing eNOS fused to green fluorescent protein (GFP), which allows the study of localization and regulation of eNOS expression. We tested the functionality of eNOS in the eNOS-GFP mice. Expression of eNOS was restricted to the endothelial lining of blood vessels in various tissues tested, without appreciable expression in non-endothelial cells. Activity of the enzyme was confirmed by assaying the conversion of L-arginine to L-citrulline. NO production in isolated vessels was increased in transgenic mice when compared to non-transgenic control animals (4.88 +/- 0.59 and 2.48 +/- 0.47 micro mol/L NO, respectively, P < 0.005). Both the mean aortic pressure and the pulmonary artery pressure were reduced in eNOS-GFP mice (both approximately 30%, P < 0.05). Plasma cholesterol levels were also slightly reduced ( approximately 20%, P < 0.05). In conclusion, eNOS-GFP mice express functional eNOS and provide a unique model to study regulation of eNOS activity or eNOS-mediated vascular events, including response to ischemia, response to differences in shear stress, angiogenesis and vasculogenesis, and to study the subcellular distribution in relation with functional responses to these events.


Subject(s)
Endothelium, Vascular/metabolism , Luminescent Proteins/genetics , Nitric Oxide Synthase/genetics , Recombinant Fusion Proteins/metabolism , Animals , Aorta/drug effects , Aorta/physiology , Blood Pressure/drug effects , Cholesterol/blood , Endothelium, Vascular/enzymology , Green Fluorescent Proteins , Hemodynamics/drug effects , Humans , Mice , Mice, Transgenic , Nitric Oxide/biosynthesis , Pulmonary Artery/drug effects , Pulmonary Artery/physiology , Recombinant Fusion Proteins/pharmacology , Tissue Distribution
17.
J Lipid Res ; 44(8): 1462-9, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12754275

ABSTRACT

Hypertriglyceridemia (HTG) is associated with insulin resistance, increased cholesteryl ester transfer (CET), and low HDL cholesterol. Phospholipid transfer protein (PLTP) may be involved in these relationships. Associations between CET, lipids, insulin resistance, CETP and PLTP activities, and PLTP mass were investigated in 18 HTG patients and 20 controls. Effects of 6 weeks of bezafibrate treatment were studied in HTG patients. HTG patients had higher serum triglycerides, insulin resistance, free fatty acid (FFA), and CET, lower levels of HDL cholesterol (-44%) and PLTP mass (-54%), and higher CETP (+20%) and PLTP activity (+48%) than controls. Bezafibrate reduced triglycerides, CET (-37%), insulin resistance (-53%), FFA (-48%), CETP activity (-12%), PLTP activity (-8%), and increased HDL cholesterol (+27%), whereas PLTP mass remained unchanged. Regression analysis showed a positive contribution of PLTP mass (P = 0.001) but not of PLTP activity to HDL cholesterol, whereas insulin resistance positively contributed to PLTP activity (P < 0.01). Bezafibrate-induced change in CET and HDL cholesterol correlated with changes in CETP activity and FFAs, but not with change in PLTP activity. Bezafibrate-induced change in PLTP activity correlated with change in FFAs (r = 0.455, P = 0.058). We propose that elevated PLTP activity in HTG is related to insulin resistance and not to increased PLTP mass. Bezafibrate-induced diminished insulin resistance is associated with a reduction of CET and PLTP activity.


Subject(s)
Bezafibrate/pharmacology , Bezafibrate/therapeutic use , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Hypertriglyceridemia/drug therapy , Hypertriglyceridemia/metabolism , Insulin Resistance/physiology , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Phospholipid Transfer Proteins , Adrenergic beta-Antagonists/pharmacology , Adrenergic beta-Antagonists/therapeutic use , Adult , Blood Glucose/analysis , Cholesterol Esters/metabolism , Cholesterol, HDL/metabolism , Cross-Over Studies , Data Interpretation, Statistical , Double-Blind Method , Female , Humans , Insulin/blood , Linear Models , Male , Middle Aged , Molecular Weight
18.
J Biol Chem ; 277(50): 48938-43, 2002 Dec 13.
Article in English | MEDLINE | ID: mdl-12372822

ABSTRACT

Plasma phospholipid transfer protein (PLTP) is thought to be involved in the remodeling of high density lipoproteins (HDL), which are atheroprotective. It is also involved in the metabolism of very low density lipoproteins (VLDL). Hence, PLTP is thought to be an important factor in lipoprotein metabolism and the development of atherosclerosis. We have overexpressed PLTP in mice heterozygous for the low density lipoprotein (LDL) receptor, a model for atherosclerosis. We show that increased PLTP activity results in a dose-dependent decrease in HDL, and a moderate stimulation of VLDL secretion (

Subject(s)
Arteriosclerosis/blood , Carrier Proteins/blood , Membrane Proteins/blood , Phospholipid Transfer Proteins , Animals , Cholesterol, HDL/blood , Cholesterol, VLDL/blood , Cholesterol, VLDL/metabolism , Diet , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Risk Factors
19.
J Lipid Res ; 43(11): 1875-80, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12401886

ABSTRACT

Two lipid transfer proteins are active in human plasma, cholesteryl ester transfer protein (CETP), and phospholipid transfer protein (PLTP). Mice by nature do not express CETP. Additional inactivation of the PLTP gene resulted in reduced secretion of VLDL and subsequently in decreased susceptibility to diet-induced atherosclerosis. The aim of this study is to assess possible effects of differences in PLTP expression on VLDL secretion in mice that are proficient in CETP and PLTP. We compared human CETP transgenic (huCETPtg) mice with mice expressing both human lipid transfer proteins (huCETPtg/huPLTPtg). Plasma cholesterol in huCETPtg mice was 1.5-fold higher compared with huCETPtg/huPLTPtg mice (P < 0.001). This difference was mostly due to a lower HDL level in the huCETPtg/huPLTPtg mice, which subsequently could lead to the somewhat decreased CETP activity and concentration that was found in huCETPtg/huPLTPtg mice (P < 0.05). PLTP activity was 2.8-fold increased in these animals (P < 0.001). The human PLTP concentration was 5 microg/ml. Moderate overexpression of PLTP resulted in a 1.5-fold higher VLDL secretion compared with huCETPtg mice (P < 0.05). The composition of nascent VLDL was similar in both strains. These results indicate that elevated PLTP activity in huCETPtg mice results in an increase in VLDL secretion. In addition, PLTP overexpression decreases plasma HDL cholesterol as well as CETP.


Subject(s)
Carrier Proteins/blood , Glycoproteins , Lipoproteins, VLDL/metabolism , Membrane Proteins/blood , Phospholipid Transfer Proteins , Up-Regulation , Animals , Carrier Proteins/genetics , Cholesterol Ester Transfer Proteins , Humans , Lipoproteins, VLDL/biosynthesis , Lipoproteins, VLDL/blood , Mice , Mice, Transgenic , Phospholipids/blood
SELECTION OF CITATIONS
SEARCH DETAIL
...