1
|
Ao L, Noordam R, Rensen PCN, van Heemst D, Willems van Dijk K. The role of genetically-influenced phospholipid transfer protein activity in lipoprotein metabolism and coronary artery disease. J Clin Lipidol 2024; 18:e579-e587. [PMID: 38906750 DOI: 10.1016/j.jacl.2024.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 02/20/2024] [Accepted: 03/26/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Phospholipid transfer protein (PLTP) transfers surface phospholipids between lipoproteins and as such plays a role in lipoprotein metabolism, but with unclear effects on coronary artery disease (CAD) risk. We aimed to investigate the associations of genetically-influenced PLTP activity with 1-H nuclear magnetic resonance (1H-NMR) metabolomic measures and with CAD. Furthermore, using factorial Mendelian randomization (MR), we examined the potential additional effect of genetically-influenced PLTP activity on CAD risk on top of genetically-influenced low-density lipoprotein-cholesterol (LDL-C) lowering. METHODS Using data from UK Biobank, genetic scores for PLTP activity and LDL-C were calculated and dichotomised based on the median, generating four groups with combinations of high/low PLTP activity and high/low LDL-C levels for the factorial MR. Linear and logistic regressions were performed on 168 metabolomic measures (N = 58,514) and CAD (N = 318,734, N-cases=37,552), respectively, with results expressed as β coefficients (in standard deviation units) or odds ratios (ORs) and 95% confidence interval (CI). RESULTS Irrespective of the genetically-influenced LDL-C, genetically-influenced low PLTP activity was associated with a higher high-density lipoprotein (HDL) particle concentration (β [95% CI]: 0.03 [0.01, 0.05]), smaller HDL size (-0.14 [-0.15, -0.12]) and higher triglyceride (TG) concentration (0.04 [0.02, 0.05]), but not with CAD (OR 0.99 [0.97, 1.02]). In factorial MR analyses, genetically-influenced low PLTP activity and genetically-influenced low LDL-C had independent associations with metabolomic measures, and genetically-influenced low PLTP activity did not show an additional effect on CAD risk. CONCLUSIONS Low PLTP activity associates with higher HDL particle concentration, smaller HDL particle size and higher TG concentration, but no association with CAD risk was observed.
Collapse
Affiliation(s)
- Linjun Ao
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands (MMed Ao and Dr Willems van Dijk).
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands (Drs Noordam and van Heemst)
| | - Patrick C N Rensen
- Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands (Drs Rensen and Willems van Dijk); Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands (Drs Rensen and Willems van Dijk)
| | - Diana van Heemst
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, the Netherlands (Drs Noordam and van Heemst)
| | - Ko Willems van Dijk
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands (MMed Ao and Dr Willems van Dijk); Department of Internal Medicine, Division of Endocrinology, Leiden University Medical Center, Leiden, the Netherlands (Drs Rensen and Willems van Dijk); Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands (Drs Rensen and Willems van Dijk)
| |
Collapse
|
2
|
Arndt L, Hernandez-Resendiz I, Moos D, Dokas J, Müller S, Jeromin F, Wagner R, Ceglarek U, Heid IM, Höring M, Liebisch G, Stadler SC, Burkhardt R. Trib1 Deficiency Promotes Hyperlipidemia, Inflammation, and Atherosclerosis in LDL Receptor Knockout Mice. Arterioscler Thromb Vasc Biol 2023; 43:979-994. [PMID: 37078290 DOI: 10.1161/atvbaha.122.318137] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/04/2023] [Indexed: 04/21/2023]
Abstract
BACKGROUND Genetic variants at the TRIB1 gene locus are strongly associated with plasma lipid traits and the risk of coronary artery disease in humans. Here, we analyzed the consequences of Trib1 deficiency on lipid metabolism and atherosclerotic lesion formation in atherosclerosis-susceptible Ldlr-/- mice. METHODS Trib1-/- mice were crossed onto the Ldlr-/- background to generate double-knockout mice (Trib1-/-Ldlr-/-) and fed a semisynthetic, modified AIN76 diet (0.02% cholesterol and 4.3% fat) until 20 weeks of age. RESULTS Trib1-/-Ldlr-/- mice had profoundly larger (5.8-fold) and more advanced atherosclerotic lesions at the aortic root as compared with Trib1+/+Ldlr-/- controls. Further, we observed significantly elevated plasma total cholesterol and triglyceride levels in Trib1-/-Ldlr-/- mice, resulting from higher VLDL (very-low-density lipoprotein) secretion. Lipidomics analysis revealed that loss of Trib1 altered hepatic lipid composition, including the accumulation of cholesterol and proinflammatory ceramide species, which was accompanied by signs of hepatic inflammation and injury. Concomitantly, we detected higher plasma levels of IL (interleukin)-6 and LCN2 (lipocalin 2), suggesting increased systemic inflammation in Trib1-/-Ldlr-/- mice. Hepatic transcriptome analysis demonstrated significant upregulation of key genes controlling lipid metabolism and inflammation in Trib1-/-Ldlr-/- mice. Further experiments suggested that these effects may be mediated through pathways involving a C/EPB (CCAAT/enhancer binding protein)-PPARγ (peroxisome proliferator-activated receptor γ) axis and JNK (c-Jun N-terminal kinase) signaling. CONCLUSIONS We provide experimental evidence that Trib1 deficiency promotes atherosclerotic lesion formation in a complex manner that includes the modulation of lipid metabolism and inflammation.
Collapse
Affiliation(s)
- Lilli Arndt
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Germany (L.A., D.M., J.D., S.M., F.J., R.W., U.C.)
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Germany (L.A., I.H.-R., M.H., G.L., S.C.S., R.B.)
| | - Ileana Hernandez-Resendiz
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Germany (L.A., I.H.-R., M.H., G.L., S.C.S., R.B.)
| | - Doreen Moos
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Germany (L.A., D.M., J.D., S.M., F.J., R.W., U.C.)
| | - Janine Dokas
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Germany (L.A., D.M., J.D., S.M., F.J., R.W., U.C.)
| | - Silvana Müller
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Germany (L.A., D.M., J.D., S.M., F.J., R.W., U.C.)
| | - Franziska Jeromin
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Germany (L.A., D.M., J.D., S.M., F.J., R.W., U.C.)
| | - Richard Wagner
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Germany (L.A., D.M., J.D., S.M., F.J., R.W., U.C.)
| | - Uta Ceglarek
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Germany (L.A., D.M., J.D., S.M., F.J., R.W., U.C.)
| | - Iris M Heid
- Department of Genetic Epidemiology, University of Regensburg, Germany (I.M.H.)
| | - Marcus Höring
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Germany (L.A., I.H.-R., M.H., G.L., S.C.S., R.B.)
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Germany (L.A., I.H.-R., M.H., G.L., S.C.S., R.B.)
| | - Sonja C Stadler
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Germany (L.A., I.H.-R., M.H., G.L., S.C.S., R.B.)
| | - Ralph Burkhardt
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Germany (L.A., I.H.-R., M.H., G.L., S.C.S., R.B.)
| |
Collapse
|
3
|
Gautier T, Deckert V, Nguyen M, Desrumaux C, Masson D, Lagrost L. New therapeutic horizons for plasma phospholipid transfer protein (PLTP): Targeting endotoxemia, infection and sepsis. Pharmacol Ther 2021; 236:108105. [PMID: 34974028 DOI: 10.1016/j.pharmthera.2021.108105] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/10/2021] [Accepted: 12/27/2021] [Indexed: 12/13/2022]
Abstract
Phospholipid Transfer Protein (PLTP) transfers amphiphilic lipids between circulating lipoproteins and between lipoproteins, cells and tissues. Indeed, PLTP is a major determinant of the plasma levels, turnover and functionality of the main lipoprotein classes: very low-density lipoproteins (VLDL), low-density lipoproteins (LDL) and high-density lipoproteins (HDL). To date, most attention has been focused on the role of PLTP in the context of cardiometabolic diseases, with additional insights in neurodegenerative diseases and immunity. Importantly, beyond its influence on plasma triglyceride and cholesterol transport, PLTP plays a key role in the modulation of the immune response, with immediate relevance to a wide range of inflammatory diseases including bacterial infection and sepsis. Indeed, emerging evidence supports the role of PLTP, in the context of its association with lipoproteins, in the neutralization and clearance of bacterial lipopolysaccharides (LPS) or endotoxins. LPS are amphipathic molecules originating from Gram-negative bacteria which harbor major pathogen-associated patterns, triggering an innate immune response in the host. Although the early inflammatory reaction constitutes a key step in the anti-microbial defense of the organism, it can lead to a dysregulated inflammatory response and to hemodynamic disorders, organ failure and eventually death. Moreover, and in addition to endotoxemia and acute inflammation, small amounts of LPS in the circulation can induce chronic, low-grade inflammation with long-term consequences in several metabolic disorders such as atherosclerosis, obesity and diabetes. After an updated overview of the role of PLTP in lipid transfer, lipoprotein metabolism and related diseases, current knowledge of its impact on inflammation, infection and sepsis is critically appraised. Finally, the relevance of PLTP as a new player and novel therapeutic target in the fight against inflammatory diseases is considered.
Collapse
Affiliation(s)
- Thomas Gautier
- INSERM, LNC UMR1231, Dijon, France; University of Bourgogne and Franche-Comté, LNC UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France.
| | - Valérie Deckert
- INSERM, LNC UMR1231, Dijon, France; University of Bourgogne and Franche-Comté, LNC UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France
| | - Maxime Nguyen
- INSERM, LNC UMR1231, Dijon, France; University of Bourgogne and Franche-Comté, LNC UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France; Service Anesthésie-Réanimation Chirurgicale, Dijon University Hospital, Dijon, France
| | - Catherine Desrumaux
- INSERM, U1198, Montpellier, France; Faculty of Sciences, Université Montpellier, Montpellier, France
| | - David Masson
- INSERM, LNC UMR1231, Dijon, France; University of Bourgogne and Franche-Comté, LNC UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France; Plateau Automatisé de Biochimie, Dijon University Hospital, Dijon, France
| | - Laurent Lagrost
- INSERM, LNC UMR1231, Dijon, France; University of Bourgogne and Franche-Comté, LNC UMR1231, Dijon, France; FCS Bourgogne-Franche Comté, LipSTIC LabEx, Dijon, France; Service de la Recherche, Dijon University Hospital, Dijon, France.
| |
Collapse
|
4
|
Mulholland M, Jakobsson G, Lei Y, Sundius L, Ljungcrantz I, Rattik S, Tietge UJF, Engelbertsen D. IL-2Rβγ signalling in lymphocytes promotes systemic inflammation and reduces plasma cholesterol in atherosclerotic mice. Atherosclerosis 2021; 326:1-10. [PMID: 33945906 DOI: 10.1016/j.atherosclerosis.2021.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 04/08/2021] [Accepted: 04/21/2021] [Indexed: 11/20/2022]
Abstract
BACKGROUND AND AIMS The relationship between inflammation and lipid metabolism is complex and bidirectional. Lymphocyte-driven inflammation has been shown to modulate both atherosclerotic plaque development and cholesterol levels, but the mechanisms are incompletely understood. METHODS The cardiometabolic effects of IL-2Rβγ signalling in atherosclerotic Apoe-/- mice were investigated by treatment with an agonistic IL-2Rβγ-targeting IL-2/anti-IL-2 complex or a monoclonal anti-CD122 (IL-2Rβ) blocking antibody. RESULTS Administration of IL-2Rβγ agonistic IL-2/anti-IL-2 complexes to Apoe-/- mice augmented opposing arms of the adaptive immune system. Expansion of effector/memory T cells and increased levels of circulating pro-inflammatory cytokines were observed along with elevated levels of regulatory T cells and IL-10. Notably, IL-2/anti-IL-2 treatment did not affect plaque size but decreased levels of plasma cholesterol. The cholesterol lowering effect of IL-2Rβγ agonism was not affected by anti-CD8 or anti-NK1.1 depleting antibody treatment but was contingent on the presence of adaptive immunity. Expression of multiple liver X receptor (LXR)-related genes, including Pltp and Srebp1c in the liver, was decreased by IL-2/anti-IL-2 treatment. Although IL-2Rβγ agonism lowered cholesterol levels, blocking IL-2Rβγ signalling using an anti-CD122 monoclonal antibody did not impact cholesterol levels or plaque burden in Apoe-/- mice. CONCLUSIONS Elevated IL-2Rβγ signalling results in activation of both inflammatory and regulatory lymphocytes with a net zero effect on atherosclerosis and decreased plasma cholesterol levels. Changes in cholesterol levels were associated with reductions in hepatic LXR-related gene expression. Further studies are needed to investigate the clinical significance of IL-2 mediated modulation of hepatic LXR signalling in inflammatory disorders.
Collapse
Affiliation(s)
- Megan Mulholland
- Department of Clinical Sciences, Lund University, Skåne University Hospital, 20502, Malmö, Sweden.
| | - Gabriel Jakobsson
- Department of Clinical Sciences, Lund University, Skåne University Hospital, 20502, Malmö, Sweden
| | - Yu Lei
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, S-14183, Stockholm, Sweden
| | - Lena Sundius
- Department of Clinical Sciences, Lund University, Skåne University Hospital, 20502, Malmö, Sweden
| | - Irena Ljungcrantz
- Department of Clinical Sciences, Lund University, Skåne University Hospital, 20502, Malmö, Sweden
| | - Sara Rattik
- Department of Clinical Sciences, Lund University, Skåne University Hospital, 20502, Malmö, Sweden
| | - Uwe J F Tietge
- Division of Clinical Chemistry, Department of Laboratory Medicine, Karolinska Institutet, S-14183, Stockholm, Sweden; Clinical Chemistry, Karolinska University Laboratory, Karolinska University Hospital, SE-141 86, Stockholm, Sweden
| | - Daniel Engelbertsen
- Department of Clinical Sciences, Lund University, Skåne University Hospital, 20502, Malmö, Sweden.
| |
Collapse
|
5
|
Russo-Savage L, Schulman IG. Liver X receptors and liver physiology. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166121. [PMID: 33713792 DOI: 10.1016/j.bbadis.2021.166121] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 12/29/2022]
Abstract
The liver x receptors LXRα (NR1H3) and LXRβ (NR1H2) are members of the nuclear hormone receptor superfamily of ligand dependent transcription factors that regulate transcription in response to the direct binding of cholesterol derivatives. Studies using genetic knockouts and synthetic ligands have defined the LXRs as important modulators of lipid homeostasis throughout the body. This review focuses on the control of cholesterol and fatty acid metabolism by LXRs in the liver and how modifying LXR activity can influence the pathology of liver diseases.
Collapse
Affiliation(s)
- Lillian Russo-Savage
- Department of Pharmacology, University of Virginia, School of Medicine, United States of America
| | - Ira G Schulman
- Department of Pharmacology, University of Virginia, School of Medicine, United States of America.
| |
Collapse
|
6
|
Jiang XC, Yu Y. The Role of Phospholipid Transfer Protein in the Development of Atherosclerosis. Curr Atheroscler Rep 2021; 23:9. [PMID: 33496859 DOI: 10.1007/s11883-021-00907-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2021] [Indexed: 12/11/2022]
Abstract
PURPOSE OF REVIEW Phospholipid transfer protein (PLTP), a member of lipid transfer protein family, is an important protein involved in lipid metabolism in the circulation. This article reviews recent PLTP research progresses, involving lipoprotein metabolism and atherogenesis. RECENT FINDINGS PLTP activity influences atherogenic and anti-atherogenic lipoprotein levels. Human serum PLTP activity is a risk factor for human cardiovascular disease and is an independent predictor of all-cause mortality. PLTP deficiency reduces VLDL and LDL levels and attenuates atherosclerosis in mouse models, while PLTP overexpression exerts an opposite effect. Both PLTP deficiency and overexpression result in reduction of HDL which has different size, inflammatory index, and lipid composition. Moreover, although both PLTP deficiency and overexpression reduce cholesterol efflux capacity, but this effect has no impact in macrophage reverse cholesterol transport in mice. Furthermore, PLTP activity is related with metabolic syndrome, thrombosis, and inflammation. PLTP could be target for the treatment of dyslipidemia and atherosclerosis, although some potential off-target effects should be noted.
Collapse
Affiliation(s)
- Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Health Sciences University, 450 Clarkson Ave, Brooklyn, NY, USA.
| | - Yang Yu
- Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271000, People's Republic of China
| |
Collapse
|
7
|
Impact of Phospholipid Transfer Protein in Lipid Metabolism and Cardiovascular Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1276:1-13. [PMID: 32705590 DOI: 10.1007/978-981-15-6082-8_1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PLTP plays an important role in lipoprotein metabolism and cardiovascular disease development in humans; however, the mechanisms are still not completely understood. In mouse models, PLTP deficiency reduces cardiovascular disease, while its overexpression induces it. Therefore, we used mouse models to investigate the involved mechanisms. In this chapter, the recent main progresses in the field of PLTP research are summarized, and our focus is on the relationship between PLTP and lipoprotein metabolism, as well as PLTP and cardiovascular diseases.
Collapse
|
8
|
Song G, Zong C, Shao M, Yu Y, Liu Q, Wang H, Qiu T, Jiao P, Guo Z, Lee P, Luo Y, Jiang XC, Qin S. Phospholipid transfer protein (PLTP) deficiency attenuates high fat diet induced obesity and insulin resistance. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1305-1313. [PMID: 31220615 DOI: 10.1016/j.bbalip.2019.06.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 06/07/2019] [Accepted: 06/14/2019] [Indexed: 01/09/2023]
Abstract
Increased phospholipid transfer protein (PLTP) activity has been found to be associated with obesity, and metabolic syndrome in humans. However, whether or not PLTP has a direct effect on insulin sensitivity and obesity is largely unknown. Here we analyzed the effect by using PLTP knockout (PLTP-/-) mouse model. Although, PLTP-/- mice have normal body-weight-gain under chow diet, these mice were protected from high-fat-diet-induced obesity and insulin resistance, compared with wild type mice. In order to understand the mechanism, we evaluated insulin receptor and Akt activation and found that PLTP deficiency significantly enhanced phosphorylated insulin receptor and Akt levels in high-fat-diet fed mouse livers, adipose tissues, and muscles after insulin stimulation, while total Akt and insulin receptor levels were unchanged. Moreover, we found that the PLTP deficiency induced significantly more GLUT4 protein in the plasma membranes of adipocytes and muscle cells after insulin stimulation. Finally, we found that PLTP-deficient hepatocytes had less sphingomyelins and free cholesterols in the lipid rafts and plasma membranes than that of controls and this may provide a molecular basis for PLTP deficiency-mediated increase in insulin sensitivity. We have concluded that PLTP deficiency leads to an improvement in tissue and whole-body insulin sensitivity through modulating lipid levels in the plasma membrane, especially in the lipid rafts.
Collapse
Affiliation(s)
- Guohua Song
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China.
| | - Chuanlong Zong
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
| | - Mingzhu Shao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
| | - Yang Yu
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
| | - Qian Liu
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
| | - Hui Wang
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
| | - Tingting Qiu
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
| | - Peng Jiao
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
| | - Zheng Guo
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China
| | - Phoebe Lee
- Downstate Medical Center State University of New York, NY, USA
| | - Yi Luo
- Downstate Medical Center State University of New York, NY, USA
| | | | - Shucun Qin
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of Atherosclerosis, Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong, China.
| |
Collapse
|
9
|
Nass KJ, van den Berg EH, Gruppen EG, Dullaart RPF. Plasma lecithin:cholesterol acyltransferase and phospholipid transfer protein activity independently associate with nonalcoholic fatty liver disease. Eur J Clin Invest 2018; 48:e12988. [PMID: 29947103 DOI: 10.1111/eci.12988] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 06/25/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a highly prevalent condition which contributes to atherogenic apolipoprotein B dyslipoproteinemias. Lecithin:cholesterol acyltransferase (LCAT) and phospholipid transfer protein (PLTP) are both synthesized by the liver and are important in lipid metabolism. Here, we interrogated the impact of NAFLD on plasma LCAT and PLTP activities. METHODS Plasma LCAT activity (exogenous substrate assay) and PLTP activity (phospholipid vesicles-HDL assay) were determined in 348 subjects (279 men; 81 subjects with type 2 diabetes (T2DM); 123 with metabolic syndrome (MetS)). A Fatty Liver Index (FLI) ≥60 was used as a proxy of NAFLD. Insulin resistance was determined by homoeostasis model assessment (HOMA-IR). RESULTS A total of 147 participants had an FLI ≥60 coinciding with T2DM and MetS (P < 0.001 for each). Plasma LCAT activity and PLTP activity were on average 12% and 5% higher, respectively, in subjects with an FLI ≥ 60 (P < 0.001 for each). In age- and sex-adjusted partial linear regression analysis, LCAT activity and PLTP activity were positively related to various obesity measures and HOMA-IR (P < 0.001 for each). In multivariable linear regression analyses adjusted for age and sex, LCAT activity was associated with an FLI ≥ 60 independent of T2DM and MetS, the waist/hip ratio, or HOMA-IR (β = 0.307 to 0.366, P < 0001 for all models). PLTP activity was also associated with an FLI ≥ 60 independent of these variables (β = 0.151 to 0223, P = 0.013 to 0.001). CONCLUSION NAFLD, as inferred from an FLI≥60, confers higher plasma LCAT and to a lesser extent PLTP activity, even when taking account of T2DM, MetS, central obesity and insulin resistance.
Collapse
Affiliation(s)
- Karlijn J Nass
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Eline H van den Berg
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Eke G Gruppen
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Robin P F Dullaart
- Department of Endocrinology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
10
|
Yu Y, Lei X, Jiang H, Li Z, Creemers JWM, Zhang M, Qin S, Jin W, Jiang X. Prodomain of Furin Promotes Phospholipid Transfer Protein Proteasomal Degradation in Hepatocytes. J Am Heart Assoc 2018; 7:e008526. [PMID: 29680823 PMCID: PMC6015287 DOI: 10.1161/jaha.118.008526] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 03/22/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND Phospholipid transfer protein (PLTP) is one of the major modulators of lipoprotein metabolism and atherosclerosis development; however, little is known about the regulation of PLTP. The effect of hepatic prodomain of furin (profurin) expression on PLTP processing and function is investigated. METHODS AND RESULTS We used adenovirus expressing profurin in mouse liver to evaluate PLTP activity, mass, and plasma lipid levels. We coexpressed PLTP and profurin in human hepatoma cell line cells and studied their interaction. We found profurin expression significantly reduced plasma lipids, plasma PLTP activity, and mass in all tested mouse models, compared with controls. Moreover, the expression of profurin dramatically reduced liver PLTP activity and protein level. We further explored the mechanism using in vivo and ex vivo approaches. We found that profurin can interact with intracellular PLTP and promote its ubiquitination and proteasomal degradation, resulting in less PLTP secretion from the hepatocytes. Furin does not cleave PLTP; instead, it forms a complex with PLTP, likely through its prodomain. CONCLUSIONS Our study reveals that hepatic PLTP protein is targeted for proteasomal degradation by profurin expression, which could be a novel posttranslational mechanism underlying PLTP regulation.
Collapse
Affiliation(s)
- Yang Yu
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of AtherosclerosisTaishan Medical UniversityTaianChina
| | - Xia Lei
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
| | - Hui Jiang
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
| | - Zhiqiang Li
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
| | - John W. M. Creemers
- Laboratory of Biochemical NeuroendocrinologyDepartment of Human GeneticsHerestraat 49 bus 6023000 LeuvenBelgium
| | - Ming Zhang
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
| | - Shucun Qin
- Key Laboratory of Atherosclerosis in Universities of Shandong and Institute of AtherosclerosisTaishan Medical UniversityTaianChina
| | - Weijun Jin
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
| | - Xian‐Cheng Jiang
- Department of Cell BiologyState University of New York Downstate Medical CenterBrooklynNY
- Molecular and Cellular Cardiology ProgramVeterans Affair New York Harbor Healthcare SystemBrooklynNY
| |
Collapse
|
11
|
Gruppen EG, Kersten S, Dullaart RPF. Plasma angiopoietin-like 4 is related to phospholipid transfer protein activity in diabetic and non-diabetic subjects: role of enhanced low grade inflammation. Lipids Health Dis 2018; 17:60. [PMID: 29587751 PMCID: PMC5870514 DOI: 10.1186/s12944-018-0717-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/21/2018] [Indexed: 01/22/2023] Open
Abstract
Background Angiopoietin-like 4 (ANGPTL4) inhibits lipoprotein lipase, whereas phospholipid transfer protein (PLTP) enhances hepatic triglyceride secretion. Both factors may be upregulated by inflammatory pathways. Since the extent to which these circulating factors are interrelated is unknown, we determined the relationship between plasma ANGPTL4 and PLTP activity, and assessed whether such a relationship could be explained by high sensitivity C-reactive protein (hsCRP) levels as a marker of low-grade chronic inflammation. Methods Fasting plasma ANGPTL4, PLTP activity (liposome-vesicle high density lipoprotein system) and hsCRP were measured in 41 type 2 diabetic (T2DM) subjects and 36 non-diabetic subjects. Results Plasma ANGPTL4 and PLTP activity were increased in T2DM (p < 0.001 for each), coinciding with elevated hsCRP, triglycerides and non-esterified fatty acids (NEFA) (p = 0.031 to 0.001). In univariate analysis, ANGTLP4 was correlated with PLTP activity (Rs = 0.309, p = 0.006), whereas both factors were related to hsCRP and NEFA levels (Rs = 0.304 to 0.411, p < 0.01 to < 0.001). In multivariable linear regression analysis adjusting for age, sex, glucose, total cholesterol, triglycerides and NEFA, ANGPTL4 and PLTP activity each remained positively associated with hsCRP (β = 0.315, p = 0.003 and β = 0.299, p = 0.034, respectively). Plasma ANGPTL4 remained positively associated with PLTP activity when taking account of age, sex, glucose, total cholesterol, triglycerides and NEFA (β = 0.315, p = 0.003). Notably, this association disappeared after further adjustment for hsCRP (β = 0.131, p = 0.25). Conclusions In conclusion, plasma ANGPTL4 and PLTP activity are interrelated, which may at least in part be explained by low-grade chronic inflammation. A pro-inflammatory state could affect triglyceride metabolism via concerted effects on ANGPTL4 and PLTP.
Collapse
Affiliation(s)
- Eke G Gruppen
- Department of Endocrinology, University of Groningen and University Medical Center, P.O. Box 301, 9700 RB, Groningen, The Netherlands
| | - Sander Kersten
- Division of Human Nutrition, Wageningen University, Wageningen, The Netherlands
| | - Robin P F Dullaart
- Department of Endocrinology, University of Groningen and University Medical Center, P.O. Box 301, 9700 RB, Groningen, The Netherlands.
| |
Collapse
|
12
|
Jiang XC. Phospholipid transfer protein: its impact on lipoprotein homeostasis and atherosclerosis. J Lipid Res 2018; 59:764-771. [PMID: 29438986 DOI: 10.1194/jlr.r082503] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/10/2018] [Indexed: 12/25/2022] Open
Abstract
Phospholipid transfer protein (PLTP) is one of the major modulators of lipoprotein metabolism and atherosclerosis development in humans; however, we still do not quite understand the mechanisms. In mouse models, PLTP overexpression induces atherosclerosis, while its deficiency reduces it. Thus, mouse models were used to explore the mechanisms. In this review, I summarize the major progress made in the PLTP research field and emphasize its impact on lipoprotein metabolism and atherosclerosis, as well as its regulation.
Collapse
Affiliation(s)
- Xian-Cheng Jiang
- Department of Cell Biology, Downstate Medical Center, State University of New York, Brooklyn, NY
| |
Collapse
|
13
|
Dullaart RPF, Garcia E, Jeyarajah E, Gruppen EG, Connelly MA. Plasma phospholipid transfer protein activity is inversely associated with betaine in diabetic and non-diabetic subjects. Lipids Health Dis 2016; 15:143. [PMID: 27581838 PMCID: PMC5007837 DOI: 10.1186/s12944-016-0313-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 08/23/2016] [Indexed: 12/24/2022] Open
Abstract
Background The choline metabolite, betaine, plays a role in lipid metabolism, and may predict the development of cardiovascular disease and type 2 diabetes mellitus (T2DM). Phospholipid transfer protein (PLTP) and lecithin:cholesterol acyltransferase (LCAT) require phosphatidylcholine as substrate, raising the possibility that there is an intricate relationship of these protein factors with choline metabolism. Here we determined the relationships of PLTP and LCAT activity with betaine in subjects with and without T2DM. Methods Plasma betaine (nuclear magnetic resonance spectroscopy), PLTP activity (liposome-vesicle HDL system), LCAT activity (exogenous substrate assay) and (apo)lipoproteins were measured in 65 type 2 diabetic (T2DM) and in 55 non-diabetic subjects. Results PLTP and LCAT activity were elevated in T2DM (p < 0.05), whereas the difference in betaine was not significant. In age-, sex- and diabetes status-controlled correlation analysis, betaine was inversely correlated with triglycerides and positively with HDL cholesterol (p < 0.05 to 0.01). PLTP and LCAT activity were positively correlated with triglycerides and inversely with HDL cholesterol (p < 0.05 to 0.001). PLTP (r = −0.245, p = 0.006) and LCAT activity (r = −0.195, p = 0.035) were correlated inversely with betaine. The inverse association of PLTP activity with betaine remained significant after additional adjustment for body mass index and lipoprotein variables (β = −0.179, p = 0.034), whereas its association with LCAT activity lost significance (β = −0.056, p = 0.44). Conclusions Betaine may influence lipoprotein metabolism via an effect on PLTP activity.
Collapse
Affiliation(s)
- R P F Dullaart
- Department of Endocrinology, University Medical Center Groningen, University of Groningen, P.O. Box 30.001, Groningen, 9700 RB, The Netherlands.
| | - Erwin Garcia
- LipoScience, Laboratory Corporation of America® Holdings, Raleigh, NC, USA
| | - Elias Jeyarajah
- LipoScience, Laboratory Corporation of America® Holdings, Raleigh, NC, USA
| | - Eke G Gruppen
- Departments of Endocrinology and Nephrology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - Margery A Connelly
- LipoScience, Laboratory Corporation of America® Holdings, Raleigh, NC, USA
| |
Collapse
|
14
|
Jiang H, Yazdanyar A, Lou B, Chen Y, Zhao X, Li R, Hoang Bui H, Kuo MS, Navab M, Qin S, Li Z, Jin W, Jiang XC. Adipocyte phospholipid transfer protein and lipoprotein metabolism. Arterioscler Thromb Vasc Biol 2014; 35:316-22. [PMID: 25477345 DOI: 10.1161/atvbaha.114.303764] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Phospholipid transfer protein (PLTP) is highly expressed in adipose tissues. Thus, the effect of adipose tissue PLTP on plasma lipoprotein metabolism was examined. APPROACH AND RESULTS We crossed PLTP-Flox-ΔNeo and adipocyte protein 2 (aP2)-Cre recombinase (Cre) transgenic mice to create PLTP-Flox-ΔNeo/aP2-Cre mice that have a 90 and a 60% reduction in PLTP mRNA in adipose tissue and macrophages, respectively. PLTP ablation resulted in a significant reduction in plasma PLTP activity (22%), high-density lipoprotein-cholesterol (21%), high-density lipoprotein-phospholipid (20%), and apolipoprotein A-I (33%) levels, but had no effect on nonhigh-density lipoprotein levels in comparison with those of PLTP-Flox-ΔNeo controls. To eliminate possible effects of PLTP ablation by macrophages, we lethally irradiated PLTP-Flox-ΔNeo/aP2-Cre mice and PLTP-Flox-ΔNeo mice, and then transplanted wild-type mouse bone marrow into them to create wild-type→PLTP-Flox-ΔNeo/aP2-Cre and wild-type→PLTP-Flox-ΔNeo mice. Thus, we constructed a mouse model (wild-type→PLTP-Flox-ΔNeo/aP2-Cre) with PLTP deficiency in adipocytes but not in macrophages. These knockout mice also showed significant decreases in plasma PLTP activity (19%) and cholesterol (18%), phospholipid (17%), and apolipoprotein A-I (26%) levels. To further investigate the mechanisms behind the reduction in plasma apolipoprotein A-I and high-density lipoprotein lipids, we measured apolipoprotein A-I-mediated cholesterol efflux in adipose tissue explants and found that endogenous and exogenous PLTP significantly increased cholesterol efflux from the explants. CONCLUSIONS Adipocyte PLTP plays a small but significant role in plasma PLTP activity and promotes cholesterol efflux from adipose tissues.
Collapse
Affiliation(s)
- Hui Jiang
- From the Department of Cell Biology, State University of New York, Downstate Medical Center, Brooklyn (H.J., A.Y., Y.C., X.Z., R.L., Z.L., W.J., X.C.J.); Fudan University, Shanghai, China (B.L., Y.C.); Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, New York (Z.L., X.C.J); Institute of Atherosclerosis, Taishan Medical University, Taian, China (X.Z., S.Q.); Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (H.H.B., M.S.K.); and Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA (M.N.)
| | - Amirfarbod Yazdanyar
- From the Department of Cell Biology, State University of New York, Downstate Medical Center, Brooklyn (H.J., A.Y., Y.C., X.Z., R.L., Z.L., W.J., X.C.J.); Fudan University, Shanghai, China (B.L., Y.C.); Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, New York (Z.L., X.C.J); Institute of Atherosclerosis, Taishan Medical University, Taian, China (X.Z., S.Q.); Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (H.H.B., M.S.K.); and Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA (M.N.)
| | - Bin Lou
- From the Department of Cell Biology, State University of New York, Downstate Medical Center, Brooklyn (H.J., A.Y., Y.C., X.Z., R.L., Z.L., W.J., X.C.J.); Fudan University, Shanghai, China (B.L., Y.C.); Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, New York (Z.L., X.C.J); Institute of Atherosclerosis, Taishan Medical University, Taian, China (X.Z., S.Q.); Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (H.H.B., M.S.K.); and Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA (M.N.)
| | - Yunqin Chen
- From the Department of Cell Biology, State University of New York, Downstate Medical Center, Brooklyn (H.J., A.Y., Y.C., X.Z., R.L., Z.L., W.J., X.C.J.); Fudan University, Shanghai, China (B.L., Y.C.); Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, New York (Z.L., X.C.J); Institute of Atherosclerosis, Taishan Medical University, Taian, China (X.Z., S.Q.); Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (H.H.B., M.S.K.); and Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA (M.N.)
| | - Xiaomin Zhao
- From the Department of Cell Biology, State University of New York, Downstate Medical Center, Brooklyn (H.J., A.Y., Y.C., X.Z., R.L., Z.L., W.J., X.C.J.); Fudan University, Shanghai, China (B.L., Y.C.); Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, New York (Z.L., X.C.J); Institute of Atherosclerosis, Taishan Medical University, Taian, China (X.Z., S.Q.); Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (H.H.B., M.S.K.); and Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA (M.N.)
| | - Ruohan Li
- From the Department of Cell Biology, State University of New York, Downstate Medical Center, Brooklyn (H.J., A.Y., Y.C., X.Z., R.L., Z.L., W.J., X.C.J.); Fudan University, Shanghai, China (B.L., Y.C.); Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, New York (Z.L., X.C.J); Institute of Atherosclerosis, Taishan Medical University, Taian, China (X.Z., S.Q.); Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (H.H.B., M.S.K.); and Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA (M.N.)
| | - Hai Hoang Bui
- From the Department of Cell Biology, State University of New York, Downstate Medical Center, Brooklyn (H.J., A.Y., Y.C., X.Z., R.L., Z.L., W.J., X.C.J.); Fudan University, Shanghai, China (B.L., Y.C.); Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, New York (Z.L., X.C.J); Institute of Atherosclerosis, Taishan Medical University, Taian, China (X.Z., S.Q.); Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (H.H.B., M.S.K.); and Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA (M.N.)
| | - Ming-Shang Kuo
- From the Department of Cell Biology, State University of New York, Downstate Medical Center, Brooklyn (H.J., A.Y., Y.C., X.Z., R.L., Z.L., W.J., X.C.J.); Fudan University, Shanghai, China (B.L., Y.C.); Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, New York (Z.L., X.C.J); Institute of Atherosclerosis, Taishan Medical University, Taian, China (X.Z., S.Q.); Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (H.H.B., M.S.K.); and Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA (M.N.)
| | - Mohamad Navab
- From the Department of Cell Biology, State University of New York, Downstate Medical Center, Brooklyn (H.J., A.Y., Y.C., X.Z., R.L., Z.L., W.J., X.C.J.); Fudan University, Shanghai, China (B.L., Y.C.); Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, New York (Z.L., X.C.J); Institute of Atherosclerosis, Taishan Medical University, Taian, China (X.Z., S.Q.); Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (H.H.B., M.S.K.); and Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA (M.N.)
| | - Shucun Qin
- From the Department of Cell Biology, State University of New York, Downstate Medical Center, Brooklyn (H.J., A.Y., Y.C., X.Z., R.L., Z.L., W.J., X.C.J.); Fudan University, Shanghai, China (B.L., Y.C.); Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, New York (Z.L., X.C.J); Institute of Atherosclerosis, Taishan Medical University, Taian, China (X.Z., S.Q.); Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (H.H.B., M.S.K.); and Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA (M.N.)
| | - Zhiqiang Li
- From the Department of Cell Biology, State University of New York, Downstate Medical Center, Brooklyn (H.J., A.Y., Y.C., X.Z., R.L., Z.L., W.J., X.C.J.); Fudan University, Shanghai, China (B.L., Y.C.); Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, New York (Z.L., X.C.J); Institute of Atherosclerosis, Taishan Medical University, Taian, China (X.Z., S.Q.); Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (H.H.B., M.S.K.); and Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA (M.N.)
| | - Weijun Jin
- From the Department of Cell Biology, State University of New York, Downstate Medical Center, Brooklyn (H.J., A.Y., Y.C., X.Z., R.L., Z.L., W.J., X.C.J.); Fudan University, Shanghai, China (B.L., Y.C.); Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, New York (Z.L., X.C.J); Institute of Atherosclerosis, Taishan Medical University, Taian, China (X.Z., S.Q.); Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (H.H.B., M.S.K.); and Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA (M.N.)
| | - Xian-Cheng Jiang
- From the Department of Cell Biology, State University of New York, Downstate Medical Center, Brooklyn (H.J., A.Y., Y.C., X.Z., R.L., Z.L., W.J., X.C.J.); Fudan University, Shanghai, China (B.L., Y.C.); Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, New York (Z.L., X.C.J); Institute of Atherosclerosis, Taishan Medical University, Taian, China (X.Z., S.Q.); Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN (H.H.B., M.S.K.); and Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA (M.N.).
| |
Collapse
|
15
|
Sun X, Luo W, Tan X, Li Q, Zhao Y, Zhong W, Sun X, Brouwer C, Zhou Z. Increased plasma corticosterone contributes to the development of alcoholic fatty liver in mice. Am J Physiol Gastrointest Liver Physiol 2013; 305:G849-61. [PMID: 24113770 PMCID: PMC3882437 DOI: 10.1152/ajpgi.00139.2013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Ethanol ingestion increases endogenous glucocorticoid levels in both humans and rodents. The present study aimed to define a mechanistic link between the increased glucocorticoids and alcoholic fatty liver in mice. Plasma corticosterone levels were not affected in mice on a 2-wk ethanol diet regimen but significantly increased upon 4 wk of ethanol ingestion. Accordingly, hepatic triglyceride levels were not altered after 2 wk of ethanol ingestion but were elevated at 4 wk. Based on the observation that 2 wk of ethanol ingestion did not significantly increase endogenous corticosterone levels, we administered exogenous glucocorticoids along with the 2-wk ethanol treatment to determine whether the elevated glucocorticoid contributes to the development of alcoholic fatty liver. Mice were subjected to ethanol feeding for 2 wk with or without dexamethasone administration. Hepatic triglyceride contents were not affected by either ethanol or dexamethasone alone but were significantly increased by administration of both. Microarray and protein level analyses revealed two distinct changes in hepatic lipid metabolism in mice administered with both ethanol and dexamethasone: accelerated triglyceride synthesis by diacylglycerol O-acyltransferase 2 and suppressed fatty acid β-oxidation by long-chain acyl-CoA synthetase 1, carnitine palmitoyltransferase 1a, and acyl-CoA oxidase 1. A reduction of hepatic peroxisome proliferation activator receptor-α (PPAR-α) was associated with coadministration of ethanol and dexamethasone. These findings suggest that increased glucocorticoid levels may contribute to the development of alcoholic fatty liver, at least partially, through hepatic PPAR-α inactivation.
Collapse
Affiliation(s)
- Xiuhua Sun
- Ctr. for Translational & Biomedical Research, The Univ. of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC 28081.
| | - Weijun Luo
- 3Bioinformatics Services Division, UNC-Charlotte, ,4Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, Kannapolis;
| | - Xiaobing Tan
- 1Center for Translational and Biomedical Research,
| | - Qiong Li
- 1Center for Translational and Biomedical Research,
| | - Yantao Zhao
- 5Department of Bioinformatics and Genomics, The University of North Carolina at Charlotte, North Carolina
| | - Wei Zhong
- 1Center for Translational and Biomedical Research,
| | - Xinguo Sun
- 1Center for Translational and Biomedical Research,
| | - Cory Brouwer
- 3Bioinformatics Services Division, UNC-Charlotte, ,4Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, Kannapolis;
| | - Zhanxiang Zhou
- 1Center for Translational and Biomedical Research, ,2Department of Nutrition, The University of North Carolina at Greensboro,
| |
Collapse
|
16
|
Yazdanyar A, Quan W, Jin W, Jiang XC. Liver-specific phospholipid transfer protein deficiency reduces high-density lipoprotein and non-high-density lipoprotein production in mice. Arterioscler Thromb Vasc Biol 2013; 33:2058-64. [PMID: 23846500 DOI: 10.1161/atvbaha.113.301628] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The liver is one of the critical organs for lipoprotein metabolism and a major source for phospholipid transfer protein (PLTP) expression. The effect of liver-specific PLTP deficiency on plasma lipoprotein production and metabolism in mice was investigated. APPROACH AND RESULTS We created a liver-specific PLTP-deficient mouse model. We measured plasma high-density lipoprotein (HDL) and apolipoprotein B (apoB)-containing lipoprotein (or non-HDL) levels and their production rates. We found that hepatic ablation of PLTP leads to a significant decrease in plasma PLTP activity, HDL lipids, non-HDL lipids, apoAI, and apoB levels. In addition, nuclear magnetic resonance examination of lipoproteins showed that the deficiency decreases HDL and apoB-containing lipoprotein particle numbers, as well as very low-density lipoprotein particle size, which was confirmed by electron microscopy. Moreover, HDL particles from the deficient mice are lipid-poor ones. To unravel the mechanism, we evaluated the apoB and triglyceride production rates. We found that hepatic PLTP deficiency significantly decreases apoB and triglyceride secretion rates. To investigate the role of liver PLTP on HDL production, we set up primary hepatocyte culture studies and found that the PLTP-deficient hepatocytes produce less nascent HDL. Furthermore, we found that exogenous PLTP promotes nascent HDL production through an ATP-binding cassette A 1-mediated pathway. CONCLUSIONS Liver-specific PLTP deficiency significantly reduces plasma HDL and apoB-containing lipoprotein levels. Reduction of production rates of both particles is one of the mechanisms.
Collapse
Affiliation(s)
- Amirfarbod Yazdanyar
- Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY 11203, USA
| | | | | | | |
Collapse
|
17
|
Zhu L, Brown WC, Cai Q, Krust A, Chambon P, McGuinness OP, Stafford JM. Estrogen treatment after ovariectomy protects against fatty liver and may improve pathway-selective insulin resistance. Diabetes 2013; 62:424-34. [PMID: 22966069 PMCID: PMC3554377 DOI: 10.2337/db11-1718] [Citation(s) in RCA: 245] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pathway-selective insulin resistance where insulin fails to suppress hepatic glucose production but promotes liver fat storage may underlie glucose and lipid abnormalities after menopause. We tested the mechanisms by which estrogen treatment may alter the impact of a high-fat diet (HFD) when given at the time of ovariectomy (OVX) in mice. Female C57BL/6J mice underwent sham operation, OVX, or OVX with estradiol (E2) treatment and were fed an HFD. Hyperinsulinemic-euglycemic clamps were used to assess insulin sensitivity, tracer incorporation into hepatic lipids, and liver triglyceride export. OVX mice had increased adiposity that was prevented with E2 at the time of OVX. E2 treatment increased insulin sensitivity with OVX and HFD. In sham and OVX mice, HFD feeding induced fatty liver, and insulin reduced hepatic apoB100 and liver triglyceride export. E2 treatment reduced liver lipid deposition and prevented the decrease in liver triglyceride export during hyperinsulinemia. In mice lacking the liver estrogen receptor α, E2 after OVX limited adiposity but failed to improve insulin sensitivity, to limit liver lipid deposition, and to prevent insulin suppression of liver triglyceride export. In conclusion, estrogen treatment may reverse aspects of pathway-selective insulin resistance by promoting insulin action on glucose metabolism but limiting hepatic lipid deposition.
Collapse
Affiliation(s)
- Lin Zhu
- Tennessee Valley Healthcare System Nashville, Tennessee
| | - William C. Brown
- Tennessee Valley Healthcare System Nashville, Tennessee
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Qing Cai
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Andrée Krust
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Pierre Chambon
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Illkirch, France
| | - Owen P. McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - John M. Stafford
- Tennessee Valley Healthcare System Nashville, Tennessee
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee
- Case Western Reserve Medical Center, Cleveland, Ohio
- Corresponding author: John M. Stafford,
| |
Collapse
|
18
|
Jiang XC, Jin W, Hussain MM. The impact of phospholipid transfer protein (PLTP) on lipoprotein metabolism. Nutr Metab (Lond) 2012; 9:75. [PMID: 22897926 PMCID: PMC3495888 DOI: 10.1186/1743-7075-9-75] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Accepted: 06/30/2012] [Indexed: 02/05/2023] Open
Abstract
It has been reported that phospholipid transfer protein (PLTP) is an independent risk factor for human coronary artery disease. In mouse models, it has been demonstrated that PLTP overexpression induces atherosclerosis, while its deficiency reduces it. PLTP is considered a promising target for pharmacological intervention to treat atherosclerosis. However, we must still answer a number of questions before its pharmaceutical potential can be fully explored. In this review, we summarized the recent progresses made in the PLTP research field and focused on its effect on apoB-containing- triglyceride-rich particle and HDL metabolism.
Collapse
Affiliation(s)
- Xian-Cheng Jiang
- Department of Cell Biology, Downstate Medical Center, State University of New York, 450 Clarkson Ave,, Box 5, Brooklyn, NY, 11203, USA.
| | | | | |
Collapse
|
19
|
Yazdanyar A, Jiang XC. Liver phospholipid transfer protein (PLTP) expression with a PLTP-null background promotes very low-density lipoprotein production in mice. Hepatology 2012; 56:576-84. [PMID: 22367708 PMCID: PMC3409695 DOI: 10.1002/hep.25648] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Accepted: 01/31/2012] [Indexed: 12/07/2022]
Abstract
UNLABELLED It is known that plasma phospholipid transfer protein (PLTP) activity influences lipoprotein metabolism. The liver is one of the major sites of lipoprotein production and degradation, as well as of PLTP expression. To address the impact of liver-expressed PLTP on lipoprotein metabolism, we created a mouse model that expresses PLTP in the liver acutely and specifically, with a PLTP-null background. This approach in mouse model preparations can also be used universally for evaluating the function of many other genes in the liver. We found that liver PLTP expression dramatically increases plasma levels of non-high-density lipoprotein (HDL) cholesterol (2.7-fold, P < 0.0001), non-HDL phospholipid (2.5-fold, P < 0.001), and triglyceride (51%, P < 0.01), but has no significant influence on plasma HDL lipids compared with controls. Plasma apolipoprotein (apo)B levels were also significantly increased in PLTP-expressing mice (2.2-fold, P < 0.001), but those of apoA-I were not. To explore the mechanism involved, we examined the lipidation and secretion of nascent very low-density lipoprotein (VLDL), finding that liver PLTP expression significantly increases VLDL lipidation in hepatocyte microsomal lumina, and also VLDL secretion into the plasma. CONCLUSION It is possible to prepare a mouse model that expresses the gene of interest only in the liver, but not in other tissues. Our results suggest, for the first time, that the major function of liver PLTP is to drive VLDL production and makes a small contribution to plasma PLTP activity.
Collapse
Affiliation(s)
| | - Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Medical Center
- To whom correspondence may be addressed at Downstate Medical Center, 450 Clarkson Ave. Box 5 Brooklyn, NY 11203, tel. (718) 270-6701, Fax (718) 270-3732,
| |
Collapse
|
20
|
Dullaart RPF, Vergeer M, de Vries R, Kappelle PJWH, Dallinga-Thie GM. Type 2 diabetes mellitus interacts with obesity and common variations in PLTP to affect plasma phospholipid transfer protein activity. J Intern Med 2012; 271:490-8. [PMID: 21973210 DOI: 10.1111/j.1365-2796.2011.02465.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND Phospholipid transfer protein (PLTP) is an emerging cardiometabolic risk marker that is important in high-density lipoprotein (HDL) and triglyceride metabolism. Plasma PLTP activity is elevated in type 2 diabetes mellitus, whereas glucose may regulate PLTP gene transcription in vitro. Of interest, common PLTP variations that predict cardiovascular disease have been identified recently. We investigated whether the diabetic state is able to amplify relationships between obesity and PLTP gene variations with circulating PLTP levels. SUBJECTS AND METHODS Plasma PLTP activity (using a phospholipid vesicles-HDL system), PLTP gene score [number of PLTP activity-decreasing alleles based on two tagging polymorphisms (rs378114 and rs60- 65904)] and waist circumference were determined in two Dutch cohorts comprising 237 patients with type 2 diabetes and 78 control subjects. RESULTS Patients with diabetes were more obese (P < 0.001 for prevalence of increased waist circumference) and had 13% higher plasma PLTP activity (P < 0.001). PLTP gene score was not different in diabetic and control subjects (P = 0.40). PLTP activity was highest in patients with diabetes with an enlarged waist and lowest in control subjects with a normal waist circumference (P < 0.001). Multiple linear regression analysis revealed a positive interaction between diabetes status and waist circumference on PLTP activity (β = 0.200, P = 0.005). Furthermore, diabetes status (β = -0.485, P = 0.046) or HbA1c (β = -0.240, P = 0.035) interacted with PLTP gene score to affect PLTP activity. CONCLUSIONS Type 2 diabetes and enlarged waist circumference interact to impact on plasma PLTP activity. Diabetes may also amplify the association between plasma PLTP activity and common PLTP gene variations. Our findings support the hypothesis that diabetes-environment and diabetes-gene interactions govern plasma PLTP activity.
Collapse
Affiliation(s)
- R P F Dullaart
- Department of Endocrinology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | | | | | | | | |
Collapse
|
21
|
Besler C, Lüscher TF, Landmesser U. Molecular mechanisms of vascular effects of High-density lipoprotein: alterations in cardiovascular disease. EMBO Mol Med 2012; 4:251-68. [PMID: 22431312 PMCID: PMC3376856 DOI: 10.1002/emmm.201200224] [Citation(s) in RCA: 152] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2011] [Revised: 01/28/2012] [Accepted: 02/03/2012] [Indexed: 12/12/2022] Open
Abstract
Low high-density lipoprotein (HDL)-cholesterol levels are associated with an increased risk of coronary artery disease (CAD) and myocardial infarction, which has triggered the hypothesis that HDL, in contrast to low-density lipoprotein (LDL), acts as an anti-atherogenic lipoprotein. Moreover, experimental studies have identified potential anti-atherogenic properties of HDL, including promotion of macrophage cholesterol efflux and direct endothelial-protective effects of HDL, such as stimulation of endothelial nitric oxide production and repair, anti-apoptotic, anti-inflammatory and anti-thrombotic properties. Studies in gene-targeted mice, however, have also indicated that increasing HDL-cholesterol plasma levels can either limit (e.g. apolipoprotein A-I) or accelerate (e.g. Scavenger receptor class B type I) atherosclerosis. Moreover, vascular effects of HDL have been observed to be heterogenous and are altered in patients with CAD or diabetes, a condition that has been termed 'HDL dysfunction'. These alterations in biological functions of HDL may need to be taken into account for HDL-targeted therapies and considering raising of HDL-cholesterol levels alone is likely not sufficient in this respect. It will therefore be important to further determine, which biological functions of HDL are critical for its anti-atherosclerotic properties, as well as how these can be measured and targeted.
Collapse
Affiliation(s)
- Christian Besler
- Department of Cardiology, Cardiovascular Center, University Hospital Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
22
|
Plasma PLTP (phospholipid-transfer protein): an emerging role in 'reverse lipopolysaccharide transport' and innate immunity. Biochem Soc Trans 2011; 39:984-8. [PMID: 21787334 DOI: 10.1042/bst0390984] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Plasma PLTP (phospholipid-transfer protein) is a member of the lipid transfer/LBP [LPS (lipopolysaccharide)-binding protein] family, which constitutes a superfamily of genes together with the short and long PLUNC (palate, lung and nasal epithelium clone) proteins. Although PLTP was studied initially for its involvement in the metabolism of HDL (high-density lipoproteins) and reverse cholesterol transport (i.e. the metabolic pathway through which cholesterol excess can be transported from peripheral tissues back to the liver for excretion in the bile), it displays a number of additional biological properties. In particular, PLTP can modulate the lipoprotein association and metabolism of LPS that are major components of Gram-negative bacteria. The delayed association of LPS with lipoproteins in PLTP-deficient mice results in a prolonged residence time, in a higher toxicity of LPS aggregates and in a significant increase in LPS-induced mortality as compared with wild-type mice. It suggests that PLTP may play a pivotal role in inflammation and innate immunity through its ability to accelerate the 'reverse LPS transport' pathway.
Collapse
|
23
|
de Vries R, Perton FG, van Tol A, Dullaart RPF. Carotid intima media thickness is related positively to plasma pre ß-high density lipoproteins in non-diabetic subjects. Clin Chim Acta 2011; 413:473-7. [PMID: 22100832 DOI: 10.1016/j.cca.2011.11.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Revised: 10/27/2011] [Accepted: 11/03/2011] [Indexed: 10/15/2022]
Abstract
BACKGROUND Lipid-poor or lipid-free high density lipoprotein (HDL) particles, designated pre ß-HDL, stimulate removal of cell-derived cholesterol to the extracellular compartment, which is an initial step in the reverse cholesterol transport pathway. Pre ß-HDL levels may be elevated in subjects with established cardiovascular disease. We determined the relationship of carotid intima media thickness (IMT), a marker of subclinical atherosclerosis, with pre ß-HDL in subjects without clinically manifest cardiovascular disease. METHODS IMT and plasma pre ß-HDL, assayed by crossed immuno-electrophoresis, were determined in 70 non-diabetic subjects (aged 56±9 years; non-smokers only; 27 women). RESULTS IMT was correlated positively with pre ß-HDL, both expressed as plasma apolipoprotein (apo) A-I concentration (r=0.271, p=0.023) and as% of apo A-I (r=0.341, p=0.004). In contrast, IMT was correlated inversely with HDL cholesterol (r=-0.253, p=0.035). IMT was also related positively to pre ß-HDL after adjustment for age, sex, systolic blood pressure (in apoA-I concentration, ß=0.203, p=0.043; in% of plasma apoA-I, ß=0.235, p=0.023). IMT remained associated with pre ß-HDL after additional adjustment for either body mass index, plasma glucose, cholesterol, triglycerides, HDL cholesterol, apoA-I and apoB. CONCLUSION Subclinical atherosclerosis may relate to higher plasma pre ß-HDL independently of apoA-I and HDL cholesterol levels.
Collapse
Affiliation(s)
- Rindert de Vries
- Department of Endocrinology, University Medical Center Groningen and University of Groningen, Groningen, The Netherlands
| | | | | | | |
Collapse
|
24
|
Masson D, Deckert V, Gautier T, Klein A, Desrumaux C, Viglietta C, Pais de Barros JP, Le Guern N, Grober J, Labbé J, Ménétrier F, Ripoll PJ, Leroux-Coyau M, Jolivet G, Houdebine LM, Lagrost L. Worsening of diet-induced atherosclerosis in a new model of transgenic rabbit expressing the human plasma phospholipid transfer protein. Arterioscler Thromb Vasc Biol 2011; 31:766-74. [PMID: 21252068 DOI: 10.1161/atvbaha.110.215756] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Plasma phospholipid transfer protein (PLTP) is involved in intravascular lipoprotein metabolism. PLTP is known to act through 2 main mechanisms: by remodeling high-density lipoproteins (HDL) and by increasing apolipoprotein (apo) B-containing lipoproteins. The aim of this study was to generate a new model of human PLTP transgenic (HuPLTPTg) rabbit and to determine whether PLTP expression modulates atherosclerosis in this species that, unlike humans and mice, displays naturally very low PLTP activity. METHODS AND RESULTS In HuPLTPTg rabbits, the human PLTP cDNA was placed under the control of the human eF1-α gene promoter, resulting in a widespread tissue expression pattern and in increased plasma PLTP. The HuPLTPTg rabbits showed a significant increase in the cholesterol content of the plasma apoB-containing lipoprotein fractions, with a more severe trait when animals were fed a cholesterol-rich diet. In contrast, HDL cholesterol level was not modified in HuPLTPTg rabbits. Formation of aortic fatty streaks was increased in hypercholesterolemic HuPLTPTg animals as compared with nontransgenic littermates. CONCLUSIONS Human PLTP expression in HuPLTPTg rabbit worsens atherosclerosis as a result of increased levels of atherogenic apoB-containing lipoproteins but not of alterations in their antioxidative protection or in cholesterol content of plasma HDL.
Collapse
Affiliation(s)
- David Masson
- Institut National de la Santé et de la Recherche Médicale, Université de Bourgogne, UMR866, Dijon, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Schizophrenia is one of the most severe psychiatric disorders affecting 1% of the world population. There is yet no empirical method to validate the diagnosis of the disease. The identification of an underlying molecular alteration could lead to an improved disease understanding and may yield an objective panel of biomarkers to aid in the diagnosis of this devastating disease. Presented is the largest reported liquid chromatography-mass spectrometry-based proteomic profiling study investigating serum samples taken from first-onset drug-naive patients compared with samples collected from healthy volunteers. The results of this large-scale study are presented along with enzyme-linked immunosorbent assay-based validation data.
Collapse
|
26
|
Liu HR, Wu G, Zhou B, Chen BS. Low cholesteryl ester transfer protein and phospholipid transfer protein activities are the factors making tree shrew and beijing duck resistant to atherosclerosis. Lipids Health Dis 2010; 9:114. [PMID: 20937151 PMCID: PMC2964723 DOI: 10.1186/1476-511x-9-114] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Accepted: 10/12/2010] [Indexed: 11/20/2022] Open
Abstract
Background Tree shrew and beijing duck are regarded as animal models resistant to atherosclerosis (AS). This study was carried out to discover the potential mechanism. Methods Blood samples were collected from healthy men and male animals. Plasma lipid profile and activities of cholesteryl ester transfer protein (CETP) and phospholipid transfer protein (PLTP) were measured, compared and analyzed in human, tree shrew, and Beijing duck. Results The results showed that there were species differences on plasma lipid profile and activities of CETP and PLTP in the three species. Compared with human, tree shrew and beijing duck had higher high density lipoprotein cholesterol (HDL-C)/total cholesterol (TC) and HDL-C/low density lipoprotein cholesterol (LDL-C) ratios, but lower CETP and PLTP activities. In the three species, CETP and PLTP activities were negatively related with the ratio of HDL-C/LDL-C. Conclusions The present study suggested that low plasma CETP and PLTP activities may lead to a high HDL-C/LDL-C ratio and a high resistance to AS finally in tree shrew and beijing duck. Moreover, low PLTP activity may also make the animals resistant to AS by the relative high vitamin E content of apoB-containing lipoproteins and high anti-inflammatory and antioxidative properties of HDL particles. A detailed study in the future is recommended.
Collapse
Affiliation(s)
- Hui-rong Liu
- College of Life Sciences, Inner Mongolia Agricultural University, Hohhot 010018, China.
| | | | | | | |
Collapse
|
27
|
Trevaskis NL, Shanker RM, Charman WN, Porter CJH. The Mechanism of Lymphatic Access of Two Cholesteryl Ester Transfer Protein Inhibitors (CP524,515 and CP532,623) and Evaluation of Their Impact on Lymph Lipoprotein Profiles. Pharm Res 2010; 27:1949-64. [DOI: 10.1007/s11095-010-0199-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2010] [Accepted: 06/16/2010] [Indexed: 10/19/2022]
|
28
|
Trevaskis NL, McEvoy CL, McIntosh MP, Edwards GA, Shanker RM, Charman WN, Porter CJH. The Role of the Intestinal Lymphatics in the Absorption of Two Highly Lipophilic Cholesterol Ester Transfer Protein Inhibitors (CP524,515 and CP532,623). Pharm Res 2010; 27:878-93. [DOI: 10.1007/s11095-010-0083-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Accepted: 02/09/2010] [Indexed: 01/01/2023]
|
29
|
Schlitt A, Schwaab B, Fingscheidt K, Lackner KJ, Heine GH, Vogt A, Buerke M, Maegdefessel L, Raaz U, Werdan K, Jiang XC. Serum phospholipid transfer protein activity after a high fat meal in patients with insulin-treated type 2 diabetes. Lipids 2010; 45:129-35. [PMID: 20108050 DOI: 10.1007/s11745-010-3384-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2009] [Accepted: 01/04/2010] [Indexed: 11/25/2022]
Abstract
Plasma phospholipid transfer protein (PLTP) mediates both net transfer and exchange of phospholipids between different lipoproteins. Animal studies have shown that it is closely related to the development of atherosclerosis. Although many studies have indicated that PLTP activity is increased in diabetes mellitus, the role of PLTP in diabetes is still unclear. To evaluate the influence of a high-fat meal on PLTP activity, 50 nondiabetic patients with coronary heart disease (CHD), 50 insulin-treated Type 2 diabetics, and 50 healthy controls were included. We determined PLTP activity before and 4 and 8 h after a high-fat meal. As expected, serum PLTP activity was significantly higher in CHD patients than in healthy controls (71.0 +/- 46.2 vs. 54.0 +/- 33.8 pmol/microl/h, P = 0.032) at baseline. More importantly, we found that serum PLTP activity increased to its maximum 4 h after fat loading and then decreased to nearly basal levels after 8 h both in controls and CHD patients. In contrast, PLTP activity continuously increased during this time period in the diabetic patients. With regards to the data from this study we hypothesize that serum PLTP is involved in the clearance of postprandial lipoproteins and this process is attenuated in diabetes. Since postprandial lipoproteins are atherogenic, the delay in clearance of these particles could play an important role in the development of atherosclerosis in patients with diabetes mellitus.
Collapse
Affiliation(s)
- Axel Schlitt
- Department of Medicine III, Martin Luther-University, Halle, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Okazaki H, Goldstein JL, Brown MS, Liang G. LXR-SREBP-1c-phospholipid transfer protein axis controls very low density lipoprotein (VLDL) particle size. J Biol Chem 2009; 285:6801-10. [PMID: 20037162 DOI: 10.1074/jbc.m109.079459] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Liver X receptors (LXRs) activate triglyceride synthesis in liver directly and indirectly by inducing sterol regulatory element-binding protein-1c (SREBP-1c). When administered to wild-type mice, the LXR activator T0901317 produces a mild and transient hypertriglyceridemia. Here, we show that T0901317 produces massive hypertriglyceridemia when given to mice lacking low density lipoprotein (LDL) receptors (Ldlr(-/-) mice). Triglycerides ranged from 4000 to 6000 mg/dl, and the plasma turned milky. The median diameter of VLDL particles, measured by electron microscopy, increased from 43 to 112 nm, 87% exceeding 80 nm, the size of chylomicrons. Hypertriglyceridemia was prevented in Ldlr(-/-) recipient mice that lacked SREBP-1c (Ldlr(-/-);Srebp-1c(-/-) double knock-out mice). In Ldlr(-/-) mice, T0901317 increased mRNAs not only for enzymes of fatty acid and triglyceride synthesis, but also for phospholipid transfer protein (PLTP), which transfers phospholipids into nascent VLDL, allowing particle expansion. The PLTP increase was blunted in Ldlr(-/-);Srebp-1c(-/-) animals. When Ldlr(-/-);Srebp-1c(-/-) mice received an adenovirus encoding Pltp, the hypertriglyceridemic response to T0901317 was partially restored and the VLDL size increased. We conclude that LXR agonists activate triglyceride synthesis and Pltp transcription by activating Srebp-1c. In concert with the increase in TG synthesis, the increased PLTP permits triglyceride incorporation into abnormally large VLDL, which are removed from plasma by LDL receptors. In the absence of LDL receptors, the large VLDLs accumulate and produce massive hypertriglyceridemia.
Collapse
Affiliation(s)
- Hiroaki Okazaki
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, Texas 75390-9046, USA
| | | | | | | |
Collapse
|
31
|
Chen X, Sun A, Mansoor A, Zou Y, Ge J, Lazar JM, Jiang XC. Plasma PLTP activity is inversely associated with HDL-C levels. Nutr Metab (Lond) 2009; 6:49. [PMID: 19948027 PMCID: PMC2793253 DOI: 10.1186/1743-7075-6-49] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2009] [Accepted: 11/30/2009] [Indexed: 12/13/2022] Open
Abstract
Phospholipid transfer protein (PLTP) is an important modulator of lipoprotein metabolism, including interparticle phospholipid transfer, remodeling of HDL, cholesterol and phospholipid efflux from peripheral tissues, and the production of hepatic VLDL. PLTP also plays an important role in inflammation and oxidative stress. Accordingly, PLTP has been implicated in the development of atherosclerosis. In this study, we evaluated the association between PLTP activity and lipoprotein metabolism in a Chinese patients cohort with or without coronary heart disease (CHD group n = 407, control group n = 215), the PLTP activity was measured and PLTP genotyping was screened for sequence anomalies by PCR. We found that human plasma PLTP activity was negatively associated with plasma HDL and apoA-I levels, and positively associated with plasma TG, apoB and apoE levels. We also found that PLTP rs2294213 polymorphism was tended to be associated with increased plasma PLTP activity.
Collapse
Affiliation(s)
- Xueying Chen
- Institute of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Aijun Sun
- Institute of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Ather Mansoor
- Division of Cardiovascular Medicine, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Yunzeng Zou
- Institute of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Junbo Ge
- Institute of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Jason M Lazar
- Division of Cardiovascular Medicine, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| |
Collapse
|
32
|
van Haperen R, Samyn H, van Gent T, Zonneveld AJ, Moerland M, Grosveld F, Jansen H, Dallinga-Thie GM, van Tol A, de Crom R. Novel roles of hepatic lipase and phospholipid transfer protein in VLDL as well as HDL metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2009; 1791:1031-6. [PMID: 19524061 DOI: 10.1016/j.bbalip.2009.06.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Revised: 05/15/2009] [Accepted: 06/04/2009] [Indexed: 10/20/2022]
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.
Collapse
Affiliation(s)
- Rien van Haperen
- Department of Cell Biology and Genetics, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Oka SI, Yoshihara E, Bizen-Abe A, Liu W, Watanabe M, Yodoi J, Masutani H. Thioredoxin binding protein-2/thioredoxin-interacting protein is a critical regulator of insulin secretion and peroxisome proliferator-activated receptor function. Endocrinology 2009; 150:1225-34. [PMID: 18974273 DOI: 10.1210/en.2008-0646] [Citation(s) in RCA: 76] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The feeding-fasting nutritional transition triggers a dynamic change in metabolic pathways and is a model for understanding how these pathways are mutually organized. The targeted disruption of the thioredoxin binding protein-2 (TBP-2)/thioredoxin-interacting protein (Txnip)/VDUP1 gene in mice results in lethality with hypertriglyceridemia and hypoglycemia during fasting. To investigate the molecular mechanism of the nutritional transition and the role of TBP-2, microarray analyses were performed using the liver of TBP-2(-/-) mice in the fed and fasted states. We found that the fasting-induced reduction in the expression of lipogenic genes targeted by insulin (SREBP-1), such as FASN and THRSP, was abolished in TBP-2(-/-) mice, and the expression of lipoprotein lipase is down-regulated, which was consistent with the lipoprotein profile. TBP-2(-/-) mice also exhibited enhanced glucose-induced insulin secretion and sensitivity. Another feature of the hepatic gene expression in fed TBP-2(-/-) mice was the augmented expression of peroxisome proliferator activated receptor (PPAR) target genes, such as CD36, FABP2, ACOT1, and FGF21, to regulate fatty acid consumption. In TBP-2(-/-) mice, PPARalpha expression was elevated in the fed state, whereas the fasting-induced up-regulation of PPARalpha was attenuated. We also detected an increased expression of PPARgamma coactivator-1alpha protein in fed TBP-2(-/-) mice. TBP-2 overexpression significantly inhibited PPARalpha-mediated transcriptional activity induced by a specific PPARalpha ligand in vitro. These results suggest that TBP-2 is a key regulator of PPARalpha expression and signaling, and coordinated regulation of PPARalpha and insulin secretion by TBP-2 is crucial in the feeding-fasting nutritional transition.
Collapse
Affiliation(s)
- Shin-ichi Oka
- Department of Biological Responses, Institute for Virus Research, Kyoto University, Kyoto, Japan
| | | | | | | | | | | | | |
Collapse
|
34
|
Schlitt A, Blankenberg S, Bickel C, Lackner KJ, Heine GH, Buerke M, Werdan K, Maegdefessel L, Raaz U, Rupprecht HJ, Munzel T, Jiang XC. PLTP activity is a risk factor for subsequent cardiovascular events in CAD patients under statin therapy: the AtheroGene study. J Lipid Res 2008; 50:723-9. [PMID: 19001358 DOI: 10.1194/jlr.m800414-jlr200] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Phospholipid transferprotein (PLTP) mediates both net transfer and exchange of phospholipids between different lipoproteins. Although many studies have investigated the role of PLTP in atherogenesis, the role of PLTP in atherosclerotic diseases is unclear. We investigated the association of serum PLTP activity with the incidence of a combined endpoint (myocardial infarction and cardiovascular death) and its relation to other markers of atherosclerosis in 1,085 patients with angiographically documented coronary artery disease (CAD). In the median follow-up of 5.1 years, 156 patients had suffered from the combined endpoint of myocardial infarction or cardiovascular death including 47 of 395 patients who were on statins at baseline. In Kaplan-Meyer analyses serum PLTP activity was not associated with the combined endpoint in all patients. However, in the subgroup of patients receiving statins at baseline, PLTP was shown to be a significant predictor of cardiovascular outcome (P = 0.019), and this also remained stable in univariate (P = 0.027) and multivariate cox regression analyses (P = 0.041) including potential confounders (classical risk factors, HDL cholesterol (HDL-C), and others). We showed in our study that, under statin treatment, high plasma PLTP activity was related to fatal and nonfatal cardiovascular events in CAD patients.
Collapse
Affiliation(s)
- Axel Schlitt
- Department of Medicine III, Martin Luther-University Halle-Wittenberg, Germany.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
PURPOSE OF REVIEW This review summarizes the role of phosphatidylcholine metabolism in plasma lipoprotein homeostasis. RECENT FINDINGS While it was previously known that phosphatidylcholine biosynthesis was required for normal hepatic VLDL secretion, recent studies have shown that both phosphatidylcholine biosynthetic pathways (the cytidine 5'-diphosphocholine and the phosphatidylethanolamine methylation pathways) are required. In addition, a requirement of acyl-coenzyme A synthetase 3, but not acyl-coenzyme A synthetase 1 or 4, for phosphatidylcholine synthesis and VLDL secretion is now documented. ABCA1 has been implicated in the transfer of phosphatidylcholine to apolipoproteinA-1 both during and after secretion of apolipoproteinA-1. Other studies have introduced the concept of reverse phosphatidylcholine transport in which both HDL and LDL supply phosphatidylcholine to the liver. An unexpected finding is that half of the phosphatidylcholine delivered to liver from lipoproteins is converted into triacylglycerol. SUMMARY The liver is both a donor of phosphatidylcholine during the assembly and secretion of lipoproteins as well as a recipient of phosphatidylcholine from plasma lipoproteins.
Collapse
Affiliation(s)
- Dennis E Vance
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
36
|
Vergeer M, Dallinga-Thie GM, Dullaart RPF, van Tol A. Evaluation of phospholipid transfer protein as a therapeutic target. ACTA ACUST UNITED AC 2008. [DOI: 10.2217/17460875.3.3.327] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
37
|
van Haperen R, Samyn H, Moerland M, van Gent T, Peeters M, Grosveld F, van Tol A, de Crom R. Elevated expression of phospholipid transfer protein in bone marrow derived cells causes atherosclerosis. PLoS One 2008; 3:e2255. [PMID: 18509527 PMCID: PMC2386265 DOI: 10.1371/journal.pone.0002255] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2008] [Accepted: 03/25/2008] [Indexed: 02/02/2023] Open
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.
Collapse
Affiliation(s)
- Rien van Haperen
- Department of Cell Biology and Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Hannelore Samyn
- Department of Cell Biology and Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Matthijs Moerland
- Department of Cell Biology and Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Teus van Gent
- Department of Cell Biology and Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Marian Peeters
- Department of Cell Biology and Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Frank Grosveld
- Department of Cell Biology and Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Arie van Tol
- Department of Cell Biology and Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Rini de Crom
- Department of Cell Biology and Genetics, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Vascular Surgery, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- * E-mail:
| |
Collapse
|
38
|
Moerland M, Samyn H, van Gent T, van Haperen R, Dallinga-Thie G, Grosveld F, van Tol A, de Crom R. Acute elevation of plasma PLTP activity strongly increases pre-existing atherosclerosis. Arterioscler Thromb Vasc Biol 2008; 28:1277-82. [PMID: 18421000 DOI: 10.1161/atvbaha.108.165084] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
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.
Collapse
Affiliation(s)
- Matthijs Moerland
- Department Cell Biology & Genetics/Vascular Surgery, Erasmus University Medical Center, Dr Molewaterplein 50, 3015 GE Rotterdam, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Dullaart RPF, de Vries R, Dallinga-Thie GM, Sluiter WJ, van Tol A. Phospholipid transfer protein activity is determined by type 2 diabetes mellitus and metabolic syndrome, and is positively associated with serum transaminases. Clin Endocrinol (Oxf) 2008; 68:375-81. [PMID: 17877759 DOI: 10.1111/j.1365-2265.2007.03049.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
BACKGROUND The extent to which plasma phospholipid transfer protein (PLTP) activity is affected by type 2 diabetes mellitus (DM) and metabolic syndrome (MetS) is still unknown. PLTP is synthesized in the liver, and elevated serum transaminases are considered to predict nonalcoholic fatty liver disease (NAFLD). In this study, we examined the relationship between plasma PLTP activity and liver enzymes in subjects with and without DM and MetS. DESIGN Plasma PLTP activity, serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were measured in 71 subjects without DM or MetS, 21 without DM but with MetS, 26 with DM but without MetS and 55 with DM and MetS (WHO and NCEP-ATP III criteria). RESULTS After controlling for age, sex and alcohol intake, PLTP activity was positively related to both MetS (P < 0.001) and DM (P = 0.001). Serum ALT (P = 0.006) and AST (P = 0.04) were both associated with MetS, but only ALT was associated with DM (P < 0.001). In multiple linear regression models, serum ALT and AST were positively and independently associated with PLTP activity (P < 0.01 for all), even when the presence of MetS and DM was taken into account, as well as after controlling for glycated haemoglobin (HbA(1c)), insulin resistance, triglycerides, free fatty acids (FFA), C-reactive protein (CRP), leptin and adiponectin. CONCLUSIONS Plasma PLTP activity is determined by MetS and by diabetes per se. Serum transaminases are independently associated with PLTP activity. We suggest that this lipid transfer protein may be a marker for NAFLD.
Collapse
Affiliation(s)
- Robin P F Dullaart
- Department of Endocrinology, University of Groningen and University Medical Centre Groningen, Groningen, The Netherlands.
| | | | | | | | | |
Collapse
|
40
|
Liu R, Iqbal J, Yeang C, Wang DQH, Hussain MM, Jiang XC. Phospholipid Transfer Protein–Deficient Mice Absorb Less Cholesterol. Arterioscler Thromb Vasc Biol 2007; 27:2014-21. [PMID: 17641249 DOI: 10.1161/atvbaha.107.149914] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Phospholipid transfer protein (PLTP) plays an important role in lipoprotein metabolism and atherosclerosis. PLTP gene knockout (KO) mice show significant reduction of plasma cholesterol levels. Because small intestine is one of the major tissue expressing PLTP, we hypothesize that PLTP deficient small intestine absorbs less cholesterol, thus contributing to the diminishing of cholesterol levels in the plasma.
Methods and Results—
We used dual-labeled cholesterol/sitostanol feeding approach to study cholesterol absorption in PLTP KO and WT mice. We found that PLTP KO mice absorb significant less cholesterol than WT mice. Primary enterocytes isolated from PLTP KO enterocytes took up significant less cholesterol. Moreover, we observed that Niemann-Pick C1-like 1 (NPC1L1) mRNA levels were significantly decreased in the small intestine of PLTP KO mice. Next, we studied the secretion of cholesterol by enterocytes. The amounts of cholesterol transported to plasma and liver were significantly reduced in PLTP KO mice, compared with WT animals. Studies with isolated PLTP KO enterocytes revealed that the secretion of cholesterol via chylomicron and intestinal-HDL was significantly reduced. Furthermore, ATP-binding cassette transporters (ABC) A1 mRNA and microsomal triglyceride transfer protein (MTP) activity levels were significantly decreased in PLTP KO small intestine.
Conclusion—
These results indicate that PLTP deficiency results in reduced cholesterol uptake as well as secretion by the intestine. We suggest that PLTP could be a useful target to lower plasma cholesterol levels, thus reducing atherosclerosis.
Collapse
Affiliation(s)
- Ruijie Liu
- Department of Anatomy and Cell Biology, SUNY Downstate Medical Center, 450 Clarkson Ave, Box 5, Brooklyn, NY 11203, USA
| | | | | | | | | | | |
Collapse
|
41
|
Dallinga-Thie GM, Dullaart RPF, van Tol A. Concerted actions of cholesteryl ester transfer protein and phospholipid transfer protein in type 2 diabetes: effects of apolipoproteins. Curr Opin Lipidol 2007; 18:251-7. [PMID: 17495597 DOI: 10.1097/mol.0b013e3280e12685] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Type 2 diabetes frequently coincides with dyslipidemia, characterized by elevated plasma triglycerides, low high-density lipoprotein cholesterol levels and the presence of small dense low-density lipoprotein particles. Plasma lipid transfer proteins play an essential role in lipoprotein metabolism. It is thus vital to understand their pathophysiology and determine which factors influence their functioning in type 2 diabetes. RECENT FINDINGS Cholesteryl ester transfer protein-mediated transfer is increased in diabetic patients and contributes to low plasma high-density lipoprotein cholesterol levels. Apolipoproteins A-I, A-II and E are components of the donor lipoprotein particles that participate in the transfer of cholesteryl esters from high-density lipoprotein to apolipoprotein B-containing lipoproteins. Current evidence for functional roles of apolipoproteins C-I, F and A-IV as modulators of cholesteryl ester transfer is discussed. Phospholipid transfer protein activity is increased in diabetic patients and may contribute to hepatic very low-density lipoprotein synthesis and secretion and vitamin E transfer. Apolipoprotein E could stimulate the phospholipid transfer protein-mediated transfer of surface fragments of triglyceride-rich lipoproteins to high-density lipoprotein, and promote high-density lipoprotein remodelling. SUMMARY Both phospholipid and cholesteryl ester transfer proteins are important in very low and high-density lipoprotein metabolism and display concerted actions in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Geesje M Dallinga-Thie
- Department of Vascular Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | | | | |
Collapse
|
42
|
Moerland M, Anghelescu N, Samyn H, van Haperen R, van Gent T, Strouboulis J, van Tol A, Grosveld F, de Crom R. Inducible expression of phospholipid transfer protein (PLTP) in transgenic mice: acute effects of PLTP on lipoprotein metabolism. Transgenic Res 2007; 16:503-13. [PMID: 17437182 DOI: 10.1007/s11248-007-9094-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Accepted: 03/19/2007] [Indexed: 10/23/2022]
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.
Collapse
Affiliation(s)
- Matthijs Moerland
- Department of Cell Biology and Genetics, Erasmus University Medical Center, 3000 CA, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Vikstedt R, Ye D, Metso J, Hildebrand RB, Van Berkel TJC, Ehnholm C, Jauhiainen M, Van Eck M. Macrophage Phospholipid Transfer Protein Contributes Significantly to Total Plasma Phospholipid Transfer Activity and Its Deficiency Leads to Diminished Atherosclerotic Lesion Development. Arterioscler Thromb Vasc Biol 2007; 27:578-86. [PMID: 17170377 DOI: 10.1161/01.atv.0000254815.49414.be] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Objective—
Systemic phospholipid transfer protein (PLTP) deficiency in mice is associated with a decreased susceptibility to atherosclerosis, whereas overexpression of human PLTP in mice increases atherosclerotic lesion development. PLTP is also expressed by macrophage-derived foam cells in human atherosclerotic lesions, but the exact role of macrophage PLTP in atherosclerosis is unknown.
Methods and Results—
To clarify the role of macrophage PLTP in atherogenesis, PLTP was selectively disrupted in hematopoietic cells, including macrophages, by transplantation of bone marrow from PLTP knockout (PLTP
−/−
) mice into irradiated low-density lipoprotein receptor knockout mice. Selective deficiency of macrophage PLTP (PLTP
−M/−M
) resulted in a 29% (
P
<0.01 for difference in lesion area) reduction in aortic root lesion area as compared with mice possessing functional macrophage PLTP (384±36*10
3
μm
2
in the PLTP
−M/−M
group (n=10), as compared with 539±35*10
3
μm
2
in the PLTP
+M/+M
group (n=14)) after 9 weeks of Western-type diet feeding. The decreased lesion size in the PLTP
−M/−M
group coincided with significantly lower serum total cholesterol, free cholesterol, and triglyceride levels in these mice. Furthermore, plasma PLTP activity in the PLTP
−M/−M
group was 2-fold (
P
<0.001) lower than that in the PLTP
+M/+M
group.
Conclusion—
Macrophage PLTP is a significant contributor to plasma PLTP activity and deficiency of PLTP in macrophages leads to lowered atherosclerotic lesion development in low-density lipoprotein receptor knockout mice on Western-type diet.
Collapse
Affiliation(s)
- Riikka Vikstedt
- National Public Health Institute, Department of Molecular Medicine, Biomedicum, Helsinki, Finland
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Lewis GF. Are the lipid-modifying effects of fibrates mediated by alterations in plasma lipid transfer activity? Clin Sci (Lond) 2007; 111:185-7. [PMID: 16761953 DOI: 10.1042/cs20060140] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Fenofibrate, a PPAR-alpha (peroxisome-proliferator-activated receptor-alpha) agonist, has been shown to modify plasma lipid and lipoprotein composition and metabolism by a variety of mechanisms. In addition, fenofibrate has been shown to increase the activity of PLTP (phospholipid transfer protein) and to reduce the activity of CETP (cholesteryl ester transfer protein). It is not known, however, whether the changes in PLTP and CETP plasma activity play an active role in the lipid changes observed with fenofibrate therapy, and this is investigated by Watts and co-workers in the present issue of Clinical Science.
Collapse
Affiliation(s)
- Gary F Lewis
- Toronto General Hospital, 200 Elizabeth Street, EN12-218, Toronto, ON, Canada M5G 2C4.
| |
Collapse
|
45
|
Siggins S, Bykov I, Hermansson M, Somerharju P, Lindros K, Miettinen TA, Jauhiainen M, Olkkonen VM, Ehnholm C. Altered hepatic lipid status and apolipoprotein A-I metabolism in mice lacking phospholipid transfer protein. Atherosclerosis 2007; 190:114-23. [PMID: 16554055 DOI: 10.1016/j.atherosclerosis.2006.02.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2005] [Revised: 02/10/2006] [Accepted: 02/15/2006] [Indexed: 11/19/2022]
Abstract
The effect of PLTP deficiency on hepatic lipid status and apolipoprotein A-I (apoA-I) biosynthesis in PLTP knockout (PLTP-KO) mice was investigated. PLTP-KO mice exhibited a marked reduction in HDL levels, but also increased triglycerides (TG), phospholipids (PL), and cholesterol in very-low-density lipoproteins (VLDL). Both male and female PLTP-KO mice displayed increased hepatic PL and decreased TG, and in the females, increased hepatic cholesterol was also detected. Primary hepatocytes from PLTP-KO mice displayed a different PL molecular species composition to the wild type (WT) controls, with prominent changes being a reduction of long chain fatty acid-containing and an increase of medium chain mono- or di-unsaturated fatty acid containing PL species. Cultured PLTP-KO hepatocytes synthesized and secreted apoA-I in similar quantities as the WT cells. However, the apoA-I secreted by PLTP-KO hepatocytes contained less choline PL, differing also in phosphatidylcholine/sphingomyelin ratio and fatty acyl species composition when compared to apoA-I from WT hepatocytes. Furthermore, the PLTP-KO-derived PL-deficient apoA-I was less stable in the hepatocyte culture medium than that produced by WT cells. These results demonstrate a complex regulatory role of PLTP in serum and liver lipid homeostasis, as well as in the formation of nascent apoA-I-PL complexes from the liver.
Collapse
Affiliation(s)
- Sarah Siggins
- Department of Molecular Medicine, National Public Health Institute, Biomedicum, P.O. Box 104, FI-00251 Helsinki, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Dullaart RPF, de Vries R, Dallinga-Thie GM, van Tol A, Sluiter WJ. Plasma cholesteryl ester transfer protein mass and phospholipid transfer protein activity are associated with leptin in type 2 diabetes mellitus. Biochim Biophys Acta Mol Cell Biol Lipids 2007; 1771:113-8. [PMID: 17185032 DOI: 10.1016/j.bbalip.2006.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2006] [Revised: 11/02/2006] [Accepted: 11/10/2006] [Indexed: 10/23/2022]
Abstract
Adipose tissue contributes to plasma levels of lipid transfer proteins and is also the major source of plasma adipokines. We hypothesized that plasma cholesteryl ester transfer protein (CETP) mass, phospholipid transfer protein (PLTP) activity and cholesteryl ester transfer (CET, a measure of CETP action) are determined by adipokine levels. In this study, relationships of plasma CETP mass, PLTP activity and CET with leptin, resistin and adiponectin were analyzed in type 2 diabetic patients and control subjects. Plasma PLTP activity (P<0.001), CET (P<0.001), leptin (P=0.003), resistin (P<0.001), high sensitive C-reactive protein (P=0.005), and insulin resistance (HOMA(ir)) (P<0.001) were higher, whereas HDL cholesterol (P<0.001) and plasma adiponectin (P<0.001) were lower in 83 type 2 diabetic patients (32 females) than in 83 sex-matched control subjects. Multiple linear regression analysis demonstrated that in diabetic patients plasma leptin levels were related to plasma CETP mass (P=0.018) and PLTP activity (P<0.001), but not to the other adipokines measured. Plasma CET was inversely correlated with adiponectin in univariate analysis, but this association disappeared in multivariate models that included plasma lipids and CETP. In conclusion, both plasma CETP mass and PLTP activity are associated with plasma leptin in type 2 diabetes. The elevated CET in these patients is not independently related to any of the measured plasma adipokines.
Collapse
Affiliation(s)
- R P F Dullaart
- Department of Endocrinology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | | | | | | | | |
Collapse
|
47
|
Liu R, Hojjati MR, Devlin CM, Hansen IH, Jiang XC. Macrophage phospholipid transfer protein deficiency and ApoE secretion: impact on mouse plasma cholesterol levels and atherosclerosis. Arterioscler Thromb Vasc Biol 2006; 27:190-6. [PMID: 17038631 DOI: 10.1161/01.atv.0000249721.96666.e5] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE PLTP and apoE play important roles in lipoprotein metabolism and atherosclerosis. It is known that formation of macrophage-derived foam cells (which highly express PLTP and apoE) is the critical step in the process of atherosclerosis. We investigated the relationship between PLTP and apoE in macrophages and the atherogenic relevance in a mouse model. METHODS AND RESULTS We transplanted PLTP-deficient mouse bone marrow into apoE-deficient mice (PLTP-/- --> apoE-/-), creating a mouse model with PLTP deficiency and apoE expression exclusively in the macrophages. We found that PLTP-/- --> apoE-/- mice have significantly lower PLTP activity, compared with controls (WT --> apoE-/-; 20%, P<0.01). On a Western diet, PLTP-/- --> apoE-/- mice have significantly lower plasma apoE than that of WT --> apoE-/- mice (63%, P<0.001), and PLTP-deficient macrophages secrete significantly less apoE than WT macrophages (44%, P<0.01). Moreover, PLTP-/- --> apoE-/- mice have significantly higher plasma cholesterol (98%, P<0.001) and phospholipid (107%, P<0.001) than that of WT --> apoE-/- mice, thus increasing atherosclerotic lesions in the aortic arch and root (403%, P<0.001), as well as the entire aorta (298%, P<0.001). CONCLUSIONS Macrophage PLTP deficiency causes a significant reduction of apoE secretion from the cells, and this in turn promotes the accumulation of cholesterol in the circulation and accelerates the development of atherosclerosis.
Collapse
Affiliation(s)
- Ruijie Liu
- Department of Anatomy and Cell Biology, SUNY Downstate Medical Center, 450 Clarkson Ave, Brooklyn, NY 11203, USA
| | | | | | | | | |
Collapse
|
48
|
Watts GF, Ji J, Chan DC, Ooi EMM, Johnson AG, Rye KA, Barrett PHR. Relationships between changes in plasma lipid transfer proteins and apolipoprotein B-100 kinetics during fenofibrate treatment in the metabolic syndrome. Clin Sci (Lond) 2006; 111:193-9. [PMID: 16700661 DOI: 10.1042/cs20060072] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The aim of the present study was to investigate the association between changes in apoB (apolipoprotein B-100) kinetics and plasma PLTP (phospholipid transfer protein) and CETP (cholesteryl ester transfer protein) activities in men with MetS (the metabolic syndrome) treated with fenofibrate. Eleven men with MetS underwent a double-blind cross-over treatment with fenofibrate (200 mg/day) or placebo for 5 weeks. Compared with placebo, fenofibrate significantly increased the FCRs (fractional catabolic rates) of apoB in VLDL (very-low-density lipoprotein), IDL (intermediate-density lipoprotein) and LDL (low-density lipoprotein) (all P<0.01), with no significant reduction (−8%; P=0.131) in VLDL-apoB PR (production rate), but an almost significant increase (+15%, P=0.061) in LDL-apoB PR. Fenofibrate significantly lowered plasma TG [triacylglycerol (triglyceride); P<0.001], the VLDL-TG/apoB ratio (P=0.003) and CETP activity (P=0.004), but increased plasma HDL (high-density lipoprotein)-cholesterol concentration (P<0.001) and PLTP activity (P=0.03). The increase in PLTP activity was positively associated with the increase in both LDL-apoB FCR (r=0.641, P=0.034) and PR (r=0.625, P=0.040), and this was independent of the fall in plasma CETP activity and lathosterol level. The decrease in CETP activity was positively associated with the decrease in VLDL-apoB PR (r=0.615, P=0.044), but this association was not robust and not independent of changes in PLTP activity and lathosterol levels. Hence, in MetS, the effects of fenofibrate on plasma lipid transfer protein activities, especially PLTP activity, may partially explain the associated changes in apoB kinetics.
Collapse
Affiliation(s)
- Gerald F Watts
- Lipoprotein Research Unit, School of Medicine and Pharmacology, University of Western Australia, Perth, WA 6000 Australia.
| | | | | | | | | | | | | |
Collapse
|
49
|
Lie J, Moerland M, van Gent T, van Haperen R, Scheek L, Sadeghi-Niaraki F, de Crom R, van Tol A. Sex differences in atherosclerosis in mice with elevated phospholipid transfer protein activity are related to decreased plasma high density lipoproteins and not to increased production of triglycerides. Biochim Biophys Acta Mol Cell Biol Lipids 2006; 1761:1070-7. [PMID: 16935026 DOI: 10.1016/j.bbalip.2006.06.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2006] [Revised: 06/14/2006] [Accepted: 06/27/2006] [Indexed: 10/24/2022]
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.
Collapse
Affiliation(s)
- Jessica Lie
- Department of Biochemistry, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Valenta DT, Ogier N, Bradshaw G, Black AS, Bonnet DJ, Lagrost L, Curtiss LK, Desrumaux CM. Atheroprotective Potential of Macrophage-Derived Phospholipid Transfer Protein in Low-Density Lipoprotein Receptor-Deficient Mice Is Overcome by Apolipoprotein AI Overexpression. Arterioscler Thromb Vasc Biol 2006; 26:1572-8. [PMID: 16675720 DOI: 10.1161/01.atv.0000225700.43836.ae] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective—
Using bone marrow transplantation, we assessed the impact of macrophage-derived phospholipid transfer protein (PLTP) on lesion development in hypercholesterolemic mice that expressed either normal levels of mouse apolipoprotein AI (apoAI) or elevated levels of only human apoAI.
Methods and Results—
Bone marrow transplantations were performed in low-density lipoprotein receptor-deficient mice (LDLr−/−) that expressed either normal levels of mouse apoAI (
ms
apoAI) or high levels of only human apoAI (
ms
apoAI−/−, LDLr−/−,
hu
apoAITg). Mice were lethally irradiated, reconstituted with either PLTP-expressing or PLTP-deficient bone marrow cells, and fed a high-fat diet over 16 weeks. Macrophage PLTP deficiency increased atherosclerosis in LDLr−/− mice with minimal changes in total plasma cholesterol levels. In contrast, the extent of atherosclerosis in
ms
apoAI−/−, LDLr−/−,
hu
apoAITg mice was not significantly different between groups that had received PLTP−/− or PLTP+/+ bone marrow. In vitro studies indicated that PLTP deficiency led to a significant decrease in α-tocopherol content and increased oxidative stress in bone marrow cells.
Conclusions—
Our observations suggest an atheroprotective role of macrophage-derived PLTP in mice with normal apoAI plasma levels. The atheroprotective properties of macrophage-derived PLTP were not observable in the presence of elevated plasma concentrations of apoAI.
Collapse
Affiliation(s)
- David T Valenta
- Department of Immunology, The Scripps Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | |
Collapse
|