1
|
Andersen CJ, Fernandez ML. Emerging Biomarkers and Determinants of Lipoprotein Profiles to Predict CVD Risk: Implications for Precision Nutrition. Nutrients 2024; 17:42. [PMID: 39796476 PMCID: PMC11722654 DOI: 10.3390/nu17010042] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 12/23/2024] [Accepted: 12/24/2024] [Indexed: 01/13/2025] Open
Abstract
Biomarkers constitute a valuable tool to diagnose both the incidence and the prevalence of chronic diseases and may help to inform the design and effectiveness of precision nutrition interventions. Cardiovascular disease (CVD) continues to be the foremost cause of death all over the world. While the reasons that lead to increased risk for CVD are multifactorial, dyslipidemias, plasma concentrations of specific lipoproteins, and dynamic measures of lipoprotein function are strong biomarkers to predict and document coronary heart disease incidence. The aim of this review is to provide a comprehensive evaluation of the biomarkers and emerging approaches that can be utilized to characterize lipoprotein profiles as predictive tools for assessing CVD risk, including the assessment of traditional clinical lipid panels, measures of lipoprotein efflux capacity and inflammatory and antioxidant activity, and omics-based characterization of lipoprotein composition and regulators of lipoprotein metabolism. In addition, we discuss demographic, genetic, metagenomic, and lifestyle determinants of lipoprotein profiles-such as age, sex, gene variants and single-nucleotide polymorphisms, gut microbiome profiles, dietary patterns, physical inactivity, obesity status, smoking and alcohol intake, and stress-which are likely to be essential factors to explain interindividual responses to precision nutrition recommendations to mitigate CVD risk.
Collapse
Affiliation(s)
- Catherine J. Andersen
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA;
| | - Maria Luz Fernandez
- Department of Nutritional Sciences, University of Connecticut, Storrs, CT 06269, USA;
- School of Nutrition and Wellness, University of Arizona, Tucson, AZ 85712, USA
| |
Collapse
|
2
|
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
|
3
|
Pan X. Cholesterol Metabolism in Chronic Kidney Disease: Physiology, Pathologic Mechanisms, and Treatment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1372:119-143. [PMID: 35503178 PMCID: PMC11106795 DOI: 10.1007/978-981-19-0394-6_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
High plasma levels of lipids and/or lipoproteins are risk factors for atherosclerosis, nonalcoholic fatty liver disease (NAFLD), obesity, and diabetes. These four conditions have also been identified as risk factors leading to the development of chronic kidney disease (CKD). Although many pathways that generate high plasma levels of these factors have been identified, most clinical and physiologic dysfunction results from aberrant assembly and secretion of lipoproteins. The results of several published studies suggest that elevated levels of low-density lipoprotein (LDL)-cholesterol are a risk factor for atherosclerosis, myocardial infarction, coronary artery calcification associated with type 2 diabetes, and NAFLD. Cholesterol metabolism has also been identified as an important pathway contributing to the development of CKD; clinical treatments designed to alter various steps of the cholesterol synthesis and metabolism pathway are currently under study. Cholesterol synthesis and catabolism contribute to a multistep process with pathways that are regulated at the cellular level in renal tissue. Cholesterol metabolism may also be regulated by the balance between the influx and efflux of cholesterol molecules that are capable of crossing the membrane of renal proximal tubular epithelial cells and podocytes. Cellular accumulation of cholesterol can result in lipotoxicity and ultimately kidney dysfunction and failure. Thus, further research focused on cholesterol metabolism pathways will be necessary to improve our understanding of the impact of cholesterol restriction, which is currently a primary intervention recommended for patients with dyslipidemia.
Collapse
Affiliation(s)
- Xiaoyue Pan
- Department of Foundations of Medicine, New York University Long Island School of Medicine, Mineola, NY, USA.
| |
Collapse
|
4
|
Zhao Q, Wang J, Miao Z, Zhang NR, Hennessy S, Small DS, Rader DJ. A Mendelian randomization study of the role of lipoprotein subfractions in coronary artery disease. eLife 2021; 10:e58361. [PMID: 33899735 PMCID: PMC8163505 DOI: 10.7554/elife.58361] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 04/23/2021] [Indexed: 12/26/2022] Open
Abstract
Recent genetic data can offer important insights into the roles of lipoprotein subfractions and particle sizes in preventing coronary artery disease (CAD), as previous observational studies have often reported conflicting results. We used the LD score regression to estimate the genetic correlation of 77 subfraction traits with traditional lipid profile and identified 27 traits that may represent distinct genetic mechanisms. We then used Mendelian randomization (MR) to estimate the causal effect of these traits on the risk of CAD. In univariable MR, the concentration and content of medium high-density lipoprotein (HDL) particles showed a protective effect against CAD. The effect was not attenuated in multivariable analyses. Multivariable MR analyses also found that small HDL particles and smaller mean HDL particle diameter may have a protective effect. We identified four genetic markers for HDL particle size and CAD. Further investigations are needed to fully understand the role of HDL particle size.
Collapse
Affiliation(s)
- Qingyuan Zhao
- Statistical Laboratory, University of CambridgeCambridgeUnited Kingdom
| | - Jingshu Wang
- Department of Statistics, University of ChicagoChicagoUnited States
| | - Zhen Miao
- Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Nancy R Zhang
- Department of Statistics, University of PennsylvaniaPhiladelphiaUnited States
| | - Sean Hennessy
- Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Dylan S Small
- Department of Statistics, University of PennsylvaniaPhiladelphiaUnited States
| | - Daniel J Rader
- Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
5
|
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
|
6
|
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
|
7
|
Zhao XM, Wang Y, Yu Y, Jiang H, Babinska A, Chen XY, He KG, Min XD, Han JJ, Yang CX, Deng K, Xue J, Zhang X, Song GH, Qin SC, Jiang XC. Plasma Phospholipid Transfer Protein Promotes Platelet Aggregation. Thromb Haemost 2018; 118:2086-2097. [PMID: 30419596 DOI: 10.1055/s-0038-1675228] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
It remains unclear whether plasma phospholipid transfer protein (PLTP) is involved in hyper-coagulation or hypo-coagulation. This study investigated the direct effect of PLTP on platelet aggregation and the underlying mechanism. Washed platelets from humans or mice and mouse platelet-rich plasma and human recombinant PLTP were isolated. PLTP is present in human platelets. We assessed adenosine diphosphate (ADP)-, collagen- and thrombin-induced platelet aggregation, phosphatidylserine externalization and photothrombosis-induced cerebral infarction in mice. PLTP over-expression increased platelet aggregation, while PLTP deficiency had the opposing reaction. Human recombinant PLTP increased both mouse and human platelet aggregation in a dose-dependent manner. Phosphatidylserine externalization provides a water/lipid surface for the interaction of coagulation factors, which accelerates thrombosis. Compared with wild-type controls, platelets from PLTP transgenic mice had significantly more phosphatidylserine on the exterior surface of the plasma membrane, whereas platelets from PLTP-deficient mice had significantly less phosphatidylserine on the surface, thus PLTP influences fibrinogen binding on the plasma membrane. Moreover, recombinant PLTP together with ADP significantly increased phosphatidylserine exposure on the plasma membrane of PLTP-deficient platelets, thereby increasing fibrinogen binding. PLTP over-expression significantly accelerated the incidence of photothrombosis-induced infarction in mice, whereas PLTP deficiency significantly reduced the frequency of infarction. We concluded that PLTP promotes phosphatidylserine externalization at the plasma membrane of platelets and accelerates ADP- or collagen-induced platelet aggregation. This effect plays an important role in the initiation of thrombin generation and platelet aggregation under sheer stress conditions. Thus, PLTP is involved in hyper-coagulation. Therefore, PLTP inhibition could be a novel approach for countering thrombosis.
Collapse
Affiliation(s)
- Xiao-Min Zhao
- The Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Yun Wang
- The Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Yang Yu
- The Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Hui Jiang
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York, United States
| | - Anna Babinska
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York, United States
| | - Xiu-Yu Chen
- The Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Ke-Gui He
- The Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Xiang-Dong Min
- The Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Ji-Ju Han
- The Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Chen-Xi Yang
- The Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Kevin Deng
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York, United States
| | - Jing Xue
- Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, China
| | - Xiangjian Zhang
- Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Hebei Key Laboratory of Vascular Homeostasis, Shijiazhuang, China
| | - Guo-Hua Song
- The Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Shu-Cun Qin
- The Institute of Atherosclerosis, Taishan Medical University, Taian, China
| | - Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, New York, United States.,Molecular and Cellular Cardiology Program, VA New York Harbor Healthcare System, Brooklyn, New York, United States
| |
Collapse
|
8
|
Phospholipid transfer protein and alpha-1 antitrypsin regulate Hck kinase activity during neutrophil degranulation. Sci Rep 2018; 8:15394. [PMID: 30337619 PMCID: PMC6193999 DOI: 10.1038/s41598-018-33851-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 10/07/2018] [Indexed: 01/21/2023] Open
Abstract
Excessive neutrophil degranulation is a common feature of many inflammatory disorders, including alpha-1 antitrypsin (AAT) deficiency. Our group has demonstrated that phospholipid transfer protein (PLTP) prevents neutrophil degranulation but serine proteases, which AAT inhibits, cleave PLTP in diseased airways. We propose to identify if airway PLTP activity can be restored by AAT augmentation therapy and how PLTP subdues degranulation of neutrophils in AAT deficient subjects. Airway PLTP activity was lower in AAT deficient patients but elevated in the airways of patients on augmentation therapy. Functional AAT protein (from PiMM homozygotes) prevented PLTP cleavage unlike its mutated ZZ variant (PiZZ). PLTP lowered leukotriene B4 induced degranulation of primary, secondary and tertiary granules from neutrophils from both groups (n = 14/group). Neutrophils isolated from Pltp knockout mice have enhance neutrophil degranulation. Both AAT and PLTP reduced neutrophil degranulation and superoxide production, possibly though their inhibition of the Src tyrosine kinase, Hck. Src kinase inhibitors saracatinib and dasatinib reduced neutrophil degranulation and superoxide production. Therefore, AAT protects PLTP from proteolytic cleavage and both AAT and PLTP mediate degranulation, possibly via Hck tyrosine kinase inhibition. Deficiency of AAT could contribute to reduced lung PLTP activity and elevated neutrophil signaling associated with lung disease.
Collapse
|
9
|
Zhang M, Zhai X, Li J, Albers JJ, Vuletic S, Ren G. Structural basis of the lipid transfer mechanism of phospholipid transfer protein (PLTP). Biochim Biophys Acta Mol Cell Biol Lipids 2018; 1863:1082-1094. [PMID: 29883800 PMCID: PMC6114099 DOI: 10.1016/j.bbalip.2018.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 05/10/2018] [Accepted: 06/01/2018] [Indexed: 12/14/2022]
Abstract
Human phospholipid transfer protein (PLTP) mediates the transfer of phospholipids among atheroprotective high-density lipoproteins (HDL) and atherogenic low-density lipoproteins (LDL) by an unknown mechanism. Delineating this mechanism would represent the first step towards understanding PLTP-mediated lipid transfers, which may be important for treating lipoprotein abnormalities and cardiovascular disease. Here, using various electron microscopy techniques, PLTP is revealed to have a banana-shaped structure similar to cholesteryl ester transfer protein (CETP). We provide evidence that PLTP penetrates into the HDL and LDL surfaces, respectively, and then forms a ternary complex with HDL and LDL. Insights into the interaction of PLTP with lipoproteins at the molecular level provide a basis to understand the PLTP-dependent lipid transfer mechanisms for dyslipidemia treatment.
Collapse
Affiliation(s)
- Meng Zhang
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States
| | - Xiaobo Zhai
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States
| | - Jinping Li
- Department of Biomedical Science, Mercer University School of Medicine, Savannah, GA 31404, United States
| | - John J Albers
- Northwest Lipid Metabolism and Diabetes Research Laboratories, Seattle, WA 98109, United States
| | - Simona Vuletic
- Northwest Lipid Metabolism and Diabetes Research Laboratories, Seattle, WA 98109, United States.
| | - Gang Ren
- The Molecular Foundry, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, United States.
| |
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
|
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
|
12
|
Deguchi H, Wolfbauer G, Cheung MC, Banerjee Y, Elias DJ, Fernández JA, Albers JJ, Griffin JH. Inhibition of thrombin generation in human plasma by phospholipid transfer protein. Thromb J 2015; 13:24. [PMID: 26185485 PMCID: PMC4504036 DOI: 10.1186/s12959-015-0054-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 05/20/2015] [Indexed: 01/10/2023] Open
Abstract
Background Plasma phospholipid transfer protein (PLTP) transfers lipids between donors and acceptors (e.g., from HDL to VLDL) and modulates lipoprotein composition, size, and levels. No study has reported an assessment of the effects of PLTP on blood clotting reactions, such as reflected in thrombin generation assays, or on the association of venous thrombosis (VTE) risk with PLTP activity. Methods The in vitro effects of PLTP on blood coagulation reactions and the correlations between plasma PLTP activity levels and VTE were studied. Results Recombinant (r) PLTP concentration-dependently inhibited plasma thrombin generation and factor XII-dependent kallikrein generation when sulfatide was used to stimulate factor XII autoactivation in plasma. However, rPLTP did not inhibit thrombin generation in plasma induced by factor XIa or tissue factor, implicating an effect of PLTP on contact activation reactions. In purified systems, rPLTP inhibited factor XII autoactivation stimulated by sulfatide in the presence of VLDL. In surface plasmon resonance studies, purified factor XII bound to immobilized rPLTP, implying that rPLTP inhibits factor XII-dependent contact activation by binding factor XII in the presence of lipoproteins. Analysis of plasmas from 40 male patients with unprovoked VTE and 40 matched controls indicated that low PLTP lipid transfer activity (≤25th percentile) was associated with an increased risk of VTE after adjustment for body mass index, plasma lipids, and two known thrombophilic genetic risk factors. Conclusion These data imply that PLTP may be an antithrombotic plasma protein by inhibiting generation of prothrombotic factor XIIa in the presence of VLDL. This newly discovered anticoagulant activity of PLTP merits further clinical and biochemical studies. Electronic supplementary material The online version of this article (doi:10.1186/s12959-015-0054-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Hiroshi Deguchi
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, MEM180, 10550 North Torrey Pines Rd., La Jolla, CA 92037 USA
| | - Gertrud Wolfbauer
- Division of Metabolism, Endocrinology, and Nutrition, Northwest Lipid Metabolism and Diabetes Research Laboratories, Department of Medicine, University of Washington, Seattle, WA 98109 USA
| | - Marian C Cheung
- Division of Metabolism, Endocrinology, and Nutrition, Northwest Lipid Metabolism and Diabetes Research Laboratories, Department of Medicine, University of Washington, Seattle, WA 98109 USA
| | - Yajnavalka Banerjee
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, MEM180, 10550 North Torrey Pines Rd., La Jolla, CA 92037 USA ; Current Address: Department of Biochemistry, College of Medicine and Health Sciences, SQ University, Muscat, Oman
| | - Darlene J Elias
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, MEM180, 10550 North Torrey Pines Rd., La Jolla, CA 92037 USA
| | - José A Fernández
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, MEM180, 10550 North Torrey Pines Rd., La Jolla, CA 92037 USA
| | - John J Albers
- Division of Metabolism, Endocrinology, and Nutrition, Northwest Lipid Metabolism and Diabetes Research Laboratories, Department of Medicine, University of Washington, Seattle, WA 98109 USA
| | - John H Griffin
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, MEM180, 10550 North Torrey Pines Rd., La Jolla, CA 92037 USA
| |
Collapse
|
13
|
Kim DS, Burt AA, Ranchalis JE, Vuletic S, Vaisar T, Li WF, Rosenthal EA, Dong W, Eintracht JF, Motulsky AG, Brunzell JD, Albers JJ, Furlong CE, Jarvik GP. PLTP activity inversely correlates with CAAD: effects of PON1 enzyme activity and genetic variants on PLTP activity. J Lipid Res 2015; 56:1351-62. [PMID: 26009633 DOI: 10.1194/jlr.p058032] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Indexed: 01/07/2023] Open
Abstract
Recent studies have failed to demonstrate a causal cardioprotective effect of HDL cholesterol levels, shifting focus to the functional aspects of HDL. Phospholipid transfer protein (PLTP) is an HDL-associated protein involved in reverse cholesterol transport. This study sought to determine the genetic and nongenetic predictors of plasma PLTP activity (PLTPa), and separately, to determine whether PLTPa predicted carotid artery disease (CAAD). PLTPa was measured in 1,115 European ancestry participants from a case-control study of CAAD. A multivariate logistic regression model was used to elucidate the relationship between PLTPa and CAAD. Separately, a stepwise linear regression determined the nongenetic clinical and laboratory characteristics that best predicted PLTPa. A final stepwise regression considering both nongenetic and genetic variables identified the combination of covariates that explained maximal PLTPa variance. PLTPa was significantly associated with CAAD (7.90 × 10(-9)), with a 9% decrease in odds of CAAD per 1 unit increase in PLTPa (odds ratio = 0.91). Triglyceride levels (P = 0.0042), diabetes (P = 7.28 × 10(-5)), paraoxonase 1 (PON1) activity (P = 0.019), statin use (P = 0.026), PLTP SNP rs4810479 (P = 6.38 × 10(-7)), and PCIF1 SNP rs181914932 (P = 0.041) were all significantly associated with PLTPa. PLTPa is significantly inversely correlated with CAAD. Furthermore, we report a novel association between PLTPa and PON1 activity, a known predictor of CAAD.
Collapse
Affiliation(s)
- Daniel Seung Kim
- Division of Medical Genetics, Department of Medicine, University of Washington School of Medicine, Seattle, WA Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA Department of Biostatistics, University of Washington School of Public Health, Seattle, WA
| | - Amber A Burt
- Division of Medical Genetics, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Jane E Ranchalis
- Division of Medical Genetics, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Simona Vuletic
- Northwest Lipid Metabolism and Diabetes Research Laboratories, Seattle, WA Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Tomas Vaisar
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Wan-Fen Li
- Division of Medical Genetics, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Elisabeth A Rosenthal
- Division of Medical Genetics, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Weijiang Dong
- Northwest Lipid Metabolism and Diabetes Research Laboratories, Seattle, WA Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA Department of Human Anatomy and Histology and Embryology, Xi'an Jiaotong University School of Medicine, Xi'an 710061, People's Republic of China
| | - Jason F Eintracht
- Department of General Medicine, Virginia Mason Medical Center, Seattle, WA
| | - Arno G Motulsky
- Division of Medical Genetics, Department of Medicine, University of Washington School of Medicine, Seattle, WA Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA
| | - John D Brunzell
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - John J Albers
- Northwest Lipid Metabolism and Diabetes Research Laboratories, Seattle, WA Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, University of Washington School of Medicine, Seattle, WA
| | - Clement E Furlong
- Division of Medical Genetics, Department of Medicine, University of Washington School of Medicine, Seattle, WA Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA
| | - Gail P Jarvik
- Division of Medical Genetics, Department of Medicine, University of Washington School of Medicine, Seattle, WA Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA
| |
Collapse
|
14
|
Elevated baseline plasma phospholipid protein (PLTP) levels are an independent predictor of long-term all-cause mortality in patients with diabetes mellitus and known or suspected coronary artery disease. Atherosclerosis 2015; 239:503-8. [PMID: 25710294 DOI: 10.1016/j.atherosclerosis.2015.02.017] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 01/11/2015] [Accepted: 02/08/2015] [Indexed: 12/13/2022]
Abstract
OBJECTIVES To investigate the long-term prognostic significance of baseline plasma PLTP levels in a group of well-characterized male patients with diabetes mellitus and known or suspected coronary artery disease referred for coronary angiography. BACKGROUND PLTP is a plasma protein that mediates the net transfer and exchange of phospholipids between lipoproteins. It has been implicated in the pathogenesis of atherosclerosis and elevated plasma levels have been reported in patients with diabetes mellitus. METHODS Baseline plasma PLTP levels were measured in 154 male patients with diabetes mellitus who were referred for coronary angiography and followed prospectively for 5 years for the development of all-cause mortality. RESULTS After adjustment for a variety of baseline clinical, angiographic and laboratory parameters, plasma PLTP levels (analyzed as a continuous variable) were an independent predictor of all-cause mortality at 5 years (HR, 1.55; 95% CI, 1.22-2.00; P = 0.0009). Furthermore, in 3 additional multivariate models that also included a wide variety of contemporary biomarkers with established prognostic efficacy (i.e., ST2, GDF-15, Cystatin C, Fibrinogen, and NT-proBNP), PLTP remained an independent predictor of all-cause mortality at 5 years. CONCLUSIONS Elevated baseline plasma levels of PLTP are associated with an increased risk of long-term all-cause mortality in patients with diabetes and known or suspected coronary disease. Furthermore, this association is independent of a variety of clinical, angiographic, and laboratory variables, including a whole host of contemporary biomarkers with established prognostic efficacy.
Collapse
|
15
|
Zhang K, Liu X, Yu Y, Luo T, Wang L, Ge C, Liu X, Song J, Jiang X, Zhang Y, Qin S, Zhang M. Phospholipid transfer protein destabilizes mouse atherosclerotic plaque. Arterioscler Thromb Vasc Biol 2014; 34:2537-44. [PMID: 25324570 DOI: 10.1161/atvbaha.114.303966] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
OBJECTIVE Phospholipid transfer protein (PLTP) accelerates the development of atherosclerosis in mouse models. We examined the role of PLTP in atherosclerotic plaque stability. APPROACH AND RESULTS We prepared apolipoprotein E and PLTP double-knockout (PLTP(-/-)ApoE(-/-)) mice. PLTP deficiency significantly decreased lesion size and reduced monocyte/macrophage infiltration, as well as macrophage apoptosis in lesion areas. Moreover, it increased fibrous content in plaques, which suggests that PLTP may affect atherosclerotic plaque stability. Importantly, PLTP overexpression mediated by adenovirus had the reverse effect. It promoted the accumulation of reactive oxygen species in macrophages, which could lead to cell apoptosis and increased the production of inflammatory cytokines and chemokines. PLTP overexpression could promote receptor-interacting protein 3 recruitment of macrophages in cytoplasm, which could induce reactive oxygen species, thus inducing atherogenesis. CONCLUSIONS PLTP plays an important role in modulating the stability of atherosclerotic plaques. The receptor-interacting protein 3- reactive oxygen species signal pathway could be involved in this PLTP-mediated process.
Collapse
Affiliation(s)
- Ke Zhang
- From The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, People's Republic of China (K.Z., X.L., L.W., C.G., X.L., J.S., Y.Z., M.Z.); The Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, Shandong, People's Republic of China (Y.Y., T.L., S.Q.); and Department of Cell Biology, State University of New York, Downstate Medical Center, New York (X.J.)
| | - Xiaoling Liu
- From The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, People's Republic of China (K.Z., X.L., L.W., C.G., X.L., J.S., Y.Z., M.Z.); The Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, Shandong, People's Republic of China (Y.Y., T.L., S.Q.); and Department of Cell Biology, State University of New York, Downstate Medical Center, New York (X.J.)
| | - Yang Yu
- From The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, People's Republic of China (K.Z., X.L., L.W., C.G., X.L., J.S., Y.Z., M.Z.); The Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, Shandong, People's Republic of China (Y.Y., T.L., S.Q.); and Department of Cell Biology, State University of New York, Downstate Medical Center, New York (X.J.)
| | - Tian Luo
- From The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, People's Republic of China (K.Z., X.L., L.W., C.G., X.L., J.S., Y.Z., M.Z.); The Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, Shandong, People's Republic of China (Y.Y., T.L., S.Q.); and Department of Cell Biology, State University of New York, Downstate Medical Center, New York (X.J.)
| | - Lin Wang
- From The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, People's Republic of China (K.Z., X.L., L.W., C.G., X.L., J.S., Y.Z., M.Z.); The Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, Shandong, People's Republic of China (Y.Y., T.L., S.Q.); and Department of Cell Biology, State University of New York, Downstate Medical Center, New York (X.J.)
| | - Chen Ge
- From The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, People's Republic of China (K.Z., X.L., L.W., C.G., X.L., J.S., Y.Z., M.Z.); The Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, Shandong, People's Republic of China (Y.Y., T.L., S.Q.); and Department of Cell Biology, State University of New York, Downstate Medical Center, New York (X.J.)
| | - Xinxin Liu
- From The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, People's Republic of China (K.Z., X.L., L.W., C.G., X.L., J.S., Y.Z., M.Z.); The Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, Shandong, People's Republic of China (Y.Y., T.L., S.Q.); and Department of Cell Biology, State University of New York, Downstate Medical Center, New York (X.J.)
| | - Jiantao Song
- From The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, People's Republic of China (K.Z., X.L., L.W., C.G., X.L., J.S., Y.Z., M.Z.); The Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, Shandong, People's Republic of China (Y.Y., T.L., S.Q.); and Department of Cell Biology, State University of New York, Downstate Medical Center, New York (X.J.)
| | - Xiancheng Jiang
- From The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, People's Republic of China (K.Z., X.L., L.W., C.G., X.L., J.S., Y.Z., M.Z.); The Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, Shandong, People's Republic of China (Y.Y., T.L., S.Q.); and Department of Cell Biology, State University of New York, Downstate Medical Center, New York (X.J.)
| | - Yun Zhang
- From The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, People's Republic of China (K.Z., X.L., L.W., C.G., X.L., J.S., Y.Z., M.Z.); The Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, Shandong, People's Republic of China (Y.Y., T.L., S.Q.); and Department of Cell Biology, State University of New York, Downstate Medical Center, New York (X.J.)
| | - Shucun Qin
- From The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, People's Republic of China (K.Z., X.L., L.W., C.G., X.L., J.S., Y.Z., M.Z.); The Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, Shandong, People's Republic of China (Y.Y., T.L., S.Q.); and Department of Cell Biology, State University of New York, Downstate Medical Center, New York (X.J.)
| | - Mei Zhang
- From The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, People's Republic of China (K.Z., X.L., L.W., C.G., X.L., J.S., Y.Z., M.Z.); The Key Laboratory of Atherosclerosis in Universities of Shandong, Institute of Atherosclerosis, Taishan Medical University, Taian, Shandong, People's Republic of China (Y.Y., T.L., S.Q.); and Department of Cell Biology, State University of New York, Downstate Medical Center, New York (X.J.).
| |
Collapse
|
16
|
Brehm A, Geraghty P, Campos M, Garcia-Arcos I, Dabo AJ, Gaffney A, Eden E, Jiang XC, D'Armiento J, Foronjy R. Cathepsin G degradation of phospholipid transfer protein (PLTP) augments pulmonary inflammation. FASEB J 2014; 28:2318-31. [PMID: 24532668 DOI: 10.1096/fj.13-246843] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Phospholipid transfer protein (PLTP) regulates phospholipid transport in the circulation and is highly expressed within the lung epithelium, where it is secreted into the alveolar space. Since PLTP expression is increased in chronic obstructive pulmonary disease (COPD), this study aimed to determine how PLTP affects lung signaling and inflammation. Despite its increased expression, PLTP activity decreased by 80% in COPD bronchoalveolar lavage fluid (BALF) due to serine protease cleavage, primarily by cathepsin G. Likewise, PLTP BALF activity levels decreased by 20 and 40% in smoke-exposed mice and in the media of smoke-treated small airway epithelial (SAE) cells, respectively. To assess how PLTP affected inflammatory responses in a lung injury model, PLTP siRNA or recombinant protein was administered to the lungs of mice prior to LPS challenge. Silencing PLTP at baseline caused a 68% increase in inflammatory cell infiltration, a 120 and 340% increase in ERK and NF-κB activation, and increased MMP-9, IL1β, and IFN-γ levels after LPS treatment by 39, 140, and 190%, respectively. Conversely, PLTP protein administration countered these effects in this model. Thus, these findings establish a novel anti-inflammatory function of PLTP in the lung and suggest that proteolytic cleavage of PLTP by cathepsin G may enhance the injurious inflammatory responses that occur in COPD.
Collapse
Affiliation(s)
- Anthony Brehm
- 2Department of Medicine, St. Luke's Roosevelt, Mt. Sinai Health System, Antenucci Bldg., 432 West 58th St., Rm. 311, New York, NY 10019, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Loria P, Marchesini G, Nascimbeni F, Ballestri S, Maurantonio M, Carubbi F, Ratziu V, Lonardo A. Cardiovascular risk, lipidemic phenotype and steatosis. A comparative analysis of cirrhotic and non-cirrhotic liver disease due to varying etiology. Atherosclerosis 2014; 232:99-109. [PMID: 24401223 DOI: 10.1016/j.atherosclerosis.2013.10.030] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 10/23/2013] [Accepted: 10/24/2013] [Indexed: 02/08/2023]
Abstract
BACKGROUND Liver regulates lipid metabolism in health and disease states. Nevertheless, the entity of cardiovascular risk (CVR) resulting from dysregulation of lipid metabolism secondary to liver disease is poorly characterized. AIM AND METHODS To review, based on a PubMed literature search, the features and the determinants of serum lipid phenotype and its correlation with hepatic steatosis, insulin resistance (IR) and CVR across the wide spectrum of the most common chronic liver diseases due to different etiologies. RESULTS Alcoholic liver disease (ALD) is associated with steatosis, IR and a typical lipid profile. The relationship between alcohol intake, incident type 2 diabetes (T2D) and CVR describes a J-shaped curve. Non-alcoholic fatty liver disease (NAFLD), and probably nonalcoholic steatohepatitis (NASH) in particular, is associated with IR, atherogenic dyslipidemia and increased CVR independent of traditional risk factors. Moreover, NASH-cirrhosis and T2D contribute to increasing CVR in liver transplant recipients. HBV infection is generally free from IR, steatosis and CVR. HCV-associated dysmetabolic syndrome, featuring steatosis, hypocholesterolemia and IR, appears to be associated with substantially increased CVR. Hyperlipidemia is an almost universal finding in primary biliary cirrhosis, a condition typically spared from steatosis and associated with neither subclinical atherosclerosis nor excess CVR. Finally, little is known on CVR in patients with hepatocellular carcinoma. CONCLUSIONS CVR is increased in ALD, NAFLD and chronic HCV infection, all conditions featuring IR and steatosis. Therefore, irrespective of serum lipid phenotype, hepatic steatosis and IR may be major shared determinants in amplifying CVR in common liver disease due to varying etiology.
Collapse
Affiliation(s)
- P Loria
- University of Modena and Reggio Emilia, Italy; Azienda USL MODENA, Italy.
| | | | - F Nascimbeni
- University of Modena and Reggio Emilia, Italy; Azienda USL MODENA, Italy
| | - S Ballestri
- University of Modena and Reggio Emilia, Italy; Azienda USL MODENA, Italy
| | - M Maurantonio
- University of Modena and Reggio Emilia, Italy; Azienda USL MODENA, Italy
| | - F Carubbi
- University of Modena and Reggio Emilia, Italy; Azienda USL MODENA, Italy
| | | | - A Lonardo
- University of Modena and Reggio Emilia, Italy; Azienda USL MODENA, Italy.
| |
Collapse
|
18
|
Phospholipid transfer protein, an emerging cardiometabolic risk marker: Is it time to intervene? Atherosclerosis 2013; 228:38-41. [DOI: 10.1016/j.atherosclerosis.2013.01.043] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 01/28/2013] [Indexed: 12/13/2022]
|
19
|
Plasma lipid transfer proteins and cardiovascular disease. The Framingham Heart Study. Atherosclerosis 2013; 228:230-6. [PMID: 23477743 DOI: 10.1016/j.atherosclerosis.2013.01.046] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Revised: 01/30/2013] [Accepted: 01/31/2013] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Cholesteryl ester transfer protein (CETP) and phospholipid transfer protein (PLTP) are two genetically-related plasma proteins involved in the exchange of cholesteryl esters and phospholipids between high-density lipoproteins (HDL) and other lipoproteins. Although low CETP and high PLTP activity both result in higher concentrations of plasma HDL-cholesterol (HDL-C), there is no evidence that either of these changes is associated with a decrease in cardiovascular disease (CVD) in a general population. METHODS Plasma CETP and PLTP activities, measured by homogenous fluorometric assays using synthetic donor particle substrates, were related to the incidence of a first CVD event in Framingham Heart Study Offspring participants without CVD (n = 2679, mean age 59 y, 56% women) attending the 6th examination cycle (1995-98). Because of an effect modification by sex for both CETP and PLTP, analyzes were stratified by sex. RESULTS During follow-up (mean 10.4 years) 187 participants experienced a first CVD event. In sex-specific Cox models, both CETP and PLTP as continuous and as binary variables were associated with significantly increased CVD in men, but not women. In men compared to a referent group with CETP ≥ median and PLTP < median, the multivariable-adjusted hazard ratio (HR) for new CVD events was significantly greater with either the combination of high CETP and high PLTP (HR 2.27, 95% CI 1.23-4.20); low CETP and low PLTP (HR 2.23, 95% CI 1.19-4.17); or low CETP and high PLTP (HR 2.85, 95% CI 1.53-5.31). In contrast, in women the multivariable-adjusted HR for new CVD events was non-significant and virtually equal to "1.0" with all combinations of high and low CETP or PLTP values. CONCLUSIONS Lower plasma CETP or higher PLTP activity was each associated with a significantly increased risk of CVD. Inexplicably, the increase in CVD associated with both lipid transfer proteins was confined to men.
Collapse
|
20
|
Elevated expression of PLTP is atherogenic in apolipoprotein E deficient mice. Atherosclerosis 2012; 227:37-42. [PMID: 23313246 DOI: 10.1016/j.atherosclerosis.2012.11.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 11/01/2012] [Accepted: 11/25/2012] [Indexed: 11/23/2022]
Abstract
OBJECTIVE Plasma phospholipid transfer protein (PLTP) plays a key role in lipoprotein metabolism. Its exact function in the development of atherosclerosis is still under debate however. We studied the effect of elevated PLTP expression in one of the most commonly used models of atherosclerosis, the ApoE deficient mouse. METHODS Experiment 1: Plasma PLTP activity, total cholesterol, HDL cholesterol and atherosclerosis development was measured in ApoE deficient mice with or without elevated expression of PLTP. Experiment 2: The same parameters were measured in ApoE deficient mice after bone marrow transplantation from wild type mice or mice with elevated PLTP expression. Experiment 3: Similar to experiment 2, but using donor mice with an ApoE deficient background. RESULTS Experiment 1: ApoE deficient mice have more than two times more atherosclerosis when overexpressing PLTP and a strongly decreased plasma level of HDL. Experiment 2: Bone marrow transplantation with ApoE proficient cells results in a strong reduction of plasma cholesterol in ApoE deficient acceptor mice. Still, elevated PLTP in bone marrow derived cells evoke a reduction of HDL cholesterol and increased atherosclerosis. Experiment 3: Bone marrow transplantation with ApoE deficient cells results in much higher cholesterol levels, but here too HDL cholesterol levels are reduced and atherosclerosis increased. CONCLUSION In all the models with ApoE deficiency, elevated PLTP expression causes higher levels of diet-induced atherosclerosis coinciding with decreased plasma levels of HDL cholesterol.
Collapse
|
21
|
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
|
22
|
Al-Zoairy R, Melmer A, Ress C, Laimer M, Kaser S, Ebenbichler C. Lipid profile changes after pronounced weight loss induced by bariatric surgery. ACTA ACUST UNITED AC 2012. [DOI: 10.2217/clp.12.9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
23
|
Stratton SA, Barton MC. p53-Mediated regulation of hepatic lipid metabolism: forging links between metabolism, atherogenesis, and cancer. J Hepatol 2012; 56:518-9. [PMID: 22051552 DOI: 10.1016/j.jhep.2011.10.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2011] [Accepted: 10/22/2011] [Indexed: 12/04/2022]
|
24
|
de Vries R, Kappelle PJ, Dallinga-Thie GM, Dullaart RP. Plasma phospholipid transfer protein activity is independently determined by obesity and insulin resistance in non-diabetic subjects. Atherosclerosis 2011; 217:253-9. [DOI: 10.1016/j.atherosclerosis.2011.03.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 03/03/2011] [Accepted: 03/14/2011] [Indexed: 12/13/2022]
|
25
|
Karalis IK, Bergheanu SC, Wolterbeek R, Dallinga-Thie GM, Hattori H, van Tol A, Liem AH, Wouter Jukema J. Effect of increasing doses of Rosuvastatin and Atorvastatin on apolipoproteins, enzymes and lipid transfer proteins involved in lipoprotein metabolism and inflammatory parameters. Curr Med Res Opin 2010; 26:2301-13. [PMID: 20731529 DOI: 10.1185/03007995.2010.509264] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
UNLABELLED This paper contains detailed results of a sub-population of the prospective randomized RADAR (Rosuvastatin and Atorvastatin in different Dosages And Reverse cholesterol transport) study. OBJECTIVE Statin treatment results in substantially decreased incidence of cardiovascular events but the exact pathophysiological mechanism of their beneficial effect is yet unclear. We aimed to examine the effects of up-titrated doses of two widely used statins (atorvastatin (ATOR) and rosuvastatin (ROSU)) on parameters involved in lipoprotein metabolism, in patients with low high density lipoprotein cholesterol values (HDL-C). RESEARCH DESIGN AND METHODS In this RADAR substudy, 80 patients, aged 40-80 years, with known cardiovascular disease and low HDL-C (<1.0 mmol/l), were randomized to receive, after an initial 6 week dietary run-in phase, either ATOR 20 mg (n = 41) or ROSU 10 mg (n = 39). The doses were up-titrated (in 6 week intervals) to 80 mg of ATOR or 40 mg of ROSU at 12 weeks. Serum lipoproteins and lipoprotein metabolism parameters were measured at baseline and at 6 and 18 weeks of follow up. RESULTS Both statins significantly reduced total cholesterol (TChol) and non-HDL-C values with ROSU being more effective for the doses studied (p < 0.05). No statistically significant effect on HDL-C was observed for either statin. Apolipoproteins (apo) B, CI, CIII, AV and E were significantly reduced in both groups (p < 0.05), while the ratio of HDL particles containing both apoAI and apoAII (LpAI-AII) over HDL containing apoAI alone (LpAI) was changed for both statins with the decrease of LpAI being more prominent in the ATOR group (p = 0.028). Cholesterol ester transfer protein (CETP) mass and activity, phospholipid transfer protein (PLTP) activity and lipoprotein-associated phospholipase A2 (Lp-PLA2) mass and activity were all significantly reduced in both treatment groups over the follow-up period (p < 0.001). ATOR displayed a more prominent decrease of PLTP activity compared to ROSU (p = 0.043), while ROSU displayed a more prominent decrease of Lp-PLA2 activity compared to ATOR (p = 0.04). Both statins effectively reduced, in a dose-dependent way, high sensitivity C-reactive protein values over time, while no effect on the levels of circulating inter cellular adhesion molecule 1 (cICAM-1) was observed. CONCLUSIONS The effects of statin treatment extend further and beyond a mere TChol and LDL cholesterol reduction, as demonstrated by the aforementioned alterations of lipoproteins, enzymes and lipid transfer proteins involved in lipoprotein metabolism and pro-atherogenic and inflammatory molecules. ROSU and ATOR displayed a similar pattern of effect on lipid metabolism with discrete differences in the magnitude of this effect in certain variables. Despite the limitations of small population size and lack of clinical end points, reported data provide an insight for the possible pathophysiological mechanisms implicated in the effect of increasing dosages of different statin treatments.
Collapse
|
26
|
Vergeer M, Boekholdt SM, Sandhu MS, Ricketts SL, Wareham NJ, Brown MJ, de Faire U, Leander K, Gigante B, Kavousi M, Hofman A, Uitterlinden AG, van Duijn CM, Witteman JCM, Jukema JW, Schadt EE, van der Schoot E, Kastelein JJP, Khaw KT, Dullaart RPF, van Tol A, Trip MD, Dallinga-Thie GM. Genetic variation at the phospholipid transfer protein locus affects its activity and high-density lipoprotein size and is a novel marker of cardiovascular disease susceptibility. Circulation 2010; 122:470-7. [PMID: 20644014 DOI: 10.1161/circulationaha.109.912519] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND In contrast to clear associations between variants in genes participating in low-density lipoprotein metabolism and cardiovascular disease risk, such associations for high-density lipoprotein (HDL)-related genes are not well supported by recent large studies. We aimed to determine whether genetic variants at the locus encoding phospholipid transfer protein (PLTP), a protein involved in HDL remodeling, underlie altered PLTP activity, HDL particle concentration and size, and cardiovascular disease risk. METHODS AND RESULTS We assessed associations between 6 PLTP tagging single nucleotide polymorphisms and PLTP activity in 2 studies (combined n=384) and identified 2 variants that show reproducible associations with altered plasma PLTP activity. A gene score based on these variants is associated with lower hepatic PLTP transcription (P=3.2x10(-18)) in a third study (n=957) and with an increased number of HDL particles of smaller size (P=3.4x10(-17)) in a fourth study (n=3375). In a combination of 5 cardiovascular disease case-control studies (n=4658 cases and 11 459 controls), a higher gene score was associated with a lower cardiovascular disease risk (per-allele odds ratio, 0.94; 95% confidence interval, 0.90 to 0.98; P=1.2x10(-3); odds ratio for highest versus lowest gene score, 0.69; 95% confidence interval, 0.55 to 0.86; P=1.0x10(-3)). CONCLUSIONS A gene score based on 2 PLTP single nucleotide polymorphisms is associated with lower PLTP transcription and activity, an increased number of HDL particles, smaller HDL size, and decreased risk of cardiovascular disease. These findings indicate that PLTP is a proatherogenic entity and suggest that modulation of specific elements of HDL metabolism may offer cardiovascular benefit.
Collapse
Affiliation(s)
- Menno Vergeer
- Department of Vascular Medicine, Academic Medical Center, 1105 AZ Amsterdam, the Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
|
28
|
Current World Literature. Curr Opin Support Palliat Care 2010; 4:111-20. [DOI: 10.1097/spc.0b013e32833a1dfc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
29
|
Miyazaki O, Fukamachi I. Preβ1-HDL, a key element of reverse cholesterol transport: its potential as a biomarker. ACTA ACUST UNITED AC 2010. [DOI: 10.2217/clp.10.17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
30
|
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
|
31
|
Strang AC, Hovingh GK, Stroes ES, Kastelein JJ. The genetics of high-density lipoprotein metabolism: clinical relevance for therapeutic approaches. Am J Cardiol 2009; 104:22E-31E. [PMID: 19895941 DOI: 10.1016/j.amjcard.2009.09.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The risk for coronary artery disease (CAD) is inversely correlated with high-density lipoprotein (HDL) cholesterol plasma levels. These plasma HDL cholesterol levels are influenced by the activity of a number of enzymes and receptors, and therefore, variations in the genes encoding for these proteins may consequently result in an altered CAD risk. Identification of such pivotal players in HDL cholesterol metabolism that are also strongly associated with CAD risk is crucial for the materialization of novel therapeutic modalities. A large amount of knowledge has been obtained by studies involving families with extreme HDL phenotypes specific to molecular defects. In fact, thus far, monogenetic defects have been described in the genes coding for apolipoprotein A-I, adenosine triphosphate-binding cassette transporter A1, cholesterol ester transfer protein, the lack of endothelial lipase (LIPG), phospholipid transfer protein, and lecithin-cholesterol acyltransferase. Despite the fact that the total number of carriers of such mutations is rather small, much can be gained by extensively studying the metabolic and vascular consequences of these mutations. Surrogate markers for atherosclerosis have proved to be useful to overcome this sample size limitation and have been widely exploited to study families with decreased or increased HDL cholesterol levels in order to correlate HDL cholesterol phenotypes to atherosclerotic burden in cases and controls. Apart from such extreme phenotype approaches, novel population-based genome-wide association studies have been used to decipher the link between genetic loci and HDL cholesterol levels, and the identification of novel HDL cholesterol-related genes is eagerly awaited. These might be instrumental in the ongoing fight against atherosclerosis.
Collapse
|
32
|
Jarvik GP, Rajagopalan R, Rosenthal EA, Wolfbauer G, McKinstry L, Vaze A, Brunzell J, Motulsky AG, Nickerson DA, Heagerty PJ, Wijsman EM, Albers JJ. Genetic and nongenetic sources of variation in phospholipid transfer protein activity. J Lipid Res 2009; 51:983-90. [PMID: 19965587 DOI: 10.1194/jlr.m000125] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Phospholipid transfer protein (PLTP) belongs to the lipid transfer/lipopolysaccharide-binding protein gene family. Expression of PLTP has been implicated in the development of atherosclerosis. We evaluated the effects of PLTP region tagging single nucleotide polymorphisms (SNPs) on the prediction of both carotid artery disease (CAAD) and PLTP activity. CAAD effects were evaluated in 442 Caucasian male subjects with severe CAAD and 497 vascular disease-free controls. SNP prediction of PLTP transfer activity was evaluated in both a subsample of 87 subjects enriched for an allele of interest and in a confirmation sample of 210 Caucasian males and females. Hemoglobin A1c or insulin level predicted 11-14% of age- and sex-adjusted PLTP activity. PLTP SNPs that predicted approximately 11-30% of adjusted PLTP activity variance were identified in the two cohorts. For rs6065904, the allele that was associated with CAAD was also associated with elevated PLTP activity in both cohorts. SNPs associated with PLTP activity also predicted variation in LDL-cholesterol and LDL-B level only in the replication cohort. These results demonstrate that PLTP activity is strongly influenced by PLTP region polymorphisms and metabolic factors.
Collapse
Affiliation(s)
- Gail P Jarvik
- Department of Medicine (Division of Medical Genetics), University of Washington, Seattle, WA, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Quintão ECR, Cazita PM. Lipid transfer proteins: past, present and perspectives. Atherosclerosis 2009; 209:1-9. [PMID: 19733354 DOI: 10.1016/j.atherosclerosis.2009.08.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2009] [Revised: 07/02/2009] [Accepted: 08/03/2009] [Indexed: 11/15/2022]
Abstract
Lipid transfer proteins (PLTP and CETP) play roles in atherogenesis by modifying the arterial intima cholesterol content via altering the concentration and function of plasma lipoproteins and influencing inflammation. In this regard, endotoxins impair the reverse cholesterol transport (RCT) system in an endotoxemic rodent model, supporting a pro-inflammatory role of HDL reported in chronic diseases where atherosclerosis is premature. High PLTP activity related to atherosclerosis in some clinical studies, but the mechanisms involved could not be ascertained. In experimental animals the relation of elevated plasma PLTP concentration with atherosclerosis was confounded by HDL-C lowering and by unfavorable effects on several inflammatory markers. Coincidently, PLTP also increases in human experimental endotoxemia and in clinical sepsis. Human population investigations seem to favor low CETP as atheroprotective; this is supported by animal models where overexpression of huCETP is atherogenic, most likely due to increased concentration of apoB-lipoprotein-cholesterol. Thus, in spite of CETP facilitating the HDL-C-mediated RCT, the reduction of apoB-LP-cholesterol concentration is the probable antiatherogenic mechanism of CETP inhibition. On the other hand, experimental huCETP expression protects mice from the harmful effects of a bacterial polysaccharide infusion and the mortality rate of severely ill patients correlates with reduction of the plasma CETP concentration. Thus, the roles played by PLTP and CETP on atherosclerosis and acute inflammation seem contradictory. Therefore, the biological roles of PLTP and CETP must be carefully monitored when investigating drugs that inhibit their activity in the prevention of atherosclerosis.
Collapse
Affiliation(s)
- Eder C R Quintão
- Lipids Lab, LIM 10, Faculty of Medical Sciences, University of São Paulo, SP, Brazil.
| | | |
Collapse
|