1
|
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: 17] [Impact Index Per Article: 5.7] [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
|
2
|
Plasma phospholipid transfer protein (PLTP) as an emerging determinant of the adaptive immune response. Cell Mol Immunol 2018; 15:1077-1079. [PMID: 29735978 DOI: 10.1038/s41423-018-0036-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 04/12/2018] [Indexed: 11/08/2022] Open
|
3
|
Daguindau E, Gautier T, Chagué C, Pais de Barros JP, Deckert V, Lagrost L, Saas P. Is It Time to Reconsider the Lipopolysaccharide Paradigm in Acute Graft-Versus-Host Disease? Front Immunol 2017; 8:952. [PMID: 28848554 PMCID: PMC5553011 DOI: 10.3389/fimmu.2017.00952] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 07/25/2017] [Indexed: 12/16/2022] Open
Affiliation(s)
- Etienne Daguindau
- Université Bourgogne Franche-Comté, EFS Bourgogne Franche-Comté, INSERM, UMR1098, Besançon, France
- Department of Hematology, University Hospital of Besançon, Besançon, France
- LipSTIC LabEx, FHU INCREASE, Besançon, France
| | - Thomas Gautier
- INSERM, Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France
- LipSTIC LabEx, Dijon, France
| | - Cécile Chagué
- Université Bourgogne Franche-Comté, EFS Bourgogne Franche-Comté, INSERM, UMR1098, Besançon, France
- Department of Hematology, University Hospital of Besançon, Besançon, France
| | - Jean-Paul Pais de Barros
- INSERM, Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France
- LipSTIC LabEx, Dijon, France
| | - Valérie Deckert
- INSERM, Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France
- LipSTIC LabEx, Dijon, France
| | - Laurent Lagrost
- INSERM, Université Bourgogne Franche-Comté, LNC UMR1231, Dijon, France
- LipSTIC LabEx, Dijon, France
- University Hospital of Dijon, Dijon, France
| | - Philippe Saas
- Université Bourgogne Franche-Comté, EFS Bourgogne Franche-Comté, INSERM, UMR1098, Besançon, France
- Department of Hematology, University Hospital of Besançon, Besançon, France
- LipSTIC LabEx, FHU INCREASE, Besançon, France
| |
Collapse
|
4
|
Abdala-Valencia H, Soveg F, Cook-Mills JM. γ-Tocopherol supplementation of allergic female mice augments development of CD11c+CD11b+ dendritic cells in utero and allergic inflammation in neonates. Am J Physiol Lung Cell Mol Physiol 2016; 310:L759-71. [PMID: 26801566 DOI: 10.1152/ajplung.00301.2015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 01/20/2016] [Indexed: 11/22/2022] Open
Abstract
γ-Tocopherol increases responses to allergen challenge in allergic adult mice, but it is not known whether γ-tocopherol regulates the development of allergic disease. Development of allergic disease often occurs early in life. In clinical studies and animal models, offspring of allergic mothers have increased responsiveness to allergen challenge. Therefore, we determined whether γ-tocopherol augments development of allergic responses in offspring of allergic female mice. Allergic female mice were supplemented with γ-tocopherol starting at mating. The pups from allergic mothers developed allergic lung responses, whereas pups from saline-treated mothers did not respond to allergen challenge. The γ-tocopherol supplementation of allergic female mice increased the numbers of eosinophils twofold in the pup bronchoalveolar lavage and lungs after allergen challenge. There was also about a twofold increase in pup lung CD11b(+) subsets of CD11c(+) dendritic cells and in numbers of these dendritic cells expressing the transcription factor IRF4. There was no change in several CD11b(-) dendritic cell subsets. Furthermore, maternal supplementation with γ-tocopherol increased the number of fetal liver CD11b(+)CD11c(+) dendritic cells twofold in utero. In the pups, γ-tocopherol increased lung expression of the inflammatory mediators CCL11, amphiregulin, activin A, and IL-5. In conclusion, maternal supplementation with γ-tocopherol increased fetal development of subsets of dendritic cells that are critical for allergic responses and increased development of allergic responses in pups from allergic mothers. These results have implications for supplementation of allergic mothers with γ-tocopherol in prenatal vitamins.
Collapse
Affiliation(s)
- Hiam Abdala-Valencia
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Frank Soveg
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Joan M Cook-Mills
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
5
|
Lee SD, Tontonoz P. Liver X receptors at the intersection of lipid metabolism and atherogenesis. Atherosclerosis 2015; 242:29-36. [PMID: 26164157 PMCID: PMC4546914 DOI: 10.1016/j.atherosclerosis.2015.06.042] [Citation(s) in RCA: 108] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 06/19/2015] [Accepted: 06/22/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Stephen D Lee
- Howard Hughes Medical Institute, Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095, USA
| | - Peter Tontonoz
- Howard Hughes Medical Institute, Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA 90095, USA.
| |
Collapse
|
6
|
Plasma phospholipid transfer protein (PLTP) modulates adaptive immune functions through alternation of T helper cell polarization. Cell Mol Immunol 2015; 13:795-804. [PMID: 26320740 DOI: 10.1038/cmi.2015.75] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/01/2015] [Accepted: 07/01/2015] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Plasma phospholipid transfer protein (PLTP) is a key determinant of lipoprotein metabolism, and both animal and human studies converge to indicate that PLTP promotes atherogenesis and its thromboembolic complications. Moreover, it has recently been reported that PLTP modulates inflammation and immune responses. Although earlier studies from our group demonstrated that PLTP can modify macrophage activation, the implication of PLTP in the modulation of T-cell-mediated immune responses has never been investigated and was therefore addressed in the present study. Approach and results: In the present study, we demonstrated that PLTP deficiency in mice has a profound effect on CD4+ Th0 cell polarization, with a shift towards the anti-inflammatory Th2 phenotype under both normal and pathological conditions. In a model of contact hypersensitivity, a significantly impaired response to skin sensitization with the hapten-2,4-dinitrofluorobenzene (DNFB) was observed in PLTP-deficient mice compared to wild-type (WT) mice. Interestingly, PLTP deficiency in mice exerted no effect on the counts of total white blood cells, lymphocytes, granulocytes, or monocytes in the peripheral blood. Moreover, PLTP deficiency did not modify the amounts of CD4+ and CD8+ T lymphocyte subsets. However, PLTP-deficiency, associated with upregulation of the Th2 phenotype, was accompanied by a significant decrease in the production of the pro-Th1 cytokine interleukin 18 by accessory cells. CONCLUSIONS For the first time, this work reports a physiological role for PLTP in the polarization of CD4+ T cells toward the pro-inflammatory Th1 phenotype.
Collapse
|
7
|
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: 0.9] [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
|
8
|
Abdala-Valencia H, Berdnikovs S, Soveg FW, Cook-Mills JM. α-Tocopherol supplementation of allergic female mice inhibits development of CD11c+CD11b+ dendritic cells in utero and allergic inflammation in neonates. Am J Physiol Lung Cell Mol Physiol 2014; 307:L482-96. [PMID: 25015974 DOI: 10.1152/ajplung.00132.2014] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
α-Tocopherol blocks responses to allergen challenge in allergic adult mice, but it is not known whether α-tocopherol regulates the development of allergic disease. Development of allergic disease often occurs early in life. In clinical studies and animal models, offspring of allergic mothers have increased responsiveness to allergen challenge. Therefore, we determined whether α-tocopherol blocked development of allergic responses in offspring of allergic female mice. Allergic female mice were supplemented with α-tocopherol starting at mating. The pups from allergic mothers developed allergic lung responses, whereas pups from saline-treated mothers did not respond to the allergen challenge, and α-tocopherol supplementation of allergic female mice resulted in a dose-dependent reduction in eosinophils in the pup bronchoalveolar lavage and lungs after allergen challenge. There was also a reduction in pup lung CD11b(+) dendritic cell subsets that are critical to development of allergic responses, but there was no change in several CD11b(-) dendritic cell subsets. Furthermore, maternal supplementation with α-tocopherol reduced the number of fetal liver CD11b(+) dendritic cells in utero. In the pups, there was reduced allergen-induced lung mRNA expression of IL-4, IL-33, TSLP, CCL11, and CCL24. Cross-fostering pups at the time of birth demonstrated that α-tocopherol had a regulatory function in utero. In conclusion, maternal supplementation with α-tocopherol reduced fetal development of subsets of dendritic cells that are critical for allergic responses and reduced development of allergic responses in pups from allergic mothers. These results have implications for supplementation of allergic mothers with α-tocopherol.
Collapse
Affiliation(s)
- Hiam Abdala-Valencia
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Sergejs Berdnikovs
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Frank W Soveg
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Joan M Cook-Mills
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| |
Collapse
|
9
|
Phospholipid transfer protein (PLTP) deficiency impaired blood–brain barrier integrity by increasing cerebrovascular oxidative stress. Biochem Biophys Res Commun 2014; 445:352-6. [DOI: 10.1016/j.bbrc.2014.01.194] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 01/31/2014] [Indexed: 11/23/2022]
|
10
|
Li X, Wang G, Chen D, Lu Y. Binding of ascorbic acid and α-tocopherol to bovine serum albumin: a comparative study. ACTA ACUST UNITED AC 2014; 10:326-37. [DOI: 10.1039/c3mb70373h] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
11
|
|
12
|
Fanali G, Fasano M, Ascenzi P, Zingg JM, Azzi A. α-Tocopherol binding to human serum albumin. Biofactors 2013; 39:294-303. [PMID: 23355326 DOI: 10.1002/biof.1070] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 11/02/2012] [Indexed: 11/12/2022]
Abstract
Given the ability of human serum albumin (HSA) to bind hydrophobic ligands, the binding mode of α-tocopherol, the most representative member of the vitamin E family, is reported. α-Tocopherol binds to HSA with Kd0 = (7.0 ± 3.0) × 10(-6) M (pH 7.2, 25.0°C). Competitive and allosteric modulation of α-tocopherol binding to full-length and truncated (Asp1-Glu382) HSA by endogenous and exogenous ligands suggests that it accommodates preferentially in the FA3-FA4 site. As HSA is taken up into cells, colocalizes with the α-tocopherol transfer protein, and contributes to ligand secretion via ABCA1, it might participate in the distribution of α-tocopherol between plasma, cells, and tissues.
Collapse
Affiliation(s)
- Gabriella Fanali
- Division of Biomedical Sciences, Department of Theoretical and Applied Sciences, Center of Neuroscience, University of Insubria, Busto Arsizio (VA), Italy.
| | | | | | | | | |
Collapse
|
13
|
Desrumaux C, Pisoni A, Meunier J, Deckert V, Athias A, Perrier V, Villard V, Lagrost L, Verdier JM, Maurice T. Increased amyloid-β peptide-induced memory deficits in phospholipid transfer protein (PLTP) gene knockout mice. Neuropsychopharmacology 2013; 38:817-25. [PMID: 23303044 PMCID: PMC3671992 DOI: 10.1038/npp.2012.247] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Oxidative stress is recognized as one of the earliest and most intense pathological processes in Alzheimer's disease (AD), and the antioxidant vitamin E has been shown to efficiently prevent amyloid plaque formation and neurodegeneration. Plasma phospholipid transfer protein (PLTP) has a major role in vitamin E transfers in vivo, and PLTP deficiency in mice is associated with reduced brain vitamin E levels. To determine the impact of PLTP on amyloid pathology in vivo, we analyzed the vulnerability of PLTP-deficient (PLTP-KO) mice to the toxic effects induced by intracerebroventricular injection of oligomeric amyloid-β 25-35 (Aβ 25-35) peptide, a non-transgenic model of AD. Under basal conditions, PLTP-KO mice showed increased cerebral oxidative stress, increased brain Aβ 1-42 levels, and a lower expression of the synaptic function marker synaptophysin, as compared with wild-type mice. This PLTP-KO phenotype was associated with increased memory impairment 1 week after Aβ25-35 peptide injection. Restoration of brain vitamin E levels in PLTP-KO mice through a chronic dietary supplementation prevented Aβ 25-35-induced memory deficits and reduced cerebral oxidative stress and toxicity. We conclude that PLTP, through its ability to deliver vitamin E to the brain, constitutes an endogenous neuroprotective agent. Increasing PLTP activity may offer a new way to develop neuroprotective therapies.
Collapse
Affiliation(s)
- Catherine Desrumaux
- INSERM U710, Université Montpellier 2, CC105, Place Eugène Bataillon, 34095 Montpellier, Cedex 05, France.
| | - Amandine Pisoni
- INSERM U710, Montpellier, France,Université Montpellier 2, Montpellier, France,EPHE, Paris, France
| | | | | | - Anne Athias
- Lipidomics analytical platform, SFR 100, Dijon, France
| | - Véronique Perrier
- INSERM U710, Montpellier, France,Université Montpellier 2, Montpellier, France,EPHE, Paris, France
| | | | | | - Jean-Michel Verdier
- INSERM U710, Montpellier, France,Université Montpellier 2, Montpellier, France,EPHE, Paris, France
| | - Tangui Maurice
- INSERM U710, Montpellier, France,Université Montpellier 2, Montpellier, France,EPHE, Paris, France,INSERM U710, Université Montpellier 2, CC105, Place Eugène Bataillon, 34095 Montpellier, Cedex 05, France. Tel: (+33/0) 4 67 14 36 23, Fax: (+33/0) 4 67 14 92 95, E-mail: or E-mail:
| |
Collapse
|
14
|
Gao J, Zhao R, Xue Y, Niu Z, Cui K, Yu F, Zhang B, Li S. Role of enolase-1 in response to hypoxia in breast cancer: exploring the mechanisms of action. Oncol Rep 2013; 29:1322-32. [PMID: 23381546 DOI: 10.3892/or.2013.2269] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 12/06/2012] [Indexed: 11/05/2022] Open
Abstract
In the present study, we investigated the effect of reduced enolase-1 expression in human umbilical vein endothelial cells (HUVECs)/MDA-MB-231 cells on the response to hypoxia and the possible mechanisms involved. Breast cancer cells transfected with enolase-1 siRNA were injected into mice to establish a tumor-bearing mouse model, and the correlation between enolase-1 expression and breast cancer angiogenesis, as well as its effect on the efficacy of radiation therapy were assessed. HUVECs were cultured in vitro, and transfected with enolase-1 siRNA. Following stable passage, 1.0% O2 was used to induce hypoxia. The growth, proliferation, division and angiogenesis of HUVECs were observed using MTT assay, flow cytometry (FCM) and time-lapse video microscopy. The key regulatory molecules were detected using western blot analysis, two-dimensional (2-D) electrophoresis and matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF-MS). The breast cancer cell line, MDA-MB-231, was cultured in vitro, and transfected with enolase-1 siRNA. The cells were injected into nude mice, and radiation therapy was administered. Tumor growth, angiogenesis in tumor tissues and apoptosis were observed, and the expression of the endogenous hypoxia marker, hypoxia inducible factor-1α (HIF-1α), was detected using immunohistochemistry after the mice were sacrificed. A significant reduction in the hypoxia-induced apoptosis of HUVECs was observed in the control group compared with the endothelial cells transfected with enolase-1 siRNA. After the enolase-1 transfected breast cancer cells were injected into nude mice, tumor growth significantly declined, and the tumor volume and weight were reduced. Following treatment with radiation therapy, tumor size significantly decreased in both groups, and the highest reduction was observed in the transfected group. The reduction in enolase-1 expression significantly decreases the response to hypoxia and enhances the sensitivity of the cells to radiation therapy; therefore, enolase-1 may be a drug target for the treatment of breast cancer.
Collapse
Affiliation(s)
- Jie Gao
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital, Shandong Academy of Medical Sciences, Jinan 276000, PR China
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Yang J, Han Y, Ye W, Liu F, Zhuang K, Wu G. Alpha tocopherol treatment reduces the expression of Nogo-A and NgR in rat brain after traumatic brain injury. J Surg Res 2012. [PMID: 23207171 DOI: 10.1016/j.jss.2012.11.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
BACKGROUND Neurite outgrowth inhibitor-A (Nogo-A), myelin-associated glycoprotein, and oligodendrocyte myelin glycoprotein are three myelin-associated proteins that act as inhibitors to central nervous system regeneration. Neurite outgrowth inhibitor-A imposes the strongest effect on inhibiting axonal regeneration after traumatic brain injury. Alpha-tocopherol, a member of the vitamin E family, is recognized as an active antioxidative substance. Its use has not been well studied in brain injury research, especially in axonal regeneration research. METHODS We obtained 99 intact adult male Sprague-Dawley rats (200-250 g) from the Experimental Animal Center of Central South University. We used the modified method of Freeney to generate moderate brain injury in the rats. We injected 600 mg/kg α-tocopherol intraperitoneally daily as traumatic brain injury (TBI) treatment. Then, we performed behavioral tests in the corresponding time point, examined brain tissues after hematoxylin-eosin staining to identify changes in cell morphology, and performed immunohistochemical staining and quantitative real-time polymerase chain reaction to detect the expression of NoGo and Nogo receptor (NgR) in brain tissue. RESULTS For the Neurological Severity Scores of rats, there were obvious differences among the three groups at the corresponding time points. Standard hematoxylin-eosin staining showed that the brain structure of a sham-operated group of rats was clear, uniform, and compact. A TBI group exhibited hemorrhage, edema, inflammatory cell infiltration, condensed nuclei, and necrosis. We also saw glial cells and fibrous tissue proliferation. The α-tocopherol-treated TBI group had similar but less severe changes than the TBI group. Expression of Nogo-A and NgR increased after TBI compared with the sham-operated group. However, Nogo-A and NgR expression was significantly lower in the α-tocopherol-treated TBI group compared with the TBI group. Similarly, results showed that functional neurological deficits among rats in the α-tocopherol-treated TBI group were less pronounced than in the TBI group (model group). CONCLUSIONS Our data demonstrate that α-tocopherol-treated rats had reduced microscopic evidence of brain damage. Alpha-tocopherol reduced Nogo-A and NgR expression in brain tissue after traumatic brain injury and promoted nerve regeneration. Alpha-tocopherol treatment of TBI rats had a neuroprotective role in their recovery.
Collapse
Affiliation(s)
- Jinfu Yang
- Department of Neurosurgery, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | | | | | | | | | | |
Collapse
|
16
|
Tzoumaki MV, Moschakis T, Scholten E, Biliaderis CG. In vitro lipid digestion of chitin nanocrystal stabilized o/w emulsions. Food Funct 2012; 4:121-9. [PMID: 23064096 DOI: 10.1039/c2fo30129f] [Citation(s) in RCA: 141] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Chitin nanocrystals (ChN) have been shown to form stable Pickering emulsions. These oil-in-water emulsions were compared with conventional milk (whey protein isolate, WPI, and sodium caseinate, SCn) protein-stabilized emulsions in terms of their lipid digestion kinetics using an in vitro enzymatic protocol. The kinetics of fatty acid release were evaluated as well as the change in oil droplet size of the respective emulsions during lipid digestion. The interfacial pressure was measured by addition of the duodenal components using drop tensiometry and the electrical charge of the oil droplets was also assessed, in an attempt to relate the interfacial properties with the stability of the emulsions towards lipolysis. Lipid hydrolysis in the ChN-stabilized emulsion was appreciably slower and the plateau values of the total concentration of fatty acids released were much lower, compared to the WPI- and SCn-stabilized emulsions. Moreover, the ChN-stabilized emulsions were relatively stable to coalescence during lipid digestion, whereas the WPI- and SCn-stabilized emulsions exhibited a significant increase in their droplet size. On the other hand, no major differences were shown among the different emulsion samples in terms of their interfacial properties. The increased stability of the ChN-stabilized emulsions towards lipolysis could be attributed to several underlying mechanisms: (i) strong and irreversible adsorption of the chitin nanocrystals at the interface that might inhibit an extensive displacement of the solid particles by bile salts and lipase, (ii) network formation by the nanocrystals in the bulk (continuous) phase that may reduce lipid digestion kinetics, and (iii) the ability of chitin, and consequently of ChNs, to impair pancreatic lipase activity. The finding that ChNs can be used to impede lipid digestion may have important implications for the design and fabrication of structured emulsions with controlled lipid digestibility that could provide the basis for the development of novel products that may promote satiety, reduce caloric intake and combat obesity.
Collapse
Affiliation(s)
- Maria V Tzoumaki
- Department of Food Science and Technology, School of Agriculture, Aristotle University, GR-541 24, P.O. Box 235, Thessaloniki, Greece
| | | | | | | |
Collapse
|
17
|
Eren E, Yilmaz N, Aydin O. High Density Lipoprotein and it's Dysfunction. Open Biochem J 2012; 6:78-93. [PMID: 22888373 PMCID: PMC3414806 DOI: 10.2174/1874091x01206010078] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2012] [Revised: 04/18/2012] [Accepted: 04/24/2012] [Indexed: 01/22/2023] Open
Abstract
Plasma high-density lipoprotein cholesterol(HDL-C) levels do not predict functionality and composition of high-density lipoprotein(HDL). Traditionally, keeping levels of low-density lipoprotein cholesterol(LDL-C) down and HDL-C up have been the goal of patients to prevent atherosclerosis that can lead to coronary vascular disease(CVD). People think about the HDL present in their cholesterol test, but not about its functional capability. Up to 65% of cardiovascular death cannot be prevented by putative LDL-C lowering agents. It well explains the strong interest in HDL increasing strategies. However, recent studies have questioned the good in using drugs to increase level of HDL. While raising HDL is a theoretically attractive target, the optimal approach remains uncertain. The attention has turned to the quality, rather than the quantity, of HDL-C. An alternative to elevations in HDL involves strategies to enhance HDL functionality. The situation poses an opportunity for clinical chemists to take the lead in the development and validation of such biomarkers. The best known function of HDL is the capacity to promote cellular cholesterol efflux from peripheral cells and deliver cholesterol to the liver for excretion, thereby playing a key role in reverse cholesterol transport (RCT). The functions of HDL that have recently attracted attention include anti-inflammatory and anti-oxidant activities. High antioxidant and anti-inflammatory activities of HDL are associated with protection from CVD.This review addresses the current state of knowledge regarding assays of HDL functions and their relationship to CVD. HDL as a therapeutic target is the new frontier with huge potential for positive public health implications.
Collapse
Affiliation(s)
- Esin Eren
- Antalya Public Health Center of Ministry of Health, Antalya, Turkey
| | | | | |
Collapse
|
18
|
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: 44] [Impact Index Per Article: 3.4] [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
|
19
|
Albers JJ, Vuletic S, Cheung MC. Role of plasma phospholipid transfer protein in lipid and lipoprotein metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1821:345-57. [PMID: 21736953 DOI: 10.1016/j.bbalip.2011.06.013] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2011] [Revised: 06/01/2011] [Accepted: 06/14/2011] [Indexed: 12/13/2022]
Abstract
The understanding of the physiological and pathophysiological role of PLTP has greatly increased since the discovery of PLTP more than a quarter of century ago. A comprehensive review of PLTP is presented on the following topics: PLTP gene organization and structure; PLTP transfer properties; different forms of PLTP; characteristics of plasma PLTP complexes; relationship of plasma PLTP activity, mass and specific activity with lipoprotein and metabolic factors; role of PLTP in lipoprotein metabolism; PLTP and reverse cholesterol transport; insights from studies of PLTP variants; insights of PLTP from animal studies; PLTP and atherosclerosis; PLTP and signal transduction; PLTP in the brain; and PLTP in human disease. PLTP's central role in lipoprotein metabolism and lipid transport in the vascular compartment has been firmly established. However, more studies are needed to further delineate PLTP's functions in specific tissues, such as the lung, brain and adipose tissue. Furthermore, the specific role that PLTP plays in human diseases, such as atherosclerosis, cancer, or neurodegenerative disease, remains to be clarified. Exciting directions for future research include evaluation of PLTP's physiological relevance in intracellular lipid metabolism and signal transduction, which undoubtedly will advance our knowledge of PLTP functions in health and disease. This article is part of a Special Issue entitled Advances in High Density Lipoprotein Formation and Metabolism: A Tribute to John F. Oram (1945-2010).
Collapse
Affiliation(s)
- John J Albers
- Northwest Lipid Metabolism and Diabetes Research Laboratories, Division of Metabolism, Endocrinology and Nutrition, Department of Medicine, University of Washington, 401 Queen Anne Ave N, Seattle, WA 98109, USA.
| | | | | |
Collapse
|
20
|
Cheung MC, Wolfbauer G, Albers JJ. Different phospholipid transfer protein complexes contribute to the variation in plasma PLTP specific activity. Biochim Biophys Acta Mol Cell Biol Lipids 2011; 1811:343-7. [PMID: 21303701 DOI: 10.1016/j.bbalip.2011.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2010] [Revised: 01/15/2011] [Accepted: 02/01/2011] [Indexed: 11/28/2022]
Abstract
Phospholipid transfer protein (PLTP) facilitates the transfer of phospholipids among lipoproteins. Over half of the PLTP in human plasma has been found to have little phospholipid transfer activity (inactive PLTP). We recently observed that plasma PLTP specific activity is inversely correlated with high-density lipoprotein (HDL) level and particle size in healthy adults. The purpose of this study was to evaluate the factors that contribute to the variation in plasma PLTP specific activity. Analysis of the specific activity of PLTP complexes in nine plasma samples from healthy adults revealed two clusters of inactive PLTP complexes with mean molecular weights (MW) of 342kDa and 146kDa. The large and small inactive PLTP complexes represented 52±8% (range 39-63%) and 8±8% (range 1-28%) of the plasma PLTP, respectively. Active PLTP complexes had a mean MW of 207kDa and constituted 40±6% (range 33-50%) of the plasma PLTP. The specific activity of active PLTP varied from 16 to 32μmol/μg/h. These data demonstrate for the first time the existence of small inactive plasma PLTP complexes. Variation in the amount of the two clusters of inactive PLTP complexes and the specific activity of the active PLTP contribute to the variation in plasma PLTP specific activity.
Collapse
Affiliation(s)
- Marian C Cheung
- Division of Metabolism, Endocrinology and Nutrition, Northwest Lipid Metabolism and Diabetes Research Laboratories, Department of Medicine, University of Washington, Seattle, WA 98109-4517, USA
| | | | | |
Collapse
|
21
|
McCary CA, Abdala-Valencia H, Berdnikovs S, Cook-Mills JM. Supplemental and highly elevated tocopherol doses differentially regulate allergic inflammation: reversibility of α-tocopherol and γ-tocopherol's effects. THE JOURNAL OF IMMUNOLOGY 2011; 186:3674-85. [PMID: 21317387 DOI: 10.4049/jimmunol.1003037] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
We have reported that supplemental doses of the α- and γ-tocopherol isoforms of vitamin E decrease and increase, respectively, allergic lung inflammation. We have now assessed whether these effects of tocopherols are reversible. For these studies, mice were treated with Ag and supplemental tocopherols in a first phase of treatment followed by a 4-wk clearance phase, and then the mice received a second phase of Ag and tocopherol treatments. The proinflammatory effects of supplemental levels of γ-tocopherol in phase 1 were only partially reversed by supplemental α-tocopherol in phase 2, but were completely reversed by raising α-tocopherol levels 10-fold in phase 2. When γ-tocopherol levels were increased 10-fold (highly elevated tocopherol) so that the lung tissue γ-tocopherol levels were equal to the lung tissue levels of supplemental α-tocopherol, γ-tocopherol reduced leukocyte numbers in the lung lavage fluid. In contrast to the lung lavage fluid, highly elevated levels of γ-tocopherol increased inflammation in the lung tissue. These regulatory effects of highly elevated tocopherols on tissue inflammation and lung lavage fluid were reversible in a second phase of Ag challenge without tocopherols. In summary, the proinflammatory effects of supplemental γ-tocopherol on lung inflammation were partially reversed by supplemental levels of α-tocopherol but were completely reversed by highly elevated levels of α-tocopherol. Also, highly elevated levels of γ-tocopherol were inhibitory and reversible in lung lavage but, importantly, were proinflammatory in lung tissue sections. These results have implications for future studies with tocopherols and provide a new context in which to review vitamin E studies in the literature.
Collapse
Affiliation(s)
- Christine A McCary
- Allergy-Immunology Division, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | |
Collapse
|
22
|
Desrumaux C, Deckert V, Lemaire-Ewing S, Mossiat C, Athias A, Vandroux D, Dumont L, Monier S, Pais de Barros JP, Klein A, De Maistre E, Blache D, Beley A, Marie C, Garnier P, Lagrost L. Plasma phospholipid transfer protein deficiency in mice is associated with a reduced thrombotic response to acute intravascular oxidative stress. Arterioscler Thromb Vasc Biol 2010; 30:2452-7. [PMID: 20864671 DOI: 10.1161/atvbaha.110.207654] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Earlier in vitro studies suggested a putative role for the plasma phospholipid transfer protein (PLTP) in the modulation of blood coagulation. The effect of PLTP expression on blood coagulation under both basal and oxidative stress conditions was compared here in wild-type and PLTP-deficient (PLTP-/-) mice. METHODS AND RESULTS Under basal conditions, PLTP deficiency was associated with an extended tail bleeding time despite a significant depletion of vascular α-tocopherol content and an impairment of endothelial function. When acute oxidative stress was generated in vivo in the brain vasculature, the steady state levels of oxidized lipid derivatives, the extent of blood vessel occlusion, and the volume of ischemic lesions were more severe in wild-type than in PLTP-/- mice. CONCLUSIONS In addition to its recognized hyperlipidemic, proinflammatory, and proatherogenic properties, PLTP increases blood coagulation and worsens the extent of ischemic lesions in response to acute oxidative stress. Thus, PLTP arises here as a cardiovascular risk factor for the late thrombotic events occurring in the acute phase of atherosclerosis.
Collapse
Affiliation(s)
- Catherine Desrumaux
- Institut National de la Santé et de la Recherche Médicale, UMR866, Dijon, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Lemaire-Ewing S, Desrumaux C, Néel D, Lagrost L. Vitamin E transport, membrane incorporation and cell metabolism: Is alpha-tocopherol in lipid rafts an oar in the lifeboat? Mol Nutr Food Res 2010; 54:631-40. [PMID: 20166147 DOI: 10.1002/mnfr.200900445] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Vitamin E is composed of closely related compounds, including tocopherols and tocotrienols. Studies of the last decade provide strong support for a specific role of alpha-tocopherol in cell signalling and the regulation of gene expression. It produces significant effects on inflammation, cell proliferation and apoptosis that are not shared by other vitamin E isomers with similar antioxidant properties. The different behaviours of vitamin E isomers might relate, at least in part, to the specific effects they exert at the plasma membrane. alpha-Tocopherol is not randomly distributed throughout the phospholipid bilayer of biological membranes, and as compared with other isomers, it shows a propensity to associate with lipid rafts. Distinct aspects of vitamin E transport and metabolism is discussed with emphasis on the interaction between alpha-tocopherol and lipid rafts and the consequences of these interactions on cell metabolism.
Collapse
|
24
|
Cheung MC, Vaisar T, Han X, Heinecke JW, Albers JJ. Phospholipid transfer protein in human plasma associates with proteins linked to immunity and inflammation. Biochemistry 2010; 49:7314-22. [PMID: 20666409 DOI: 10.1021/bi100359f] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Phospholipid transfer protein (PLTP), which associates with apolipoprotein A-I (the major HDL protein), plays a key role in lipoprotein remodeling. Because its level in plasma increases during acute inflammation, it may also play previously unsuspected roles in the innate immune system. To gain further insight into its potential physiological functions, we isolated complexes containing PLTP from plasma by immunoaffinity chromatography and determined their composition. Shotgun proteomics revealed that only 6 of the 24 proteins detected in the complexes were apolipoproteins. The most abundant proteins were clusterin (apoJ), PLTP itself, coagulation factors, complement factors, and apoA-I. Remarkably, 20 of the 24 proteins had known protein-protein interactions. Biochemical studies confirmed two previously established interactions and identified five new ones between PLTP and proteins. Moreover, clusterin, apoA-I, and apoE preserved the lipid-transfer activity of recombinant PLTP in the absence of lipid, indicating that these interactions may have functional significance. Unexpectedly, lipids accounted for only 3% of the mass of the PLTP complexes. Collectively, our observations indicate that PLTP in human plasma resides on lipid-poor complexes dominated by clusterin and proteins implicated in host defense and inflammation. They further suggest that protein-protein interactions drive the formation of PLTP complexes in plasma.
Collapse
Affiliation(s)
- Marian C Cheung
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, School of Medicine, University of Washington, Seattle, Washington 98109, USA
| | | | | | | | | |
Collapse
|
25
|
Civelek M, Grant GR, Irolla CR, Shi C, Riley RJ, Chiesa OA, Stoeckert CJ, Karanian JW, Pritchard WF, Davies PF. Prelesional arterial endothelial phenotypes in hypercholesterolemia: universal ABCA1 upregulation contrasts with region-specific gene expression in vivo. Am J Physiol Heart Circ Physiol 2009; 298:H163-70. [PMID: 19897713 DOI: 10.1152/ajpheart.00652.2009] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Atherosclerosis originates as focal arterial lesions having a predictable distribution to regions of bifurcations, branches, and inner curvatures where blood flow characteristics are complex. Distinct endothelial phenotypes correlate with regional hemodynamics. We propose that systemic risk factors modify regional endothelial phenotype to influence focal susceptibility to atherosclerosis. Transcript profiles of freshly isolated endothelial cells from three atherosusceptible and three atheroprotected arterial regions in adult swine were analyzed to determine the initial prelesional effects of hypercholesterolemia on endothelial phenotypes in vivo. Cholesterol efflux transporter ATP-binding cassette transporter A1 (ABCA1) was upregulated at all sites in response to short-term high-fat diet. Proinflammatory and antioxidative endothelial gene expression profiles were induced in atherosusceptible and atheroprotected regions, respectively. However, markers for endoplasmic reticulum stress, a signature of susceptible endothelial phenotype, were not further enhanced by brief hypercholesterolemia. Both region-specific and ubiquitous (ABCA1) phenotype changes were identified as early prelesional responses of the endothelium to hypercholesterolemia.
Collapse
Affiliation(s)
- Mete Civelek
- Institute for Medicine and Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Berdnikovs S, Abdala-Valencia H, McCary C, Somand M, Cole R, Garcia A, Bryce P, Cook-Mills JM. Isoforms of vitamin E have opposing immunoregulatory functions during inflammation by regulating leukocyte recruitment. THE JOURNAL OF IMMUNOLOGY 2009; 182:4395-405. [PMID: 19299740 DOI: 10.4049/jimmunol.0803659] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Reports indicate contradictory outcomes for anti-inflammatory functions of the alpha-tocopherol isoform of vitamin E in clinical studies of asthma and atherosclerosis. These seemingly disparate clinical results are consistent with novel unrecognized properties of isoforms of vitamin E reported in this study. We demonstrate that the isoform d-gamma-tocopherol elevates inflammation in experimental asthma. Moreover, d-gamma-tocopherol, at as little as 10% the concentration of d-alpha-tocopherol, ablates the anti-inflammatory benefit of the d-alpha-tocopherol isoform. A mechanism for these opposing immunoregulatory functions of purified tocopherols at physiological concentrations is not through modulation of expression of several cytokines, chemokines, or adhesion molecules, but is, at least in part, by regulation of endothelial cell signals during leukocyte recruitment. These opposing regulatory functions of vitamin E isoforms have impact on interpretations of vitamin E studies. In summary, our studies with purified tocopherol isoforms alter our understanding of vitamin E regulation of vascular function and asthma.
Collapse
Affiliation(s)
- Sergejs Berdnikovs
- Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Vuletic S, Taylor BA, Tofler GH, Chait A, Marcovina SM, Schenck K, Albers JJ. SAA and PLTP activity in plasma of periodontal patients before and after full-mouth tooth extraction. Oral Dis 2009; 14:514-9. [PMID: 18826383 DOI: 10.1111/j.1601-0825.2007.01411.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE To assess whether treatment of advanced periodontal disease affects plasma levels of serum amyloid A (SAA) and phospholipid transfer protein (PLTP) activity. DESIGN We measured the levels of SAA and PLTP activity in plasma of 66 patients with advanced periodontal disease before and after treatment by full-mouth tooth extraction (FME). RESULTS At baseline, median SAA levels in our study population were within the normal range (2.7 microg ml(-1)) but SAA was elevated (>5 microg ml(-1)) in 18% of periodontitis patients. Three months after FME, SAA levels were significantly reduced (P = 0.04). SAA did not correlate with any of the periodontal disease parameters. PLTP activity was elevated in patients with periodontitis, compared to the PLTP activity reference group (age-matched systemically healthy adults, n = 29; 18 micromol ml(-1) h(-1)vs 13 micromol ml(-1) h(-1), respectively, P = 0.002). PLTP activity inversely correlated with average periodontal pocket depth (PPD) per tooth (r(s) = -0.372; P = 0.002). Three months after FME, median PLTP activity did not change significantly. CONCLUSIONS Full-mouth tooth extraction significantly reduces SAA, a marker of inflammation, while it does not affect plasma PLTP activity. However, the inverse correlation between PLTP activity and average PPD suggests that increased PLTP activity may limit periodontal tissue damage.
Collapse
Affiliation(s)
- S Vuletic
- Department of Medicine, University of Washington, Seattle, Washington 98109, United States.
| | | | | | | | | | | | | |
Collapse
|
28
|
Hacquebard M, Vandenbranden M, Malaisse WJ, Ruysschaert JM, Deckelbaum RJ, Carpentier YA. Vitamin E transfer from lipid emulsions to plasma lipoproteins: mediation by multiple mechanisms. Lipids 2008; 43:663-71. [PMID: 18509690 DOI: 10.1007/s11745-008-3184-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2007] [Accepted: 04/17/2008] [Indexed: 11/30/2022]
Abstract
The present study determined alpha-tocopherol mass transfer from an alpha-tocopherol-rich emulsion to LDL and HDL, and assessed the potential of different mechanisms to modulate alpha-tocopherol transfers. Emulsion particles rich in alpha-tocopherol were incubated in vitro with physiological concentrations of LDL or HDL. The influence of plasma proteins was assessed by adding human lipoprotein poor plasma (LPP) fraction with intact vs heat inactivated PLTP, or with a specific cholesteryl ester transfer protein (CETP) inhibitor, or by adding purified PLTP or pig LPP which lacks CETP activity. After 4 h incubation in absence of LPP, alpha-tocopherol content was increased by ~80% in LDL and ~160% in HDL. Addition of LPP markedly enhanced alpha-tocopherol transfer leading to 350-400% enrichment in LDL or HDL at 4 h. Higher (~10 fold) enrichment was achieved after 20 h incubation with LPP. Facilitation of alpha-tocopherol transfer was (i) more than 50% higher with human vs pig LPP (despite similar PLTP phospholipid transfer activity), (ii) reduced by specific CETP activity inhibition, (iii) not fully suppressed by heat inactivation, and (iv) not restored by purified PLTP. In conclusion, alpha-tocopherol content in LDL and HDL can be markedly raised by rapid transfer from an alpha-tocopherol-rich emulsion. Our results indicate that alpha-tocopherol mass transfer between emulsion particles and lipoproteins is mediated by more than one single mechanism and that this transfer may be facilitated not only by PLTP but likely also by other plasma proteins such as CETP.
Collapse
Affiliation(s)
- M Hacquebard
- Université Libre de Bruxelles, 1070, Brussels, Belgium.
| | | | | | | | | | | |
Collapse
|
29
|
Tölle M, Pawlak A, Schuchardt M, Kawamura A, Tietge UJ, Lorkowski S, Keul P, Assmann G, Chun J, Levkau B, van der Giet M, Nofer JR. HDL-associated lysosphingolipids inhibit NAD(P)H oxidase-dependent monocyte chemoattractant protein-1 production. Arterioscler Thromb Vasc Biol 2008; 28:1542-8. [PMID: 18483405 DOI: 10.1161/atvbaha.107.161042] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
OBJECTIVE High-density lipoprotein (HDL) levels are inversely proportional to the risk of atherosclerosis, but mechanisms of HDL atheroprotection remain unclear. Monocyte chemoatractant protein-1 (MCP-1) constitutes an early component of inflammatory response in atherosclerosis. Here we investigated the influence of HDL on MCP-1 production in vascular smooth muscle cells (VSMCs) and rat aortic explants. METHODS AND RESULTS HDL inhibited the thrombin-induced production of MCP-1 in a concentration-dependent manner. The HDL-dependent inhibition of MCP-1 production was accompanied by the suppression of reactive oxygen species (ROS), which regulate the MCP-1 production in VSMCs. HDL inhibited NAD(P)H oxidase, the preponderant source of ROS in the vasculature, and prevented the activation of Rac1, which precedes NAD(P)H-oxidase activation. The HDL capacity to inhibit MCP-1 production, ROS generation, and NAD(P)H-oxidase activation was emulated by sphingosine 1-phosphate (S1P) and sphingosylphosphorylcholine (SPC), two lysosphingolipids present in HDL, but not by apolipoprotein A-I. HDL-, S1P-, and SPC-induced inhibition of MCP-1 production was attenuated in VSMCs pretreated with VPC23019, an antagonist of lysosphingolipid receptors S1P(1) and S1P(3), but not by JTE013, an antagonist of S1P(2). In addition, HDL, S1P, and SPC failed to inhibit MCP1 production and ROS generation in aortas from S1P(3)- and SR-B1-deficient mice. CONCLUSIONS HDL-associated lysosphingolipids inhibit NAD(P)H oxidase-dependent ROS generation and MCP-1 production in a process that requires coordinate signaling through S1P(3) and SR-B1 receptors.
Collapse
Affiliation(s)
- Markus Tölle
- Charite - Campus Benjamin Franklin, Medizinische Klinik, Berlin, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Gohil K, Oommen S, Quach HT, Vasu VT, Aung HH, Schock B, Cross CE, Vatassery GT. Mice lacking alpha-tocopherol transfer protein gene have severe alpha-tocopherol deficiency in multiple regions of the central nervous system. Brain Res 2008; 1201:167-76. [PMID: 18299118 DOI: 10.1016/j.brainres.2008.01.044] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2007] [Revised: 01/14/2008] [Accepted: 01/15/2008] [Indexed: 01/09/2023]
Abstract
Ataxia with vitamin E deficiency is caused by mutations in alpha-tocopherol transfer protein (alpha-TTP) gene and it can be experimentally generated in mice by alpha-TTP gene inactivation (alpha-TTP-KO). This study compared alpha-tocopherol (alpha-T) concentrations of five brain regions and of four peripheral organs from 5 months old, male and female, wild-type (WT) and alpha-TTP-KO mice. All brain regions of female WT mice contained significantly higher alpha-T than those from WT males. alpha-T concentration in the cerebellum was significantly lower than that in other brain regions of WT mice. These sex and regional differences in brain alpha-T concentrations do not appear to be determined by alpha-TTP expression which was undetectable in all brain regions. All the brain regions of alpha-TTP-KO mice were severely depleted in alpha-T. The concentration of another endogenous antioxidant, total glutathione, was unaffected by gender but was decreased slightly but significantly in most brain regions of alpha-TTP-KO mice. The results show that both gender and the hepatic alpha-TTP, but not brain alpha-TTP gene expression are important in determining alpha-T concentrations within the brain. Interestingly, functional abnormality (ataxia) develops only very late in alpha-TTP-KO mice in spite of the severe alpha-tocopherol deficiency in the brain starting at an early age.
Collapse
Affiliation(s)
- Kishorchandra Gohil
- Department of Internal Medicine, Genome and Biomedical Sciences Facility, University of California, Davis, CA 95616, USA.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
The blood-brain barrier (BBB) is composed of uniquely differentiated brain microvascular endothelial cells (BMEC). Often, it is of interest to replicate these attributes in the form of an in vitro model, and such models are widely used in the research community. However, the BMEC used to create in vitro BBB models de-differentiate in culture and lose many specialized characteristics. These changes are poorly understood at a molecular level, and little is known regarding the consequences of removing BMEC from their local in vivo microenvironment. To address these issues, suppression subtractive hybridization (SSH) was used to identify 25 gene transcripts that were differentially expressed between in vivo and in vitro BMEC. Genes affected included those involved in angiogenesis, transport and neurogenesis, and real-time quantitative polymerase chain reaction (qPCR) verified transcripts were primarily and significantly downregulated. Since this quantitative gene panel represented those BMEC characteristics lost upon culture, we used it to assess how culture manipulation, specifically BMEC purification and barrier induction by hydrocortisone, influenced the quality of in vitro models. Puromycin purification of BMEC elicited minimal differences compared with untreated BMEC, as assessed by qPCR. In contrast, qPCR-based gene panel analysis after induction with hydrocortisone indicated a modest shift of 10 of the 23 genes toward a more 'in vivo-like' gene expression profile, which correlated with improved barrier phenotype. Genomic analysis of BMEC de-differentiation in culture has thus yielded a functionally diverse set of genes useful for comparing the in vitro and in vivo BBB.
Collapse
Affiliation(s)
| | - Eric V. Shusta
- To whom correspondence should be addressed: Eric V. Shusta Department of Chemical and Biological Engineering University of Wisconsin-Madison 1415 Engineering Drive Madison, WI 53706 Ph: (608) 265-5103 Fax: (608) 262-5434
| |
Collapse
|
32
|
Ciocoiu M, Badescu MM, Lupusoru EC. The intervention of antioxidant therapy on platelet adhesion and immunomodulation in experimental physical stress. Free Radic Res 2007; 41:829-38. [PMID: 17577744 DOI: 10.1080/10715760701416434] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
During effort overstress the reactive oxygen species act chiefly on unsaturated lipids, inducing the formation of certain peroxidation products. We have investigated malondialdehide (MDA), platelet adhesion index, and immunological activation parameters during effort overstress and administration of vitamins E and C. Biochemical measurements were performed on erythrocytes and heart homogenate. In the vitamin E supplemented group, the platelet adhesion index was constantly correlated with the MDA level (p < 0.001). There is a protecting effect concerning the oxidative stress in animals pretreated with vitamin E and C, which is expressed through the diminution of the MDA quantity both in the erythrocyte and in the heart. The physical effort required by swimming led to a decrease in the NBT test values and in the activity of the serum complement. The steady administration of vitamin E in the effort overstress, due to its antioxidant properties, causes the progressive decrease in peroxidation and platelet adhesion.
Collapse
Affiliation(s)
- Manuela Ciocoiu
- Department of Pathophysiology, University of Medicine and Pharmacy, Iasi, Romania.
| | | | | |
Collapse
|
33
|
Ogier N, Klein A, Deckert V, Athias A, Bessède G, Le Guern N, Lagrost L, Desrumaux C. Cholesterol accumulation is increased in macrophages of phospholipid transfer protein-deficient mice: normalization by dietary alpha-tocopherol supplementation. Arterioscler Thromb Vasc Biol 2007; 27:2407-12. [PMID: 17717294 DOI: 10.1161/atvbaha.107.151753] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Phospholipid transfer protein (PLTP) is a multifunctional, extracellular lipid transport protein that plays a major role in lipoprotein metabolism and atherosclerosis. Recent in vivo studies suggested that unlike systemic PLTP, macrophage-derived PLTP would be antiatherogenic. The present study aimed at characterizing the atheroprotective properties of macrophage-derived PLTP. METHODS AND RESULTS Peritoneal macrophages were isolated from PLTP-deficient and wild-type mice and their biochemical characteristics were compared. It is shown that macrophages isolated from PLTP-deficient mice have increased basal cholesterol content and accumulate more cholesterol in the presence of LDL compared with wild-type cells. Cholesterol parameters in macrophages of PLTP-deficient mice were normalized by dietary alpha-tocopherol supplementation. CONCLUSIONS The antiatherogenic properties of macrophage-derived PLTP are related at least in part to its ability to reduce cholesterol accumulation in macrophages through changes in the alpha-tocopherol content and oxidative status of the cells.
Collapse
Affiliation(s)
- Nicolas Ogier
- INSERM U866, Centre de Recherche, IFR Santé-STIC, Faculté de Médecine, Université de Bourgogne, Dijon, France
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Schgoer W, Mueller T, Jauhiainen M, Wehinger A, Gander R, Tancevski I, Salzmann K, Eller P, Ritsch A, Haltmayer M, Ehnholm C, Patsch JR, Foeger B. Low phospholipid transfer protein (PLTP) is a risk factor for peripheral atherosclerosis. Atherosclerosis 2007; 196:219-226. [PMID: 17553507 DOI: 10.1016/j.atherosclerosis.2007.04.046] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2006] [Revised: 04/18/2007] [Accepted: 04/27/2007] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Phospholipid transfer protein (PLTP) facilitates cholesterol efflux from cells, intravascular HDL remodelling and transfer of vitamin E and endotoxin. In humans, the relationship of PLTP to atherosclerosis is unknown. However, strong coronary risk factors like obesity, diabetes, cigarette smoking and inflammation increase circulating levels of active PLTP. The aim of the present, cross-sectional study was to analyze the relationship of PLTP to peripheral arterial disease, a marker of generalized atherosclerosis, independently of potentially confounding factors like obesity, diabetes and smoking. METHODS We performed a case control study in 153 patients with symptomatic peripheral arterial disease (PAD) and 208 controls free of vascular disease. Smokers and patients with diabetes mellitus were excluded. A lipoprotein-independent assay was used for measurement of circulating bioactive PLTP and an ELISA utilizing a monoclonal antibody was used to analyze PLTP mass. RESULTS PLTP activity was significantly decreased in patients with PAD 5.5 (4.6-6.4)(median (25th-75th percentile)) versus 5.9 (5.1-6.9) micromol/mL/h in controls (p=0.001). In contrast, PLTP mass was similar in patients with PAD 8.5 microg/mL (7.3-9.5) and in controls 8.3 microg/mL (6.9-9.7) (p=0.665). Multivariate logistic regression analysis revealed that PLTP activity is independently associated with the presence of PAD. PLTP activity was similar in patients with and without lipid-lowering drugs (p=0.396). CONCLUSION Our results show that in non-diabetic, non-smoking subjects low rather than high PLTP activity is a marker for the presence of peripheral arterial disease and that distribution of PLTP between high-activity and low-activity forms may be compromised in atherosclerosis.
Collapse
Affiliation(s)
- Wilfried Schgoer
- Department of Internal Medicine, Medical University Innsbruck, Austria
| | - Thomas Mueller
- Department of Laboratory Medicine, Konventhospital Barmherzige Brueder, Linz, Austria
| | - Matti Jauhiainen
- Department of Molecular Medicine, National Public Health Institute, Biomedicum, Helsinki, Finland
| | - Andreas Wehinger
- Department of Internal Medicine, Medical University Innsbruck, Austria; Department of Internal Medicine, Landeskrankenhaus Bregenz, Austria
| | - Roland Gander
- Department of Internal Medicine, Medical University Innsbruck, Austria
| | - Ivan Tancevski
- Department of Internal Medicine, Medical University Innsbruck, Austria
| | - Karin Salzmann
- Department of Internal Medicine, Medical University Innsbruck, Austria
| | - Philipp Eller
- Department of Internal Medicine, Medical University Innsbruck, Austria
| | - Andreas Ritsch
- Department of Internal Medicine, Medical University Innsbruck, Austria
| | - Meinhard Haltmayer
- Department of Laboratory Medicine, Konventhospital Barmherzige Brueder, Linz, Austria
| | - Christian Ehnholm
- Department of Molecular Medicine, National Public Health Institute, Biomedicum, Helsinki, Finland
| | - Josef R Patsch
- Department of Internal Medicine, Medical University Innsbruck, Austria
| | - Bernhard Foeger
- Department of Internal Medicine, Medical University Innsbruck, Austria; Department of Internal Medicine, Landeskrankenhaus Bregenz, Austria.
| |
Collapse
|
35
|
Wehinger A, Tancevski I, Schgoer W, Eller P, Hochegger K, Morak M, Hermetter A, Ritsch A, Patsch JR, Foeger B. Phospholipid Transfer Protein Augments Apoptosis in THP-1–Derived Macrophages Induced by Lipolyzed Hypertriglyceridemic Plasma. Arterioscler Thromb Vasc Biol 2007; 27:908-15. [PMID: 17272752 DOI: 10.1161/01.atv.0000259361.91267.8c] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Lipolysis of triglyceride-rich lipoproteins (TGRLPs) generates phospholipid-rich surface remnants and induces cytotoxic effects in adjacent vascular cells. We hypothesized that by integrating surface remnants into HDL, phospholipid transfer protein (PLTP) alleviates cytotoxicity. METHODS AND RESULTS To test this hypothesis and gain insight into cytotoxicity during the postprandial phase in vivo, we injected normo-TG and hyper-TG human volunteers after a standardized fat meal (postprandial sample) with heparin, thereby stimulating lipolysis (postprandial heparinized sample). Incubation of (primary) human macrophages and primary human endothelial cells with postprandial heparinized hyper-TG plasma induced pronounced cytotoxic effects that were dose dependent on the TG content of the sample. No such effects were seen with normo-TG and postprandial hyper-TG plasma. In vitro lipolysis of VLDL and chylomicrons indicated that both lipoprotein fractions can cause cytotoxicity. Interestingly, in experiments with THP-1-derived macrophages stably transfected with PLTP, PLTP substantially augmented both net phospholipid uptake and apoptotic cell death due to postprandial heparinized hyper-TG plasma. We observed that activation of caspase-3/7, poly-ADP-ribose polymerase, and enhanced bioactivity of acid sphingomyelinase may all contribute to this augmented apoptosis. CONCLUSIONS Our data show that lipolysis of TGRLPs and their remodelling by PLTP interact to disturb cellular phospholipid flux and intracellular signaling processes, ultimately leading to apoptosis in human macrophages and endothelial cells.
Collapse
Affiliation(s)
- Andreas Wehinger
- Department of Internal Medicine, Medical University Innsbruck, Innsbruck, Austria
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Heart disease is the number one cause of death in the United States and has long been recognized to be multifactorial. A growing body of evidence suggests that not only free radical-mediated reactions but also inflammatory responses play major roles in atherogenesis. Vitamin E has both antioxidant and antiinflammatory properties and is the most widely studied vitamin in clinical trials and thus will be the primary example used in this review. Clinical trials of vitamin E efficacy, in hindsight, have been overly optimistic in their expectation that a vitamin could reverse poor dietary habits and a sedentary lifestyle as well as provide benefit beyond that of pharmaceutical agents in treating heart disease. However, it is also apparent that most Americans do not consume dietary amounts adequate to meet established vitamin E requirements. In response to oxidative stressors, vitamin E can decrease biomarkers of lipid peroxidation, is itself killed, and requires optimal vitamin C status to function most effectively. Thus, adequate vitamin E intakes are clearly needed, but what is adequate for what function has yet to be defined. It is noteworthy that in most trials, biomarkers were not used nor were oxidative stress and lipid peroxidation markers used or plasma vitamin E concentrations measured.
Collapse
Affiliation(s)
- Maret G Traber
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA.
| |
Collapse
|
37
|
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: 2.7] [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
|
38
|
Cheung MC, Albers JJ. Active plasma phospholipid transfer protein is associated with apoA-I- but not apoE-containing lipoproteins. J Lipid Res 2006; 47:1315-21. [PMID: 16520487 DOI: 10.1194/jlr.m600042-jlr200] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Plasma phospholipid transfer protein (PLTP) is a multifaceted protein with diverse biological functions. It has been shown to exist in both active and inactive forms. To determine the nature of lipoproteins associated with active PLTP, plasma samples were adsorbed with anti-A-I, anti-A-II, or anti-E immunoadsorbent, and PLTP activity was measured in the resulting plasma devoid of apolipoprotein A-I (apoA-I), apoA-II, or apoE. Anti-A-I and anti-A-II immunoadsorbents removed 98 +/- 1% (n = 8) and 38 +/- 25% (n = 7) of plasma PLTP activity, respectively. In contrast, only 1 +/- 5% of plasma PLTP activity was removed by anti-E immunoadsorbent (n = 7). Dextran sulfate (DS) cellulose did not bind apoA-I, but it removed 83 +/- 5% (n = 4) of the PLTP activity in plasma. In size-exclusion chromatography, PLTP activity removed by anti-A-I or anti-A-II immunoadsorbent was associated primarily with particles of a size corresponding to HDL, whereas a substantial portion of the PLTP activity dissociated from DS cellulose was found in particles larger or smaller than HDL. These data show the following: 1) active plasma PLTP is associated primarily with apoA-I- but not apoE-containing lipoproteins; 2) active PLTP is present in HDL particles with and without apoA-II, and its distribution between these two HDL subpopulations varies widely among individuals; and 3) DS cellulose can remove active PLTP from apoA-I-containing lipoproteins, and this process creates new active PLTP-containing particles in vitro.
Collapse
Affiliation(s)
- Marian C Cheung
- Division of Metabolism, Endocrinology, and Nutrition, Department of Medicine, School of Medicine, University of Washington, Seattle, WA, USA.
| | | |
Collapse
|
39
|
Drouineaud V, Lagrost L, Klein A, Desrumaux C, Le Guern N, Athias A, Ménétrier F, Moiroux P, Sagot P, Jimenez C, Masson D, Deckert V. Phospholipid transfer protein (PLTP) deficiency reduces sperm motility and impairs fertility of mouse males. FASEB J 2006; 20:794-6. [PMID: 16467369 DOI: 10.1096/fj.05-5385fje] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Vitamin E was discovered for its implication in reproductive biology, and its transport in mammalian plasma and brain was shown to be governed by plasma phospholipid transfer protein (PLTP). We show that PLTP deficiency is associated with hypofertility of mouse males but not mouse females, and it accounts for a significant decrease in total number of pups produced over a 2-month breeding period of PLTP knocked out mice (-32%, P<0.03). PLTP is highly expressed in epididymis of mouse males, and alpha-tocopherol, the main vitamin E isomer in vivo, was significantly less abundant in cauda and caput epididymis of PLTP-deficient mice as compared with wild-type counterparts (caput: -26%, P<0.05; cauda: -21%, P<0.05). Mature spermatozoa from PLTP-deficient epididymis were shown to retain an abnormal alpha-tocopherol content. PLTP deficiency tended to reduce sperm motility as shown by a 24% reduction in spermatozoa with progressive motility (P<0.02), with no change in other sperm parameters as compared with wild-type males. Finally, in vitro fertilization rates of wild-type oocytes with spermatozoa from PLTP-deficient males were markedly reduced as compared with those measured with spermatozoa from wild-type males (-60%, P<0.05). It is concluded that PLTP is a new, key factor that determines sperm motility and male fertility.
Collapse
Affiliation(s)
- Véronique Drouineaud
- Laboratoire de Biologie de la Reproduction, EA Génétique et Reproduction 3185, CHU Dijon, Dijon, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Cheung MC, Brown BG, Marino Larsen EK, Frutkin AD, O'Brien KD, Albers JJ. Phospholipid transfer protein activity is associated with inflammatory markers in patients with cardiovascular disease. Biochim Biophys Acta Mol Basis Dis 2006; 1762:131-7. [PMID: 16216472 DOI: 10.1016/j.bbadis.2005.09.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2005] [Revised: 09/02/2005] [Accepted: 09/07/2005] [Indexed: 11/28/2022]
Abstract
Plasma phospholipid lipid transfer protein (PLTP) has several known key functions in lipoprotein metabolism. Recent studies suggest that it also may play a role in the inflammatory response. Inflammatory cell activity contributes to the development of atherosclerosis. To seek further evidence for the association of PLTP with inflammation, we studied the relationship between PLTP activity and five inflammatory markers [C-reactive protein (CRP), serum amyloid A (SAA), interleukin 6 (IL-6), white blood cells (WBC), and fibrinogen] in 93 patients with low HDL and cardiovascular disease (CVD). Plasma PLTP activity had the strongest correlation with CRP (r=0.332, P<0.001) followed by SAA (r=0.239, P=0.021). PLTP, CRP, and SAA were significantly associated with body mass index (BMI), insulin or glucose, apolipoprotein (apo) B, and/or apo E level (r=0.264-0.393, P<0.01). PLTP, SAA, and IL-6 also were associated with the concentration of HDL particles without apo A-II [Lp(A-I)](r=0.373-0.472, P<0.005, n=56), but not particles with apo A-II. Smoking was associated with increased PLTP activity, CRP, and WBC, and hypertension with increased PLTP activity. In linear models, CRP remained significantly associated with PLTP after adjustment of CVD risk factors and insulin resistance. Also, much of the variability of plasma PLTP activity was explained by CRP, BMI, Lp(A-I), smoking, glucose, and blood pressure. These findings show for the first time that plasma PLTP activity is associated positively with CRP in CVD, a state of chronic inflammation.
Collapse
Affiliation(s)
- Marian C Cheung
- Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, WA 98109-4517, USA.
| | | | | | | | | | | |
Collapse
|
41
|
Oomen PHN, van Tol A, Hattori H, Smit AJ, Scheek LM, Dullaart RPF. Human plasma phospholipid transfer protein activity is decreased by acute hyperglycaemia: studies without and with hyperinsulinaemia in Type 1 diabetes mellitus. Diabet Med 2005; 22:768-74. [PMID: 15910630 DOI: 10.1111/j.1464-5491.2005.01521.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
AIMS Little is known about the regulation of phospholipid transfer protein (PLTP), that plays a key role in lipoprotein metabolism. PLTP secretion may be up-regulated by glucose in vitro, whereas plasma PLTP activity is decreased by exogenous hyperinsulinaemia and glucose-induced hyperinsulinaemia in vivo. In the present study, we evaluated the separate effects of hyperglycaemia and hyperinsulinaemia in C-peptide-negative Type 1 diabetic patients. METHODS The protocol was carried out in 16 patients (eight females). In each individual, plasma PLTP mass and activity (measured by enzyme-linked immuno-sorbent assay and liposome-high density lipoprotein system, respectively) as well as plasma cholesteryl ester transfer protein (CETP) activity, lipids and apolipoprotein levels were determined at the end of four different glucose clamps, each lasting 210 min: standard insulin (30 mU/kg/h) and standard glucose (glucose 5.0 mmol/l) (SI-SG), standard insulin and high glucose (glucose 12 mmol/l) (SI-HG), high insulin (150 mU/kg/h) and standard glucose (HI-SG), and high insulin and high glucose (HI-HG). RESULTS Plasma lipids and (apo)lipoproteins, measured at the end of the SI-HG, HI-SG and HI-HG clamps, were not significantly different compared with the levels obtained at the end of the SI-SG clamp. Median plasma PLTP mass and activity at the end of the SI-SG clamp were 12.8 mg/l and 13.2 micromol/ml/h, respectively. Median plasma PLTP mass decreased by 9.1% at the end of the HI-HG clamp (P < 0.01), whereas the changes at the end of the SI-HG and HI-SG clamps were not significant. Median plasma PLTP activity decreased by 5.7, 4.6 and 8.6% at the end of the SI-HG, HI-SG and HI-HG clamps, respectively (all P < 0.05). Median plasma CETP activity was 177 nmol/ml/h at the end of the SI-SG clamp, and decreased by 4.9% (P < 0.05) and by 8.3% (P < 0.05) at the end of the HI-SG and the HI-HG clamps, respectively. Plasma CETP activity did not change significantly at the end of the SI-HG clamp. CONCLUSIONS The present study demonstrates that plasma PLTP activity is independently decreased by acute hyperglycaemia and hyperinsulinaemia in humans in vivo. These data do not support a direct role of short-term hyperglycaemia in up-regulating plasma PLTP levels.
Collapse
Affiliation(s)
- P H N Oomen
- Department of Endocrinology, University Hospital Groningen, Groningen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW Large-scale clinical trials have failed to demonstrate a benefit for vitamin E supplementation in cardiovascular prevention. This contrasts with previous epidemiological studies indicating that individuals with high vitamin E status benefit from protection against chronic illnesses, including cardiovascular diseases. These conflicting results suggest that the metabolism of supplemental versus naturally delivered vitamin E and their potential roles, other than a potent antioxidant action, are not fully understood. The purpose of this review is to provide an update on current knowledge on the intestinal absorption of vitamin E, its plasma transport and its supply to cells. The review will also discuss the intravascular metabolism of intravenously delivered vitamin E. RECENT FINDINGS Although the luminal digestion of vitamin E is fairly well understood, several pathways regulating net vitamin E absorption remain to be elucidated. In several cell types, cholesterol and vitamin E share common mechanisms for cellular uptake (scavenger receptor B type I and LDL receptors) and efflux (ABCA1 transporters). The role of specific binding proteins in alpha-tocopherol intracellular trafficking is increasingly being understood, leading to new insights into the non-antioxidant functions of vitamin E. SUMMARY Substantial progress has been made in characterizing the plasma transport of vitamin E and its delivery to cells. Mechanisms regulating the balance between the cellular uptake and efflux of vitamin E are under investigation. Vitamin E is not only an antioxidant but may also modulate pathways of cell signalling and gene expression. The translation of this new knowledge into clinical studies will help define future indications for vitamin E supplementation.
Collapse
Affiliation(s)
- Mirjam Hacquebard
- L. Deloyers Laboratory for Experimental Surgery, Université Libre de Bruxelles, Brussels, Belgium
| | | |
Collapse
|
43
|
Schneider M, Vergès B, Klein A, Miller ER, Deckert V, Desrumaux C, Masson D, Gambert P, Brun JM, Fruchart-Najib J, Blache D, Witztum JL, Lagrost L. Alterations in plasma vitamin E distribution in type 2 diabetic patients with elevated plasma phospholipid transfer protein activity. Diabetes 2004; 53:2633-9. [PMID: 15448094 DOI: 10.2337/diabetes.53.10.2633] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Mouse studies indicated that plasma phospholipid transfer protein (PLTP) determines the plasma distribution of vitamin E, a potent lipophilic antioxidant. Vitamin E distribution, antioxidant status, and titer of anti-oxidized LDLs (oxLDL) autoantibodies were evaluated in plasma from control subjects (n = 31) and type 2 diabetic patients (n = 31) with elevated plasma PLTP concentration. Unlike diabetic and control HDLs, which displayed similar vitamin E contents, diabetic VLDLs and diabetic LDLs contained fewer vitamin E molecules than normal counterparts. Plasma PLTP concentration in diabetic plasmas correlated negatively with vitamin E in VLDL+LDL, but positively with vitamin E in HDL, with an even stronger correlation with the VLDL+LDL-to-HDL vitamin E ratio. Circulating levels of oxLDL were significantly higher in diabetic plasmas than in control plasmas. Whereas the titer of IgG autoantibodies to modified LDL did not differ significantly between diabetic patients and control subjects, diabetic plasmas showed significantly lower levels of potentially protective IgM autoantibodies. The present observations support a pathophysiological role of PLTP in decreasing the vitamin E content of apolipoprotein B-containing lipoproteins, but not of HDL in plasma of type 2 diabetic patients, contributing to a greater potential for LDL oxidation.
Collapse
Affiliation(s)
- Martina Schneider
- Institut National de la Santé et de la Recherche Médicale, U498, Dijon Cedex, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Mardones P, Rigotti A. Cellular mechanisms of vitamin E uptake: relevance in α-tocopherol metabolism and potential implications for disease. J Nutr Biochem 2004; 15:252-60. [PMID: 15135148 DOI: 10.1016/j.jnutbio.2004.02.006] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2003] [Indexed: 01/24/2023]
Abstract
alpha-Tocopherol is an essential micronutrient involved in various oxidative stress-related processes. Because of its hydrophobic nature, alpha-tocopherol is transported in plasma lipoproteins, and the pathways involved in its cellular uptake are closely related to the lipoprotein metabolism. alpha-Tocopherol transfer from plasma to cells can occur by different mechanisms such as uptake facilitated by lipid transfer proteins and lipases, receptor-mediated lipoprotein endocytosis, and selective lipid uptake. Here we discuss recent progress in understanding the physiological and pathophysiological relevance of these different pathways for cellular uptake of vitamin E in vivo. This review is mainly focused on the role of the scavenger receptor class B type I (SR-BI) on alpha-tocopherol metabolism and its potential implications for disease conditions.
Collapse
Affiliation(s)
- Pablo Mardones
- Departamento de Gastroenterología, Facultad de Medicina, Pontificia Universidad Católica, Marcoleta 367, Santiago, Chile
| | | |
Collapse
|
45
|
Tholstrup T, Ehnholm C, Jauhiainen M, Petersen M, Høy CE, Lund P, Sandström B. Effects of medium-chain fatty acids and oleic acid on blood lipids, lipoproteins, glucose, insulin, and lipid transfer protein activities. Am J Clin Nutr 2004; 79:564-9. [PMID: 15051598 DOI: 10.1093/ajcn/79.4.564] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Dietary medium-chain fatty acids (MCFAs) are of nutritional interest because they are more easily absorbed from dietary medium-chain triacylglycerols (MCTs) than are long-chain fatty acids from, for example, vegetable oils. It has generally been claimed that MCFAs do not increase plasma cholesterol, although this claim is poorly documented. OBJECTIVE We compared the effects of a diet rich in either MCFAs or oleic acid on fasting blood lipids, lipoproteins, glucose, insulin, and lipid transfer protein activities in healthy men. DESIGN In a study with a double-blind, randomized, crossover design, 17 healthy young men replaced part of their habitual dietary fat intake with 70 g MCTs (66% 8:0 and 34% 10:0) or high-oleic sunflower oil (89.4% 18:1). Each intervention period lasted 21 d, and the 2 periods were separated by a washout period of 2 wk. Blood samples were taken before and after the intervention periods. RESULTS Compared with the intake of high-oleic sunflower oil, MCT intake resulted in 11% higher plasma total cholesterol (P = 0.0005), 12% higher LDL cholesterol (P = 0.0001), 32% higher VLDL cholesterol (P = 0.080), a 12% higher ratio of LDL to HDL cholesterol (P = 0.002), 22% higher plasma total triacylglycerol (P = 0.0361), and higher plasma glucose (P = 0.033). Plasma HDL-cholesterol and insulin concentrations and activities of cholesterol ester transfer protein and phospholipid transfer protein did not differ significantly between the diets. CONCLUSIONS Compared with fat high in oleic acid, MCT fat unfavorably affected lipid profiles in healthy young men by increasing plasma LDL cholesterol and triacylglycerol. No changes in the activities of phospholipid transfer protein and cholesterol ester transfer protein were evident.
Collapse
Affiliation(s)
- Tine Tholstrup
- Research Department of Human Nutrition, Center of Advanced Food Research, The Royal Veterinary and Agricultural University, Frederiksberg, Denmark.
| | | | | | | | | | | | | |
Collapse
|
46
|
Korstanje R, Albers JJ, Wolfbauer G, Li R, Tu AY, Churchill GA, Paigen BJ. Quantitative Trait Locus Mapping of Genes That Regulate Phospholipid Transfer Activity in SM/J and NZB/BlNJ Inbred Mice. Arterioscler Thromb Vasc Biol 2004; 24:155-60. [PMID: 14592843 DOI: 10.1161/01.atv.0000104241.44819.10] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [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), an important protein in the transfer of phospholipids between lipoprotein particles and in the remodeling of HDL, is regulated at both the transcriptional and the protein level. We performed quantitative trait locus (QTL) analysis to identify genomic loci regulating PLTP activity in mice. METHODS AND RESULTS Plasma PLTP activity was measured in 217 male F2 progeny from a SM/J x NZB/B1NJ intercross. Two QTL for plasma PLTP activity in mice fed chow (Pltpq1 and Pltpq2) were found on chromosomes 3 (34 cM, logarithm of odds [LOD] 3.5) and 10 (66 cM, LOD 4.1); two additional QTL in mice fed atherogenic diet (Pltpq3 and Pltpq4) were found on chromosomes 9 (56 cM, LOD 4.5) and 15 (34 cM, LOD 5.0); and one QTL (Pltiq1) for the inducibility of PLTP activity was found on chromosome 4 (70 cM, LOD 3.7). Several candidate genes for these 5 QTL were tested by sequence comparison and expression studies. CONCLUSIONS We identified five significant loci involved in PLTP activity in the mouse and provided supporting evidence for the candidacy of Nr1h4 and Apof as the genes underlying Pltpq2.
Collapse
|
47
|
Yang XP, Yan D, Qiao C, Liu RJ, Chen JG, Li J, Schneider M, Lagrost L, Xiao X, Jiang XC. Increased atherosclerotic lesions in apoE mice with plasma phospholipid transfer protein overexpression. Arterioscler Thromb Vasc Biol 2003; 23:1601-7. [PMID: 12855484 DOI: 10.1161/01.atv.0000085841.55248.13] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Plasma phospholipid transfer protein (PLTP) is involved in the metabolism of HDL and apolipoprotein B (apoB)-containing lipoproteins. Atherosclerosis susceptibility is decreased in mice with PLTP deficiency that is associated with decreased liver production of apoB-containing lipoproteins and increase in their antioxidant. To investigate additionally the effect of PLTP on the development of atherosclerosis, we overexpressed PLTP in mice. METHODS AND RESULTS PLTP was overexpressed in apoE knockout mice using an adenovirus-associated virus (AAV)-mediated system. Plasma PLTP activity was 1.3- to 2-fold higher in mice injected with AAV-PLTP than in mice injected with control AAV-GFP, and PLTP levels were sustained during the experiment period (4 months). We show that 2-fold increased PLTP activity results in (1) a decrease in HDL cholesterol, HDL phospholipid, and apoAI levels; (2) a decrease in vitamin E contents in total plasma and in individual lipoprotein fractions; (3) an increase in lipoprotein oxidizability as assessed by copper-induced formation of conjugated dienes; (4) an increase in autoantibodies against oxidized apoB-containing particles; and (5) an increase in atherosclerosis lesions in proximal aorta. CONCLUSIONS These observations indicate that elevated plasma PLTP levels constitute a novel, long-term risk factor for atherosclerosis.
Collapse
Affiliation(s)
- Xiao Ping Yang
- Department of Anatomy and Cell Biology, State University of New York, Downstate Medical Center, Brooklyn, NY 11203, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Desrumaux CM, Mak PA, Boisvert WA, Masson D, Stupack D, Jauhiainen M, Ehnholm C, Curtiss LK. Phospholipid transfer protein is present in human atherosclerotic lesions and is expressed by macrophages and foam cells. J Lipid Res 2003; 44:1453-61. [PMID: 12730304 DOI: 10.1194/jlr.m200281-jlr200] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Phospholipid transfer protein (PLTP) in plasma promotes phospholipid transfer from triglyceride-rich lipoproteins to HDL and plays a major role in HDL remodeling. Recent in vivo observations also support a key role for PLTP in cholesterol metabolism. Our immunohistochemical analysis of human carotid endarterectomy samples identified immunoreactive PLTP in areas that colocalized with CD68-positive macrophages, suggesting that PLTP could be produced locally by intimal macrophages. Using RT-PCR, Western blot analysis with a monoclonal anti-PLTP antibody, and a PLTP activity assay, we observed PLTP mRNA and protein expression in human macrophages. In adherent peripheral blood human macrophages, this PLTP expression was increased by culture with granulocyte macrophage colony-stimulating factor. Incubation of macrophages with acetylated-LDL induced an increase in PLTP mRNA and protein expression that paralleled cholesterol loading. PLTP expression was observed in elicited mouse peritoneal macrophages and in cultured Raw264.7 cells as well. Thus, this study demonstrates that PLTP is expressed by macrophages, is regulated by cholesterol loading, and is present in atherosclerotic lesions.
Collapse
|
49
|
Deckert V, Desrumaux C, Athias A, Duverneuil L, Palleau V, Gambert P, Masson D, Lagrost L. Prevention of LDL alpha-tocopherol consumption, cholesterol oxidation, and vascular endothelium dysfunction by polyphenolic compounds from red wine. Atherosclerosis 2002; 165:41-50. [PMID: 12208469 DOI: 10.1016/s0021-9150(02)00189-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Red wine polyphenolic compounds (RWPCs) have been demonstrated to possess antioxidant properties, and several studies have suggested that they might constitute a relevant dietary factor in the protection from coronary heart disease. The aim of the present study was to determine further the mechanism by which RWPCs can prevent the formation of vasoactive compounds in oxidized LDL. RWPCs were obtained from the Cabernet-Sauvignon grape variety. Human LDL was oxidized in the presence of CuSO(4) (ox-LDL). Vascular reactivity studies were conducted on rabbit aortic rings. RWPCs significantly reduced the formation of 7 beta-hydroxycholesterol and 7-ketocholesterol and in a lower extent the emergence of lysophosphatidylcholine in ox-LDL. The ability of RWPCs to prevent cholesterol oxide formation was directly dependent on the LDL alpha-tocopherol content. Once the LDL alpha-tocopherol has been consumed, RWPCs were no longer effective, indicating that RWPCs act by sparing endogenous alpha-tocopherol. As a consequence of the preservation of the endogenous alpha-tocopherol content of LDL, RWPCs could prevent the inhibition of the acetylcholine-mediated endothelium-dependent relaxation of rabbit aorta which was linked to a direct effect on NO release. Independently of a treatment with ox-LDL, RWPC exerted a concentration-dependent and persistent inhibitory effect on the norepinephrine-induced contraction of rabbit aorta. In conclusion, RWPCs can preserve a normal vascular reactivity by acting at different stages of the cascade that leads to lipid oxidation, endothelium dysfunction and vasospasm.
Collapse
Affiliation(s)
- Valérie Deckert
- Laboratoire de Biochimie des Lipoprotéines-INSERM U498, Faculté de Médecine, 7 Boulevard Jeanne d'Arc, BP 87900, 21079 Dijon Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
The tissue-specific gene expression at the brain microvasculature, which forms the blood-brain barrier (BBB) can be elucidated with a brain vascular genomics program, which starts with the isolation of gene products derived from purified brain microvessels. Genes commonly expressed in peripheral organs are subtracted with the suppression subtractive hybridization method using driver cDNA produced from a pool of rat liver/kidney-derived poly A+RNA. From a screen of 480 clones in the subtracted tester cDNA library, 156 clones were sequenced. The clones fell into 3 groups: known genes (51%), rat expressed sequence tags (31%), and novel rat genes not found in databases (18%). The known genes could be categorized into families of common function including vascular remodeling, signal transduction, transcription factors, biologic transport, vascular amyloid, hemostasis, myelin, lipids, secretion, cytoskeleton, and junctional complexes. Brain vascular genomics, or BBB genomics, allows for an accelerated discovery of the gene families that are differentially expressed at the microvasculature in brain.
Collapse
Affiliation(s)
- Jian Yi Li
- Department of Medicine, UCLA School of Medicine, Los Angeles, California 90024, USA.
| | | | | |
Collapse
|