1
|
Koca N, Seferoğlu M. Effects of disease-modifying therapies on lipid parameters in patients with multiple sclerosis. Mult Scler Relat Disord 2023; 77:104876. [PMID: 37423049 DOI: 10.1016/j.msard.2023.104876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 06/27/2023] [Accepted: 07/04/2023] [Indexed: 07/11/2023]
Abstract
BACKGROUND Cholesterol and lipids are essential components of nerve cells. Myelin synthesis and stabilization is a cholesterol-dependent process. It has been shown in several studies that high plasma cholesterol levels may be associated with clinical deterioration in Multiple Sclerosis (MS). There is scarce information about the effects of disease-modifying treatment (DMTs) on lipid profile. In this study, we aimed to investigate the effect of DMTs on plasma lipid profiles in MS patients. METHOD The records of 380 MS patients who were still under follow-up were analyzed in terms of age, sex, disease duration, EDSS scores, serum lipid levels, and used DMTs. The data of patients receiving Interferon (n = 53), Glatiramer acetate (n = 25), Fingolimod (n = 44), Teriflunomide (n = 24), Dimethyl fumarate (n = 7) and Ocrelizumab (n = 14) were compared with the data of control group (n = 53). RESULTS A total of 220 patients, 157 women, and 63 men, were included in the study. The average age of the participants in the study was 39.83 ± 10.21 years, mean disease duration was 8.45 ± 6.56 years, and the EDSS score was 2.25 ± 1.97. Although, Lipid parameters were higher in MS patients using Fingolimod the difference cannot reach the statistical significance. CONCLUSION No significant relationship was found between the DMTs that MS patients had been using for the last six months and their cholesterol levels.
Collapse
Affiliation(s)
- Nizameddin Koca
- University of Health Sciences, Bursa Sehir Training & Research Hospital, Department of Internal Medicine, Bursa, Turkey
| | - Meral Seferoğlu
- University of Health Sciences, Bursa Yuksek Ihtisas Training and Research Hospital, Department of Neurology, Bursa, Turkey
| |
Collapse
|
2
|
Piccoli M, Cirillo F, Ghiroldi A, Rota P, Coviello S, Tarantino A, La Rocca P, Lavota I, Creo P, Signorelli P, Pappone C, Anastasia L. Sphingolipids and Atherosclerosis: The Dual Role of Ceramide and Sphingosine-1-Phosphate. Antioxidants (Basel) 2023; 12:antiox12010143. [PMID: 36671005 PMCID: PMC9855164 DOI: 10.3390/antiox12010143] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/28/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Sphingolipids are bioactive molecules that play either pro- and anti-atherogenic roles in the formation and maturation of atherosclerotic plaques. Among SLs, ceramide and sphingosine-1-phosphate showed antithetic properties in regulating various molecular mechanisms and have emerged as novel potential targets for regulating the development of atherosclerosis. In particular, maintaining the balance of the so-called ceramide/S1P rheostat is important to prevent the occurrence of endothelial dysfunction, which is the trigger for the entire atherosclerotic process and is strongly associated with increased oxidative stress. In addition, these two sphingolipids, together with many other sphingolipid mediators, are directly involved in the progression of atherogenesis and the formation of atherosclerotic plaques by promoting the oxidation of low-density lipoproteins (LDL) and influencing the vascular smooth muscle cell phenotype. The modulation of ceramide and S1P levels may therefore allow the development of new antioxidant therapies that can prevent or at least impair the onset of atherogenesis, which would ultimately improve the quality of life of patients with coronary artery disease and significantly reduce their mortality.
Collapse
Affiliation(s)
- Marco Piccoli
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Piazza Malan 2, San Donato Milanese, 20097 Milan, Italy
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
| | - Federica Cirillo
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Piazza Malan 2, San Donato Milanese, 20097 Milan, Italy
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
| | - Andrea Ghiroldi
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Piazza Malan 2, San Donato Milanese, 20097 Milan, Italy
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
| | - Paola Rota
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20133 Milan, Italy
| | - Simona Coviello
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Piazza Malan 2, San Donato Milanese, 20097 Milan, Italy
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
| | - Adriana Tarantino
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Piazza Malan 2, San Donato Milanese, 20097 Milan, Italy
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
- Faculty of Medicine and Surgery, University Vita-Salute San Raffaele, Via Olgettina 58, 20132 Milan, Italy
| | - Paolo La Rocca
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
- Department of Biomedical Sciences for Health, University of Milan, Via Mangiagalli 31, 20133 Milan, Italy
| | - Ivana Lavota
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Piazza Malan 2, San Donato Milanese, 20097 Milan, Italy
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
| | - Pasquale Creo
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Piazza Malan 2, San Donato Milanese, 20097 Milan, Italy
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
| | - Paola Signorelli
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
- Aldo Ravelli Center for Neurotechnology and Experimental Brain Therapeutics, Department of Health Sciences, University of Milan, Via Antonio di Rudinì 8, 20142 Milan, Italy
| | - Carlo Pappone
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
- Faculty of Medicine and Surgery, University Vita-Salute San Raffaele, Via Olgettina 58, 20132 Milan, Italy
- Arrhythmology Department, IRCCS Policlinico San Donato, Piazza Malan 2, San Donato Milanese, 20097 Milan, Italy
| | - Luigi Anastasia
- Laboratory of Stem Cells for Tissue Engineering, IRCCS Policlinico San Donato, Piazza Malan 2, San Donato Milanese, 20097 Milan, Italy
- Institute for Molecular and Translational Cardiology (IMTC), San Donato Milanese, 20097 Milan, Italy
- Faculty of Medicine and Surgery, University Vita-Salute San Raffaele, Via Olgettina 58, 20132 Milan, Italy
- Correspondence: ; Tel.: +39-0226437765
| |
Collapse
|
3
|
FTY720 Reduces Lipid Accumulation by Upregulating ABCA1 through Liver X Receptor and Sphingosine Kinase 2 Signaling in Macrophages. Int J Mol Sci 2022; 23:ijms232314617. [PMID: 36498944 PMCID: PMC9740778 DOI: 10.3390/ijms232314617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 11/24/2022] Open
Abstract
Formation of foam cells as a result of excess lipid accumulation by macrophages is a pathological hallmark of atherosclerosis. Fingolimod (FTY720) is an immunosuppressive agent used in clinical settings for the treatment of multiple sclerosis and has been reported to inhibit atherosclerotic plaque development. However, little is known about the effect of FTY720 on lipid accumulation leading to foam cell formation. In this study, we investigated the effects of FTY720 on lipid accumulation in murine macrophages. FTY720 treatment reduced lipid droplet formation and increased the expression of ATP-binding cassette transporter A1 (ABCA1) in J774 mouse macrophages. FTY720 also enhanced the expression of liver X receptor (LXR) target genes such as FASN, APOE, and ABCG1. In addition, FTY720-induced upregulation of ABCA1 was abolished by knockdown of sphingosine kinase 2 (SphK2) expression. Furthermore, we found that FTY720 treatment induced histone H3 lysine 9 (H3K9) acetylation, which was lost in SphK2-knockdown cells. Taken together, FTY720 induces ABCA1 expression through SphK2-mediated acetylation of H3K9 and suppresses lipid accumulation in macrophages, which provides novel insights into the mechanisms of action of FTY720 on atherosclerosis.
Collapse
|
4
|
Tolksdorf C, Moritz E, Wolf R, Meyer U, Marx S, Bien-Möller S, Garscha U, Jedlitschky G, Rauch BH. Platelet-Derived S1P and Its Relevance for the Communication with Immune Cells in Multiple Human Diseases. Int J Mol Sci 2022; 23:ijms231810278. [PMID: 36142188 PMCID: PMC9499465 DOI: 10.3390/ijms231810278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/02/2022] [Accepted: 09/03/2022] [Indexed: 11/16/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a versatile signaling lipid involved in the regulation of numerous cellular processes. S1P regulates cellular proliferation, migration, and apoptosis as well as the function of immune cells. S1P is generated from sphingosine (Sph), which derives from the ceramide metabolism. In particular, high concentrations of S1P are present in the blood. This originates mainly from erythrocytes, endothelial cells (ECs), and platelets. While erythrocytes function as a storage pool for circulating S1P, platelets can rapidly generate S1P de novo, store it in large quantities, and release it when the platelet is activated. Platelets can thus provide S1P in a short time when needed or in the case of an injury with subsequent platelet activation and thereby regulate local cellular responses. In addition, platelet-dependently generated and released S1P may also influence long-term immune cell functions in various disease processes, such as inflammation-driven vascular diseases. In this review, the metabolism and release of platelet S1P are presented, and the autocrine versus paracrine functions of platelet-derived S1P and its relevance in various disease processes are discussed. New pharmacological approaches that target the auto- or paracrine effects of S1P may be therapeutically helpful in the future for pathological processes involving S1P.
Collapse
Affiliation(s)
- Céline Tolksdorf
- Division of Pharmacology and Toxicology, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
- Department of General Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Eileen Moritz
- Department of General Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Robert Wolf
- Department of General Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Ulrike Meyer
- Division of Pharmacology and Toxicology, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | - Sascha Marx
- Department of Neurosurgery, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Sandra Bien-Möller
- Department of General Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany
- Department of Neurosurgery, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Ulrike Garscha
- Institute of Pharmacy, University of Greifswald, 17489 Greifswald, Germany
| | - Gabriele Jedlitschky
- Department of General Pharmacology, University Medicine Greifswald, 17489 Greifswald, Germany
| | - Bernhard H. Rauch
- Division of Pharmacology and Toxicology, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
- Correspondence:
| |
Collapse
|
5
|
Keul P, Peters S, von Wnuck Lipinski K, Schröder NH, Nowak MK, Duse DA, Polzin A, Weske S, Gräler MH, Levkau B. Sphingosine-1-Phosphate (S1P) Lyase Inhibition Aggravates Atherosclerosis and Induces Plaque Rupture in ApoE−/− Mice. Int J Mol Sci 2022; 23:ijms23179606. [PMID: 36077004 PMCID: PMC9455951 DOI: 10.3390/ijms23179606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/09/2022] [Accepted: 08/20/2022] [Indexed: 11/17/2022] Open
Abstract
Altered plasma sphingosine-1-phosphate (S1P) concentrations are associated with clinical manifestations of atherosclerosis. However, whether long-term elevation of endogenous S1P is pro- or anti-atherogenic remains unclear. Here, we addressed the impact of permanently high S1P levels on atherosclerosis in cholesterol-fed apolipoprotein E-deficient (ApoE−/−) mice over 12 weeks. This was achieved by pharmacological inhibition of the S1P-degrading enzyme S1P lyase with 4-deoxypyridoxine (DOP). DOP treatment dramatically accelerated atherosclerosis development, propagated predominantly unstable plaque phenotypes, and resulted in frequent plaque rupture with atherothrombosis. Macrophages from S1P lyase-inhibited or genetically deficient mice had a defect in cholesterol efflux to apolipoprotein A-I that was accompanied by profoundly downregulated cholesterol transporters ATP-binding cassette transporters ABCA1 and ABCG1. This was dependent on S1P signaling through S1PR3 and resulted in dramatically enhanced atherosclerosis in ApoE−/−/S1PR3−/− mice, where DOP treatment had no additional effect. Thus, high endogenous S1P levels promote atherosclerosis, compromise cholesterol efflux, and cause genuine plaque rupture.
Collapse
Affiliation(s)
- Petra Keul
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Susann Peters
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Karin von Wnuck Lipinski
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Nathalie H. Schröder
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Melissa K. Nowak
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Dragos A. Duse
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Amin Polzin
- Division of Cardiology, Pulmonology, and Vascular Medicine, Heinrich Heine University Medical Center Düsseldorf, 40225 Düsseldorf, Germany
| | - Sarah Weske
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Markus H. Gräler
- Department of Anesthesiology and Intensive Care Medicine, Center for Sepsis Control and Care and Center for Molecular Biomedicine, University Hospital Jena, 07743 Jena, Germany
| | - Bodo Levkau
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
- Correspondence: ; Tel.: +49-211-88-12611
| |
Collapse
|
6
|
Vornholz L, Nienhaus F, Gliem M, Alter C, Henning C, Lang A, Ezzahoini H, Wolff G, Clasen L, Rassaf T, Flögel U, Kelm M, Gerdes N, Jander S, Bönner F. Acute Heart Failure After Reperfused Ischemic Stroke: Association With Systemic and Cardiac Inflammatory Responses. Front Physiol 2022; 12:782760. [PMID: 34992548 PMCID: PMC8724038 DOI: 10.3389/fphys.2021.782760] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 11/24/2021] [Indexed: 12/02/2022] Open
Abstract
Patients with acute ischemic stroke (AIS) present an increased incidence of systemic inflammatory response syndrome and release of Troponin T coinciding with cardiac dysfunction. The nature of the cardiocirculatory alterations remains obscure as models to investigate systemic interferences of the brain-heart-axis following AIS are sparse. Thus, this study aims to investigate acute cardiocirculatory dysfunction and myocardial injury in mice after reperfused AIS. Ischemic stroke was induced in mice by transient right-sided middle cerebral artery occlusion (tMCAO). Cardiac effects were investigated by electrocardiograms, 3D-echocardiography, magnetic resonance imaging (MRI), invasive conductance catheter measurements, histology, flow-cytometry, and determination of high-sensitive Troponin T (hsTnT). Systemic hemodynamics were recorded and catecholamines and inflammatory markers in circulating blood and myocardial tissue were determined by immuno-assay and flow-cytometry. Twenty-four hours following tMCAO hsTnT was elevated 4-fold compared to controls and predicted long-term survival. In parallel, systolic left ventricular dysfunction occurred with impaired global longitudinal strain, lower blood pressure, reduced stroke volume, and severe bradycardia leading to reduced cardiac output. This was accompanied by a systemic inflammatory response characterized by granulocytosis, lymphopenia, and increased levels of serum-amyloid P and interleukin-6. Within myocardial tissue, MRI relaxometry indicated expansion of extracellular space, most likely due to inflammatory edema and a reduced fluid volume. Accordingly, we found an increased abundance of granulocytes, apoptotic cells, and upregulation of pro-inflammatory cytokines within myocardial tissue following tMCAO. Therefore, reperfused ischemic stroke leads to specific cardiocirculatory alterations that are characterized by acute heart failure with reduced stroke volume, bradycardia, and changes in cardiac tissue and accompanied by systemic and local inflammatory responses.
Collapse
Affiliation(s)
- Lilian Vornholz
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany.,Department of Neurology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Fabian Nienhaus
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Michael Gliem
- Department of Neurology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Christina Alter
- Experimental Cardiovascular Imaging, Department of Molecular Cardiology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Carina Henning
- Department of Biology, Institute of Metabolic Physiology, Heinrich-Heine University, Düsseldorf, Germany
| | - Alexander Lang
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Hakima Ezzahoini
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Georg Wolff
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Lukas Clasen
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Essen, Germany
| | - Ulrich Flögel
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany.,Experimental Cardiovascular Imaging, Department of Molecular Cardiology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.,Cardiovascular Research Institute Düsseldorf (CARID), Heinrich Heine University, Düsseldorf, Germany
| | - Malte Kelm
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany.,Cardiovascular Research Institute Düsseldorf (CARID), Heinrich Heine University, Düsseldorf, Germany
| | - Norbert Gerdes
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Sebastian Jander
- Department of Neurology, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| | - Florian Bönner
- Division of Cardiology, Pulmonology, and Vascular Medicine, Medical Faculty, University Hospital Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
7
|
Warboys CM, Weinberg PD. S1P in the development of atherosclerosis: roles of hemodynamic wall shear stress and endothelial permeability. Tissue Barriers 2021; 9:1959243. [PMID: 34542010 DOI: 10.1080/21688370.2021.1959243] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Atherosclerosis is characterized by focal accumulations of lipid within the arterial wall, thought to arise from effects of hemodynamic wall shear stress (WSS) on endothelial permeability. Identifying pathways that mediate the effects of shear on permeability could therefore provide new therapeutic opportunities. Here, we consider whether the sphingosine-1-phosphate (S1P) pathway could constitute such a route. We review effects of S1P in endothelial barrier function, the influence of WSS on S1P production and signaling, the results of trials investigating S1P in experimental atherosclerosis in mice, and associations between S1P levels and cardiovascular disease in humans. Although it seems clear that S1P reduces endothelial permeability and responds to WSS, the evidence that it influences atherosclerosis is equivocal. The effects of specifically pro- and anti-atherosclerotic WSS profiles on the S1P pathway require investigation, as do influences of S1P on the vesicular pathways likely to dominate low-density lipoprotein transport across endothelium.
Collapse
Affiliation(s)
- Christina M Warboys
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Peter D Weinberg
- Department of Bioengineering, Imperial College London, London, UK
| |
Collapse
|
8
|
Rauma I, Huhtala H, Soilu-Hänninen M, Kuusisto H. Lipid Profile Alterations during Fingolimod Treatment in Multiple Sclerosis Patients. J Neuroimmune Pharmacol 2020; 15:567-569. [PMID: 32729054 DOI: 10.1007/s11481-020-09937-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 06/22/2020] [Indexed: 11/30/2022]
Abstract
Fingolimod reduces inflammatory activity in multiple sclerosis (MS) by acting as a functional antagonist of sphingosine 1-phosphate (S1P) receptors. It has been suggested that S1P might also contribute to the antiatherogenic effect of high-density lipoprotein (HDL). We conducted a retrospective observational study using data of 72 MS patients from two Finnish hospital districts to find out whether lipid profiles change during treatment with fingolimod. A mixed-effects model with patient as a random effect was used to analyze lipid profile alterations. We found a statistically significant elevation in both total cholesterol (0.12 mmol/L per year) and HDL (0.04 mmol/L per year) during a median follow-up of 12 months, while low-density lipoprotein (LDL) and triglycerides remained unchanged. Since the mean elevation observed in both lipid values seems to be modest, we suggest that routine lipid profile monitoring is unnecessary during fingolimod treatment in MS patients without pre-existing cardiovascular comorbidities. Graphical abstract.
Collapse
Affiliation(s)
- Ilkka Rauma
- Faculty of Medicine and Health Technology (MET), Tampere University, Arvo Ylpön Katu 34, 33520, Tampere, Finland.
| | - Heini Huhtala
- Faculty of Social Sciences (SOC), Tampere University, Arvo Ylpön Katu 34, 33520, Tampere, Finland
| | - Merja Soilu-Hänninen
- Neurocenter, Turku University Hospital, P.O. Box 52, 20521, Turku, Finland
- Department of Neurology, University of Turku, P.O. Box 52, 20521, Turku, Finland
| | - Hanna Kuusisto
- Department of Neurology, Tampere University Hospital, Teiskontie 35, 33520, Tampere, Finland
- Department of Health and Social Management, University of Eastern Finland, Yliopistonranta 1, 70210, Kuopio, Finland
| |
Collapse
|
9
|
Gisterå A, Klement ML, Polyzos KA, Mailer RKW, Duhlin A, Karlsson MCI, Ketelhuth DFJ, Hansson GK. Low-Density Lipoprotein-Reactive T Cells Regulate Plasma Cholesterol Levels and Development of Atherosclerosis in Humanized Hypercholesterolemic Mice. Circulation 2019; 138:2513-2526. [PMID: 29997115 PMCID: PMC6254780 DOI: 10.1161/circulationaha.118.034076] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Supplemental Digital Content is available in the text. Background: Atherosclerotic cardiovascular disease is a chronic inflammatory process initiated when cholesterol-carrying low-density lipoprotein (LDL) is retained in the arterial wall. CD4+ T cells, some of which recognize peptide components of LDL as antigen, are recruited to the forming lesion, resulting in T-cell activation. Although these T cells are thought to be proatherogenic, LDL immunization reduces disease in experimental animals. These seemingly contradictory findings have hampered the development of immune-based cardiovascular therapy. The present study was designed to clarify how activation of LDL-reactive T cells impacts on metabolism and vascular pathobiology. Methods: We have developed a T-cell receptor–transgenic mouse model to characterize the effects of immune reactions against LDL. Through adoptive cell transfers and cross-breeding to hypercholesterolemic mice expressing the antigenic human LDL protein apolipoprotein B-100, we evaluate the effects on atherosclerosis. Results: A subpopulation of LDL-reactive T cells survived clonal selection in the thymus, developed into T follicular helper cells in lymphoid tissues on antigen recognition, and promoted B-cell activation. This led to production of anti-LDL immunoglobulin G antibodies that enhanced LDL clearance through immune complex formation. Furthermore, the cellular immune response to LDL was associated with increased cholesterol excretion in feces and with reduced vascular inflammation. Conclusions: These data show that anti-LDL immunoreactivity evokes 3 atheroprotective mechanisms: antibody-dependent LDL clearance, increased cholesterol excretion, and reduced vascular inflammation.
Collapse
Affiliation(s)
- Anton Gisterå
- Department of Medicine, Center for Molecular Medicine, Karolinska University Hospital (A.G., M.L.K., K.A.P., R.K.W.M., D.F.J.K., G.K.H.), Karolinska Institutet, Stockholm, Sweden
| | - Maria L Klement
- Department of Medicine, Center for Molecular Medicine, Karolinska University Hospital (A.G., M.L.K., K.A.P., R.K.W.M., D.F.J.K., G.K.H.), Karolinska Institutet, Stockholm, Sweden.,Department of Immunotechnology, Lund University, Sweden (M.L.K.)
| | - Konstantinos A Polyzos
- Department of Medicine, Center for Molecular Medicine, Karolinska University Hospital (A.G., M.L.K., K.A.P., R.K.W.M., D.F.J.K., G.K.H.), Karolinska Institutet, Stockholm, Sweden
| | - Reiner K W Mailer
- Department of Medicine, Center for Molecular Medicine, Karolinska University Hospital (A.G., M.L.K., K.A.P., R.K.W.M., D.F.J.K., G.K.H.), Karolinska Institutet, Stockholm, Sweden.,Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Germany (R.K.W.M.)
| | - Amanda Duhlin
- Department of Microbiology, Tumor and Cell Biology (A.D., M.C.I.K.), Karolinska Institutet, Stockholm, Sweden
| | - Mikael C I Karlsson
- Department of Microbiology, Tumor and Cell Biology (A.D., M.C.I.K.), Karolinska Institutet, Stockholm, Sweden
| | - Daniel F J Ketelhuth
- Department of Medicine, Center for Molecular Medicine, Karolinska University Hospital (A.G., M.L.K., K.A.P., R.K.W.M., D.F.J.K., G.K.H.), Karolinska Institutet, Stockholm, Sweden
| | - Göran K Hansson
- Department of Medicine, Center for Molecular Medicine, Karolinska University Hospital (A.G., M.L.K., K.A.P., R.K.W.M., D.F.J.K., G.K.H.), Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
10
|
Liu M, Frej C, Langefeld CD, Divers J, Bowden DW, Carr JJ, Gebre AK, Xu J, Larsson B, Dahlbäck B, Freedman BI, Parks JS. Plasma apoM and S1P levels are inversely associated with mortality in African Americans with type 2 diabetes mellitus. J Lipid Res 2019; 60:1425-1431. [PMID: 31133557 DOI: 10.1194/jlr.p089409] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 04/27/2019] [Indexed: 12/21/2022] Open
Abstract
apoM is a minor HDL apolipoprotein and carrier for sphingosine-1-phosphate (S1P). HDL apoM and S1P concentrations are inversely associated with atherosclerosis progression in rodents. We evaluated associations between plasma concentrations of S1P, plasma concentrations of apoM, and HDL apoM levels with prevalent subclinical atherosclerosis and mortality in the African American-Diabetes Heart Study participants (N = 545). Associations between plasma S1P, plasma apoM, and HDL apoM with subclinical atherosclerosis and mortality were assessed using multivariate parametric, nonparametric, and Cox proportional hazards models. At baseline, participants' median (25th percentile, 75th percentile) age was 55 (49, 62) years old and their coronary artery calcium (CAC) mass score was 26.5 (0.0, 346.5). Plasma S1P, plasma apoM, and HDL apoM were not associated with CAC. After 64 (57.6, 70.3) months of follow-up, 81 deaths were recorded. Higher concentrations of plasma S1P [odds ratio (OR) = 0.14, P = 0.01] and plasma apoM (OR = 0.10, P = 0.02), but not HDL apoM (P = 0.89), were associated with lower mortality after adjusting for age, sex, statin use, CAC, kidney function, and albuminuria. We conclude that plasma S1P and apoM concentrations are inversely and independently associated with mortality, but not CAC, in African Americans with type 2 diabetes after accounting for conventional risk factors.
Collapse
Affiliation(s)
- Mingxia Liu
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Cecilia Frej
- Department of Translational Medicine Skåne University Hospital, Lund University, Malmö, Sweden
| | - Carl D Langefeld
- Division of Public Health Sciences, Department of Biostatistics and Data Science Wake Forest School of Medicine, Winston-Salem, NC
| | - Jasmin Divers
- Division of Public Health Sciences, Department of Biostatistics and Data Science Wake Forest School of Medicine, Winston-Salem, NC
| | - Donald W Bowden
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC
| | - J Jeffrey Carr
- Department of Radiology Vanderbilt University Medical Center, Nashville, TN
| | - Abraham K Gebre
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - Jianzhao Xu
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC
| | - Benny Larsson
- Department of Clinical Chemistry Skåne University Hospital, Lund, Sweden
| | - Björn Dahlbäck
- Department of Translational Medicine Skåne University Hospital, Lund University, Malmö, Sweden
| | - Barry I Freedman
- Section on Nephrology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC
| | - John S Parks
- Section on Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC .,Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC
| |
Collapse
|
11
|
Liu H, Jin H, Han J, Yue X, Yang H, Zayed MA, Gropler RJ, Tu Z. Upregulated Sphingosine 1-Phosphate Receptor 1 Expression in Human and Murine Atherosclerotic Plaques. Mol Imaging Biol 2019; 20:448-456. [PMID: 29134505 DOI: 10.1007/s11307-017-1141-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
PURPOSE Dysregulation of sphingosine 1-phosphate receptor 1 (S1PR1) signaling contributes to inflammation-related pathophysiological changes in cardiovascular diseases including atherosclerosis (AS). S1PR1-targeting compounds significantly reduce lesion size in murine models of AS. Therefore, characterization of S1PR1 expression in vitro and in vivo in atherosclerotic plaque could enable mechanistic studies and inform S1PR1 targeted therapies. PROCEDURES H&E staining and immunostaining studies were performed on variably diseased human femoral endarterectomy plaque specimens, as well as mouse aortic sections from ApoE-/- mice maintained on a high-fat diet (AS mice). In vitro autoradiography study in human femoral plaques was used to confirm the tracer specificity. Micro positron emission tomography (PET) and ex vivo autoradiography studies were conducted in AS mice and their controls using a S1PR1-specific radioligand [11C]TZ3321 for in vivo and ex vivo quantification of S1PR1 expression in mouse aortic plaques. RESULTS Increased S1PR1 expression was observed in areas of human femoral endarterectomy plaque specimens with foam cell accumulation compared with control tissue; in vitro autoradiography study indicated that SEW2781, a S1PR1 compound was able to reduce the uptake of [11C]TZ3321 by 56 %. S1PR1 levels were also upregulated in AS mouse aortic plaques. MicroPET data showed the aorta-to-blood tracer uptake ratio in AS mice was approximately 20 % higher than that in controls. Autoradiographic study also revealed elevated tracer accumulation in AS mouse aorta. CONCLUSIONS Upregulated S1PR1 expression in human and mouse atherosclerotic plaques was successfully identified by immunostaining and radioligand-based methods. This data demonstrates that [11C]TZ3321 PET provides great promise in imaging S1PR1 expression in atherosclerotic plaques.
Collapse
Affiliation(s)
- Hui Liu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hongjun Jin
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Junbin Han
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Xuyi Yue
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Hao Yang
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Mohamed A Zayed
- Department of Surgery, Section of Vascular Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Robert J Gropler
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Zhude Tu
- Department of Radiology, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
12
|
Huwiler A, Zangemeister-Wittke U. The sphingosine 1-phosphate receptor modulator fingolimod as a therapeutic agent: Recent findings and new perspectives. Pharmacol Ther 2018; 185:34-49. [DOI: 10.1016/j.pharmthera.2017.11.001] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
13
|
Estrogen receptor α/HDAC/NFAT axis for delphinidin effects on proliferation and differentiation of T lymphocytes from patients with cardiovascular risks. Sci Rep 2017; 7:9378. [PMID: 28839227 PMCID: PMC5570903 DOI: 10.1038/s41598-017-09933-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 08/01/2017] [Indexed: 12/16/2022] Open
Abstract
Delphinidin, an anthocyanin present in red wine, has been reported to preserve the integrity of endothelium via an estrogen receptor alpha (ERα)-dependent mechanism. However, the effect of delphinidin on the immune response in obesity-related inflammation remains unknown. Given the important role of T lymphocytes in obesity-related inflammation, we investigated the effect of delphinidin on proliferation and differentiation of T lymphocytes from healthy subjects and metabolic syndrome patients. Delphinidin decreased the proliferation stimulated by different agents acting through different mechanisms. This effect of delphinidin was associated with its ability to inhibit Ca2+ signaling via reduced store-operated Ca2+ entry and release, and subsequent decrease of HDAC and NFAT activations. Delphinidin also inhibited ERK1/2 activation. Pharmacological inhibition of ER with fulvestrant, or deletion of ERα, prevented the effect of delphinidin. Further, delphinidin suppressed the differentiation of T cells toward Th1, Th17 and Treg without affecting Th2 subsets. Interestingly, delphinidin inhibited both proliferation and differentiation of T cells taken from patients with cardiovascular risks associated with metabolic syndrome. Together, we propose that delphinidin, by acting on ERα via multiple cellular targets, may represent a new approach against chronic inflammation associated with T lymphocyte activation, proliferation and differentiation, in patients with cardiovascular risk factors.
Collapse
|
14
|
Mauer AS, Hirsova P, Maiers JL, Shah VH, Malhi H. Inhibition of sphingosine 1-phosphate signaling ameliorates murine nonalcoholic steatohepatitis. Am J Physiol Gastrointest Liver Physiol 2017; 312:G300-G313. [PMID: 28039158 PMCID: PMC5401989 DOI: 10.1152/ajpgi.00222.2016] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 11/28/2016] [Accepted: 12/11/2016] [Indexed: 01/31/2023]
Abstract
Nonalcoholic steatohepatitis (NASH) is a lipotoxic disorder, wherein proinflammatory lipids, such as ceramide and its derivative sphingosine 1-phosphate (S1P), contribute to macrophage-associated liver inflammation. For example, we have previously demonstrated a role for S1P in steatotic hepatocyte-derived S1P-enriched extracellular vesicles in macrophage chemotaxis in vitro. Therefore, we hypothesized that FTY720, an S1P antagonist, would ameliorate NASH by inhibiting proinflammatory monocyte chemotaxis. To test our hypothesis, NASH was established in C57BL/6 male mice by feeding a diet high in fructose, saturated fat, and cholesterol for 22 wk. Then mice received daily intraperitoneal injections of FTY720 for 2 wk before analysis of liver injury, inflammation, and fibrosis. FTY720-treated mice with NASH demonstrated improved liver histology with a significant reduction in hepatocyte ballooning and inflammatory foci. Hepatomegaly was reversed, and liver triglycerides were reduced following FTY720 administration to mice with NASH. Correspondingly, serum ALT levels, hepatic inflammatory macrophage accumulation, and the expression of Ly6C in recruited myeloid cells was reduced in FTY720-treated mice. Hepatic collagen accumulation and expression of α-smooth muscle actin were significantly lowered as well. Body composition, energy consumption and utilization, and hepatic sphingolipid composition remained unchanged following FTY720 administration. FTY720 ameliorates murine nonalcoholic steatohepatitis. Reduction in liver injury and inflammation is associated with a reduction in hepatic macrophage accumulation, likely due to dampened recruitment of circulating myeloid cells into the liver. Nonalcoholic steatohepatitis may be a novel indication for the therapeutic use of FTY720.NEW & NOTEWORTHY There are no approved pharmacologic therapies for nonalcoholic steatohepatitis (NASH), the leading cause of chronic liver disease worldwide. This study describes the use of FTY720, a novel small molecule, for the amelioration of NASH in a mouse model. We demonstrate that 2-wk administration of FTY720 to mice with NASH led to a reduction in liver injury, inflammation, and fibrosis. These data provide a preclinical rationale for studying this drug in human NASH.
Collapse
Affiliation(s)
- Amy S Mauer
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Petra Hirsova
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Jessica L Maiers
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Vijay H Shah
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Harmeet Malhi
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
15
|
Boini KM, Xia M, Koka S, Gehr TWB, Li PL. Sphingolipids in obesity and related complications. FRONT BIOSCI-LANDMRK 2017; 22:96-116. [PMID: 27814604 PMCID: PMC5844360 DOI: 10.2741/4474] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Sphingolipids are biologically active lipids ubiquitously produced in all vertebrate cells. Asides from structural components of cell membrane, sphingolipids also function as intracellular and extracellular mediators that regulate many important physiological cellular processes including cell survival, proliferation, apoptosis, differentiation, migration and immune processes. Recent studies have also indicated that disruption of sphingolipid metabolism is strongly associated with different diseases that exhibit diverse neurological and metabolic consequences. Here, we briefly summarize current evidence for understanding of sphingolipid pathways in obesity and associated complications. The regulation of sphingolipids and their enzymes may have a great impact in the development of novel therapeutic modalities for a variety of metabolic diseases.
Collapse
Affiliation(s)
- Krishna M Boini
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA, 23298, USA and Department of Nephrology, Virginia Commonwealth University, School of Medicine, Richmond, VA, 23298, USA
| | - Min Xia
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA, 23298
| | - Saisudha Koka
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, School of Medicine, Richmond, VA, 23298, USA
| | - Todd W B Gehr
- Department of Nephrology, Virginia Commonwealth University, School of Medicine, Richmond, VA, 23298, USA
| | - Pin-Lan Li
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, 410 N, 12th Street, Richmond, VA, 23298,
| |
Collapse
|
16
|
Abstract
Numerous preclinical studies indicate that sustained endothelial activation significantly contributes to tissue edema, perpetuates the inflammatory response, and exacerbates tissue injury ultimately resulting in organ failure. However, no specific therapies aimed at restoring endothelial function are available as yet. Sphingosine-1-phosphate (S1P) is emerging as a potent modulator of endothelial function and endothelial responses to injury. Recent studies indicate that S1PR are attractive targets to treat not only disorders of the arterial endothelium but also microvascular dysfunction caused by ischemic or inflammatory injury. In this article, we will review the current knowledge of the role of S1P and its receptors in endothelial function in health and disease, and we will discuss the therapeutic potential of targeting S1PR not only for disorders of the arterial endothelium but also the microvasculature. The therapeutic targeting of S1PR in the endothelium could help to bridge the gap between biomedical research in vascular biology and clinical practice.
Collapse
Affiliation(s)
- Teresa Sanchez
- Department of Pathology and Laboratory Medicine, Center for Vascular Biology, Weill Cornell Medical College, 1300 York Ave, Room A607B/Box 69, New York, NY, 10065, USA.
| |
Collapse
|
17
|
Mo ZC, Ren K, Liu X, Tang ZL, Yi GH. A high-density lipoprotein-mediated drug delivery system. Adv Drug Deliv Rev 2016; 106:132-147. [PMID: 27208399 DOI: 10.1016/j.addr.2016.04.030] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 04/13/2016] [Accepted: 04/27/2016] [Indexed: 01/08/2023]
Abstract
High-density lipoprotein (HDL) is a comparatively dense and small lipoprotein that can carry lipids as a multifunctional aggregate in plasma. Several studies have shown that increasing the levels or improving the functionality of HDL is a promising target for treating a wide variety of diseases. Among lipoproteins, HDL particles possess unique physicochemical properties, including naturally synthesized physiological components, amphipathic apolipoproteins, lipid-loading and hydrophobic agent-incorporating characteristics, specific protein-protein interactions, heterogeneity, nanoparticles, and smaller size. Recently, the feasibility and superiority of using HDL particles as drug delivery vehicles have been of great interest. In this review, we summarize the structure, constituents, biogenesis, remodeling, and reconstitution of HDL drug delivery systems, focusing on their delivery capability, characteristics, applications, manufacturing, and drug-loading and drug-targeting characteristics. Finally, the future prospects are presented regarding the clinical application and challenges of using HDL as a pharmacodelivery carrier.
Collapse
Affiliation(s)
- Zhong-Cheng Mo
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang City 421001, Hunan Province, China; Department of Histology and Embryology, University of South China, Hengyang, Hunan 421001, China
| | - Kun Ren
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang City 421001, Hunan Province, China
| | - Xing Liu
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, 100005 Beijing, China
| | - Zhen-Li Tang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang City 421001, Hunan Province, China
| | - Guang-Hui Yi
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang City 421001, Hunan Province, China.
| |
Collapse
|
18
|
Hovi A, Airas L. Effect of Fingolimod-Treatment on Blood Lipid Profiles of Multiple Sclerosis Patients. J Neuroimmune Pharmacol 2016; 11:611-612. [PMID: 27388874 DOI: 10.1007/s11481-016-9697-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 06/29/2016] [Indexed: 02/03/2023]
|
19
|
Erbel C, Rupp G, Domschke G, Linden F, Akhavanpoor M, Doesch AO, Katus HA, Gleissner CA. Differential regulation of aldose reductase expression during macrophage polarization depends on hyperglycemia. Innate Immun 2016; 22:230-7. [PMID: 26873505 DOI: 10.1177/1753425916632053] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 01/19/2016] [Indexed: 01/23/2023] Open
Abstract
Aldose reductase (AR; gene AKR1B1) is the rate-limiting enzyme of the polyol pathway and has been associated with diabetes and atherosclerosis. Here, we sought to identify the mechanisms underlying differential AR expression in human atherosclerotic plaque macrophages. In vitro, M1-polarized human monocyte-derived macrophages expressed significantly higher levels of AKR1B1 mRNA and AR protein compared with M2-polarized macrophages. AR activity was significantly higher in M1 macrophages. AKR1B1 mRNA expression correlated positively with the M1 marker TNF(r = 0.430,P = 0.006) and negatively with the M2 marker MRC1 (r = -0.443,P = 0.044). Increased AR expression in M1 macrophages depended on hyperglycemia. Concomitantly, expression of SLC2A1 (coding for the Glc transporter GLUT-1) was significantly higher in M1 than in M2 macrophages. Pharmacological inhibition of GLUT-1 using STF-32 completely abrogated Glc-induced AR up-regulation in M1 macrophages. When analyzing AR expression in post-mortem coronary artery plaque macrophages, a history of diabetes was associated with a significantly increased proportion of CD68(+)AR(++)macrophages, supporting the in vivo relevance of our in vitro findings. We demonstrate that the phenotype of atherosclerotic plaque macrophages may be affected by cardiovascular risk factors such as hyperglycemia. Our data illustrate the complex interplay between systemic and local factors in atherogenesis.
Collapse
Affiliation(s)
- Christian Erbel
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg, Heidelberg, Germany
| | - Gregor Rupp
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg, Heidelberg, Germany
| | - Gabriele Domschke
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg, Heidelberg, Germany
| | - Fabian Linden
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg, Heidelberg, Germany
| | - Mohammadreza Akhavanpoor
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg, Heidelberg, Germany
| | - Andreas O Doesch
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg, Heidelberg, Germany
| | - Hugo A Katus
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg, Heidelberg, Germany
| | - Christian A Gleissner
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg, Heidelberg, Germany
| |
Collapse
|
20
|
Potì F, Ceglarek U, Burkhardt R, Simoni M, Nofer JR. SKI-II--a sphingosine kinase 1 inhibitor--exacerbates atherosclerosis in low-density lipoprotein receptor-deficient (LDL-R-/-) mice on high cholesterol diet. Atherosclerosis 2015; 240:212-5. [PMID: 25801013 DOI: 10.1016/j.atherosclerosis.2015.03.020] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Revised: 03/05/2015] [Accepted: 03/07/2015] [Indexed: 01/06/2023]
Abstract
BACKGROUND Sphingosine 1-phosphate (S1P) is a lysosphingolipid associated with high-density lipoproteins (HDL) that contributes to their anti-atherogenic potential. We investigated whether a reduction in S1P plasma levels affects atherosclerosis in low-density lipoprotein receptor deficient (LDL-R-/-) mice. METHODS AND RESULTS LDL-R-/- mice on Western diet containing low (0.25% w/w) or high (1.25% w/w) cholesterol were treated for 16 weeks with SKI-II, a sphingosine kinase 1 inhibitor that significantly reduced plasma S1P levels. SKI-II treatment increased atherosclerotic lesions in the thoracic aorta in mice on high but not low cholesterol diet. This compound did not affect body weight, blood cell counts and plasma total and HDL cholesterol, but decreased triglycerides. In addition, mice on high cholesterol diet receiving SKI-II showed elevated levels of tumor necrosis factor-α and endothelial adhesion molecules (sICAM-1, sVCAM-1). CONCLUSION Prolonged lowering of plasma S1P produces pro-atherogenic effects in LDL-R-/- mice that are evident under condition of pronounced hypercholesterolemia.
Collapse
Affiliation(s)
- Francesco Potì
- Department of Biomedical, Metabolic and Neural Sciences - Endocrinology Section, University of Modena and Reggio Emilia, Italy
| | - Uta Ceglarek
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Ralph Burkhardt
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, University Hospital Leipzig, Leipzig, Germany
| | - Manuela Simoni
- Department of Biomedical, Metabolic and Neural Sciences - Endocrinology Section, University of Modena and Reggio Emilia, Italy
| | - Jerzy-Roch Nofer
- Department of Biomedical, Metabolic and Neural Sciences - Endocrinology Section, University of Modena and Reggio Emilia, Italy; Center for Laboratory Medicine, University Hospital Münster, Germany.
| |
Collapse
|
21
|
Borup A, Christensen PM, Nielsen LB, Christoffersen C. Apolipoprotein M in lipid metabolism and cardiometabolic diseases. Curr Opin Lipidol 2015; 26:48-55. [PMID: 25551802 DOI: 10.1097/mol.0000000000000142] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE This review will address recent findings on apolipoprotein M (apoM) and its ligand sphingosine-1-phosphate (S1P) in lipid metabolism and inflammatory diseases. RECENT FINDINGS ApoM's likely role(s) in health and disease has become more diverse after the discovery that apoM functions as a chaperone for S1P. Hence, apoM has recently been implicated in lipid metabolism, diabetes and rheumatoid arthritis through in-vivo, in-vitro and genetic association studies. It remains to be established to which degree such associations with apoM can be attributed to its ability to bind S1P. SUMMARY The apoM/S1P axis and its implications in atherosclerosis and lipid metabolism have been thoroughly studied. Owing to the discovery of the apoM/S1P axis, the scope of apoM research has broadened. ApoM and S1P have been implicated in lipid metabolism, that is by modulating HDL particles. Also, the importance in regulating endothelial function is being investigated. Furthermore, both apoM and S1P have been linked to diabetes and glucose and insulin metabolism. Finally, genetic variations in the apoM gene are associated with lipid disturbances, diabetes and rheumatoid arthritis. These findings suggest not only diverse effects of apoM, but also the important question of whether apoM mainly acts as a S1P carrier, if apoM carries other substances with biological effects as well, or whether the apoM protein has effects on its own.
Collapse
Affiliation(s)
- Anna Borup
- aDepartment of Clinical Biochemistry, Rigshospitalet bInstitute of Biomedical Sciences cInstitute of Clinical Medicine, Copenhagen University, Copenhagen, Denmark *Dr Anna Borup and Pernille M. Christensen contributed equally to the writing of this article
| | | | | | | |
Collapse
|
22
|
Akhavanpoor M, Gleissner CA, Gorbatsch S, Doesch AO, Akhavanpoor H, Wangler S, Jahn F, Lasitschka F, Katus HA, Erbel C. CCL19 and CCL21 modulate the inflammatory milieu in atherosclerotic lesions. DRUG DESIGN DEVELOPMENT AND THERAPY 2014; 8:2359-71. [PMID: 25473269 PMCID: PMC4251537 DOI: 10.2147/dddt.s72394] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Despite advances in the pharmacologic and interventional treatment of coronary artery disease, atherosclerosis remains the leading cause of death worldwide. Atherosclerosis is a chronic inflammatory disease, and elevated expression of CCL19 and CCL21 has been observed in ruptured lesions of coronary arteries of patients with myocardial infarction and carotid plaques of patients with ischemic symptoms, as well as in plasma of coronary artery disease patients. However, the exact role of CCL19 and CCL21 in atherosclerosis remains unknown. In order to identify CCL19 and CCL21 as a novel therapeutic target, we performed bone marrow transplantation as an immunomodulatory treatment concept. Bone marrow of plt/plt mice (lacking CCL19 and CCL21-Ser) was transplanted into atherogenic Ldlr(-/-) mice. The study demonstrated a significantly increased inflammatory cellular infiltration into the lesions of plt/plt/Ldlr(-/-) mice versus controls. Although the level of chemoattraction was increased, messenger ribonucleic acid and protein levels in thoracic aorta and serum of several proinflammatory cytokines (TNFα, IFNγ, IL-6, IL-12, and IL-17) were significantly reduced in plt/plt/Ldlr(-/-) versus control mice. Increased influx, accompanied by reduced activation of leukocytes in atherosclerotic lesion, was accompanied by increased plaque stability but unchanged lesion development. In conclusion, modulation of the chemokines CCL19 and CCL21 represents a potent immunoregulatory treatment approach, and thus represents a novel therapeutic target to stabilize atherosclerotic lesions.
Collapse
Affiliation(s)
- Mohammadreza Akhavanpoor
- Department of Cardiology, University of Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Christian A Gleissner
- Department of Cardiology, University of Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Stephanie Gorbatsch
- Department of Cardiology, University of Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Andreas O Doesch
- Department of Cardiology, University of Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Hamidreza Akhavanpoor
- Department of Cardiology, University of Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Susanne Wangler
- Department of Cardiology, University of Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Frederik Jahn
- Department of Cardiology, University of Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | | | - Hugo A Katus
- Department of Cardiology, University of Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Christian Erbel
- Department of Cardiology, University of Heidelberg, Germany ; DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| |
Collapse
|
23
|
The immunosuppressant FTY720 prolongs survival in a mouse model of diet-induced coronary atherosclerosis and myocardial infarction. J Cardiovasc Pharmacol 2014; 63:132-143. [PMID: 24508946 DOI: 10.1097/fjc.0000000000000031] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
FTY720, an analogue of sphingosine-1-phosphate, is cardioprotective during acute injury. Whether long-term FTY720 affords cardioprotection is unknown. Here, we report the effects of oral FTY720 on ischemia/reperfusion injury and in hypomorphic apoE mice deficient in SR-BI receptor expression (ApoeR61(h/h)/SRB1(-/- mice), a model of diet-induced coronary atherosclerosis and heart failure. We added FTY720 (0.3 mg·kg(-1)·d(-1)) to the drinking water of C57BL/6J mice. After ex vivo cardiac ischemia/reperfusion injury, these mice had significantly improved left ventricular (LV) developed pressure and reduced infarct size compared with controls. Subsequently, ApoeR61(h/h)/SRB1(-/-) mice fed a high-fat diet for 4 weeks were treated or not with oral FTY720 (0.05 mg·kg(-1)·d(-1)). This sharply reduced mortality (P < 0.02) and resulted in better LV function and less LV remodeling compared with controls without reducing hypercholesterolemia and atherosclerosis. Oral FTY720 reduced the number of blood lymphocytes and increased the percentage of CD4+Foxp3+ regulatory T cells (Tregs) in the circulation, spleen, and lymph nodes. FTY720-treated mice exhibited increased TGF-β and reduced IFN-γ expression in the heart. Also, CD4 expression was increased and strongly correlated with molecules involved in natural Treg activity, such as TGF-β and GITR. Our data suggest that long-term FTY720 treatment enhances LV function and increases longevity in mice with heart failure. These benefits resulted not from atheroprotection but from systemic immunosuppression and a moderate reduction of inflammation in the heart.
Collapse
|
24
|
Browne RW, Weinstock-Guttman B, Horakova D, Zivadinov R, Bodziak ML, Tamaño-Blanco M, Badgett D, Tyblova M, Vaneckova M, Seidl Z, Krasensky J, Bergsland N, Ramasamy DP, Hagemeier J, Havrdova E, Ramanathan M. Apolipoproteins are associated with new MRI lesions and deep grey matter atrophy in clinically isolated syndromes. J Neurol Neurosurg Psychiatry 2014; 85:859-64. [PMID: 24470599 DOI: 10.1136/jnnp-2013-307106] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
OBJECTIVES There is increasing evidence that serum lipoprotein cholesterol biomarkers are associated with disease progression in clinically isolated syndromes (CIS). Apolipoproteins (Apo) are recognition ligands that mediate the physiological interactions of cholesterol-containing lipoproteins. The objective of this study was to investigate whether serum Apo levels are associated with CIS disease progression. METHODS ApoB, ApoAI, ApoAII, ApoE and lipoprotein (a) (Lpa) levels were measured in serum samples obtained prior to the start of treatment from 181 CIS patients (123 women, 58 men, 68% women; mean age: 28.1±SD 8.1 years). All patients were treated with intramuscular interferon-β as part of the prospective study. Clinical and MRI assessments were obtained at baseline, 6, 12 and 24 months after start of interferon-β treatment. RESULTS Greater ApoB levels were associated with increased number of new T2 lesions (p<0.001) and increased number of new or enlarging T2 lesions (p<0.001) over 2 years. Each 10 mg/dL of greater baseline ApoB is associated with a 16% increase in the number of new T2 lesions over 2 years. ApoAI, ApoAII, ApoE and Lpa were not associated with T2 lesions. Greater ApoE levels were associated with greater deep grey matter atrophy (partial correlation rp=-0.28, p<0.001). Each 1 mg/dL increment in ApoE levels was associated with a 1% increase in deep grey matter atrophy over 2 years. CONCLUSIONS Serum ApoB levels are associated with new lesion accumulation whereas ApoE levels are associated with deep grey matter atrophy in high risk CIS patients treated with interferon β-1a.
Collapse
Affiliation(s)
- Richard W Browne
- Department of Biotechnical and Clinical Laboratory Sciences, State University of New York, Buffalo, New York, USA
| | | | - Dana Horakova
- Department of Neurology and Center of Clinical Neuroscience, Charles University in Prague, 1st Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Robert Zivadinov
- Department of Neurology, State University of New York, Buffalo, New York, USA Buffalo Neuroimaging Analysis Center, Department of Neurology, State University of New York, Buffalo, New York, USA
| | - Mary Lou Bodziak
- Department of Biotechnical and Clinical Laboratory Sciences, State University of New York, Buffalo, New York, USA
| | - Miriam Tamaño-Blanco
- Department of Pharmaceutical Sciences, State University of New York, Buffalo, New York, USA
| | - Darlene Badgett
- Department of Pharmaceutical Sciences, State University of New York, Buffalo, New York, USA
| | - Michaela Tyblova
- Department of Neurology and Center of Clinical Neuroscience, Charles University in Prague, 1st Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Manuela Vaneckova
- Department of Radiology, 1st Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Zdenek Seidl
- Department of Radiology, 1st Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Jan Krasensky
- Department of Radiology, 1st Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Niels Bergsland
- Buffalo Neuroimaging Analysis Center, Department of Neurology, State University of New York, Buffalo, New York, USA
| | - Deepa P Ramasamy
- Buffalo Neuroimaging Analysis Center, Department of Neurology, State University of New York, Buffalo, New York, USA
| | - Jesper Hagemeier
- Buffalo Neuroimaging Analysis Center, Department of Neurology, State University of New York, Buffalo, New York, USA
| | - Eva Havrdova
- Department of Neurology and Center of Clinical Neuroscience, Charles University in Prague, 1st Faculty of Medicine and General University Hospital, Charles University, Prague, Czech Republic
| | - Murali Ramanathan
- Department of Neurology, State University of New York, Buffalo, New York, USA Department of Pharmaceutical Sciences, State University of New York, Buffalo, New York, USA
| |
Collapse
|
25
|
COX-2 protects against atherosclerosis independently of local vascular prostacyclin: identification of COX-2 associated pathways implicate Rgl1 and lymphocyte networks. PLoS One 2014; 9:e98165. [PMID: 24887395 PMCID: PMC4041570 DOI: 10.1371/journal.pone.0098165] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Accepted: 04/29/2014] [Indexed: 12/13/2022] Open
Abstract
Cyxlo-oxygenase (COX)-2 inhibitors, including traditional nonsteroidal anti-inflammatory drugs (NSAIDs) are associated with increased cardiovascular side effects, including myocardial infarction. We and others have shown that COX-1 and not COX-2 drives vascular prostacyclin in the healthy cardiovascular system, re-opening the question of how COX-2 might regulate cardiovascular health. In diseased, atherosclerotic vessels, the relative contribution of COX-2 to prostacyclin formation is not clear. Here we have used apoE(-/-)/COX-2(-/-) mice to show that, whilst COX-2 profoundly limits atherosclerosis, this protection is independent of local prostacyclin release. These data further illustrate the need to look for new explanations, targets and pathways to define the COX/NSAID/cardiovascular risk axis. Gene expression profiles in tissues from apoE(-/-)/COX-2(-/-) mice showed increased lymphocyte pathways that were validated by showing increased T-lymphocytes in plaques and elevated plasma Th1-type cytokines. In addition, we identified a novel target gene, rgl1, whose expression was strongly reduced by COX-2 deletion across all examined tissues. This study is the first to demonstrate that COX-2 protects vessels against atherosclerotic lesions independently of local vascular prostacyclin and uses systems biology approaches to identify new mechanisms relevant to development of next generation NSAIDs.
Collapse
|
26
|
Potì F, Simoni M, Nofer JR. Atheroprotective role of high-density lipoprotein (HDL)-associated sphingosine-1-phosphate (S1P). Cardiovasc Res 2014; 103:395-404. [PMID: 24891400 DOI: 10.1093/cvr/cvu136] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Numerous epidemiological studies documented an inverse relationship between plasma high-density lipoprotein (HDL) cholesterol levels and the extent of atherosclerotic disease. However, clinical interventions targeting HDL cholesterol failed to show clinical benefits with respect to cardiovascular risk reduction, suggesting that HDL components distinct from cholesterol may account for anti-atherogenic effects attributed to this lipoprotein. Sphingosine-1-phosphate (S1P)-a lysosphingolipid exerting its biological activity via binding to specific G protein-coupled receptors and regulating a wide array of biological responses in a variety of different organs and tissues including the cardiovascular system-has been identified as an integral constituent of HDL particles. In the present review, we discuss current evidence from epidemiological studies, experimental approaches in vitro, and animal models of atherosclerosis, suggesting that S1P contributes to atheroprotective effects exerted by HDL particles.
Collapse
Affiliation(s)
- Francesco Potì
- Department of Biomedical, Metabolic and Neural Sciences-Endocrinology Section, University of Modena and Reggio Emilia, Modena, Italy
| | - Manuela Simoni
- Department of Biomedical, Metabolic and Neural Sciences-Endocrinology Section, University of Modena and Reggio Emilia, Modena, Italy
| | - Jerzy-Roch Nofer
- Department of Biomedical, Metabolic and Neural Sciences-Endocrinology Section, University of Modena and Reggio Emilia, Modena, Italy Center for Laboratory Medicine, University Hospital Münster, Albert-Schweizer-Campus 1, Geb. A1, Münster D-48149, Germany
| |
Collapse
|
27
|
Mao Y, Wang J, Zhao Y, Yan R, Li H, Chen CS, Lee RJ, Byrd JC, Lee LJ, Muthusamy N, Phelps MA. Quantification of OSU-2S, a novel derivative of FTY720, in mouse plasma by liquid chromatography-tandem mass spectrometry. J Pharm Biomed Anal 2014; 98:160-5. [PMID: 24927402 DOI: 10.1016/j.jpba.2014.05.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 05/01/2014] [Accepted: 05/06/2014] [Indexed: 02/03/2023]
Abstract
OSU-2S is a novel anti-cancer and immune modulatory agent designed specifically to avert the immunosuppressive effects and related toxicities observed in clinical studies with its predecessor analog, FTY720. To characterize its preclinical pharmacokinetics, a liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed and validated for the quantification of OSU-2S in mouse plasma. Ethyl acetate extraction of samples containing OSU-2S and the internal standard, Sph-17, was followed by separation with a 6min gradient (water/0.1% formic acid and methanol/0.1% formic acid) on a reverse-phase C18 column at room temperature. Selected reaction monitoring was used for detection on a triple quadrupole mass spectrometer with positive ionization. The assay was linear over the concentration range 3-3000ng/mL with accuracy ranging from 103 to 111%, and both within- and between-run precision (CV%) ≤11%. All stability samples were within ±15% of nominal values, and replicates were within 15% CV. The assay was successfully applied to a mouse pharmacokinetic study of OSU-2S with intravenous and intraperitoneal administration. OSU-2S non-compartmental pharmacokinetic parameters, area under the concentration-time curve, clearance, and elimination half-life were estimated at 1522hμg/L, 3.06L/h/kg and 15.6h, respectively, for intravenous injection. Systemic availability after intraperitoneal injection was approximately 46%. These data demonstrate the OSU-2S compound displays acceptable pharmacokinetic properties for further in vivo pharmacologic evaluation, which can be facilitated by the validated LC-MS/MS assay.
Collapse
Affiliation(s)
- Yicheng Mao
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; Nanoscale Science and Engineering Center for Affordable Nanoengineering of Polymeric Biomedical Devices (CANPBD), The Ohio State University, Columbus, OH 43210, USA
| | - Jiang Wang
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Yuan Zhao
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Ribai Yan
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Hao Li
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Ching-Shih Chen
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Robert J Lee
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA; Nanoscale Science and Engineering Center for Affordable Nanoengineering of Polymeric Biomedical Devices (CANPBD), The Ohio State University, Columbus, OH 43210, USA
| | - John C Byrd
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - L James Lee
- Nanoscale Science and Engineering Center for Affordable Nanoengineering of Polymeric Biomedical Devices (CANPBD), The Ohio State University, Columbus, OH 43210, USA; William G. Lowrie Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH 43210, USA
| | - Natarajan Muthusamy
- Division of Hematology, Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Mitch A Phelps
- College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
28
|
Abstract
Atherosclerosis is one of the most common causes of death and disability in the United States today despite the availability of statins, which reduce hyperlipidemia, a risk factor that predisposes individuals to this disease. Epidemiology of human populations has overwhelmingly demonstrated an inverse correlation between the concentration of plasma high-density lipoprotein (HDL) cholesterol (HDL-C) and the likelihood of developing cardiovascular disease (CVD). Decades of observations and mechanistic studies suggest that one protective function of HDL is its central role in reverse cholesterol transport. In this pathway, the ATP-binding cassette transporter A1 releases intracellular cholesterol, which is packaged with apolipoprotein A-I (apoA-I) into nascent HDL particles and released from the plasma membrane. Further lipidation and maturation of HDL occur in plasma with the eventual uptake by the liver where cholesterol is removed. It is generally accepted that CVD risk can be reduced if plasma HDL-C levels are elevated. Several different pharmacological approaches have been tried; the most popular approach targets the movement of cholesteryl ester from HDL to triglyceride-rich particles by cholesteryl ester transfer protein. Inhibition of cholesteryl ester transfer protein increases plasma HDL-C concentration; however, beneficial effects have yet to be demonstrated, likely the result of off-target effects. These revelations have led to a reevaluation of how elevating HDL concentration could decrease risk. A recent, landmark study showed that the inherent cholesterol efflux capacity of an individual's plasma was a better predictor of CVD status than overall HDL-C concentration. Even more provocative are recent studies showing that apoA-I, the principle protein component of HDL modulates cellular inflammation and oxidation. The following will review all these potential routes explaining how HDL apoA-I can reduce the risk of CVD.
Collapse
|
29
|
Abstract
PURPOSE OF REVIEW The review will address the potential roles of apolipoprotein M (apoM) as a carrier protein and modulator of sphingosine-1-phosphate (S1P) bioactivity. RECENT FINDINGS Recombinant apoM can bind small lipids such as retinoic acid, oxidized phospholipids, and S1P. Thus, the effects of apoM may be pleiotrophic. The S1P binding ability of apoM has biological impact. ApoM-bound S1P can activate S1P1 receptors on endothelial cells and deficiency of apoM abolishes the presence of S1P in HDL. In mice, the lack of apoM causes dysfunctional endothelial barrier function in the lungs. In humans, sepsis that is characterized by impaired endothelial function is associated with low plasma apoM. SUMMARY Plasma apoM is mainly bound to HDL. The roles of apoM in atherosclerosis and lipoprotein metabolism have been given much attention. New in the field is the discovery of apoM as a chaperone for S1P. S1P is a bioactive lipid with effects on angiogenesis, lymphocyte trafficking, endothelial cell migration, and inflammation. A drug targeting the S1P-system (fingolimod) is now used for treatment of multiple sclerosis. It improves the blood-brain barrier and inhibits migration of lymphocytes into the brain. Further exploration of the apoM/S1P axis may uncover its potential as a biomarker and target for new treatments.
Collapse
Affiliation(s)
- Christina Christoffersen
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| | | |
Collapse
|
30
|
Shao J, Katika MR, Schmeits PCJ, Hendriksen PJM, van Loveren H, Peijnenburg AACM, Volger OL. Toxicogenomics-based identification of mechanisms for direct immunotoxicity. Toxicol Sci 2013; 135:328-46. [PMID: 23824090 DOI: 10.1093/toxsci/kft151] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Compounds with direct immunotoxic properties, including metals, mycotoxins, agricultural pesticides, and industrial chemicals, form potential human health risks due to exposure through food, drinking water, and the environment. Insights into the mechanisms of action are currently lacking for the majority of these direct immunotoxicants. Therefore, the present work aimed to gain insights into the molecular mechanisms underlying direct immunotoxicity. To this end, we assessed in vitro the effects of 31 test compounds on the transcriptome of the human Jurkat T-cell line. These compounds included direct immunotoxicants, immunosuppressive drugs with different mode of actions, and nonimmunotoxic control chemicals. Pathway analysis of the microarray data allowed us to identify canonical pathways and Gene Ontology processes that were transcriptionally regulated in common by immunotoxicants (1) with structural similarities, such as tributyltin chloride and tributyltin oxide that activated the retinoic acid/X receptor signaling pathway and (2) without structural similarities, such as As2O3, dibutyltin chloride, diazinon, MeHg, ochratoxin A (OTA), S9-treated OTA, S9-treated cyclophosphamide, and S9-treated benzo[a]pyrene, which activated unfolded protein response, and FTY720, lindane, and propanil, which activated the cholesterol biosynthesis pathway. In addition, processes uniquely affected by individual immunotoxicants were identified, such as the induction of Notch receptor signaling and the downregulation of acute-phase response genes by OTA. These findings were validated by quantitative real-time PCR analysis of genes involved in these processes. Our study indicated that diverse modes of action are involved in direct immunotoxicity and that a set of pathways or genes, rather than one single gene, can be used to screen compounds for direct immunotoxicity.
Collapse
Affiliation(s)
- Jia Shao
- * RIKILT-Institute of Food Safety, Wageningen University and Research Centre, 6700 AE Wageningen, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
31
|
Lietz M, Berges A, Lebrun S, Meurrens K, Steffen Y, Stolle K, Schueller J, Boue S, Vuillaume G, Vanscheeuwijck P, Moehring M, Schlage W, De Leon H, Hoeng J, Peitsch M. Cigarette-smoke-induced atherogenic lipid profiles in plasma and vascular tissue of apolipoprotein E-deficient mice are attenuated by smoking cessation. Atherosclerosis 2013; 229:86-93. [DOI: 10.1016/j.atherosclerosis.2013.03.036] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 02/28/2013] [Accepted: 03/26/2013] [Indexed: 11/16/2022]
|
32
|
The apolipoprotein m-sphingosine-1-phosphate axis: biological relevance in lipoprotein metabolism, lipid disorders and atherosclerosis. Int J Mol Sci 2013; 14:4419-31. [PMID: 23439550 PMCID: PMC3634416 DOI: 10.3390/ijms14034419] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 01/17/2013] [Accepted: 02/05/2013] [Indexed: 01/27/2023] Open
Abstract
Apolipoprotein M (apoM) is a plasma apolipoprotein that mainly associates with high-density lipoproteins. Hence, most studies on apoM so far have investigated its effect on and association with lipid metabolism and atherosclerosis. The insight into apoM biology recently took a major turn. ApoM was identified as a carrier of the bioactive lipid sphingosine-1-phosphate (S1P). S1P activates five different G-protein-coupled receptors, known as the S1P-receptors 1–5 and, hence, affects a wide range of biological processes, such as lymphocyte trafficking, angiogenesis, wound repair and even virus suppression and cancer. The ability of apoM to bind S1P is due to a lipophilic binding pocket within the lipocalin structure of the apoM molecule. Mice overexpressing apoM have increased plasma S1P concentrations, whereas apoM-deficient mice have decreased S1P levels. ApoM-S1P is able to activate the S1P-receptor-1, affecting the function of endothelial cells, and apoM-deficient mice display impaired endothelial permeability in the lung. This review will focus on the putative biological roles of the new apoM–S1P axis in relation to lipoprotein metabolism, lipid disorders and atherosclerosis.
Collapse
|
33
|
Klingenberg R, Gerdes N, Badeau RM, Gisterå A, Strodthoff D, Ketelhuth DFJ, Lundberg AM, Rudling M, Nilsson SK, Olivecrona G, Zoller S, Lohmann C, Lüscher TF, Jauhiainen M, Sparwasser T, Hansson GK. Depletion of FOXP3+ regulatory T cells promotes hypercholesterolemia and atherosclerosis. J Clin Invest 2013; 123:1323-34. [PMID: 23426179 DOI: 10.1172/jci63891] [Citation(s) in RCA: 276] [Impact Index Per Article: 25.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 12/20/2012] [Indexed: 12/17/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease promoted by hyperlipidemia. Several studies support FOXP3-positive regulatory T cells (Tregs) as inhibitors of atherosclerosis; however, the mechanism underlying this protection remains elusive. To define the role of FOXP3-expressing Tregs in atherosclerosis, we used the DEREG mouse, which expresses the diphtheria toxin (DT) receptor under control of the Treg-specific Foxp3 promoter, allowing for specific ablation of FOXP3+ Tregs. Lethally irradiated, atherosclerosis-prone, low-density lipoprotein receptor-deficient (Ldlr(-/-)) mice received DEREG bone marrow and were injected with DT to eliminate FOXP3(+) Tregs. Depletion of Tregs caused a 2.1-fold increase in atherosclerosis without a concomitant increase in vascular inflammation. These mice also exhibited a 1.7-fold increase in plasma cholesterol and an atherogenic lipoprotein profile with increased levels of VLDL. Clearance of VLDL and chylomicron remnants was hampered, leading to accumulation of cholesterol-rich particles in the circulation. Functional and protein analyses complemented by gene expression array identified reduced protein expression of sortilin-1 in liver and increased plasma enzyme activity of lipoprotein lipase, hepatic lipase, and phospholipid transfer protein as mediators of the altered lipid phenotype. These results demonstrate that FOXP3(+) Tregs inhibit atherosclerosis by modulating lipoprotein metabolism.
Collapse
Affiliation(s)
- Roland Klingenberg
- Center for Molecular Medicine, Department of Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Sphingosine-1-phosphate (S1P) regulates important functions in cardiac and vascular homeostasis. It has been implied to play causal roles in the pathogenesis of many cardiovascular disorders such as coronary artery disease, atherosclerosis, myocardial infarction, and heart failure. The majority of S1P in plasma is associated with high-density lipoproteins (HDL), and their S1P content has been shown to be responsible, at least in part, for several of the beneficial effects of HDL on cardiovascular risk. The attractiveness of S1P-based drugs for potential cardiovascular applications is increasing in the wake of the clinical approval of FTY720, but answers to important questions on the effects of S1P in cardiovascular biology and medicine must still be found. This chapter focuses on the current understanding of the role of S1P and its receptors in cardiovascular physiology, pathology, and disease.
Collapse
Affiliation(s)
- Bodo Levkau
- University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
35
|
Hornemann T, Worgall TS. Sphingolipids and atherosclerosis. Atherosclerosis 2012; 226:16-28. [PMID: 23075523 DOI: 10.1016/j.atherosclerosis.2012.08.041] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 08/06/2012] [Accepted: 08/22/2012] [Indexed: 11/19/2022]
Abstract
The atherosclerotic lesion contains a high amount of sphingolipids, a large group of structurally diverse lipids that regulate distinct biological functions beyond their role as structural membrane components. Assessment of their role in atherogenesis has been enabled after genes that regulate their metabolism had been identified and facilitated by the more wide availability of mass spectrometry. Here we discuss recent mechanistic insights obtained in animal and epidemiological studies that have greatly enhanced our understanding of mechanisms how sphingolipids affect the atherosclerotic process.
Collapse
Affiliation(s)
- Thorsten Hornemann
- Inst. for Clinical Chemistry, University Hospital Zuerich, Raemistrasse 100, 8091 Zuerich, Switzerland.
| | | |
Collapse
|
36
|
Huang K, Li SQ, Wang WJ, Liu LS, Jiang YG, Feng PN, Wang YQ, Wang SM. Oral FTY720 administration induces immune tolerance and inhibits early development of atherosclerosis in apolipoprotein E-deficient mice. Int J Immunopathol Pharmacol 2012; 25:397-406. [PMID: 22697071 DOI: 10.1177/039463201202500209] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Orally administered immunomodulatory drugs have recently demonstrated the ability to induce an oral tolerance via inhibition of effector T cells and induction of certain subsets of regulatory T cells (Tregs) which have the potential to prevent several autoimmune diseases. In the present study, we hypothesized that short-term, low-dose, oral FTY720 administration may induce latency-associated peptide (LAP) Tregs and CD4(+) Foxp3(+) Tregs in atherogenesis, potentially resulting in remission of early development of atherosclerosis in apolipoprotein E-deficient (APOE(-/-)) mice. FTY720 was orally administered to APOE(-/-) mice 4 weeks of age on a high-cholesterol diet and atherosclerosis was assessed at 8 weeks of age. Oral administration of FTY720 significantly reduced atherosclerotic lesion formation compared with control mice. We observed a significant increase in LAP(+) and Foxp3(+) cells in the CD4+T-cell population of FTY720-treated mice in association with increased production of the anti-inflammatory cytokine transforming growth factor-β (TGF-β) as well as suppressed T-helper type 1 immune responses. Our findings reveal that short-term, low-dose oral FTY720 treatment had great benefits in inhibiting early development of atherosclerosis in mice via induction of a regulatory T-cell response and inhibition of effector T responses. These findings suggest that oral immune modulation may represent an attractive therapeutic approach to atherosclerosis.
Collapse
Affiliation(s)
- K Huang
- Department of Vascular and Thyroid Surgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Bikman BT. A role for sphingolipids in the pathophysiology of obesity-induced inflammation. Cell Mol Life Sci 2012; 69:2135-46. [PMID: 22294100 PMCID: PMC11114706 DOI: 10.1007/s00018-012-0917-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2011] [Revised: 01/01/2012] [Accepted: 01/04/2012] [Indexed: 12/20/2022]
Abstract
Following the initial discovery that adipose tissue actively synthesizes and secretes cytokines, obesity-induced inflammation has been implicated in the etiology of a host of disease states related to obesity, including cardiovascular disease and type II diabetes. Interestingly, a growing body of evidence similarly implicates sphingolipids as prime instigators in these same diseases. From the recent discovery that obesity-related inflammatory pathways modulate sphingolipid metabolism comes a novel perspective—sphingolipids may act as the dominant mediators of deleterious events stemming from obesity-induced inflammation. This paradigm may identify sphingolipids as an effective target for future therapeutics aimed at ameliorating diseases associated with chronic inflammation.
Collapse
Affiliation(s)
- Benjamin T Bikman
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA.
| |
Collapse
|
38
|
Karliner JS. Sphingosine kinase and sphingosine 1-phosphate in the heart: a decade of progress. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1831:203-12. [PMID: 22735359 DOI: 10.1016/j.bbalip.2012.06.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Revised: 06/14/2012] [Accepted: 06/17/2012] [Indexed: 12/11/2022]
Abstract
Activation of sphingosine kinase/sphingosine 1-phosphate (SK/S1P)-mediated signaling has emerged as a critical cardioprotective pathway in response to acute ischemia/reperfusion injury. S1P is released in both ischemic pre- and post-conditioning. Application of exogenous S1P to cultured cardiac myocytes subjected to hypoxia or treatment of isolated hearts either before ischemia or at the onset of reperfusion exerts prosurvival effects. Synthetic congeners of S1P such as FTY720 mimic these responses. Gene targeted mice null for the SK1 isoform whose hearts are subjected to ischemia/reperfusion injury exhibit increased infarct size and respond poorly either to ischemic pre- or postconditioning. Measurements of cardiac SK activity and S1P parallel these observations. Experiments in SK2 knockout mice have revealed that this isoform is necessary for survival in the heart. High density lipoprotein (HDL) is a major carrier of S1P, and studies of hearts in which selected S1P receptors have been inhibited implicate the S1P cargo of HDL in cardioprotection. Inhibition of S1P lyase, an endogenous enzyme that degrades S1P, also leads to cardioprotection. These observations have considerable relevance for future therapeutic approaches to acute and chronic myocardial injury. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.
Collapse
|
39
|
Poti F, Costa S, Bergonzini V, Galletti M, Pignatti E, Weber C, Simoni M, Nofer JR. Effect of sphingosine 1-phosphate (S1P) receptor agonists FTY720 and CYM5442 on atherosclerosis development in LDL receptor deficient (LDL-R⁻/⁻) mice. Vascul Pharmacol 2012; 57:56-64. [PMID: 22459073 DOI: 10.1016/j.vph.2012.03.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2011] [Revised: 02/12/2012] [Accepted: 03/10/2012] [Indexed: 11/17/2022]
Abstract
OBJECTIVES Sphingosine 1-phosphate (S1P)--a lysosphingolipid present in HDL--exerts atheroprotective effects in vitro, while FTY720, a non-selective S1P mimetic inhibits atherosclerosis in LDL receptor-deficient (LDL-R⁻/⁻) mice under conditions of severe hypercholesterolemia. We here examined the effect of FTY720 and a selective S1P receptor type 1 agonist CYM5442 on atherosclerosis in moderately hypercholesterolemic LDL-R⁻/⁻ mice. METHODS AND RESULTS LDL-R⁻/⁻ mice fed Western diet (0.25% cholesterol) were given FTY720 (0.4 mg/kg/day) or CYM5442 (2.0 mg/kg/day) for 18 weeks. FTY720 but not CYM5422 persistently lowered blood lymphocytes, depleted CD4⁺ and CD8⁺ T cells in spleen and lymph nodes, and reduced splenocyte IL-2 secretion. However, both compounds reduced the activity of splenic and peritoneal macrophages as inferred from the down-regulated CD68 and MHC-II expression in CD11b⁺ cells and the reduced IL-6 secretion in response to LPS, respectively. CYM5442 and FTY720 reduced weight gain, white adipose tissue depots and fasting glucose suggesting improvement of metabolic control, but failed to influence atherosclerosis in LDL-R⁻/⁻ mice. CONCLUSION Despite down-regulating macrophage function and--in case of FTY720--altering lymphocyte distribution CYM5442 and FTY720 fail to affect atherosclerosis in moderately hypercholesterolemic LDL-R⁻/⁻ mice. We hypothesize that S1P mimetics exert atheroprotective effects only under conditions of increased cholesterol burden exacerbating vascular inflammation.
Collapse
MESH Headings
- Animals
- Antigens, CD/immunology
- Antigens, CD/metabolism
- Antigens, Differentiation, Myelomonocytic/immunology
- Antigens, Differentiation, Myelomonocytic/metabolism
- Atherosclerosis/drug therapy
- Atherosclerosis/immunology
- Atherosclerosis/metabolism
- Body Weight/immunology
- CD11b Antigen/immunology
- CD11b Antigen/metabolism
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Female
- Fingolimod Hydrochloride
- Genes, MHC Class II/immunology
- Indans/pharmacology
- Interleukin-2/immunology
- Interleukin-2/metabolism
- Interleukin-6/immunology
- Interleukin-6/metabolism
- Lymph Nodes/drug effects
- Lymph Nodes/immunology
- Lymph Nodes/metabolism
- Lymphocyte Activation/immunology
- Lymphocytes/drug effects
- Lymphocytes/immunology
- Lymphocytes/metabolism
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Mice
- Mice, Inbred C57BL
- Oxadiazoles/pharmacology
- Proprotein Convertases/immunology
- Proprotein Convertases/metabolism
- Propylene Glycols/immunology
- Propylene Glycols/pharmacology
- Receptors, LDL/immunology
- Receptors, LDL/metabolism
- Receptors, Lysosphingolipid/agonists
- Receptors, Lysosphingolipid/immunology
- Receptors, Lysosphingolipid/metabolism
- Serine Endopeptidases/immunology
- Serine Endopeptidases/metabolism
- Sphingosine/analogs & derivatives
- Sphingosine/immunology
- Sphingosine/pharmacology
- Spleen/drug effects
- Spleen/immunology
- Spleen/metabolism
Collapse
Affiliation(s)
- Francesco Poti
- Department of Medicine, Endocrinology, Metabolism and Geriatrics, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Shimizu T, De Wispelaere A, Winkler M, D'Souza T, Caylor J, Chen L, Dastvan F, Deou J, Cho A, Larena-Avellaneda A, Reidy M, Daum G. Sphingosine-1-phosphate receptor 3 promotes neointimal hyperplasia in mouse iliac-femoral arteries. Arterioscler Thromb Vasc Biol 2012; 32:955-61. [PMID: 22308044 DOI: 10.1161/atvbaha.111.241034] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE The objective of this study was to define a role for sphingosine-1-phosphate receptor 3 (S1PR3) in intimal hyperplasia. METHODS AND RESULTS A denudation model of the iliac-femoral artery in wild-type and S1PR3-null mice was used to define a role for S1PR3 in the arterial injury response because we found in humans and mice that expression of S1PR3 was higher in these arteries compared with carotid arteries. At 28 days after surgery, wild-type arteries formed significantly larger lesions than S1PR3-null arteries. Bromodeoxyuridine labeling experiments demonstrated that on injury, wild-type arteries exhibited higher medial as well as intimal proliferation than S1PR3-null arteries. Because S1PR3 expression in vitro was low, we expressed S1PR3 in S1PR3-null smooth muscle cells (SMCs) using retroviral-mediated gene transfer to study the effects of S1PR3 on cell functions and signaling. SMCs expressing S1PR3, but not vector-transfected controls, responded to sphingosine-1-phosphate stimulation with activation of Rac, Erk, and Akt. SMCs expressing S1PR3 also migrated more. CONCLUSIONS In humans and mice, S1PR3 expression was higher in iliac-femoral arteries compared with carotid arteries. S1PR3 promoted neointimal hyperplasia on denudation of iliac-femoral arteries in mice, likely by stimulating cell migration and proliferation through activation of signaling pathways involving Erk, Akt, and Rac.
Collapse
Affiliation(s)
- Takuya Shimizu
- Department of Pathology, University of Washington, Seattle, 98109, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Schuchardt M, Tölle M, Prüfer J, van der Giet M. Pharmacological relevance and potential of sphingosine 1-phosphate in the vascular system. Br J Pharmacol 2011; 163:1140-62. [PMID: 21309759 DOI: 10.1111/j.1476-5381.2011.01260.x] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) was identified as a crucial molecule for regulating immune responses, inflammatory processes as well as influencing the cardiovascular system. S1P mediates differentiation, proliferation and migration during vascular development and homoeostasis. S1P is a naturally occurring lipid metabolite and is present in human blood in nanomolar concentrations. S1P is not only involved in physiological but also in pathophysiological processes. Therefore, this complex signalling system is potentially interesting for pharmacological intervention. Modulation of the system might influence inflammatory, angiogenic or vasoregulatory processes. S1P activates G-protein coupled receptors, namely S1P(1-5) , whereas only S1P(1-3) is present in vascular cells. S1P can also act as an intracellular signalling molecule. This review highlights the pharmacological potential of S1P signalling in the vascular system by giving an overview of S1P-mediated processes in endothelial cells (ECs) and vascular smooth muscle cells (VSMCs). After a short summary of S1P metabolism and signalling pathways, the role of S1P in EC and VSMC proliferation and migration, the cause of relaxation and constriction of arterial blood vessels, the protective functions on endothelial apoptosis, as well as the regulatory function in leukocyte adhesion and inflammatory responses are summarized. This is followed by a detailed description of currently known pharmacological agonists and antagonists as new tools for mediating S1P signalling in the vasculature. The variety of effects influenced by S1P provides plenty of therapeutic targets currently under investigation for potential pharmacological intervention.
Collapse
Affiliation(s)
- Mirjam Schuchardt
- Charité- Universitätsmedizin Berlin, CharitéCentrum 10, Department of Nephrology, Campus Benjamin Franklin, Hindenburgdamm 30, Berlin, Germany
| | | | | | | |
Collapse
|
42
|
von Vietinghoff S, Koltsova EK, Mestas J, Diehl CJ, Witztum JL, Ley K. Mycophenolate mofetil decreases atherosclerotic lesion size by depression of aortic T-lymphocyte and interleukin-17-mediated macrophage accumulation. J Am Coll Cardiol 2011; 57:2194-204. [PMID: 21596236 DOI: 10.1016/j.jacc.2010.12.030] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2010] [Revised: 11/24/2010] [Accepted: 12/15/2010] [Indexed: 12/21/2022]
Abstract
OBJECTIVES This study tested whether immunosuppression with mycophenolate mofetil (MMF) inhibits atherosclerosis development in apolipoprotein-E-deficient (Apoe(-/-)) mice and investigated the mechanism. BACKGROUND Chronic vascular inflammation involving both innate and adaptive immunity is central in the development of atherosclerosis, but immunosuppressive treatment is not uniformly beneficial. The immunosuppressive MMF targets lymphocyte proliferation by inhibiting inosine-monophosphate dehydrogenase. METHODS Young and aged Apoe(-/-) mice were treated with 30 mg/kg daily MMF during 12 and 3 weeks of a high-fat diet, respectively. Aortic lesion size and composition was investigated by histology and flow cytometry; soluble inflammatory mediators were investigated by enzyme-linked immunosorbent assay. RESULTS Macroscopic and histologic aortic atherosclerotic lesions were significantly decreased in both MMF-treated groups. While systemic immunoglobulin G directed against low-density lipoproteins was not significantly altered, the T-cell cytokine interleukin (IL)-17 was significantly reduced in plasma of MMF-treated mice and supernatants from their aortas after T-cell stimulation. The MMF treatment decreased aortic αβ T-cell receptor(+) lymphocyte proliferation and cell numbers. Also, aortic contents of CD11b(+)CD11c(+) cells and their proliferation were reduced in MMF-treated Apoe(-/-) mice. The IL-17 supplementation restored the number of proliferating aortic CD11b(+)CD11c(+) cells in MMF-treated mice. The IL-17 receptor A was highly expressed on circulating monocytes that are macrophage progenitors. Genetic deletion of IL-17 receptor A or IL-17A reduced inflammatory peritoneal CD11b(+)CD11c(+) macrophage accumulation. CONCLUSIONS The lymphocyte-directed immunosuppressant MMF that curbs IL-17 production was a successful antiatherosclerotic treatment. Our data delineate a role for IL-17 in CD11b(+)CD11c(+) cell accumulation.
Collapse
|
43
|
Sphingosine-1-Phosphate-Specific G Protein-Coupled Receptors as Novel Therapeutic Targets for Atherosclerosis. Pharmaceuticals (Basel) 2011. [PMCID: PMC4052545 DOI: 10.3390/ph4010117] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory process involving complex interactions of modified lipoproteins, monocyte-derived macrophages or foam cells, lymphocytes, endothelial cells (ECs), and vascular smooth muscle cells. Sphingosine-1-phosphate (S1P), a biologically active blood-borne lipid mediator, exerts pleiotropic effects such as cell proliferation, migration and cell-cell adhesion in a variety of cell types via five members of S1P-specific high-affinity G protein-coupled receptors (S1P1-S1P5). Among them, S1P1, S1P2 and S1P3 are major receptor subtypes which are widely expressed in various tissues. Available evidence suggest that S1P and HDL-bound S1P exert atheroprotective effects including inhibition of leukocyte adhesion and stimulation of endothelial nitric oxide synthase (eNOS) in endothelial cells (ECs) through the activation of Gi signaling pathway via S1P3 and probably S1P1, although there is still controversy. FTY720, the phosphorylation product of which is a high-affinity agonist for all S1P receptors except S1P2 and act as an immunosuppressant by downregulating S1P1 on lymphocytes, inhibits atherosclerosis in LDL receptor-null mice and apoE-null mice through the inhibition of lymphocyte and macrophage functions and probably stimulation of EC functions, without influencing plasma lipid concentrations. In contrast to S1P1 and S1P3, S1P2 facilitates atherosclerosis by activating G12/13-Rho-Rho kinase (ROCK) in apoE-null mice. S1P2 mediates transmigration of monocytes into the arterial intima, oxidized LDL accumulation and cytokine secretion in monocyte-derived macrophages, and eNOS inhibition and cytokine secretion in ECs through Rac inhibition, NF-κB activation and 3′-specific phosphoinositide phosphatase (PTEN) stimulation downstream of G12/13-Rho-ROCK. Systemic long-term administration of a selective S1P2-blocker remarkably inhibits atherosclerosis without overt toxicity. Thus, multiple S1P receptors positively and negatively regulate atherosclerosis through multitudes of mechanisms. Considering the essential and multi-faceted role of S1P2 in atherogenesis and the impact of S1P2 inactivation on atherosclerosis, S1P2 is a particularly promising therapeutic target for atherosclerosis.
Collapse
|
44
|
Keul P, Lucke S, von Wnuck Lipinski K, Bode C, Gräler M, Heusch G, Levkau B. Sphingosine-1-phosphate receptor 3 promotes recruitment of monocyte/macrophages in inflammation and atherosclerosis. Circ Res 2010; 108:314-23. [PMID: 21164103 DOI: 10.1161/circresaha.110.235028] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE The role of sphingosine-1-phosphate (S1P) and its receptors in the pathogenesis of atherosclerosis has not been investigated. OBJECTIVE We hypothesized that the S1P receptor 3 (S1P(3)) plays a causal role in the pathogenesis of atherosclerosis. METHODS AND RESULTS We examined atherosclerotic lesion development in mice deficient for S1P(3) and apolipoprotein (Apo)E. Although S1P(3) deficiency did not affect lesion size after 25 or 45 weeks of normal chow diet, it resulted in a dramatic reduction of the monocyte/macrophage content in lesions of S1P(3)(-/-)/ApoE(-/-) double knockout mice. To search for putative defects in monocyte/macrophage recruitment, we examined macrophage-driven inflammation during thioglycollate-induced peritonitis. Elicited peritoneal macrophages were reduced in S1P(3)-deficient mice and expressed lower levels of tumor necrosis factor-α and monocyte chemoattractant protein-1. Bone marrow-derived S1P(3)-deficient macrophages produced less MCP-1 in response to lipopolysaccharide stimulation. In vitro, S1P was chemotactic for wild-type but not S1P(3)-deficient peritoneal macrophages. In vivo, S1P concentration increased rapidly in the peritoneal cavity after initiation of peritonitis. Treatment with the S1P analog FTY720 attenuated macrophage recruitment to the peritoneum. Studies in bone marrow chimeras showed that S1P(3) in both hematopoietic and nonhematopoietic cells contributed to monocyte/macrophage accumulation in atherosclerotic lesions. Finally, S1P(3) deficiency increased the smooth muscle cell content of atherosclerotic lesions and enhanced neointima formation after carotid ligation arguing for an antiproliferative/antimigratory role of S1P(3) in the arterial injury response. CONCLUSIONS Our data suggest that S1P(3) mediates the chemotactic effect of S1P in macrophages in vitro and in vivo and plays a causal role in atherosclerosis by promoting inflammatory monocyte/macrophage recruitment and altering smooth muscle cell behavior.
Collapse
Affiliation(s)
- Petra Keul
- Institute for Pathophysiology, University Hospital Essen, Hufelandstrasse 55, 45122 Essen, Germany
| | | | | | | | | | | | | |
Collapse
|
45
|
Glaros E, Kim W, Garner B. Myriocin-mediated up-regulation of hepatocyte apoA-I synthesis is associated with ERK inhibition. Clin Sci (Lond) 2010; 118:727-36. [PMID: 20102334 PMCID: PMC2860698 DOI: 10.1042/cs20090452] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2009] [Revised: 12/14/2009] [Accepted: 01/26/2010] [Indexed: 02/05/2023]
Abstract
Sphingolipids including sphingomyelin have been implicated as potential atherogenic lipids. Studies in apoE (apolipoprotein E)-null mice have revealed that the serine palmitoyltransferase inhibitor myriocin reduces plasma levels of sphingomyelin, ceramide, sphingosine-1-phosphate and glycosphingolipids and that this is associated with potent inhibition of atherosclerosis. Interestingly, hepatic apoA-I (apolipoprotein A-I) synthesis and plasma HDL (high-density lipoprotein)-cholesterol levels were also increased in apoE-null mice treated with myriocin. Since myriocin is a known inhibitor of ERK (extracellular-signal-related kinase) phosphorylation, we assessed the possibility that myriocin may be acting to increase hepatic apoA-I production via this pathway. To address this, HepG2 cells and primary mouse hepatocytes were treated with 200 muM myriocin for up to 48 h. Myriocin increased apoA-I mRNA and protein levels by approx. 3- and 2-fold respectively. Myriocin also increased apoA-I secretion up to 3.5-fold and decreased ERK phosphorylation by approx. 70%. Similar findings were obtained when primary hepatocytes were isolated from apoE-null mice that were treated with myriocin (intraperitoneal injection at a dose of 0.3 mg/kg body weight). Further experiments revealed that the MEK (mitogen-activated protein kinase/ERK kinase) inhibitor PD98059 potently inhibited ERK phosphorylation, as expected, and increased primary hepatocyte apoA-I production by 3-fold. These results indicate that ERK phosphorylation plays a role in regulating hepatic apoA-I expression and suggest that the anti-atherogenic mechanism of action for myriocin may be linked to this pathway.
Collapse
Key Words
- apolipoprotein a-i (apoa-i)
- atherosclerosis
- extracellular-signal regulated kinase (erk)
- hepatocyte
- myriocin
- sphingolipid
- 4-aap, 4-aminoantipyrine
- apoa-i, apolipoprotein a-i
- apoe, apolipoprotein-e
- daos, n-ethyl-n-(2-hydroxy-3-sulfopropyl)-3,5-dimethoxyaniline sodium salt
- dmem, dulbecco's modified eagle's medium
- erk, extracellular-signal-related kinase
- fcs, fetal calf serum
- gcs, glucosylceramide synthase
- gsl, glycosphingolipid
- hdl, high-density lipoprotein
- hrp, horseradish peroxidase
- i.p., intraperitoneal(ly)
- ldl, low-density lipoprotein
- mapk, mitogen-activated protein kinase
- mek, mapk/erk kinase
- mtt, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2h-tetrazolium bromide
- qpcr, quantitative pcr
- s1p, sphingosine 1-phosphate
- sm, sphingomyelin
- smase, sphingomyelinase
- spt, serine palmitoyltransferase
Collapse
Affiliation(s)
- Elias N. Glaros
- *Prince of Wales Medical Research Institute, Sydney, NSW 2031, Australia
- †School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Woojin S. Kim
- *Prince of Wales Medical Research Institute, Sydney, NSW 2031, Australia
- †School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
| | - Brett Garner
- *Prince of Wales Medical Research Institute, Sydney, NSW 2031, Australia
- †School of Medical Sciences, Faculty of Medicine, University of New South Wales, Sydney, NSW 2052, Australia
- ‡School of Biological Sciences, University of Wollongong, Wollongong, NSW 2522, Australia
| |
Collapse
|
46
|
Erbel C, Chen L, Bea F, Wangler S, Celik S, Lasitschka F, Wang Y, Böckler D, Katus HA, Dengler TJ. Inhibition of IL-17A attenuates atherosclerotic lesion development in apoE-deficient mice. THE JOURNAL OF IMMUNOLOGY 2010; 183:8167-75. [PMID: 20007582 DOI: 10.4049/jimmunol.0901126] [Citation(s) in RCA: 267] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The importance of an (auto)immune response in atherogenesis is becoming increasingly well understood. IL-17A-expressing T cells modulate immune cell trafficking, initiating inflammation and cytokine production in (auto)immune diseases. In human carotid artery plaques, we previously showed the presence of IL-17A-producing T cells and IL-23; however, IL-17A effects on atherogenesis have not been studied. Aortic root sections from 8-wk-old apolipoprotein E-deficient mice fed a standard chow diet were examined after 12 wk for lesion area, plaque composition, cellular infiltration, cytokine expression, and apoptosis. The treatment group (n = 15) received anti-IL-17A Ab and the controls (n = 10) received irrelevant Abs. Inhibition of IL-17A markedly reduced atherosclerotic lesion area (p < 0.001), maximal stenosis (p < 0.001), and vulnerability of the lesion. IL-17A mAb-treated mice showed reduced cellular infiltration, down-regulation of activation markers on endothelium and immune cells (e.g., VCAM-1), and reduced cytokine/chemokine secretion (e.g., IL6, TNFalpha, CCL5). To investigate possible mechanisms, different atherogenic cell types (e.g., macrophages, dendritic cells, HUVECs, vascular smooth muscle cells) were stimulated with IL-17A in addition to TNF-alpha, IFN-gamma, or LPS to induce cellular activation or apoptosis in vitro. Stimulation with IL-17A induced proinflammatory changes in several atherogenic cell types and apoptotic cell death in murine cells. Functional blockade of IL-17A reduces atherosclerotic lesion development and decreases plaque vulnerability, cellular infiltration, and tissue activation in apolipoprotein E-deficient mice. The present data support a pathogenic role of IL-17A in the development of atherosclerosis by way of its widespread proinflammatory and proapoptotic effects on atherogenic cells.
Collapse
Affiliation(s)
- Christian Erbel
- Department of Cardiology, University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Blom T, Bäck N, Mutka AL, Bittman R, Li Z, de Lera A, Kovanen PT, Diczfalusy U, Ikonen E. FTY720 stimulates 27-hydroxycholesterol production and confers atheroprotective effects in human primary macrophages. Circ Res 2010; 106:720-9. [PMID: 20056921 DOI: 10.1161/circresaha.109.204396] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
RATIONALE The synthetic sphingosine analog FTY720 is undergoing clinical trials as an immunomodulatory compound, acting primarily via sphingosine 1-phosphate receptor activation. Sphingolipid and cholesterol homeostasis are closely connected but whether FTY720 affects atherogenesis in humans is not known. OBJECTIVE We examined the effects of FTY720 on the processing of scavenged lipoprotein cholesterol in human primary monocyte-derived macrophages. METHODS AND RESULTS FTY720 did not affect cholesterol uptake but inhibited its delivery to the endoplasmic reticulum, reducing cellular free cholesterol cytotoxicity. This was accompanied by increased levels of Niemann-Pick C1 protein (NPC1) and ATP-binding cassette transporter (ABC)A1 proteins and increased efflux of endosomal cholesterol to apolipoprotein A-I. These effects were not dependent on sphingosine 1-phosphate receptor activation. Instead, FTY720 stimulated the production of 27-hydroxycholesterol, an endogenous ligand of the liver X receptor, leading to liver X receptor-induced upregulation of ABCA1. Fluorescently labeled FTY720 was targeted to late endosomes, and the FTY720-induced upregulation of ABCA1 was NPC1-dependent, but the endosomal exit of FTY720 itself was not. CONCLUSIONS We conclude that FTY720 decreases cholesterol toxicity in primary human macrophages by reducing the delivery of scavenged lipoprotein cholesterol to the endoplasmic reticulum and facilitating its release to physiological extracellular acceptors. Furthermore, FTY720 stimulates 27-hydroxycholesterol production, providing an explanation for the atheroprotective effects and identifying a novel mechanism by which FTY720 modulates signaling.
Collapse
Affiliation(s)
- Tomas Blom
- Institute of Biomedicine/Anatomy, Haartmaninkatu 8, University of Helsinki, Finland
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Klingenberg R, Hansson GK. Treating inflammation in atherosclerotic cardiovascular disease: emerging therapies. Eur Heart J 2009; 30:2838-44. [PMID: 19880848 DOI: 10.1093/eurheartj/ehp477] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis constitutes the underlying disease to the clinical manifestations of myocardial infarction, stroke, and gangrene. Despite the success of statins, prevention of clinical events of atherosclerosis remains a major challenge in current-day cardiology. Research into the inflammatory nature of atherosclerosis has led to improved mechanistic understanding of its pathogenesis and to the identification of novel therapeutic targets discussed in this review. Recent genetic and epidemiological data document shared pathologies of chronic inflammatory diseases and atherosclerosis. Anti-inflammatory treatment regimens used in these diseases, including tumor necrosis factor-alpha blockade, IL-1 receptor antagonism, and leukotriene blockade may be beneficial also in patients with coronary artery disease. Enhancing inherent atheroprotective immunity by expansion of regulatory T cells may emerge as a future therapeutic strategy. Immunization strategies directed against atherosclerosis-related antigens such as epitopes within the low-density lipoprotein particle have been extensively studied in animal models and may enter the clinical stage. Success of these novel therapies will be critically dependent on the adequate identification of patients and choice of appropriate clinical endpoints.
Collapse
Affiliation(s)
- Roland Klingenberg
- Department of Cardiology, University Hospital Zurich, CH-8091 Zurich, Switzerland.
| | | |
Collapse
|
49
|
Gereke M, Jung S, Buer J, Bruder D. Alveolar Type II Epithelial Cells Present Antigen to CD4+T Cells and Induce Foxp3+Regulatory T Cells. Am J Respir Crit Care Med 2009; 179:344-55. [DOI: 10.1164/rccm.200804-592oc] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
50
|
Sattler K, Levkau B. Sphingosine-1-phosphate as a mediator of high-density lipoprotein effects in cardiovascular protection. Cardiovasc Res 2009; 82:201-11. [PMID: 19233866 DOI: 10.1093/cvr/cvp070] [Citation(s) in RCA: 149] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) has gained special attention in the high-density lipoprotein (HDL) field because HDL is the most prominent plasma carrier of S1P and because the S1P content of HDL may be responsible for many of the pleiotropic functions of HDL. This revelation has come from the evidence that HDL employ S1P receptors and signalling pathways to implement several HDL-ascribed biological effects as diverse as endothelial nitric oxide production, vasodilation, survival, and cardioprotection. This review focuses on HDL effects that are completely or partially mediated by the S1P content of the HDL particle and differentiates them from genuine HDL effects that are S1P-independent. In addition, the functional properties of 'free', HDL-unbound S1P are sometimes different from or even contrary to those of HDL-associated S1P. The nature of the physical interactions between HDL and local and systemic S1P production will be discussed as well as their consequences for organ function. Finally, we will elucidate the potential benefits and limitations of S1P analogues as a new class of functional HDL mimetics for cardiovascular therapy.
Collapse
Affiliation(s)
- Katherine Sattler
- Institute of Pathophysiology, Zentrum für Innere Medizin, Universitätsklinikum Essen, Hufelandstr. 55, 45122 Essen, Germany
| | | |
Collapse
|