Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Acta Neuropathol ; 124(3): 397-410, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22810490

ABSTRACT

Alterations in sphingolipid metabolism are described to contribute to various neurological disorders. We here determined the expression of enzymes involved in the sphingomyelin cycle and their products in postmortem brain tissue of multiple sclerosis (MS) patients. In parallel, we investigated the effect of the sphingosine-1 receptor agonist Fingolimod (Gilenya(®)) on sphingomyelin metabolism in reactive astrocytes and determined its functional consequences for the process of neuro-inflammation. Our results demonstrate that in active MS lesions, marked by large number of infiltrated immune cells, an altered expression of enzymes involved in the sphingomyelin cycle favors enhanced ceramide production. We identified reactive astrocytes as the primary cellular source of enhanced ceramide production in MS brain samples. Astrocytes isolated from MS lesions expressed enhanced mRNA levels of the ceramide-producing enzyme acid sphingomyelinase (ASM) compared to astrocytes isolated from control white matter. In addition, TNF-α treatment induced ASM mRNA and ceramide levels in astrocytes isolated from control white matter. Incubation of astrocytes with Fingolimod prior to TNF-α treatment reduced ceramide production and mRNA expression of ASM to control levels in astrocytes. Importantly, supernatants derived from reactive astrocytes treated with Fingolimod significantly reduced transendothelial monocyte migration. Overall, the present study demonstrates that reactive astrocytes represent a possible additional cellular target for Fingolimod in MS by directly reducing the production of pro-inflammatory lipids and limiting subsequent transendothelial leukocyte migration.


Subject(s)
Astrocytes/drug effects , Blood-Brain Barrier/drug effects , Ceramides/metabolism , Immunosuppressive Agents/pharmacology , Multiple Sclerosis/physiopathology , Propylene Glycols/pharmacology , Sphingosine/analogs & derivatives , Adult , Aged , Aged, 80 and over , Astrocytes/metabolism , Astrocytes/pathology , Blood-Brain Barrier/pathology , Blood-Brain Barrier/physiopathology , Cell Movement/drug effects , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Fingolimod Hydrochloride , Humans , Male , Middle Aged , Monocytes/drug effects , Monocytes/metabolism , Monocytes/pathology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Sphingomyelins/metabolism , Sphingosine/pharmacology
2.
J Neuroinflammation ; 9: 133, 2012 Jun 20.
Article in English | MEDLINE | ID: mdl-22715976

ABSTRACT

BACKGROUND: The sphingosine 1-phosphate (S1P) receptor modulator FTY720P (Gilenya®) potently reduces relapse rate and lesion activity in the neuroinflammatory disorder multiple sclerosis. Although most of its efficacy has been shown to be related to immunosuppression through the induction of lymphopenia, it has been suggested that a number of its beneficial effects are related to altered endothelial and blood-brain barrier (BBB) functionality. However, to date it remains unknown whether brain endothelial S1P receptors are involved in the maintenance of the function of the BBB thereby mediating immune quiescence of the brain. Here we demonstrate that the brain endothelial receptor S1P5 largely contributes to the maintenance of brain endothelial barrier function. METHODS: We analyzed the expression of S1P5 in human post-mortem tissues using immunohistochemistry. The function of S1P5 at the BBB was assessed in cultured human brain endothelial cells (ECs) using agonists and lentivirus-mediated knockdown of S1P5. Subsequent analyses of different aspects of the brain EC barrier included the formation of a tight barrier, the expression of BBB proteins and markers of inflammation and monocyte transmigration. RESULTS: We show that activation of S1P5 on cultured human brain ECs by a selective agonist elicits enhanced barrier integrity and reduced transendothelial migration of monocytes in vitro. These results were corroborated by genetically silencing S1P5 in brain ECs. Interestingly, functional studies with these cells revealed that S1P5 strongly contributes to brain EC barrier function and underlies the expression of specific BBB endothelial characteristics such as tight junctions and permeability. In addition, S1P5 maintains the immunoquiescent state of brain ECs with low expression levels of leukocyte adhesion molecules and inflammatory chemokines and cytokines through lowering the activation of the transcription factor NFκB. CONCLUSION: Our findings demonstrate that S1P5 in brain ECs contributes to optimal barrier formation and maintenance of immune quiescence of the barrier endothelium.


Subject(s)
Blood-Brain Barrier/cytology , Blood-Brain Barrier/immunology , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Immunity, Cellular , Receptors, Lysosphingolipid/physiology , Aged, 80 and over , Blood-Brain Barrier/metabolism , Cell Line , Cells, Cultured , Endothelium, Vascular/metabolism , Gene Knockdown Techniques , Humans , Immunity, Cellular/genetics , Lentivirus/genetics , Male , Receptors, Lysosphingolipid/deficiency , Receptors, Lysosphingolipid/genetics
3.
Glia ; 58(12): 1465-76, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20648639

ABSTRACT

Sphingolipids are a class of biologically active lipids that have a role in multiple biological processes including inflammation. Sphingolipids exert their functions by direct signaling or through signaling by their specific receptors. Phosphorylated FTY720 (FTY720P) is a sphingosine 1-phosphate (S1P) analogue that is currently in trial for treatment of multiple sclerosis (MS), which targets all S1P receptors but S1P(2). To date, however, it remains unknown whether FTY720P may exert direct anti-inflammatory effects within the central nervous system (CNS), because data concerning S1P receptor expression and regulation under pathological conditions in the human brain are lacking. To investigate potential regulation of S1P receptors in the human brain during MS, we performed immunohistochemical analysis of S1P receptor 1 and 3 expression in well-characterized MS lesions. A strong increase in S1P receptor 1 and 3 expression on reactive astrocytes was detected in active and chronic inactive MS lesions. In addition, we treated primary cultures of human astrocytes with the proinflammatory cytokine tumor necrosis factor-alpha to identify the regulation of S1P(1/3) on astrocytes under pathological conditions. Importantly, we demonstrate that FTY720P exerts an anti-inflammatory action on human astrocytes by limiting secretion of proinflammatory cytokines. Our data demonstrate that reactive astrocytes in MS lesions and cultured under proinflammatory conditions strongly enhance expression of S1P receptors 1 and 3. Results from this study indicate that astrocytes may act as a yet-unknown target within the CNS for the anti-inflammatory effects observed after FTY720P administration in the treatment of MS.


Subject(s)
Multiple Sclerosis/physiopathology , Receptors, Lysosphingolipid/metabolism , Up-Regulation/physiology , Adult , Aged , Aged, 80 and over , Astrocytes/metabolism , Brain/cytology , Cells, Cultured , Chemokines/metabolism , Enzyme-Linked Immunosorbent Assay/methods , Female , Fingolimod Hydrochloride , Humans , Immunosuppressive Agents/pharmacology , Male , Middle Aged , Propylene Glycols/pharmacology , Receptors, Lysosphingolipid/genetics , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Sphingosine-1-Phosphate Receptors , T-Lymphocytes/metabolism , Up-Regulation/drug effects
4.
J Clin Invest ; 118(8): 2979-85, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18654662

ABSTRACT

Novel biomarkers, such as circulating (auto)antibody signatures, may improve early detection and treatment of ruptured atherosclerotic lesions and accompanying cardiovascular events, such as myocardial infarction. Using a phage-display library derived from cDNAs preferentially expressed in ruptured peripheral human atherosclerotic plaques, we performed serological antigen selection to isolate displayed cDNA products specifically interacting with antibodies in sera from patients with proven ruptured peripheral atherosclerotic lesions. Two cDNA products were subsequently evaluated on a validation series of patients with peripheral atherosclerotic lesions, healthy controls, and patients with coronary artery disease at different stages. Our biomarker set was able to discriminate between patients with peripheral ruptured lesions and patients with peripheral stable plaques with 100% specificity and 76% sensitivity. Furthermore, 93% of patients with an acute myocardial infarction (AMI) tested positive for our biomarkers, whereas all patients with stable angina pectoris tested negative. Moreover, 90% of AMI patients who initially tested negative for troponin T, for which a positive result is known to indicate myocardial infarction, tested positive for our biomarkers upon hospital admission. In conclusion, antibody profiling constitutes a promising approach for noninvasive diagnosis of atherosclerotic lesions, because a positive serum response against a set of 2 cDNA products showed a strong association with the presence of ruptured peripheral atherosclerotic lesions and myocardial infarction.


Subject(s)
Atherosclerosis/diagnosis , Atherosclerosis/pathology , Autoantibodies , Myocardial Infarction/diagnosis , Myocardial Infarction/pathology , Aged , Angina Pectoris/blood , Angina Pectoris/pathology , Antigens/immunology , Atherosclerosis/blood , Autoantibodies/blood , Biomarkers/blood , Case-Control Studies , Cohort Studies , Coronary Artery Disease/blood , Coronary Artery Disease/pathology , Cross-Sectional Studies , DNA, Complementary/isolation & purification , Female , Humans , Male , Middle Aged , Myocardial Infarction/blood , Peptide Library , Peripheral Vascular Diseases/blood , Peripheral Vascular Diseases/pathology , Reproducibility of Results , Rupture, Spontaneous/pathology , Sensitivity and Specificity
5.
FASEB J ; 21(13): 3666-76, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17586731

ABSTRACT

The blood-brain barrier (BBB) prevents the entrance of circulating molecules and immune cells into the central nervous system. The barrier is formed by specialized brain endothelial cells that are interconnected by tight junctions (TJ). A defective function of the BBB has been described for a variety of neuroinflammatory diseases, indicating that proper regulation is essential for maintaining brain homeostasis. Under pathological conditions, reactive oxygen species (ROS) significantly contribute to BBB dysfunction and inflammation in the brain by enhancing cellular migration. However, a detailed study about the molecular mechanism by which ROS alter BBB integrity has been lacking. Here we demonstrate that ROS alter BBB integrity, which is paralleled by cytoskeleton rearrangements and redistribution and disappearance of TJ proteins claudin-5 and occludin. Specific signaling pathways, including RhoA and PI3 kinase, mediated observed processes and specific inhibitors of these pathways prevented ROS-induced monocyte migration across an in vitro model of the BBB. Interestingly, these processes were also mediated by protein kinase B (PKB/Akt), a previously unknown player in cytoskeleton and TJ dynamics that acted downstream of RhoA and PI3 kinase. Our study reveals new insights into molecular mechanisms underlying BBB regulation and provides novel opportunities for the treatment of neuroinflammatory diseases.


Subject(s)
Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Tight Junctions/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cell Line, Transformed , Humans , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...