Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Evi Schmid is active.

Publication


Featured researches published by Evi Schmid.


Circulation Research | 2012

The Inflammatory Chemokine CXC Motif Ligand 16 Triggers Platelet Activation and Adhesion Via CXC Motif Receptor 6–Dependent Phosphatidylinositide 3-Kinase/Akt Signaling

Oliver Borst; Patrick Münzer; Sergios Gatidis; Eva-Maria Schmidt; Tanja Schönberger; Evi Schmid; Syeda T. Towhid; Konstantinos Stellos; Peter Seizer; Andreas E. May; Florian Lang; Meinrad Gawaz

Rationale: The recently discovered chemokine CXC motif ligand 16 (CXCL16) is highly expressed in atherosclerotic lesions and is a potential pathogenic mediator in coronary artery disease. Objective: The aim of this study was to test the role of CXCL16 on platelet activation and vascular adhesion, as well as the underlying mechanism and signaling pathway. Methods and Results: Reverse-transcriptase polymerase chain reaction, Western blotting, confocal microscopy, and flow cytometry revealed that CXCL16-specific receptor, CXC motif receptor 6, is highly expressed in platelets. According to flow cytometry and confocal microscopy, stimulation of platelets with CXCL16 induced platelet degranulation, integrin &agr;IIb&bgr;3 activation, and shape change. CXCL16 increased Akt phosphorylation (Thr308/Ser473), an effect abrogated by phosphatidylinositide 3-kinase inhibitors wortmannin (100 nmol/L) and LY294002 (25 µmol/L). The phosphatidylinositide 3-kinase inhibitors and Akt inhibitor SH-6 (20 µmol/L) further diminished CXCL16-induced platelet activation. CXCL16-mediated platelet degranulation, integrin &agr;IIb&bgr;3 activation, and Akt phosphorylation were blunted in platelets lacking CXCL16-specific receptor CXC motif receptor 6. CXCL16-induced platelet activation was abrogated in Akt1- or Akt2-deficient platelets. CXCL16 enhanced platelet adhesion to endothelium in vitro after high arterial shear stress (2000−s) and to injured vascular wall in vivo after carotid ligation. CXCL16-induced stimulation of platelet adhesion again was prevented by phosphatidylinositide 3-kinase and Akt inhibitors. Apyrase and antagonists of platelet purinergic receptors P2Y1 (MRS2179, 100 µmol/L) and especially P2Y12 (Cangrelor, 10 µmol/L) blunted CXCL16-triggered platelet activation as well as CXCL16-induced platelet adhesion under high arterial shear stress in vitro and after carotid ligation in vivo. Conclusions: The inflammatory chemokine CXCL16 triggers platelet activation and adhesion via CXC motif receptor 6–dependent phosphatidylinositide 3-kinase/Akt signaling and paracrine activation, suggesting a decisive role for CXCL16 in linking vascular inflammation and thrombo-occlusive diseases.Rationale: The recently discovered chemokine CXCL16 is highly expressed in atherosclerotic lesions and a potential pathogenic mediator in coronary artery disease. Objective: To test the role of CXCL16 on platelet activation and vascular adhesion as well as the underlying mechanism and signaling pathway. Methods and Results: RT-PCR, western blotting, confocal microscopy and flow cytometry revealed that CXCL16-specific receptor CXCR6 is highly expressed on platelets. According to flow cytometry and confocal microscopy stimulation of platelets with CXCL16 induced platelet degranulation, integrin α IIb β 3 activation and shape change. CXCL16 increased Akt phosphorylation (Thr 308 /Ser 473 ), an effect abrogated by phosphatidylinositide 3-kinase (PI3K) inhibitors wortmannin (100nM) and LY294002 (25 µM). The PI3K inhibitors and Akt inhibitor SH-6 (20 µM) further diminished CXCL16-induced platelet activation. CXCL16-mediated platelet degranulation, integrin α IIb β 3 activation and Akt phosphorylation were blunted in platelets lacking CXCL16-specific receptor CXCR6. CXCL16-induced platelet activation was abrogated in Akt1- or Akt2-deficient platelets. CXCL16 enhanced platelet adhesion to endothelium in vitro following high arterial shear stress (2000 -s ) and to injured vascular wall in vivo following carotis ligation. CXCL16-induced stimulation of platelet adhesion was again prevented by PI3K and Akt inhibitors. Apyrase and antagonists of platelet purinergic receptors P 2 Y 1 (MRS2179, 100µM) and especially P 2 Y 12 (Cangrelor, 10µM) blunted CXCL16-triggered platelet activation as well as CXCL16-induced platelet adhesion under high arterial shear stress in vitro and after carotis ligation in vivo. Conclusions: The inflammatory chemokine CXCL16 triggers platelet activation and adhesion via CXCR6-dependent PI3K/Akt signaling and paracrine activation suggesting a decisive role for CXCL16 in linking vascular inflammation and thrombo-occlusive diseases.


Blood | 2012

The serum- and glucocorticoid-inducible kinase 1 (SGK1) influences platelet calcium signaling and function by regulation of Orai1 expression in megakaryocytes

Oliver Borst; Eva-Maria Schmidt; Patrick Münzer; Tanja Schönberger; Syeda T. Towhid; Margitta Elvers; Christina Leibrock; Evi Schmid; Anja Eylenstein; Dietmar Kuhl; Andreas E. May; Meinrad Gawaz; Florian Lang

Platelets are activated on increase of cytosolic Ca2+ activity ([Ca2+](i)), accomplished by store-operated Ca2+ entry (SOCE) involving the pore-forming ion channel subunit Orai1. Here, we show, for the first time, that the serum- and glucocorticoid-inducible kinase 1 (SGK1) is expressed in platelets and megakaryocytes. SOCE and agonist-induced [Ca2+](i) increase are significantly blunted in platelets from SGK1 knockout mice (sgk1(-/-)). Similarly, Ca2+ -dependent degranulation, integrin α(IIb)β3 activation, phosphatidylserine exposure, aggregation, and in vitro thrombus formation were significantly impaired in sgk1(-/-) platelets, whereas tail bleeding time was not significantly enhanced. Platelet and megakaryocyte Orai1 transcript levels and membrane protein abundance were significantly reduced in sgk1(-/-) mice. In human megakaryoblastic cells (MEG-01), transfection with constitutively active (S422D)SGK1 but not with inactive (K127N)SGK1 significantly enhanced Orai1 expression and SOCE, while effects reversed by the SGK1 inhibitor GSK650394 (1μM). Transfection of MEG-01 cells with (S422D)SGK1 significantly increased phosphorylation of IκB kinase α/β and IκBα resulting in nuclear translocation of NF-κB subunit p65. Treatment of (S422D)SGK1-transfected MEG-01 cells with the IκB kinase inhibitor BMS-345541 (10μM) abolished SGK1-induced increase of Orai1 expression and SOCE. The present observations unravel SGK1 as novel regulator of platelet function, effective at least in part by NF-κB-dependent transcriptional up-regulation of Orai1 in megakaryocytes and increasing platelet SOCE.


Circulation Research | 2012

The Inflammatory Chemokine CXCL16 Triggers Platelet Activation and Adhesion via CXCR6-Dependent PI3K/Akt Signaling

Oliver Borst; Patrick Münzer; Sergios Gatidis; Eva-Maria Schmidt; Tanja Schönberger; Evi Schmid; Syeda T. Towhid; Konstantinos Stellos; Peter Seizer; Andreas E. May; Florian Lang; Meinrad Gawaz

Rationale: The recently discovered chemokine CXC motif ligand 16 (CXCL16) is highly expressed in atherosclerotic lesions and is a potential pathogenic mediator in coronary artery disease. Objective: The aim of this study was to test the role of CXCL16 on platelet activation and vascular adhesion, as well as the underlying mechanism and signaling pathway. Methods and Results: Reverse-transcriptase polymerase chain reaction, Western blotting, confocal microscopy, and flow cytometry revealed that CXCL16-specific receptor, CXC motif receptor 6, is highly expressed in platelets. According to flow cytometry and confocal microscopy, stimulation of platelets with CXCL16 induced platelet degranulation, integrin &agr;IIb&bgr;3 activation, and shape change. CXCL16 increased Akt phosphorylation (Thr308/Ser473), an effect abrogated by phosphatidylinositide 3-kinase inhibitors wortmannin (100 nmol/L) and LY294002 (25 µmol/L). The phosphatidylinositide 3-kinase inhibitors and Akt inhibitor SH-6 (20 µmol/L) further diminished CXCL16-induced platelet activation. CXCL16-mediated platelet degranulation, integrin &agr;IIb&bgr;3 activation, and Akt phosphorylation were blunted in platelets lacking CXCL16-specific receptor CXC motif receptor 6. CXCL16-induced platelet activation was abrogated in Akt1- or Akt2-deficient platelets. CXCL16 enhanced platelet adhesion to endothelium in vitro after high arterial shear stress (2000−s) and to injured vascular wall in vivo after carotid ligation. CXCL16-induced stimulation of platelet adhesion again was prevented by phosphatidylinositide 3-kinase and Akt inhibitors. Apyrase and antagonists of platelet purinergic receptors P2Y1 (MRS2179, 100 µmol/L) and especially P2Y12 (Cangrelor, 10 µmol/L) blunted CXCL16-triggered platelet activation as well as CXCL16-induced platelet adhesion under high arterial shear stress in vitro and after carotid ligation in vivo. Conclusions: The inflammatory chemokine CXCL16 triggers platelet activation and adhesion via CXC motif receptor 6–dependent phosphatidylinositide 3-kinase/Akt signaling and paracrine activation, suggesting a decisive role for CXCL16 in linking vascular inflammation and thrombo-occlusive diseases.Rationale: The recently discovered chemokine CXCL16 is highly expressed in atherosclerotic lesions and a potential pathogenic mediator in coronary artery disease. Objective: To test the role of CXCL16 on platelet activation and vascular adhesion as well as the underlying mechanism and signaling pathway. Methods and Results: RT-PCR, western blotting, confocal microscopy and flow cytometry revealed that CXCL16-specific receptor CXCR6 is highly expressed on platelets. According to flow cytometry and confocal microscopy stimulation of platelets with CXCL16 induced platelet degranulation, integrin α IIb β 3 activation and shape change. CXCL16 increased Akt phosphorylation (Thr 308 /Ser 473 ), an effect abrogated by phosphatidylinositide 3-kinase (PI3K) inhibitors wortmannin (100nM) and LY294002 (25 µM). The PI3K inhibitors and Akt inhibitor SH-6 (20 µM) further diminished CXCL16-induced platelet activation. CXCL16-mediated platelet degranulation, integrin α IIb β 3 activation and Akt phosphorylation were blunted in platelets lacking CXCL16-specific receptor CXCR6. CXCL16-induced platelet activation was abrogated in Akt1- or Akt2-deficient platelets. CXCL16 enhanced platelet adhesion to endothelium in vitro following high arterial shear stress (2000 -s ) and to injured vascular wall in vivo following carotis ligation. CXCL16-induced stimulation of platelet adhesion was again prevented by PI3K and Akt inhibitors. Apyrase and antagonists of platelet purinergic receptors P 2 Y 1 (MRS2179, 100µM) and especially P 2 Y 12 (Cangrelor, 10µM) blunted CXCL16-triggered platelet activation as well as CXCL16-induced platelet adhesion under high arterial shear stress in vitro and after carotis ligation in vivo. Conclusions: The inflammatory chemokine CXCL16 triggers platelet activation and adhesion via CXCR6-dependent PI3K/Akt signaling and paracrine activation suggesting a decisive role for CXCL16 in linking vascular inflammation and thrombo-occlusive diseases.


Journal of Biological Chemistry | 2012

Transcription Factor NF-κB Regulates Expression of Pore-forming Ca2+ Channel Unit, Orai1, and Its Activator, STIM1, to Control Ca2+ Entry and Affect Cellular Functions

Anja Eylenstein; Sebastian Schmidt; Shuchen Gu; Wenting Yang; Evi Schmid; Eva-Maria Schmidt; Ioana Alesutan; Kalina Szteyn; Ivonne Regel; Ekaterina Shumilina; Florian Lang

The serum and glucocorticoid-inducible kinase SGK1 increases the activity of Orai1, the pore forming unit of store-operated Ca2+ entry, and thus influences Ca2+-dependent cellular functions such as migration. SGK1 further regulates transcription factor nuclear factor κB (NF-κB). This study explored whether SGK1 influences transcription of Orai1 and/or STIM1, the Orai1-activating Ca2+ sensor. Orai1 and STIM1 transcript levels were decreased in mast cells from SGK1 knock-out mice and increased in HEK293 cells transfected with active S422DSGK1 but not with inactive K127NSGK1 or in S422DSGK1-transfected cells treated with the NF-κB inhibitor Wogonin (100 μm). Treatment with the stem cell factor enhanced transcript levels of STIM1 and Orai1 in sgk1+/+ but not in sgk1−/− mast cells and not in sgk1+/+ cells treated with Wogonin. Orai1 and STIM1 transcript levels were further increased in sgk1+/+ and sgk1−/− mast cells by transfection with active NF-κB subunit p65 as well as in HEK293 cells by transfection with NF-κB subunits p65/p50 or p65/p52. They were decreased by silencing of NF-κB subunits p65, p50, or p52 or by NF-κB inhibitor Wogonin (100 μm). Luciferase assay and chromatin immunoprecipitation defined NF-κB-binding sites in promoter regions accounting for NF-κB sensitive genomic regulation of STIM1 and Orai1. Store-operated Ca2+ entry was similarly increased by overexpression of p65/p50 or p65/p52 and decreased by treatment with Wogonin. Transfection of HEK293 cells with p65/p50 or p65/p52 further augmented migration. The present observations reveal powerful genomic regulation of Orai1/STIM1 by SGK1-dependent NF-κB signaling.


Cellular Physiology and Biochemistry | 2012

SGK3 regulates Ca(2+) entry and migration of dendritic cells.

Evi Schmid; Madhuri Bhandaru; Meerim K. Nurbaeva; Wenting Yang; Kalina Szteyn; Antonella Russo; Christina Leibrock; Leonid Tyan; David A. Pearce; Ekaterina Shumilina; Florian Lang

Background/Aims: Dendritic cells (DCs) are antigen-presenting cells linking innate and adaptive immunity. DC maturation and migration are governed by alterations of cytosolic Ca<sup>2+</sup> concentrations ([Ca<sup>2+</sup>]<sub>i</sub>). Ca<sup>2+</sup> entry is in part accomplished by store-operated Ca<sup>2+</sup> (SOC) channels consisting of the membrane pore-forming subunit Orai and the ER Ca<sup>2+</sup> sensing subunit STIM. Moreover, DC functions are under powerful regulation of the phosphatidylinositol-3-kinase (PI3K) pathway, which suppresses proinflammatory cytokine production but supports DC migration. Downstream targets of PI3K include serum- and glucocorticoid-inducible kinase isoform SGK3. The present study explored, whether SGK3 participates in the regulation of [Ca<sup>2+</sup>]<sub>i</sub> and Ca<sup>2+</sup>-dependent functions of DCs, such as maturation and migration. Methods/ Results: Experiments were performed with bone marrow derived DCs from gene targeted mice lacking SGK3 (sgk3<sup>-/-</sup>) and DCs from their wild type littermates (sgk3<sup>+/+</sup>). Maturation, phagocytosis and cytokine production were similar in sgk3<sup>-/-</sup> and sgk3<sup>+/+</sup> DCs. However, SOC entry triggered by intracellular Ca<sup>2+</sup> store depletion with the endosomal Ca<sup>2+</sup> ATPase inhibitor thapsigargin (1 µM) was significantly reduced in sgk3<sup>-/-</sup> compared to sgk3<sup>+/+</sup> DCs. Similarly, bacterial lipopolysaccharide (LPS, 1 µg/ml)- and chemokine CXCL12 (300 ng/ml)- induced increase in [Ca<sup>2+</sup>]<sub>i</sub> was impaired in sgk3<sup>-/-</sup> DCs. Moreover, currents through SOC channels were reduced in sgk3<sup>-/-</sup> DCs. STIM2 transcript levels and protein abundance were significantly lower in sgk3<sup>-/-</sup> DCs than in sgk3<sup>+/+</sup> DCs, whereas Orai1, Orai2, STIM1 and TRPC1 transcript levels and/or protein abundance were similar in sgk3-/- and sgk3<sup>+/+</sup> DCs. Migration of both, immature DCs towards CXCL12 and LPS-matured DCs towards CCL21 was reduced in sgk3<sup>-/-</sup> as compared to sgk3<sup>+/+</sup> DCs. Migration of sgk3<sup>+/+</sup> DCs was further sensitive to SOC channel inhibitor 2-APB (50 µM) and to STIM1/STIM2 knock-down. Conclusion: SGK3 contributes to the regulation of store-operated Ca<sup>2+</sup> entry into and migration of dendritic cells, effects at least partially mediated through SGK3-dependent upregulation of STIM2 expression.


Cellular Physiology and Biochemistry | 2012

Expression and functional significance of the Ca(2+)-activated Cl(-) channel ANO6 in dendritic cells.

Kalina Szteyn; Evi Schmid; Meerim K. Nurbaeva; Wenting Yang; Patrick Münzer; Karl Kunzelmann; Florian Lang; Ekaterina Shumilina

Background/Aims: Migration of dendritic cells (DCs), antigen presenting cells that link innate and adaptive immunity, is critical for initiation of immune responses. DC migration is controlled by the activity of different ion channels, which mediate Ca2+ flux or set the membrane potential. Moreover, cell migration requires local volume changes at the leading and rear end of travelling cells, which might be mediated by the fluxes of osmotically active solutes, including Cl-. The present study explored the functional expression, regulation and role of Cl- channels in mouse bone marrow-derived DCs. Methods/Results: In whole-cell patch clamp experiments we detected outwardly rectifying Cl- currents which were activated by elevation of cytosolic Ca2+, triggered either by ionomycin in the presence of extracellular Ca2+ or mobilization of Ca2+ by IP3 Most importantly, Ca2+-activated Cl- channels (CaCCs) were activated by CCL21 (75 ng/ml), an agonist of the chemokine receptor CCR7. The currents showed sensitivity to Cl- channel blockers such as tannic acid (10 µM), digallic acid (100 µM) and more specific CaCC blockers niflumic acid (300 µM) and AO1 (20 µM). According to RT-PCR and Western blot data, Anoctamin 6 (ANO6) is expressed in DCs. Knock-down of ANO6 with siRNA led to inhibition of CaCC currents in DCs. Moreover, chemokine-induced migration of both immature and LPS-matured DCs was reduced upon ANO6 knock-down. Conclusion: Our data identify ANO6 as a Ca2+-activated Cl- channel in mouse DCs, show its activation upon chemokine receptor ligation and establish an important role of ANO6 in chemokine-induced DC migration.


Cellular Physiology and Biochemistry | 2013

Stimulation of platelet death by vancomycin.

Syeda T. Towhid; Eva-Maria Schmidt; Alexander Tolios; Patrick Münzer; Evi Schmid; Oliver Borst; Meinrad Gawaz; Evi Stegmann; Florian Lang

Background/Aims: Side effects of vancomycin, a widely used antibiotic, include thrombocytopenia. The vancomycin-induced thrombocytopenia has been attributed to immune reactions. At least in theory, thrombocytopenia could result in part from the triggering of apoptosis, which results in cell shrinkage and cell membrane scrambling with subsequent phosphatidylserine exposure at the cell surface. The cell membrane scrambling could be initiated by a signaling involving increase of cytosolic Ca2+ activity, ceramide formation, mitochondrial depolarization and/or caspase activation. Vancomycin has indeed been shown to trigger neutrophil apoptosis. An effect of vancomycin on platelet apoptosis has, however, never been tested. The present study thus explored the effect of vancomycin on platelet activation and apoptosis. Methods: Human blood platelets were exposed to vancomycin and forward scatter was utilized to estimate cell volume, annexin V-binding to quantify phosphatidylserine (PS) exposure, Fluo-3 AM fluorescence to estimate cytosolic Ca2+ activity ([Ca2+]i), antibodies to quantify ceramide formation and immunofluorescence to quantify protein abundance of active caspase-3. Results: A 30 minutes exposure to vancomycin (≥1 µg/ ml) decreased cell volume, triggered annexin V-binding, increased [Ca2+]i, activated caspase 3, stimulated ceramide formation, triggered release of thromboxane B2, and upregulated surface expression of CD62P (P-selectin) as well as activated integrin αllbβ3. Annexin V-binding and upregulation of CD62P (P-selectin) and integrin αllbβ3 was significantly blunted by removal of extracellular Ca2+. Annexin V-binding was not significantly blunted by pan-caspase inhibitor zVAD-FMK (1 µM). In conclusion, vancomycin results in platelet activation and suicidal platelet death with increase of [Ca2+]i, caspase-3 activation, cell membrane scrambling and cell shrinkage. Activation and cell membrane scrambling required the presence of Ca2+, but not activation of caspases. Conclusion: Vancomycin exposure leads to platelet activation and apoptosis.


The FASEB Journal | 2012

Enhanced Ca2+ entry and Na+/Ca2+ exchanger activity in dendritic cells from AMP-activated protein kinase-deficient mice

Meerim K. Nurbaeva; Evi Schmid; Kalina Szteyn; Wenting Yang; Benoit Viollet; Ekaterina Shumilina; Florian Lang

In dendritic cells (DCs), chemotactic chemokines, such as CXCL12, rapidly increase cytosolic Ca2+ concentrations ([Ca2+]i) by triggering Ca2+ release from intracellular stores followed by store‐operated Ca2+ (SOC) entry. Increase of [Ca2+]i is blunted and terminated by Ca2+ extrusion, accomplished by K+independent Na+/Ca2+ exchangers (NCXs) and K+‐dependent Na+/Ca2+ exchangers (NCKXs). Increased [Ca2+]i activates energy‐sensing AMP‐activated protein kinase (AMPK), which suppresses proinflammatory responses of DCs and macrophages. The present study explored whether AMPK participates in the regulation of DC [Ca2+]i and migration. DCs were isolated from AMPKα1‐deficient (ampk–/–) mice and, as control, from their wild‐type (ampk+/+) littermates. AMPKα1, Orai1‐2, STIM1‐2, and mitochondrial calcium uniporter protein expression was determined by Western blotting, [Ca2+]i by Fura‐2 fluorescence, SOC entry by inhibition of endosomal Ca2+ ATPase with thapsigargin (1 μM), Na+/Ca2+ exchanger activity from increase of [Ca2+]i, and respective whole‐cell current in patch clamp following removal of extracellular Na+. Migration was quantified utilizing transwell chambers. AMPKα1 protein is expressed in ampk+/+ DCs but not in ampk–/– DCs. CXCL12 (300 ng/ml)‐induced increase of [Ca2+]i, SOC entry, Orai 1 protein abundance, NCX, and NCKX were all significantly higher in ampk–/– DCs than in ampk+/+ DCs. NCX and NCKX currents were similarly increased in ampk–/– DCs. Moreover, CXCL12 (50 ng/ml)‐induced DC migration was enhanced in ampk–/– DCs. AMPK thus inhibits SOC entry, Na+/Ca2+ exchangers, and migration of DCs.—Nurbaeva, M. K., Schmid, E., Szteyn, K., Yang, W., Viollet, B., Shumilina, E., Lang, F. Enhanced Ca2+ entry and Na+/Ca2+ exchanger activity in dendritic cells from AMP‐activated protein kinase‐deficient mice. FASEB J. 26, 3049–3058 (2012). www.fasebj.org


Arteriosclerosis, Thrombosis, and Vascular Biology | 2014

Acid Sphingomyelinase Regulates Platelet Cell Membrane Scrambling, Secretion, and Thrombus Formation

Patrick Münzer; Oliver Borst; Britta Walker; Evi Schmid; Marion A. H. Feijge; Judith M. E. M. Cosemans; Madhumita Chatterjee; Eva-Maria Schmidt; Sebastian Schmidt; Syeda T. Towhid; Christina Leibrock; Margitta Elvers; Martin Schaller; Peter Seizer; Klaus Ferlinz; Andreas E. May; Erich Gulbins; Johan W. M. Heemskerk; Meinrad Gawaz; Florian Lang

Objective— Platelet activation is essential for primary hemostasis and acute thrombotic vascular occlusions. On activation, platelets release their prothrombotic granules and expose phosphatidylserine, thus fostering thrombin generation and thrombus formation. In other cell types, both degranulation and phosphatidylserine exposure are modified by sphingomyelinase-dependent formation of ceramide. The present study thus explored whether acid sphingomyelinase participates in the regulation of platelet secretion, phosphatidylserine exposure, and thrombus formation. Approach and Results— Collagen-related peptide–induced or thrombin-induced ATP release and P-selectin exposure were significantly blunted in platelets from Asm-deficient mice (Smpd1 −/−) when compared with platelets from wild-type mice (Smpd1 +/+ ). Moreover, phosphatidylserine exposure and thrombin generation were significantly less pronounced in Smpd1 −/− platelets than in Smpd1 +/+ platelets. In contrast, platelet integrin &agr;IIb&bgr;3 activation and aggregation, as well as activation-dependent Ca2+ flux, were not significantly different between Smpd1 −/− and Smpd1 +/+ platelets. In vitro thrombus formation at shear rates of 1700 s−1 and in vivo thrombus formation after FeCl3 injury were significantly blunted in Smpd1 −/− mice while bleeding time was unaffected. Asm-deficient platelets showed significantly reduced activation-dependent ceramide formation, whereas exogenous ceramide rescued diminished platelet secretion and thrombus formation caused by Asm deficiency. Treatment of Smpd1 +/+ platelets with bacterial sphingomyelinase (0.01 U/mL) increased, whereas treatment with functional acid sphingomyelinase-inhibitors, amitriptyline or fluoxetine (5 &mgr;mol/L), blunted activation-dependent platelet degranulation, phosphatidylserine exposure, and thrombus formation. Impaired degranulation and thrombus formation of Smpd1 −/− platelets were again overcome by exogenous bacterial sphingomyelinase. Conclusions— Acid sphingomyelinase is a completely novel element in the regulation of platelet plasma membrane properties, secretion, and thrombus formation.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2015

Pivotal Role of Serum- and Glucocorticoid-Inducible Kinase 1 in Vascular Inflammation and Atherogenesis

Oliver Borst; Malte Schaub; Britta Walker; Evi Schmid; Patrick Münzer; Jakob Voelkl; Ioana Alesutan; José M. Rodríguez; Sebastian Vogel; Tanja Schoenberger; Katja Metzger; Dominik Rath; Anja T. Umbach; Dietmar Kuhl; Iris Müller; Peter Seizer; Tobias Geisler; Meinrad Gawaz; Florian Lang

Objective— Atherosclerosis, an inflammatory disease of arterial vessel walls, requires migration and matrix metalloproteinase (MMP)-9–dependent invasion of monocytes/macrophages into the vascular wall. MMP-9 expression is stimulated by transcription factor nuclear factor-&kgr;B, which is regulated by inhibitor &kgr;B (I&kgr;B) and thus I&kgr;B kinase. Regulators of nuclear factor-&kgr;B include serum- and glucocorticoid-inducible kinase 1 (SGK1). The present study explored involvement of SGK1 in vascular inflammation and atherogenesis. Approach and Results— Gene-targeted apolipoprotein E (ApoE)–deficient mice without (apoe −/− sgk1 +/+) or with (apoe −/− sgk1 −/−) additional SGK1 knockout received 16-week cholesterol-rich diet. According to immunohistochemistry atherosclerotic lesions in aorta and carotid artery, vascular CD45+ leukocyte infiltration, Mac-3+ macrophage infiltration, vascular smooth muscle cell content, MMP-2, and MMP-9 positive areas in atherosclerotic tissue were significantly less in apoe −/− sgk1 −/−mice than in apoe −/− sgk1 +/+mice. As determined by Boyden chamber, thioglycollate-induced peritonitis and air pouch model, migration of SGK1-deficient CD11b+F4/80+ macrophages was significantly diminished in vitro and in vivo. Zymographic MMP-2 and MMP-9 production, MMP-9 activity and invasion through matrigel in vitro were significantly less in sgk1 −/− than in sgk1 +/+macrophages and in control plasmid–transfected or inactive K127NSGK1-transfected than in constitutively active S422DSGK1-transfected THP-1 cells. Confocal microscopy revealed reduced macrophage number and macrophage MMP-9 content in plaques of apoe −/− sgk1 −/− mice. In THP-1 cells, MMP-inhibitor GM6001 (25 &mgr;mol/L) abrogated S422DSGK1-induced MMP-9 production and invasion. According to reverse transcription polymerase chain reaction, MMP-9 transcript levels were significantly reduced in sgk1 −/−macrophages and strongly upregulated in S422DSGK1-transfected THP-1 cells compared with control plasmid–transfected or K127NSGK1-transfected THP-1 cells. According to immunoblotting and confocal microscopy, phosphorylation of I&kgr;B kinase and inhibitor &kgr;B and nuclear translocation of p50 were significantly lower in sgk1 −/−macrophages than in sgk1 +/+macrophages and significantly higher in S422DSGK1-transfected THP-1 cells than in control plasmid–transfected or K127NSGK1-transfected THP-1 cells. Treatment of S422DSGK1-transfected THP-1 cells with I&kgr;B kinase-inhibitor BMS-345541 (10 &mgr;mol/L) abolished S422DSGK1-induced increase of MMP-9 transcription and gelatinase activity. Conclusions— SGK1 plays a pivotal role in vascular inflammation during atherogenesis. SGK1 participates in the regulation of monocyte/macrophage migration and MMP-9 transcription via regulation of nuclear factor-&kgr;B.

Collaboration


Dive into the Evi Schmid's collaboration.

Top Co-Authors

Avatar

Florian Lang

Russian Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Oliver Borst

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wenting Yang

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jing Yan

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge