Network


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

Hotspot


Dive into the research topics where Britta Walker is active.

Publication


Featured researches published by Britta Walker.


Journal of Clinical Investigation | 2015

Platelet-derived HMGB1 is a critical mediator of thrombosis

Sebastian Vogel; Rebecca Bodenstein; Qiwei Chen; Susanne Feil; Robert Feil; Johannes Rheinlaender; Tilman E. Schäffer; Erwin Bohn; Julia-Stefanie Frick; Oliver Borst; Patrick Münzer; Britta Walker; Justin Markel; Gábor Csányi; Patrick J. Pagano; Patricia Loughran; Morgan E. Jessup; Simon Watkins; Grant C. Bullock; Jason L. Sperry; Brian S. Zuckerbraun; Timothy R. Billiar; Michael T. Lotze; Meinrad Gawaz; Matthew D. Neal

Thrombosis and inflammation are intricately linked in several major clinical disorders, including disseminated intravascular coagulation and acute ischemic events. The damage-associated molecular pattern molecule high-mobility group box 1 (HMGB1) is upregulated by activated platelets in multiple inflammatory diseases; however, the contribution of platelet-derived HMGB1 in thrombosis remains unexplored. Here, we generated transgenic mice with platelet-specific ablation of HMGB1 and determined that platelet-derived HMGB1 is a critical mediator of thrombosis. Mice lacking HMGB1 in platelets exhibited increased bleeding times as well as reduced thrombus formation, platelet aggregation, inflammation, and organ damage during experimental trauma/hemorrhagic shock. Platelets were the major source of HMGB1 within thrombi. In trauma patients, HMGB1 expression on the surface of circulating platelets was markedly upregulated. Moreover, evaluation of isolated platelets revealed that HMGB1 is critical for regulating platelet activation, granule secretion, adhesion, and spreading. These effects were mediated via TLR4- and MyD88-dependent recruitment of platelet guanylyl cyclase (GC) toward the plasma membrane, followed by MyD88/GC complex formation and activation of the cGMP-dependent protein kinase I (cGKI). Thus, we establish platelet-derived HMGB1 as an important mediator of thrombosis and identify a HMGB1-driven link between MyD88 and GC/cGKI in platelets. Additionally, these findings suggest a potential therapeutic target for patients sustaining trauma and other inflammatory disorders associated with abnormal coagulation.


PLOS ONE | 2014

Blood Platelets in the Progression of Alzheimer's Disease

Nina S. Gowert; Lili Donner; Madhumita Chatterjee; Yvonne S. Eisele; Seyda T. Towhid; Patrick Münzer; Britta Walker; Isabella Ogorek; Oliver Borst; Maria Grandoch; Martin Schaller; Jens W. Fischer; Meinrad Gawaz; Sascha Weggen; Florian Lang; Mathias Jucker; Margitta Elvers

Alzheimer’s disease (AD) is characterized by neurotoxic amyloid-ß plaque formation in brain parenchyma and cerebral blood vessels known as cerebral amyloid angiopathy (CAA). Besides CAA, AD is strongly related to vascular diseases such as stroke and atherosclerosis. Cerebrovascular dysfunction occurs in AD patients leading to alterations in blood flow that might play an important role in AD pathology with neuronal loss and memory deficits. Platelets are the major players in hemostasis and thrombosis, but are also involved in neuroinflammatory diseases like AD. For many years, platelets were accepted as peripheral model to study the pathophysiology of AD because platelets display the enzymatic activities to generate amyloid-ß (Aß) peptides. In addition, platelets are considered to be a biomarker for early diagnosis of AD. Effects of Aß peptides on platelets and the impact of platelets in the progression of AD remained, however, ill-defined. The present study explored the cellular mechanisms triggered by Aß in platelets. Treatment of platelets with Aß led to platelet activation and enhanced generation of reactive oxygen species (ROS) and membrane scrambling, suggesting enhanced platelet apoptosis. More important, platelets modulate soluble Aß into fibrillar structures that were absorbed by apoptotic but not vital platelets. This together with enhanced platelet adhesion under flow ex vivo and in vivo and platelet accumulation at amyloid deposits of cerebral vessels of AD transgenic mice suggested that platelets are major contributors of CAA inducing platelet thrombus formation at vascular amyloid plaques leading to vessel occlusion critical for cerebrovascular events like stroke.


Circulation Research | 2014

Macrophage Migration Inhibitory Factor Limits Activation-Induced Apoptosis of Platelets via CXCR7-Dependent Akt Signaling

Madhumita Chatterjee; Oliver Borst; Britta Walker; Anna Fotinos; Sebastian Vogel; Peter Seizer; Andreas F. Mack; Setareh Alampour-Rajabi; Dominik Rath; Tobias Geisler; Florian Lang; Harald Langer; Juergen Bernhagen; Meinrad Gawaz

Rationale: Macrophage migration inhibitory factor (MIF) is released on platelet activation. Circulating MIF could potentially regulate platelets and thereby platelet-mediated inflammatory and regenerative mechanisms. However, the effect of MIF on platelets is unknown. Objective: The present study evaluated MIF in regulating platelet survival and thrombotic potential. Methods and Results: MIF interacted with CXCR4-CXCR7 on platelets, defining CXCR7 as a hitherto unrecognized receptor for MIF on platelets. MIF internalized CXCR4, but unlike CXCL12 (SDF-1&agr;), it did not phosphorylate Erk1/2 after CXCR4 ligation because of the lack of CD74 and failed in subsequent CXCR7 externalization. MIF did not alter the activation status of platelets. However, MIF rescued platelets from activation and BH3 mimetic ABT-737–induced apoptosis in vitro via CXCR7 and enhanced circulating platelet survival when administered in vivo. The antiapoptotic effect of MIF was absent in Cxcr7−/− murine embryonic cells but pronounced in CXCR7-transfected Madin–Darby canine kidney cells. This prosurvival effect was attributed to the MIF–CXCR7–initiated PI3K-Akt pathway. MIF induced CXCR7-Akt–dependent phosphorylation of BCL-2 antagonist of cell death (BAD) both in vitro and in vivo. Consequentially, MIF failed to rescue Akt−/− platelets from thrombin-induced apoptosis when challenged ex vivo, also in prolonging platelet survival and in inducing BAD phosphorylation among Akt−/− mice in vivo. MIF reduced thrombus formation under arterial flow conditions in vitro and retarded thrombotic occlusion after FeCl3-induced arterial injury in vivo, an effect mediated through CXCR7. Conclusion: MIF interaction with CXCR7 modulates platelet survival and thrombotic potential both in vitro and in vivo and thus could regulate thrombosis and inflammation.


Kidney & Blood Pressure Research | 2012

Enhanced FGF23 Serum Concentrations and Phosphaturia in Gene Targeted Mice Expressing WNK-Resistant Spak

Ganesh Pathare; Michael Föller; Diana Michael; Britta Walker; Michael Hierlmeier; Julia G. Mannheim; Bernd J. Pichler; Florian Lang

Background: The WNK-dependent STE20/SPS1-related proline/alanine-rich kinase (SPAK) regulates the renal thiazide sensitive NaCl cotransporter (NCC) and the renal furosemide sensitive Na+,K+,2Cl- cotransporter (NKCC2) and thus participates in the regulation of renal salt excretion, extracellular fluid volume and blood pressure. Inhibition of NCC leads to anticalciuria. Moreover, NCC is also expressed in osteoblasts where it is implicated in the regulation of bone mineralization. Osteoblasts further influence mineral metabolism by releasing the phosphaturic hormone FGF23. The present study explored, whether SPAK participates in the regulation of calcium-phosphate homeostasis. Methods: FGF23 serum levels and phosphate homeostasis were analyzed in gene targeted mice expressing SPAK resistant to WNK-dependent activation (spaktg/tg) and in mice expressing wild type SPAK (spakwt/wt). Results: Serum FGF23 level was significantly higher, urinary phosphate excretion significantly larger and serum phosphate concentration significantly lower in spaktg/tg mice than in spakwt/wt mice. Urinary calcium excretion was significantly decreased in spaktg/tg mice. Serum levels of calcitriol and PTH were not significantly different between the genotypes. Bone density was significantly increased in spaktg/tg mice compared to spakwt/wt mice. Treatment of spakwt/wt mice with HCT increased FGF23 serum levels, and led to phosphaturia and hypophosphatemia. Conclusions: SPAK is a strong regulator of FGF23 formation, bone mineralization and renal Ca2+ and phosphate excretion.


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.


The FASEB Journal | 2014

1,25( OH)(2) vitamin D-3-dependent inhibition of platelet Ca2+ signaling and thrombus formation in klotho-deficient mice

Oliver Borst; Patrick Münzer; Evi Schmid; Eva Maria Schmidt; Antonella Russo; Britta Walker; Wenting Yang; Christina Leibrock; Kalina Szteyn; Sebastian Schmidt; Margitta Elvers; Caterina Faggio; Ekaterina Shumilina; Makoto Kuro-O; Meinrad Gawaz; Florian Lang

Platelets are activated by increased cytosolic Ca2+ concentration ([Ca2+]i) following store‐operated calcium entry (SOCE) accomplished by calcium‐release‐activated calcium (CRAC) channel moiety Orai1 and its regulator STIM1. In other cells, Ca2+ transport is regulated by 1,25(OH)2 vitamin D3 [1,25(OH)2D3]. 1,25(OH)2D3 formation is inhibited by klotho and excessive in klotho‐deficient mice (kl/kl). The present study explored the effect of klotho deficiency on platelet Ca2+ signaling and activation. Platelets and megakaryocytes isolated from WT and kl/kl‐mice were analyzed by RT‐PCR, Western blotting, confocal microscopy, Fura‐2‐fluorescence, patch clamp, flow cytometry, aggregometry, and flow chamber. STIM1/Orai1 transcript and protein levels, SOCE, agonist‐induced [Ca2+]i increase, activation‐dependent degranulation, integrin αIIbβ3 activation and aggregation, and thrombus formation were significantly blunted in kl/kl platelets (by 27–90%). STIM1/Orai1 transcript and protein levels, as well as CRAC currents, were significantly reduced in kl/kl megakaryocytes (by 38–73%) and 1,25(OH)2D3‐treated WT megakaryocytes. Nuclear NF‐κB subunit p50/p65 abundance was significantly reduced in kl/kl‐megakaryocytes (by 51–76%). Transfection with p50/p65 significantly increased STIM1/Orai1 transcript and protein levels in megakaryocytic MEG‐01 cells (by 46–97%). Low‐vitamin D diet (LVD) of kl/kl mice normalized plasma 1,25(OH)2D3 concentration and function of platelets and megakaryocytes. Klotho deficiency inhibits platelet Ca2+ signaling and activation, an effect at least partially due to 1,25(OH)2D3‐dependent down‐regulation of NF‐κB activity and STIM1/Orai1 expression in megakaryocytes.—Borst, O., Münzer, P., Schmid, E., Schmidt, E.‐M., Russo, A., Walker, B., Yang, W., Leibrock, C., Szteyn, K., Schmidt, S., Elvers, M., Faggio, C., Shumilina, E., Kuro‐o, M., Gawaz, M., Lang, F. 1,25(OH)2 vitamin D3‐dependent inhibition of platelet Ca2+ signaling and thrombus formation in klotho‐deficient mice. FASEB J. 28, 2108–2119 (2014). www.fasebj.org


Cellular Physiology and Biochemistry | 2013

Skepinone-L, a Novel Potent and Highly Selective Inhibitor of p38 MAP Kinase, Effectively Impairs Platelet Activation and Thrombus Formation

Oliver Borst; Britta Walker; Patrick Münzer; Antonella Russo; Evi Schmid; Caterina Faggio; Boris Bigalke; Stefan Laufer; Meinrad Gawaz; Florian Lang

Background/Aims: Platelets are critically important for primary haemostasis and the major players in thrombotic vascular occlusion. Platelets are activated by agonists, such as thrombin and collagen-related peptide as well as second-wave mediators including thromboxane A2 via different intracellular signaling pathways resulting in degranulation, aggregation and thrombus formation. Platelet activation is paralleled by phosphorylation and activation of p38 MAPK. The limited specificity of hitherto known p38 MAPK inhibitors precluded safe conclusions on the precise role of p38 MAPK in the regulation of platelet function. The present study examined the impact of Skepinone-L, a novel and highly selective inhibitor of p38 mitogen-activated protein kinase (p38 MAPK), on platelet activation and thrombus formation. Methods: Experiments were performed in freshly isolated human platelets. Protein phosphorylation was quantified by Western blotting, thromboxane B2 synthesis by enzyme immunoassay, ATP release by ChronoLume luciferin assay, cytosolic Ca2+ concentration by Fura-2 fluorescence-measurements, platelet aggregation by a light transmissions measurement and in vitro thrombus formation by a flow chamber. Results: Skepinone-L (1 μM) virtually abrogated the phosphorylation of platelet p38 MAPK substrate Hsp27 following stimulation with CRP (1 μg/ml), thrombin (5 mU/ml) or thromboxane A2 analogue U-46619 (1 μM). Furthermore, Skepinone-L significantly blunted activation-dependent platelet secretion and aggregation following threshold concentrations of CRP, thrombin and thromboxane A2 analogue U-46619. Skepinone-L did not impair platelet Ca2+ signaling but prevented agonist-induced thromboxane A2 synthesis through abrogation of p38 MAPK-dependent phosphorylation of platelet cytosolic phospholipase A2 (cPLA2). Skepinone-L further markedly blunted thrombus formation under low (500-s) and high (1700-s) arterial shear rates. Conclusions: The present study discloses a powerful inhibiting effect of p38 MAPK-blocker Skepinone-L on platelet activation and thrombus formation.


American Journal of Physiology-cell Physiology | 2014

Dynamic adhesion of eryptotic erythrocytes to immobilized platelets via platelet phosphatidylserine receptors

Britta Walker; Syeda T. Towhid; Evi Schmid; Sascha M. Hoffmann; Majed Abed; Patrick Münzer; Sebastian Vogel; Felix Neis; Sara Y. Brucker; Meinrad Gawaz; Oliver Borst; Florian Lang

Glucose depletion of erythrocytes triggers suicidal erythrocyte death or eryptosis, which leads to cell membrane scrambling with phosphatidylserine exposure at the cell surface. Eryptotic erythrocytes adhere to endothelial cells by a mechanism involving phosphatidylserine at the erythrocyte surface and CXCL16 as well as CD36 at the endothelial cell membrane. Nothing has hitherto been known about an interaction between eryptotic erythrocytes and platelets, the decisive cells in primary hemostasis and major players in thrombotic vascular occlusion. The present study thus explored whether and how glucose-depleted erythrocytes adhere to platelets. To this end, adhesion of phosphatidylserine-exposing erythrocytes to platelets under flow conditions was examined in a flow chamber model at arterial shear rates. Platelets were immobilized on collagen and further stimulated with adenosine diphosphate (ADP, 10 μM) or thrombin (0.1 U/ml). As a result, a 48-h glucose depletion triggered phosphatidylserine translocation to the erythrocyte surface and augmented the adhesion of erythrocytes to immobilized platelets, an effect significantly increased upon platelet stimulation. Adherence of erythrocytes to platelets was blunted by coating of erythrocytic phosphatidylserine with annexin V or by neutralization of platelet phosphatidylserine receptors CXCL16 and CD36 with respective antibodies. In conclusion, glucose-depleted erythrocytes adhere to platelets. The adhesive properties of platelets are augmented by platelet activation. Erythrocyte adhesion to immobilized platelets requires phosphatidylserine at the erythrocyte surface and CXCL16 as well as CD36 expression on platelets. Thus platelet-mediated erythrocyte adhesion may foster thromboocclusive complications in diseases with stimulated phosphatidylserine exposure of erythrocytes.


Kidney & Blood Pressure Research | 2013

PKB/SGK-Resistant GSK-3 Signaling Following Unilateral Ureteral Obstruction

Jakob Voelkl; Sobuj Mia; Adrian Meissner; Mohamed Siyabeldin E. Ahmed; Martina Feger; Bernat Elvira; Britta Walker; Dario R. Alessi; Ioana Alesutan; Florian Lang

Background/Aims: Renal tissue fibrosis contributes to the development of end-stage renal disease. Causes for renal tissue fibrosis include obstructive nephropathy. The development of renal fibrosis following unilateral ureteral obstruction (UUO) is blunted in gene-targeted mice lacking functional serum- and glucocorticoid-inducible kinase SGK1. Similar to Akt isoforms, SGK1 phosphorylates and thus inactivates glycogen synthase kinase GSK-3. The present study explored whether PKB/SGK-dependent phoshorylation of GSK-3α/β impacts on pro-fibrotic signaling following UUO. Methods: UUO was induced in mice carrying a PKB/SGK-resistant GSK-3α/β (gsk-3KI) and corresponding wild-type mice (gsk-3WT). Three days after the obstructive injury, expression of fibrosis markers in kidney tissues was analyzed by quantitative RT-PCR and western blotting. Results: GSK-3α and GSK-3β phosphorylation was absent in both, the non-obstructed and the obstructed kidney tissues from gsk-3KI mice but was increased by UUO in kidney tissues from gsk-3WT mice. Expression of α-smooth muscle actin, type I collagen and type III collagen in the non-obstructed kidney tissues was not significantly different between gsk-3KI mice and gsk-3WT mice but was significantly less increased in the obstructed kidney tissues from gsk-3KI mice than from gsk-3WT mice. After UUO treatment, renal β-catenin protein abundance and renal expression of the β-catenin sensitive genes: c-Myc, Dkk1, Twist and Lef1 were again significantly less increased in kidney tissues from gsk-3KI mice than from gsk-3WT mice. Conclusions: PKB/SGK-dependent phosphorylation of glycogen synthase kinase GSK-3 contributes to the pro-fibrotic signaling leading to renal tissue fibrosis in obstructive nephropathy.

Collaboration


Dive into the Britta Walker's collaboration.

Top Co-Authors

Avatar

Florian Lang

University of Tübingen

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

Evi Schmid

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter Seizer

University of Tübingen

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge