Network


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

Hotspot


Dive into the research topics where Wendy Theelen is active.

Publication


Featured researches published by Wendy Theelen.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2012

Contribution of Platelet CX 3 CR1 to Platelet–Monocyte Complex Formation and Vascular Recruitment During Hyperlipidemia

Otilia Postea; Elena M. Vasina; Sandra Cauwenberghs; Delia Projahn; Elisa A. Liehn; Dirk Lievens; Wendy Theelen; Birgit Kramp; Elena Butoi; Oliver Soehnlein; Johan W. M. Heemskerk; Andreas Ludwig; Christian Weber; Rory R. Koenen

Objective—The chemokine receptor CX3CR1 is an inflammatory mediator in vascular diseases. On platelets, its ligation with fractalkine (CX3CL1) induces platelet activation followed by leukocyte recruitment to activated endothelium. Here, we evaluated the expression and role of platelet-CX3CR1 during hyperlipidemia and vascular injury. Methods and Results—The existence of CX3CR1 on platelets at mRNA and protein level was analyzed by RT-PCR, quantitative (q)PCR, FACS analysis, and Western blot. Elevated CX3CR1 expression was detected on human platelets after activation and, along with increased binding of CX3CL1, platelet CX3CR1 was also involved in the formation of platelet–monocyte complexes. Interestingly, the expression of CX3CR1 was elevated on platelets from hyperlipidemic mice. Accordingly, CX3CL1-binding and the number of circulating platelet–monocyte complexes were increased. In addition, CX3CR1 supported monocyte arrest on inflamed smooth muscle cells in vitro, whereas CX3CR1-deficient platelets showed decreased adhesion to the denuded vessel wall in vivo. Conclusion—Platelets in hyperlipidemic mice display increased CX3CR1-expression and assemble with circulating monocytes. The formation of platelet–monocyte complexes and the detection of platelet-bound CX3CL1 on inflamed smooth muscle cells suggest a significant involvement of the CX3CL1–CX3CR1 axis in platelet accumulation and monocyte recruitment at sites of arterial injury in atherosclerosis.


Arteriosclerosis, Thrombosis, and Vascular Biology | 2014

Deficiency of Endothelial Cxcr4 Reduces Reendothelialization and Enhances Neointimal Hyperplasia After Vascular Injury in Atherosclerosis-Prone Mice

Heidi Noels; Baixue Zhou; Pathricia V. Tilstam; Wendy Theelen; Xiaofeng Li; Lukas Pawig; Corinna Schmitz; Shamima Akhtar; Sakine Simsekyilmaz; Erdenechimeg Shagdarsuren; Andreas Schober; Ralf H. Adams; Jürgen Bernhagen; Elisa A. Liehn; Yvonne Döring; Christian Weber

Objective—The Cxcl12/Cxcr4 chemokine ligand/receptor axis mediates the mobilization of smooth muscle cell progenitors, driving injury-induced neointimal hyperplasia. This study aimed to investigate the role of endothelial Cxcr4 in neointima formation. Approach and Results—&bgr;-Galactosidase staining using bone marrow x kinase (Bmx)-CreERT2 reporter mice and double immunofluorescence revealed an efficient and endothelial-specific deletion of Cxcr4 in Bmx-CreERT2+ compared with Bmx-CreERT2− Cxcr4-floxed apolipoprotein E–deficient (Apoe−/−) mice (referred to as Cxcr4EC-KOApoE−/− and Cxcr4EC-WT ApoE−/−, respectively). Endothelial Cxcr4 deficiency significantly increased wire injury–induced neointima formation in carotid arteries from Cxcr4EC-KOApoE−/− mice. The lesions displayed a higher number of macrophages, whereas the smooth muscle cell and collagen content were reduced. This was associated with a significant reduction in reendothelialization and endothelial cell proliferation in injured Cxcr4EC-KOApoE−/− carotids compared with Cxcr4EC-WTApoE−/− controls. Furthermore, stimulation of human aortic endothelial cells with chemokine (C-X-C motif) ligand 12 (CXCL12) significantly enhanced their wound-healing capacity in an in vitro scratch assay, an effect that could be reversed with the CXCR4 antagonist AMD3100. Also, flow cytometric analysis showed a reduced mobilization of Sca1+Flk1+Cd31+ and of Lin−Sca1+ progenitors in Cxcr4EC-KO ApoE−/− mice after vascular injury, although Cxcr4 surface expression was unaltered. No differences could be detected in plasma concentrations of Cxcl12, vascular endothelial growth factor, sphingosine 1-phosphate, or Flt3 (fms-related tyrosine kinase 3) ligand, all cytokines with an established role in progenitor cell mobilization. Nonetheless, double immunofluorescence revealed a significant reduction in local endothelial Cxcl12 staining in injured carotids from Cxcr4EC-KOApoE−/− mice. Conclusions—Endothelial Cxcr4 is crucial for efficient reendothelialization after vascular injury through endothelial wound healing and proliferation, and through the mobilization of Sca1+Flk1+Cd31+ cells, often referred to as circulating endothelial progenitor cells.


Circulation Research | 2014

Deficiency of the Sialyltransferase St3Gal4 Reduces Ccl5-Mediated Myeloid Cell Recruitment and Arrest Short Communication

Yvonne Döring; Heidi Noels; Manuela Mandl; Birgit Kramp; Carlos Neideck; Dirk Lievens; Maik Drechsler; Remco T.A. Megens; Pathricia V. Tilstam; Marcella Langer; Helene Hartwig; Wendy Theelen; Jamey D. Marth; Markus Sperandio; Oliver Soehnlein; Christian Weber

Rationale: Sialylation by &agr;2,3-sialyltransferases has been shown to be a crucial glycosylation step in the generation of functional selectin ligands. Recent evidence suggests that sialylation also affects the binding of chemokines to their corresponding receptor. Objective: Because the chemokine receptors for Ccl5 and Ccl2 are important in atherogenic recruitment of neutrophils and monocytes, we here investigated the role of &agr;2,3-sialyltransferase IV (ST3Gal-IV) in Ccl5- and Ccl2-mediated myeloid cell arrest and further studied its relevance in a mouse model of atherosclerosis. Methods and Results: St3Gal4-deficient myeloid cells showed a reduced binding of Ccl5 and an impaired Ccl5-triggered integrin activation. Correspondingly, Ccl5-induced arrest on tumor necrosis factor-&agr;–stimulated endothelium was almost completely abrogated, as observed in flow chamber adhesion assays and during ex vivo perfusion or intravital microscopy of carotid arteries. Moreover, Ccl5-triggered neutrophil and monocyte extravasation into the peritoneal cavity was severely reduced in St3Gal4−/− mice. In contrast, St3Gal4 deficiency did not significantly affect Ccl2 binding and only marginally decreased Ccl2-induced flow arrest of myeloid cells. In agreement with the crucial role of leukocyte accumulation in atherogenesis, and the importance of Ccl5 chemokine receptors mediating myeloid cell recruitment to atherosclerotic vessels, St3Gal4 deficiency drastically reduced the size, stage, and inflammatory cell content of atherosclerotic lesions in Apoe−/− mice on high-fat diet. Conclusions: In summary, these findings identify ST3Gal-IV as a promising target to reduce inflammatory leukocyte recruitment and arrest.


Circulation Research | 2014

Deficiency of the Sialyltransferase St3Gal4 Reduces Ccl5-Mediated Myeloid Cell Recruitment and Arrest

Yvonne Döring; Heidi Noels; Manuela Mandl; Birgit Kramp; Carlos Neideck; Dirk Lievens; Maik Drechsler; Remco T.A. Megens; Pathricia V. Tilstam; Marcella Langer; Helene Hartwig; Wendy Theelen; Jamey D. Marth; Markus Sperandio; Oliver Soehnlein; Christian Weber

Rationale: Sialylation by &agr;2,3-sialyltransferases has been shown to be a crucial glycosylation step in the generation of functional selectin ligands. Recent evidence suggests that sialylation also affects the binding of chemokines to their corresponding receptor. Objective: Because the chemokine receptors for Ccl5 and Ccl2 are important in atherogenic recruitment of neutrophils and monocytes, we here investigated the role of &agr;2,3-sialyltransferase IV (ST3Gal-IV) in Ccl5- and Ccl2-mediated myeloid cell arrest and further studied its relevance in a mouse model of atherosclerosis. Methods and Results: St3Gal4-deficient myeloid cells showed a reduced binding of Ccl5 and an impaired Ccl5-triggered integrin activation. Correspondingly, Ccl5-induced arrest on tumor necrosis factor-&agr;–stimulated endothelium was almost completely abrogated, as observed in flow chamber adhesion assays and during ex vivo perfusion or intravital microscopy of carotid arteries. Moreover, Ccl5-triggered neutrophil and monocyte extravasation into the peritoneal cavity was severely reduced in St3Gal4−/− mice. In contrast, St3Gal4 deficiency did not significantly affect Ccl2 binding and only marginally decreased Ccl2-induced flow arrest of myeloid cells. In agreement with the crucial role of leukocyte accumulation in atherogenesis, and the importance of Ccl5 chemokine receptors mediating myeloid cell recruitment to atherosclerotic vessels, St3Gal4 deficiency drastically reduced the size, stage, and inflammatory cell content of atherosclerotic lesions in Apoe−/− mice on high-fat diet. Conclusions: In summary, these findings identify ST3Gal-IV as a promising target to reduce inflammatory leukocyte recruitment and arrest.


PLOS ONE | 2014

Bone Marrow-Specific Knock-In of a Non-Activatable Ikkα Kinase Mutant Influences Haematopoiesis but Not Atherosclerosis in Apoe-Deficient Mice

Pathricia V. Tilstam; Marion J. J. Gijbels; Mohamed Habbeddine; Céline Cudejko; Yaw Asare; Wendy Theelen; Baixue Zhou; Yvonne Döring; Maik Drechsler; Lukas Pawig; Sakine Simsekyilmaz; Rory R. Koenen; Menno P.J. de Winther; Toby Lawrence; Jürgen Bernhagen; Alma Zernecke; Christian Weber; Heidi Noels

Background The Ikkα kinase, a subunit of the NF-κB-activating IKK complex, has emerged as an important regulator of inflammatory gene expression. However, the role of Ikkα-mediated phosphorylation in haematopoiesis and atherogenesis remains unexplored. In this study, we investigated the effect of a bone marrow (BM)-specific activation-resistant Ikkα mutant knock-in on haematopoiesis and atherosclerosis in mice. Methods and Results Apolipoprotein E (Apoe)-deficient mice were transplanted with BM carrying an activation-resistant Ikkα gene (IkkαAA/AAApoe−/−) or with Ikkα+/+Apoe−/− BM as control and were fed a high-cholesterol diet for 8 or 13 weeks. Interestingly, haematopoietic profiling by flow cytometry revealed a significant decrease in B-cells, regulatory T-cells and effector memory T-cells in IkkαAA/AAApoe−/− BM-chimeras, whereas the naive T-cell population was increased. Surprisingly, no differences were observed in the size, stage or cellular composition of atherosclerotic lesions in the aorta and aortic root of IkkαAA/AAApoe−/− vs Ikkα+/+Apoe−/− BM-transplanted mice, as shown by histological and immunofluorescent stainings. Necrotic core sizes, apoptosis, and intracellular lipid deposits in aortic root lesions were unaltered. In vitro, BM-derived macrophages from IkkαAA/AAApoe−/− vs Ikkα+/+Apoe−/− mice did not show significant differences in the uptake of oxidized low-density lipoproteins (oxLDL), and, with the exception of Il-12, the secretion of inflammatory proteins in conditions of Tnf-α or oxLDL stimulation was not significantly altered. Furthermore, serum levels of inflammatory proteins as measured with a cytokine bead array were comparable. Conclusion Our data reveal an important and previously unrecognized role of haematopoietic Ikkα kinase activation in the homeostasis of B-cells and regulatory T-cells. However, transplantation of IkkαAA mutant BM did not affect atherosclerosis in Apoe−/− mice. This suggests that the diverse functions of Ikkα in haematopoietic cells may counterbalance each other or may not be strong enough to influence atherogenesis, and reveals that targeting haematopoietic Ikkα kinase activity alone does not represent a therapeutic approach.


PLOS ONE | 2016

Targeting In-Stent-Stenosis with RGD- and CXCL1-Coated Mini-Stents in Mice

Sakine Simsekyilmaz; Elisa A. Liehn; Stefan Weinandy; Fabian Schreiber; Remco T.A. Megens; Wendy Theelen; Ralf Smeets; Stefan Jockenhövel; Thomas Gries; Martin Möller; Doris Klee; Christian Weber; Alma Zernecke

Atherosclerotic lesions that critically narrow the artery can necessitate an angioplasty and stent implantation. Long-term therapeutic effects, however, are limited by excessive arterial remodeling. We here employed a miniaturized nitinol-stent coated with star-shaped polyethylenglycole (star-PEG), and evaluated its bio-functionalization with RGD and CXCL1 for improving in-stent stenosis after implantation into carotid arteries of mice. Nitinol foils or stents (bare metal) were coated with star-PEG, and bio-functionalized with RGD, or RGD/CXCL1. Cell adhesion to star-PEG-coated nitinol foils was unaltered or reduced, whereas bio-functionalization with RGD but foremost RGD/CXCL1 increased adhesion of early angiogenic outgrowth cells (EOCs) and endothelial cells but not smooth muscle cells when compared with bare metal foils. Stimulation of cells with RGD/CXCL1 furthermore increased the proliferation of EOCs. In vivo, bio-functionalization with RGD/CXCL1 significantly reduced neointima formation and thrombus formation, and increased re-endothelialization in apoE-/- carotid arteries compared with bare-metal nitinol stents, star-PEG-coated stents, and stents bio-functionalized with RGD only. Bio-functionalization of star-PEG-coated nitinol-stents with RGD/CXCL1 reduced in-stent neointima formation. By supporting the adhesion and proliferation of endothelial progenitor cells, RGD/CXCL1 coating of stents may help to accelerate endothelial repair after stent implantation, and thus may harbor the potential to limit the complication of in-stent restenosis in clinical approaches.


Circulation | 2017

Vascular CXCR4 Limits Atherosclerosis by Maintaining Arterial Integrity: Evidence from Mouse and Human Studies.

Yvonne Döring; Heidi Noels; Emiel P. C. van der Vorst; Carlos Neideck; Virginia Egea; Maik Drechsler; Manuela Mandl; Lukas Pawig; Yvonne Jansen; Katrin Schröder; Kiril Bidzhekov; Remco T.A. Megens; Wendy Theelen; Barbara Mara Klinkhammer; Peter Boor; Leon J. Schurgers; Rick van Gorp; Christian Ries; Pascal Kusters; Allard C. van der Wal; Tilman M. Hackeng; Gabor Gäbel; Ralf P. Brandes; Oliver Soehnlein; Esther Lutgens; Dietmar Vestweber; Daniel Teupser; Lesca M. Holdt; Daniel J. Rader; Danish Saleheen

Background: The CXCL12/CXCR4 chemokine ligand/receptor axis controls (progenitor) cell homeostasis and trafficking. So far, an atheroprotective role of CXCL12/CXCR4 has only been implied through pharmacological intervention, in particular, because the somatic deletion of the CXCR4 gene in mice is embryonically lethal. Moreover, cell-specific effects of CXCR4 in the arterial wall and underlying mechanisms remain elusive, prompting us to investigate the relevance of CXCR4 in vascular cell types for atheroprotection. Methods: We examined the role of vascular CXCR4 in atherosclerosis and plaque composition by inducing an endothelial cell (BmxCreERT2-driven)–specific or smooth muscle cell (SMC, SmmhcCreERT2- or TaglnCre-driven)–specific deficiency of CXCR4 in an apolipoprotein E–deficient mouse model. To identify underlying mechanisms for effects of CXCR4, we studied endothelial permeability, intravital leukocyte adhesion, involvement of the Akt/WNT/&bgr;-catenin signaling pathway and relevant phosphatases in VE-cadherin expression and function, vascular tone in aortic rings, cholesterol efflux from macrophages, and expression of SMC phenotypic markers. Finally, we analyzed associations of common genetic variants at the CXCR4 locus with the risk for coronary heart disease, along with CXCR4 transcript expression in human atherosclerotic plaques. Results: The cell-specific deletion of CXCR4 in arterial endothelial cells (n=12–15) or SMCs (n=13–24) markedly increased atherosclerotic lesion formation in hyperlipidemic mice. Endothelial barrier function was promoted by CXCL12/CXCR4, which triggered Akt/WNT/&bgr;-catenin signaling to drive VE-cadherin expression and stabilized junctional VE-cadherin complexes through associated phosphatases. Conversely, endothelial CXCR4 deficiency caused arterial leakage and inflammatory leukocyte recruitment during atherogenesis. In arterial SMCs, CXCR4 sustained normal vascular reactivity and contractile responses, whereas CXCR4 deficiency favored a synthetic phenotype, the occurrence of macrophage-like SMCs in the lesions, and impaired cholesterol efflux. Regression analyses in humans (n=259 796) identified the C-allele at rs2322864 within the CXCR4 locus to be associated with increased risk for coronary heart disease. In line, C/C risk genotype carriers showed reduced CXCR4 expression in carotid artery plaques (n=188), which was furthermore associated with symptomatic disease. Conclusions: Our data clearly establish that vascular CXCR4 limits atherosclerosis by maintaining arterial integrity, preserving endothelial barrier function, and a normal contractile SMC phenotype. Enhancing these beneficial functions of arterial CXCR4 by selective modulators might open novel therapeutic options in atherosclerosis.


Scientific Reports | 2018

Reduced post-operative DPP4 activity associated with worse patient outcome after cardiac surgery

Heidi Noels; Wendy Theelen; Marieke Sternkopf; Vera Jankowski; Julia Moellmann; Sandra Kraemer; Michael Lehrke; Nikolaus Marx; Lukas Martin; Gernot Marx; Joachim Jankowski; Andreas Goetzenich; Christian Stoppe

Cardiac surgery with cardiopulmonary bypass (CPB) triggers myocardial ischemia/reperfusion injury contributing to organ dysfunction. Preclinical studies revealed that dipeptidyl peptidase (DPP4) inhibition is protective during myocardial infarction. Here, we assessed for the first time the relation of peri-operative DPP4-activity in serum of 46 patients undergoing cardiac surgery with patients’ post-operative organ dysfunction during intensive care unit (ICU) stay. Whereas a prior myocardial infarction significantly reduced pre-operative DDP4-activity, patients with preserved left ventricular function showed an intra-operative decrease of DPP4-activity. The latter correlated with aortic cross clamping time, indicative for the duration of surgery-induced myocardial ischemia. As underlying mechanism, mass-spectrometry revealed increased DPP4 oxidation by cardiac surgery, with DPP4 oxidation reducing DPP4-activity in vitro. Further, post-operative DPP4-activity was negatively correlated with the extent of post-operative organ injury as measured by SAPS II and SOFA scoring, circulating levels of creatinine and lactate, as well as patients’ stay on the ICU. In conclusion, cardiac surgery reduces DPP4-activity through oxidation, with low post-operative DPP4-activity being associated with organ dysfunction and worse outcome of patients during the post-operative ICU stay. This likely reflects the severity of myocardial ischemia/reperfusion injury and may suggest potential beneficial effects of anti-oxidative treatments during cardiac surgery.


PLOS ONE | 2018

Prolyl carboxypeptidase activity in the circulation and its correlation with body weight and adipose tissue in lean and obese subjects

Kaat Kehoe; Heidi Noels; Wendy Theelen; Emilie De Hert; Shenguan Xu; An Verrijken; Thierry Arnould; Erik Fransen; Nina Hermans; Anne-Marie Lambeir; Per Venge; Luc Van Gaal; Ingrid De Meester

Background Prolyl carboxypeptidase (PRCP) is involved in the regulation of body weight, likely by hydrolysing alpha-melanocyte-stimulating hormone and apelin in the hypothalamus and in the periphery. A link between PRCP protein concentrations in plasma and metabolic disorders has been reported. In this study, we investigated the distribution of circulating PRCP activity and assessed its relation with body weight and adipose tissue in obese patients and patients who significantly lost weight. Methods PRCP activity was measured using reversed-phase high-performance liquid chromatography in different isolated blood fractions and primary human cells to investigate the distribution of circulating PRCP. PRCP activity was measured in serum of individuals (n = 75) categorized based on their body mass index (BMI < 25.0; 25.0–29.9; 30.0–39.9; ≥ 40.0 kg/m2) and the diagnosis of metabolic syndrome. Differences in serum PRCP activity were determined before and six months after weight loss, either by diet (n = 45) or by bariatric surgery (n = 24). Potential correlations between serum PRCP activity and several metabolic and biochemical parameters were assessed. Additionally, plasma PRCP concentrations were quantified using a sensitive ELISA in the bariatric surgery group. Results White blood cells and plasma contributed the most to circulating PRCP activity. Serum PRCP activity in lean subjects was 0.83 ± 0.04 U/L and increased significantly with a rising BMI (p<0.001) and decreased upon weight loss (diet, p<0.05; bariatric surgery, p<0.001). The serum PRCP activity alteration reflected body weight changes and was found to be positively correlated with several metabolic parameters, including: total, abdominal and visceral adipose tissue. Plasma PRCP concentration was found to be significantly correlated to serum PRCP activity (0.865; p<0.001). Additionally, a significant decrease (p<0.001) in plasma PRCP protein concentration (mean ± SD) before (18.2 ± 3.7 ng/mL) and 6 months after bariatric surgery (15.7 ± 2.7 ng/mL) was found. Conclusion Our novel findings demonstrate that white blood cells and plasma contributed the most to circulating PRCP activity. Additionally, we have shown that there were significant correlations between serum PRCP activity and various metabolic parameters, and that plasma PRCP concentration was significantly correlated to serum PRCP activity. These novel findings on PRCP activity in serum support further investigation of its in vivo role and involvement in several metabolic diseases.


Circulation | 2017

Vascular CXCR4 Limits Atherosclerosis by Maintaining Arterial IntegrityClinical Perspective: Evidence From Mouse and Human Studies

Yvonne Döring; Heidi Noels; Emiel P. C. van der Vorst; Carlos Neideck; Virginia Egea; Maik Drechsler; Manuela Mandl; Lukas Pawig; Yvonne Jansen; Katrin Schröder; Kiril Bidzhekov; Remco T.A. Megens; Wendy Theelen; Barbara Mara Klinkhammer; Peter Boor; Leon J. Schurgers; Rick van Gorp; Christian Ries; Pascal Kusters; Allard C. van der Wal; Tilman M. Hackeng; Gabor Gäbel; Ralf P. Brandes; Oliver Soehnlein; Esther Lutgens; Dietmar Vestweber; Daniel Teupser; Lesca M. Holdt; Daniel J. Rader; Danish Saleheen

Background: The CXCL12/CXCR4 chemokine ligand/receptor axis controls (progenitor) cell homeostasis and trafficking. So far, an atheroprotective role of CXCL12/CXCR4 has only been implied through pharmacological intervention, in particular, because the somatic deletion of the CXCR4 gene in mice is embryonically lethal. Moreover, cell-specific effects of CXCR4 in the arterial wall and underlying mechanisms remain elusive, prompting us to investigate the relevance of CXCR4 in vascular cell types for atheroprotection. Methods: We examined the role of vascular CXCR4 in atherosclerosis and plaque composition by inducing an endothelial cell (BmxCreERT2-driven)–specific or smooth muscle cell (SMC, SmmhcCreERT2- or TaglnCre-driven)–specific deficiency of CXCR4 in an apolipoprotein E–deficient mouse model. To identify underlying mechanisms for effects of CXCR4, we studied endothelial permeability, intravital leukocyte adhesion, involvement of the Akt/WNT/&bgr;-catenin signaling pathway and relevant phosphatases in VE-cadherin expression and function, vascular tone in aortic rings, cholesterol efflux from macrophages, and expression of SMC phenotypic markers. Finally, we analyzed associations of common genetic variants at the CXCR4 locus with the risk for coronary heart disease, along with CXCR4 transcript expression in human atherosclerotic plaques. Results: The cell-specific deletion of CXCR4 in arterial endothelial cells (n=12–15) or SMCs (n=13–24) markedly increased atherosclerotic lesion formation in hyperlipidemic mice. Endothelial barrier function was promoted by CXCL12/CXCR4, which triggered Akt/WNT/&bgr;-catenin signaling to drive VE-cadherin expression and stabilized junctional VE-cadherin complexes through associated phosphatases. Conversely, endothelial CXCR4 deficiency caused arterial leakage and inflammatory leukocyte recruitment during atherogenesis. In arterial SMCs, CXCR4 sustained normal vascular reactivity and contractile responses, whereas CXCR4 deficiency favored a synthetic phenotype, the occurrence of macrophage-like SMCs in the lesions, and impaired cholesterol efflux. Regression analyses in humans (n=259 796) identified the C-allele at rs2322864 within the CXCR4 locus to be associated with increased risk for coronary heart disease. In line, C/C risk genotype carriers showed reduced CXCR4 expression in carotid artery plaques (n=188), which was furthermore associated with symptomatic disease. Conclusions: Our data clearly establish that vascular CXCR4 limits atherosclerosis by maintaining arterial integrity, preserving endothelial barrier function, and a normal contractile SMC phenotype. Enhancing these beneficial functions of arterial CXCR4 by selective modulators might open novel therapeutic options in atherosclerosis.

Collaboration


Dive into the Wendy Theelen's collaboration.

Top Co-Authors

Avatar

Heidi Noels

RWTH Aachen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lukas Pawig

RWTH Aachen University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jamey D. Marth

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Katrin Schröder

Goethe University Frankfurt

View shared research outputs
Top Co-Authors

Avatar

Peter Boor

RWTH Aachen University

View shared research outputs
Top Co-Authors

Avatar

Ralf P. Brandes

Goethe University Frankfurt

View shared research outputs
Researchain Logo
Decentralizing Knowledge