Potential cell-specific functions of CXCR4 in atherosclerosis

Similar documents
DECLARATION OF CONFLICT OF INTEREST. No disclosures

Role of Inflammation in Pulmonary Hypertension

Supplementary Materials for

Cytokines, adhesion molecules and apoptosis markers. A comprehensive product line for human and veterinary ELISAs

DECLARATION OF CONFLICT OF INTEREST. No conflicts of interest

Role of Inflammatory and Progenitor Cells in Pulmonary Vascular Remodeling: Potential Role for Targeted Therapies. Traditional Hypothesis Stress

Adipose Tissue as an Endocrine Organ. Abdel Moniem Ibrahim, MD Professor of Physiology Cairo University

Endothelial Injury and Repair as a Working Paradigm

Microparticles- Signaling in Atherothrombosis Agneta Siegbahn, MD, PhD, FESC Professor in Coagulation Science

Crosstalk between Adiponectin and IGF-IR in breast cancer. Prof. Young Jin Suh Department of Surgery The Catholic University of Korea

Cardiovascular Division, Brigham and Women s Hospital, Harvard Medical School

Leptin deficiency suppresses progression of atherosclerosis in apoe-deficient mice

Role of apolipoprotein B-containing lipoproteins in the development of atherosclerosis Jan Borén MD, PhD

Chapter 3, Part A (Pages 37-45): Leukocyte Migration into Tissues

ONLINE SUPPLEMENT MATERIAL. CD70 limits atherosclerosis and promotes macrophage function.

Paracrine Mechanisms in Adult Stem Cell Signaling and Therapy

The recruitment of leukocytes and plasma proteins from the blood to sites of infection and tissue injury is called inflammation

Basic Mechanisms of Atherosclerosis and Plaque Rupture: Clinical Implications

Subject Index. Bcl-2, apoptosis regulation Bone marrow, polymorphonuclear neutrophil release 24, 26

Roles of Flow Mechanics in Vascular Cell Biology in Health and Disease

Atherosclerosis is the primary cause of mortality and

Question 1. Kupffer cells, microglial cells and osteoclasts are all examples of what type of immune system cell?

Journal Club Semmler Lorenz

Arteriosclerosis & Atherosclerosis

Endotoxin Induces Toll-Like Receptor 4 Expression in Vascular Cells: A Novel Mechanism Involved in Vascular Inflammation

Dyslipidemia Endothelial dysfunction Free radicals Immunologic

Summary and concluding remarks

ROLE OF INFLAMMATION IN HYPERTENSION. Dr Barasa FA Physician Cardiologist Eldoret

Basis of Immunology and

The Study of Endothelial Function in CKD and ESRD

Lymphoid architecture & Leukocyte recirculation. Thursday Jan 26th, 2017

Novel function of NADPH oxidase in atherosclerosis. Yun Soo Bae Department of Life Science Ewha Womans University

Tissue renewal and Repair. Nisamanee Charoenchon, PhD Department of Pathobiology, Faculty of Science

Child born in year /3 will die before parents in US (diabetes)

Chapter 2 (pages 22 33): Cells and Tissues of the Immune System. Prepared by Kristen Dazy, MD, Scripps Clinic Medical Group

Pathology of Cardiovascular Interventions. Body and Disease 2011

ESC Congress August 2011 Paris France. VEGFs and the angiogenic paradox in diabetic patients

Tissue repair. (3&4 of 4)

Airway Inflammation in Asthma Chih-Yung Chiu 1,2, Kin-Sun Wong 2 1 Department of Pediatrics, Chang Gung Memorial Hospital, Keelung, Taiwan.

Cytokines modulate the functional activities of individual cells and tissues both under normal and pathologic conditions Interleukins,

Invited Review. Vascular smooth muscle cell proliferation in the pathogenesis of atherosclerotic cardiovascular diseases

Effects of the angiotensin II type-1 receptor antagonist telmisartan on endothelial activation induced by advanced glycation endproducts

Endothelial PGC 1 - α 1 mediates vascular dysfunction in diabetes

renoprotection therapy goals 208, 209

ROMANIAN ACADEMY INSTITUTE OF CELLULAR BIOLOGY AND PATHOLOGY NICOLAE SIMIONESCU. PhD THESIS Summary

1) Mononuclear phagocytes : 2) Regarding acute inflammation : 3) The epithelioid cells of follicular granulomas are :

ATHEROSCLEROSIS زيد ثامر جابر. Zaid. Th. Jaber

Pathology of Coronary Artery Disease

The Angiopoietin Axis in Cancer

Classification of Endothelial Dysfunction. Stefano Taddei Department of Internal Medicine University of Pisa, Italy

Introduction: 年 Fas signal-mediated apoptosis. PI3K/Akt

CD34+ Cells: A Comparison of Stem and Progenitor Cells in Cord Blood, Peripheral Blood, and the Bone Marrow

Adaptive immune responses: T cell-mediated immunity

Mesenchymal Stem Cells to Repair Vascular Damage after Chemotherapy: Past, Present and Future

BL-8040: BEST-IN-CLASS CXCR4 ANTAGONIST FOR TREATMENT OF ONCOLOGICAL MALIGNANCIES. Overview and Mechanism of Action Dr.

Chronic variable stress activates hematopoietic stem cells

Oncolytic Virotherapy: Targeting Cancer Stem Cells

Basic immunology. Lecture 9. Innate immunity: inflammation, leukocyte migration. Péter Engelmann

Cytokines. Luděk Šefc. Cytokines Protein regulators of cellular communication. Cytokines x hormones

Targeting tumour associated macrophages in anti-cancer therapies. Annamaria Gal Seminar Series on Drug Discovery Budapest 5 January 2018

Endogenous TNFα orchestrates the trafficking of neutrophils into and within lymphatic vessels during acute inflammation

Innovation Interlude: Molecular Imaging in Cardiology. Celestial Doppelgangers and Relativity

removed replaced inflammation scar tissue

The aorta is an integral part of the cardiovascular system and should not be considered as just a conduit for blood supply from the heart to the

High density lipoprotein metabolism

CYTOKINE RECEPTORS AND SIGNAL TRANSDUCTION

Effector T Cells and

Vein graft neointimal hyperplasia is exacerbated by CXCR4 signaling in vein graft-extrinsic cells

Future Applications of Contrast Echocardiography

Adipokines at the crossroad between obesity and cardiovascular disease

Pathophysiology of Lipid Disorders

Curriculum Vitae. Research Officer in Baker IDI Heart & Diabetes Institute from Aug

Chapter 7 Conclusions

Hematopoiesis. BHS Liège 27/1/2012. Dr Sonet Anne UCL Mont-Godinne

Metabolism and Atherogenic Properties of LDL

1Why lipids cannot be transported in blood alone? 2How we transport Fatty acids and steroid hormones?

The New Gold Standard for Lipoprotein Analysis. Advanced Testing for Cardiovascular Risk

Potential mechanisms for the resolution of atherosclerosis by conjugated linoleic acid (CLA)

Adenosine stimulates the recruitment of endothelial progenitor cells to the ischemic heart

Newly Recognized Components of the Innate Immune System

Follicular Lymphoma. ced3 APOPTOSIS. *In the nematode Caenorhabditis elegans 131 of the organism's 1031 cells die during development.

Prepared by Cyrus H. Nozad, MD, University of Tennessee and John Seyerle, MD, Ohio State University

LIPOPROTEIN-ASSOCIATED PHOSPHOLIPASE A 2 : EFFECTS OF LOW DENSITY LIPOPROTEIN APHERESIS

Hematopoiesis. Hematopoiesis. Hematopoiesis

Stewart et al. CD36 ligands promote sterile inflammation through assembly of a TLR 4 and 6 heterodimer

Natural Killer T cells and their functions in atherosclerosis and obesity: a therapeutic perspective for cardiovascular diseases

Potential Role of Sphingosine 1-Phosphate in the. Pathogenesis of Rheumatoid Arthritis

The 10 th International & 15 th National Congress on Quality Improvement in Clinical Laboratories

Acute Kidney Injury: Basic Research Interactive Workshop

EARLY INFLAMMATORY RESPONSES TO VASCULAR DEVICES

Bases for Immunotherapy in Multiple Myeloma

INCREASED SERUM HIGH-DENSITY LIPOPROTEIN CHOLESTEROL LEVEL IS ASSOCIATED WITH A REDUCTION IN PERIPHERAL MONOCYTE COUNT

Potential Atheroprotective Effects of Ixmyelocel-T Cellular Therapy. Kelly J. Ledford, Nikki Murphy, Frank Zeigler, Ronnda L.

Dengue pathogenesis and vaccine design

Angiostasis and Angiogenesis Regulated by Angiopoietin1-Tie2 Receptor System

Biology of Immune Aging

Role of Alpha-lipoic acid(ala) and statin in vascular smooth muscle cell proliferation

4. TEXTBOOK: ABUL K. ABBAS. ANDREW H. LICHTMAN. CELLULAR AND MOLECULAR IMMUNOLOGY. 5 TH EDITION. Chapter 2. pg

Tissue stroma as a regulator of leukocyte recruitment in inflammation

Blood Vessel Mechanics

Transcription:

Review 97 Potential cell-specific functions of CXCR4 in atherosclerosis Christian Weber 1 3 ; Yvonne Döring 1 ; Heidi Noels 4 1 Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany; 2 German Centre for Cardiovascular Research, partner site Munich Heart Alliance, Germany; 3 Cardiovascular Research Institute Maastricht, University of Maastricht, the Netherlands; 4 Institute for Molecular Cardiovascular Research, RWTH Aachen University, Aachen, Germany Keywords CXCL12, CXCR4, CXCR7 Summary The chemokine CXCL12 and its receptor CXCR4 form an important axis contributing to cellular functions in homeostasis and disease. In addition, the atypical CXCL12 receptor CXCR7 may shape the availability and function of CXCL12. Further to their role through progenitor cell mobilization, CXCL12 and CXCR4 may affect native atherogenesis by modifying atherosclerosis-relevant cellular functions. This short review intends to provide a concise summary of current knowledge with regards to cell-specific functions of CXCL12 and its receptors CXCR4 and CXCR7 with potential implications for the initiation and progression of atherosclerosis. Correspondence to: Prof. Dr. med. Christian Weber Institute for Cardiovascular Prevention (IPEK) Ludwig-Maximilians-University Munich Pettenkoferstraße 9, 80336 Munich, Germany E-mail: chweber@med.lmu.de Schlüsselwörter CXCL12, CXCR4, CXCR7 Zusammenfassung Der Chemokinrezeptor CXCR4 and sein Ligand CXCL12 bilden eine wichtige Achse in der Regulation von Zellfunktionen bei normaler Homöostase und bei Erkrankungen. Zusätzlich kann der atypische CXCL12 Rezeptor CXCR7 die Verfügbarkeit und Funktion von CXCL12 modulieren. Neben ihrer Rolle in der Mobilisierung von Stamm- und Vorläuferzellen, können CXCR4 und CXCL12 auch die Entwicklung der Atherosklerose über verschiedene Zellfunktionen beeinflussen. Dieser kurze Übersichtsartikel fasst das gegenwärtige Wissen zu den zellspezifischen Funktionen von CXCL12 und den Rezeptoren CXCR4 und CXCR7 mit möglichen Implikationen für die Entstehung und Progression der Atherosklerose zusammen Potenziell zellspezifische Funktionen des Rezeptors CXCR4 in der Atheroklerose Hämostaseologie 2016; 36: 97 102 http://dx.doi.org/10.5482/hamo-14-10-0054 received: October 19, 2014 accepted in revised form: November 27, 2014 epub ahead of print: January 14, 2015 Chemokines are a structurally related family of chemotactic cytokines that are classified in subgroups based on the position of the N-terminal cysteine residues (CC, CXC, CX3C, C). Expressed by not only activated endothelial cells and smooth muscle cells but also emigrated leukocytes, chemokines primarily direct leukocytes to sites of inflammation. Recent evidence has emerged to demonstrate that chemokines also exert functions beyond cell recruitment, e. g. controlling cell homeostasis and activation (1). For instance, the chemokine CXCL12 is crucial in controlling mobilization of haematopoietic stem cells and the homeostasis of many cell types. Expression of the CXCL12 (also known as stromal cellderived factor 1) receptor CXCR4 has been described on many cell types including monocytes and macrophages, neutrophils (2), T-cells (3), B-cells (4), mature endothelial cells (ECs) (5), and vascular smooth muscle cells (VSMCs) (6, 7). While all of these cells play distinct roles in the pathophysiology of atherosclerosis (8), little is known about the precise role of CXCR4 during individual cellular responses in this context ( Fig. 1). Macrophages, neutrophils CXCL12/CXCR4 signalling has been linked to enhanced macropinocytosis in leukocytes (9), suggesting that a lack of CXCR4 may influence (modified) lipid accumulation in macrophages and other lesional cells. In contrast, a recent study found CXCL12 to induce phagocytosis and the uptake of acetylated LDL in THP1-derived macrophages through binding CXCR7 but not CXCR4 (10). Accordingly, the CXCR7 agonist CCX771 was shown to increase the uptake of very low-density LDL (VLDL) in adipocytes and treatment of Apoe / mice with CCX771 reduced the levels of circulating VLDL and decreased atherosclerosis (11). Whether similar mechanisms can be identified in cell types other than macrophages remains to be determined, as are the exact mechanisms underlying CXCR7-mediated uptake of (modified) lipids. In contrast to human monocytes, expression of CXCR4 in mouse is higher expressed on non-classical compared to classical monocytes but this may not imply functional differences in individual mouse and human monocyte subsets (12). Treatment of mice with the CXCR4 antagonist AMD3100 induced cell egress Schattauer 2016 Hämostaseologie 2/2016

98 C. Weber; Y. Döring; H. Noels: CXCR4 in atherosclerosis A showed less CXCL12-induced rescue of platelets from activation-induced apoptosis through CXCR7 engagement (27). Likewise, MIF can limit activation-induced apoptosis of platelets via CXCR7-dependent Akt signalling, thereby reducing the thrombotic potential (28). Hence, differential regulation of CXCR4/CXCR7 surface expression on platelets upon ligand exposure at sites of platelet activation or accumulation may orchestrate platelet survival. In turn, the vascular functions mediated by platelet CXCR4 remain to be elucidated in vivo. Fig. 1 Cell type-specific roles of the CXCL12/CXCR4 axis in the context of atherosclerosis: The CXCL12 receptor CXCR4 is expressed on all cell types important in atherosclerosis, including monocytes and macrophages, neutrophils, T-cells, B-cells, endothelial cells (5) and vascular smooth muscle cells. from the bone marrow (13), which is in line with findings of increased leukocytosis, mostly neutrophil mobilization, and enhanced lesion formation in Apoe / receiving a cholesterol-rich diet for 12 weeks while treated with AMD3100. Interestingly, monocyte numbers were only moderately enhanced in these mice and lesion growth was mainly attributable to increased plaque neutrophils and enhanced apoptosis (14). Subsequently emerging evidence underscored the role of neutrophils in atherogenesis (15, 16) and it was recognized that the CXCL12/CXCR4 axis maintains neutrophil homeostasis by regulation of neutrophil release from the bone marrow in a cell-autonomous fashion (17). Furthermore, senescent neutrophils in the periphery expressing high levels of CXCR4 can home back to the bone marrow to be cleared in a rhythmic fashion (18, 19). Accordingly, neutrophil mobilization by AMD3100- mediated CXCR4 inhibition has also been attributed to arise from lung demargination and blockade of neutrophil homing to the bone marrow (20), and consequently the increase of atherosclerosis by CXCR4 blockade may be partially due to perturbing neutrophil apoptosis and senescence. Also, CXCR4 blockade through expression of CXCR4 degrakine in bone marrow cells was shown to enhance atherosclerosis by affecting neutrophil function, as reflected in a hyperactivated state, increased adhesion capacity and reduced apoptosis (21). Lymphocytes, platelets In addition to CXCL12/CXCR4 mediating lymphocyte recruitment, CXCR4 is able to mediate MIF-induced chemotaxis of B-cells and T-cells, and T-cell arrest in vitro (22, 23). Blockade of MIF in Apoe / mice on high-fat diet indeed resulted in the formation of smaller atherosclerotic lesions displaying reduced macrophage and T-cell content, supporting a role for MIF in T-cell chemotaxis in the context of atherosclerosis in vivo (22). CXCL12 has been reported to be a weak platelet agonist amplifying platelet activation, adhesion and chemokine release triggered by low dose agonists such as ADP, thrombin, or arterial flow (24, 25). Furthermore, CXCL12 gradients could induce platelet migration and transmigration in vitro involving PI3K signalling (26). In addition, CXCL12 can trigger CXCR4 internalization and cyclophilin A-dependent CXCR7 externalization on platelets, resulting in prolonged platelet survival. Mice lacking the cytosolic chaperone cyclophilin Endothelial cells Expression of CXCR4 on various types of vascular ECs has been widely reported (29), however, ECs represent a very heterogeneous population, with ECs from different anatomic sites differing in protein expression, localization and function (30), particularly when venous and arterial ECs are concerned (31). In human carotid artery specimens, CXCR4 was abundantly expressed by lesional ECs and only marginally in minimally diseased endothelium (32, 33). Similarly, Gupta et al. showed CXCR4 mrna expression in human coronary artery ECs, although it is not clear whether these ECs were derived from inflamed or steady-state endothelium (5). CXCR4 was also found to be expressed in bovine aortic ECs, where it was redistributed from a cytoplasmic granular pattern to the surface in migrating cells (34); in human aortic ECs, where it was upregulated by stimulation with VEGF or bfgf (35); and in mouse aortic endothelium, where it may be down-regulated by atheroprotective shear flow, similarly as observed in HUVECs where low shear stress led to increased apoptosis (33, 36). As angiogenesis may contribute to plaque vulnerability and hemorrhage, a potential role of CXCR4/CXCL12 therein is important to consider. Interestingly, blocking of TLR2 appears to promote the angiogenic capacity of ECs, by inducing ERK1/2 and Akt1 signalling through CXCR4, by modulating a TLR2-CXCR4 association (37). Recently, CXCL12 has been identified as a suppressor of endothelial permeability, Hämostaseologie 2/2016 Schattauer 2016

C. Weber; Y. Döring; H. Noels: CXCR4 in atherosclerosis 99 promoting endothelial barrier integrity through CXCR4 but not CXCR7 (38). Specifically, CXCL12 stimulates the CXCR4/PI3K/Rac1 signaling pathway for subsequent cytoskeletal rearrangement. As another potential mechanism for protective effects, CXCL12-CXCR4 has been shown to induce enos phosphorylation and activity, thereby increasing endothelial NO availability (39). Furthermore, apoptotic microparticles derived from distressed ECs conferred microrna-126 to resident ECs to unleash autocrine CXCL12 induction through CXCR4 by repressing its inhibitor RGS16, triggering angiogenic cell recruitment and atheroprotective endothelial regeneration (40). A role for CXCR4 signalling in endothelial recovery was furthermore supported by the observation that endothelial-specific knock-out of CXCR4 reduced reendothelialization after endothelial denudation, which was associated with reduced mobilization of endothe- lial progenitor cells and enhanced neointima formation (41). Also, the alternative CXCR4 ligand MIF has been reported to mediate EC migration and tube formation, and to drive angiogenesis in matrigel plugs in vivo (42). Furthermore, MIF stimulation of ECs induces the secretion of CCL2 and the expression of the adhesion molecule ICAM-1 (43, 44), which could contribute to atherogenesis by mediating leukocyte recruitment and their adhesion on the endothelium. However, it remains unclear to which extent these observations depend on the chemokine receptor CXCR4, as ECs also express the MIF receptors CXCR2 and CD74, the latter under inflammatory conditions (45). VSMCs Vascular smooth muscle cells (VSMCs) are highly specialized cells controlling contrac- tion and regulation of blood vessel diameter, blood pressure and blood flow. Moreover, VSMCs play a critical role in secretion of extracellular matrix components, which shape the mechanical properties of mature blood vessels. Differentiated VSMCs in adult blood vessels proliferate at very low rates and retain high plasticity, enabling changes in phenotype, referred to as phenotypic switching (41), which is considered an important pathophysiological mechanism in atherosclerosis (42). A functional CXCR4 expression in SMCs has been implied by findings that addition of CXCL12 or HIV envelope proteins specifically binding CXCR4 did induce tissue factor activity in human aortic SMCs (43). Subsequent reports conformed CXCR4 protein expression on human vein SMCs (44), CXCR4 expression (RNA, protein) on mouse medial SMCs (6), rat (7, 45) and human aortic SMCs (Weber et al., unpublished data). Anzeige

100 C. Weber; Y. Döring; H. Noels: CXCR4 in atherosclerosis Besides switching to a synthetic secretory phenotype and their presumed migration from the medial to the intimal arterial wall giving rise to atherogenesis (42), intimal SMCs can also stabilize atherosclerotic lesions by fibrous cap formation. In a rat model of diabetes, high glucose levels were shown to trigger activation, proliferation and enhanced chemotaxis of VSMCs via the CXCL12/CXCR4 axis (7). It was described that CXCL12 stimulates pro- MMP-2 expression in human aortic SMCs via CXCR4 in association with the epidermal growth factor receptor in vitro, suggesting that CXCR4 expands its signaling repertoire by cross-talking with other receptors. Notably, oxldl as another proatherogenic trigger was found to induce rat aortic SMC proliferation. This effect could even be enhanced by addition of CXCL12 concomitant with diminished SMC apoptosis, which may be beneficial through plaque stabilization or detrimental due to intimal hyperplasia (46). Likewise, CXCL12/CXCR4 signalling might be crucial in vein graft atherosclerosis and contribute to SMC-mediated vein graft neointimal hyperplasia in mice, in association with CXCR4-mediated recruitment of inflammatory (progenitor) cells (47), similar as seen for wire-induced neointima formation (48). Systemic treatment with CXCL12 promotes a more stable atherosclerotic lesion phenotype and enhances the accumulation of smooth muscle progenitor cells in these lesions without promoting atherosclerosis, representing a promising approach to treat unstable atherosclerosis (49). Outlook In order to delineate the complex cellspecific functions of CXCR4, its primary ligand CXCL12 as well as the second receptor CXCR7, it will be mandatory to create conditional inducible knock-out mouse models for all the above cell types and beyond, e. g. including adipocytes and neuroendocrine cells, where these axes may also control crucial mechanisms affecting the pathogenesis of atherosclerosis. Furthermore, a recent report using inducible SM22a-driven ROSA26 LacZ-cre mice to irreversibly label VSMCs for fate mapping suggested that VSMC have some potential to differentiate into macrophagelike foam cells in the aorta of atherosclerotic mice, based on co-staining with the myeloid markers Mac2 and CD68 (50) and in support of earlier studies suggesting trans differentiation (51). On the other hand, recent macrophage gate mapping studies indicated that plaque macrophages proliferate locally, thus not exclusively relying on monocyte recruitment, and that the macrophage pool can be reconstituted by bone marrow-derived cells, suggesting that the source is a circulating one (52). Whether or not distinct plaque macrophage populations exist that are derived from different precursor cell types, remains unclear, as does the existence of a precursor for local macrophage proliferation, which might as well be related to a bone marrowderived VSMC progenitor that has been shown to give rise to VSMC-like cells in the atherosclerotic vascular wall. As the CXCL12/CXCR4 axis is crucial in mobilizing haematopoietic and progenitor cells from the bone marrow, a role for this axis in controlling the mobilization of specific macrophage progenitor cells and thus macrophage subpopulations into atherosclerotic lesions could be hypothesized. Conflict of interest The authors declare that they have no conflict of interest. References 1. Zernecke A, Weber C. Chemokines in atherosclerosis: proceedings resumed. Arterioscler Thromb Vasc Biol 2014; 34: 742 750. 2. Bruhl H, Cohen CD, Linder S et al. Post-translational and cell type-specific regulation of CXCR4 expression by cytokines. Eur J Immunol 2003; 33: 3028 3037. 3. Murphy PM, Baggiolini M, Charo IF et al. International union of pharmacology. XXII. Nomenclature for chemokine receptors. Pharmacol Rev 2000; 52: 145 176. 4. Nie Y, Waite J, Brewer F et al. The role of CXCR4 in maintaining peripheral B cell compartments and humoral immunity. J Exp Med 2004; 200: 1145 1156. 5. Gupta SK, Lysko PG, Pillarisetti K et al. Chemokine receptors in human endothelial cells. J Biol Chem 1998; 273: 4282 4287. 6. Nemenoff RA, Simpson PA, Furgeson SB et al. Targeted deletion of PTEN in smooth muscle cells results in vascular remodeling and recruitment of progenitor cells through induction of stromal cell-derived factor-1alpha. Circ Res 2008; 102: 1036 1045. 7. Jie W, Wang X, Zhang Y et al. SDF-1alpha/CXCR4 axis is involved in glucose-potentiated proliferation and chemotaxis in rat vascular smooth muscle cells. Int J Exp Pathol 2010; 91: 436 444. 8. Weber C, Noels H. Atherosclerosis: current pathogenesis and therapeutic options. Nat Med 2011; 17: 1410 1422. 9. Tanaka G, Nakase I, Fukuda Y et al. CXCR4 stimulates macropinocytosis. Chem Biol 2012; 19: 1437 1446. 10. Ma W, Liu Y, Ellison N, Shen J. Induction of C-X-C chemokine receptor type 7 switches stromal cellderived factor-1 signaling and phagocytic activity in macrophages linked to atherosclerosis. J Biol Chem 2013; 288: 15481 15494. 11. Li X, Zhu M, Penfold ME et al. Activation of CXCR7 limits atherosclerosis and improves hyperlipidemia by increasing cholesterol uptake in adipose tissue. Circulation 2014; 129: 1244 1253. 12. Ingersoll MA, Spanbroek R, Lottaz C et al. Comparison of gene expression profiles between human and mouse monocyte subsets. Blood 2010; 115: e10 e19. 13. Schiraldi M, Raucci A, Munoz LM et al. HMGB1 promotes recruitment of inflammatory cells to damaged tissues by forming a complex with CXCL12 and signaling via CXCR4. J Exp Med 2012; 209: 551 563. 14. Zernecke A, Bot I, Djalali-Talab Y et al. Protective role of CXC receptor 4/CXC ligand 12 unveils the importance of neutrophils in atherosclerosis. Circulation research 2008; 102: 209 217. 15. Drechsler M, Megens RT, van Zandvoort M et al. Hyperlipidemia-triggered neutrophilia promotes early atherosclerosis. Circulation 2010; 122: 1837 1845. 16. Doring Y, Soehnlein O, Drechsler M et al. Hematopoietic interferon regulatory factor 8-deficiency accelerates atherosclerosis in mice. Arterioscler Thromb Vasc Biol 2012; 32: 1613 1623. 17. Eash KJ, Means JM, White DW, Link DC. CXCR4 is a key regulator of neutrophil release from the bone marrow under basal and stress granulopoiesis conditions. Blood 2009; 113: 4711 4719. 18. Martin C, Burdon PC, Bridger G et al. Chemokines acting via CXCR2 and CXCR4 control the release of neutrophils from the bone marrow and their return following senescence. Immunity 2003; 19: 583 593. 19. Casanova-Acebes M, Pitaval C, Weiss LA et al. Rhythmic modulation of the hematopoietic niche through neutrophil clearance. Cell 2013; 153: 1025 1035. 20. Devi S, Wang Y, Chew WK et al. Neutrophil mobilization via plerixafor-mediated CXCR4 inhibition arises from lung demargination and blockade of Hämostaseologie 2/2016 Schattauer 2016

C. Weber; Y. Döring; H. Noels: CXCR4 in atherosclerosis 102 neutrophil homing to the bone marrow. J Exp Med 2013; 210: 2321 2336. 21. Bot I, Daissormont IT, Zernecke A et al. CXCR4 blockade induces atherosclerosis by affecting neutrophil function. J Mol Cell Cardiol 2014; 74: 44 52. 22. Bernhagen J, Krohn R, Lue H et al. MIF is a noncognate ligand of CXC chemokine receptors in inflammatory and atherogenic cell recruitment. Nat Med 2007; 13: 587 596. 23. Klasen C, Ohl K, Sternkopf M et al. MIF promotes B cell chemotaxis through the receptors CXCR4 and CD74 and ZAP-70 signaling. J Immunol 2014; 192: 5273 5284. 24. Kowalska MA, Ratajczak J, Hoxie J et al. Megakaryocyte precursors, megakaryocytes and platelets express the HIV co-receptor CXCR4 on their surface. Br J Haematol 1999; 104: 220 229. 25. Gear AR, Suttitanamongkol S, Viisoreanu D et al. Adenosine diphosphate strongly potentiates the ability of the chemokines MDC, TARC, and SDF-1 to stimulate platelet function. Blood 2001; 97: 937 945. 26. Kraemer BF, Borst O, Gehring EM et al. PI3 kinase-dependent stimulation of platelet migration by stromal cell-derived factor 1. J Mol Med (Berl) 2010; 88: 1277 1288. 27. Chatterjee M, Seizer P, Borst O et al. SDF-1alpha induces differential trafficking of CXCR4-CXCR7 involving cyclophilin A, CXCR7 ubiquitination and promotes platelet survival. FASEB J 2014; 28: 2864 2878. 28. Chatterjee M, Borst O, Walker B et al. Macrophage migration inhibitory factor limits activation-induced apoptosis of platelets via CXCR7-dependent akt signaling. Circ Res 2014; 115: 939 949. 29. Hillyer P, Mordelet E, Flynn G, Male D. Chemokines, chemokine receptors and adhesion molecules on different human endothelia: discriminating the tissue-specific functions that affect leucocyte migration. Clin Exp Immunol 2003; 134: 431 441. 30. Aird WC. Phenotypic heterogeneity of the endothelium: I. Structure, function, and mechanisms. Circ Res 2007; 100: 158 173. 31. Dela Paz NG, D Amore PA. Arterial versus venous endothelial cells. Cell Tissue Res 2009; 335: 5 16. 32. Molino M, Woolkalis MJ, Prevost N et al. CXCR4 on human endothelial cells can serve as both a mediator of biological responses and as a receptor for HIV-2. Biochim Biophys Acta 2000; 1500: 227 240. 33. Melchionna R, Porcelli D, Mangoni A et al. Laminar shear stress inhibits CXCR4 expression on endothelial cells: functional consequences for atherogenesis. FASEB 2005; 19: 629 631. 34. Feil C, Augustin HG. Endothelial cells differentially express functional CXC-chemokine receptor-4 under the control of autocrine activity and exogenous cytokines. Biochem Biophys Res Commun 1998; 247: 38 45. 35. Salcedo R, Wasserman K, Young HA et al. Vascular endothelial growth factor and basic fibroblast growth factor induce expression of CXCR4 on human endothelial cells. Am J Pathol 1999; 154: 1125 1135. 36. Melchionna R, Romani M, Ambrosino V et al. Role of HIF-1alpha in proton-mediated CXCR4 down-regulation in endothelial cells. Cardiovasc Res 2010; 86: 293 301. 37. Wagner NM, Bierhansl L, Noldge-Schomburg G et al. Toll-like receptor 2-blocking antibodies promote angiogenesis and induce ERK1/2 and AKT signaling via CXCR4 in endothelial cells. Arterioscler Thromb Vasc Biol 2013; 33: 1943 1951. 38. Kobayashi K, Sato K, Kida T et al. Stromal cell-derived factor-1alpha/c-x-c chemokine receptor type 4 axis promotes endothelial cell barrier integrity via phosphoinositide 3-kinase and Rac1 activation. Arterioscler Thromb Vasc Biol 2014; 34: 1716 1722. 39. Chen LH, Advani SL, Thai K et al. SDF-1/CXCR4 signaling preserves microvascular integrity and renal function in chronic kidney disease. PLoS One 2014; 9: e92227. 40. Zernecke A, Bidzhekov K, Noels H et al. Delivery of microrna-126 by apoptotic bodies induces CXCL12-dependent vascular protection. Sci Signal 2009; 2: ra81. 41. Noels H, Zhou B, Tilstam PV et al. Deficiency of endothelial Cxcr4 reduces reendothelialization and enhances neointimal hyperplasia after vascular injury in atherosclerosis-prone mice. Arterioscler Thromb Vasc Biol 2014; 34: 1209 1220. 42. Amin MA, Volpert OV, Woods JM et al. Migration inhibitory factor mediates angiogenesis via mitogen-activated protein kinase and phosphatidylinositol kinase. Circ Res 2003; 93: 321 329. 43. Gregory JL, Morand EF, McKeown SJ et al. Macrophage migration inhibitory factor induces macrophage recruitment via CC chemokine ligand 2. J Immunol 2006; 177: 8072 8079. 44. Lin SG, Yu XY, Chen YX et al. De novo expression of macrophage migration inhibitory factor in atherogenesis in rabbits. Circ Res 2000; 87: 1202 1208. 45. Tillmann S, Bernhagen J, Noels H. Arrest functions of the MIF ligand/receptor axes in atherogenesis. Front Immunol 2013; 4: 115. 46. Owens GK, Kumar MS, Wamhoff BR. Molecular regulation of vascular smooth muscle cell differentiation in development and disease. Physiol Rev 2004; 84: 767 801. 47. Gomez D, Owens GK. Smooth muscle cell phenotypic switching in atherosclerosis. Cardiovasc Res 2012; 95: 156 164. 48. Schecter AD, Berman AB, Yi L et al. HIV envelope gp120 activates human arterial smooth muscle cells. Proc Natl Acad Sci USA 2001; 98: 10142 10147. 49. Li F, Guo WY, Li WJ et al. Cyclic stretch upregulates SDF-1alpha/CXCR4 axis in human saphenous vein smooth muscle cells. Biochem Biophys Res Commun 2009; 386: 247 251. 50. Pan CH, Chen CW, Sheu MJ, Wu CH. Salvianolic acid B inhibits SDF-1alpha-stimulated cell proliferation and migration of vascular smooth muscle cells by suppressing CXCR4 receptor. Vascul Pharmacol 2012; 56: 98 105. 51. Li LX, Zhang XF, Bai X, Tong Q. SDF-1 promotes ox-ldl induced vascular smooth muscle cell proliferation. Cell Biol Int 2013; 37: 988 994. 52. Zhang L, Brian L, Freedman NJ. Vein graft neointimal hyperplasia is exacerbated by CXCR4 signaling in vein graft-extrinsic cells. J Vasc Surg 2012; 56: 1390 1397. 53. Zernecke A, Schober A, Bot I et al. SDF-1alpha/ CXCR4 axis is instrumental in neointimal hyperplasia and recruitment of smooth muscle progenitor cells. Circ Res 2005; 96: 784 791. 54. Akhtar S, Gremse F, Kiessling F et al. CXCL12 promotes the stabilization of atherosclerotic lesions mediated by smooth muscle progenitor cells in Apoe-deficient mice. Arterioscler Thromb Vasc Biol 2013; 33: 679 686. 55. Feil S, Fehrenbacher B, Lukowski R et al. Transdifferentiation of vascular smooth muscle cells to macrophage-like cells during atherogenesis. Circ Res 2014; 115: 662 667. 56. Rong JX, Shapiro M, Trogan E, Fisher EA. Transdifferentiation of mouse aortic smooth muscle cells to a macrophage-like state after cholesterol loading. Proc Natl Acad Sci USA 2003; 100: 13531 13536. 57. Robbins CS, Hilgendorf I, Weber GF et al. Local proliferation dominates lesional macrophage accumulation in atherosclerosis. Nat Med 2013; 19: 1166 1172. Schattauer 2016 Hämostaseologie 2/2016