46 results on '"Megens RTA"'
Search Results
2. Mex3a Protects Against Atherosclerosis: Evidence From Mice and Humans.
- Author
-
Santovito D, Henderson JM, Bidzhekov K, Triantafyllidou V, Jansen Y, Chen Z, Farina FM, Diagel A, Aslani M, Blanchet X, Schunkert H, Megens RTA, Döring Y, Sattler M, and Weber C
- Subjects
- Animals, Humans, Mice, Mice, Knockout, Atherosclerosis prevention & control, RNA-Binding Proteins genetics, RNA-Binding Proteins metabolism
- Abstract
Competing Interests: None.
- Published
- 2024
- Full Text
- View/download PDF
3. Generation of complex bone marrow organoids from human induced pluripotent stem cells.
- Author
-
Frenz-Wiessner S, Fairley SD, Buser M, Goek I, Salewskij K, Jonsson G, Illig D, Zu Putlitz B, Petersheim D, Li Y, Chen PH, Kalauz M, Conca R, Sterr M, Geuder J, Mizoguchi Y, Megens RTA, Linder MI, Kotlarz D, Rudelius M, Penninger JM, Marr C, and Klein C
- Subjects
- Humans, Animals, Mice, Hematopoietic Stem Cells cytology, Bone Marrow metabolism, Bone Marrow Cells cytology, Bone Marrow Cells metabolism, Cell Culture Techniques methods, Mesenchymal Stem Cells cytology, Mesenchymal Stem Cells metabolism, Organoids cytology, Organoids metabolism, Induced Pluripotent Stem Cells cytology, Induced Pluripotent Stem Cells metabolism, Cell Differentiation, Hematopoiesis
- Abstract
The human bone marrow (BM) niche sustains hematopoiesis throughout life. We present a method for generating complex BM-like organoids (BMOs) from human induced pluripotent stem cells (iPSCs). BMOs consist of key cell types that self-organize into spatially defined three-dimensional structures mimicking cellular, structural and molecular characteristics of the hematopoietic microenvironment. Functional properties of BMOs include the presence of an in vivo-like vascular network, the presence of multipotent mesenchymal stem/progenitor cells, the support of neutrophil differentiation and responsiveness to inflammatory stimuli. Single-cell RNA sequencing revealed a heterocellular composition including the presence of a hematopoietic stem/progenitor (HSPC) cluster expressing genes of fetal HSCs. BMO-derived HSPCs also exhibited lymphoid potential and a subset demonstrated transient engraftment potential upon xenotransplantation in mice. We show that the BMOs could enable the modeling of hematopoietic developmental aspects and inborn errors of hematopoiesis, as shown for human VPS45 deficiency. Thus, iPSC-derived BMOs serve as a physiologically relevant in vitro model of the human BM microenvironment to study hematopoietic development and BM diseases., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
4. Identification of a non-canonical chemokine-receptor pathway suppressing regulatory T cells to drive atherosclerosis.
- Author
-
Döring Y, van der Vorst EPC, Yan Y, Neideck C, Blanchet X, Jansen Y, Kemmerich M, Bayasgalan S, Peters LJF, Hristov M, Bidzhekov K, Yin C, Zhang X, Leberzammer J, Li Y, Park I, Kral M, Nitz K, Parma L, Gencer S, Habenicht A, Faussner A, Teupser D, Monaco C, Holdt L, Megens RTA, Atzler D, Santovito D, von Hundelshausen P, and Weber C
- Abstract
CCL17 is produced by conventional dendritic cells (cDCs), signals through CCR4 on regulatory T cells (Tregs), and drives atherosclerosis by suppressing Treg functions through yet undefined mechanisms. Here we show that cDCs from CCL17-deficient mice display a pro-tolerogenic phenotype and transcriptome that is not phenocopied in mice lacking its cognate receptor CCR4. In the plasma of CCL17-deficient mice, CCL3 was the only decreased cytokine/chemokine. We found that CCL17 signaled through CCR8 as an alternate high-affinity receptor, which induced CCL3 expression and suppressed Treg functions in the absence of CCR4. Genetic ablation of CCL3 and CCR8 in CD4
+ T cells reduced CCL3 secretion, boosted FoxP3+ Treg numbers, and limited atherosclerosis. Conversely, CCL3 administration exacerbated atherosclerosis and restrained Treg differentiation. In symptomatic versus asymptomatic human carotid atheroma, CCL3 expression was increased, while FoxP3 expression was reduced. Together, we identified a non-canonical chemokine pathway whereby CCL17 interacts with CCR8 to yield a CCL3-dependent suppression of atheroprotective Tregs., Competing Interests: Competing Financial Interests The authors declare no competing financial interests.- Published
- 2024
- Full Text
- View/download PDF
5. Targeting a cell-specific microRNA repressor of CXCR4 ameliorates atherosclerosis in mice.
- Author
-
Cimen I, Natarelli L, Abedi Kichi Z, Henderson JM, Farina FM, Briem E, Aslani M, Megens RTA, Jansen Y, Mann-Fallenbuchel E, Gencer S, Duchêne J, Nazari-Jahantigh M, van der Vorst EPC, Enard W, Döring Y, Schober A, Santovito D, and Weber C
- Subjects
- Humans, Animals, Mice, Endothelial Cells metabolism, Receptors, CXCR4 metabolism, Cell Proliferation, Myocytes, Smooth Muscle metabolism, Cell Movement, MicroRNAs genetics, MicroRNAs metabolism, Atherosclerosis genetics, Plaque, Atherosclerotic pathology
- Abstract
The CXC chemokine receptor 4 (CXCR4) in endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) is crucial for vascular integrity. The atheroprotective functions of CXCR4 in vascular cells may be counteracted by atherogenic functions in other nonvascular cell types. Thus, strategies for cell-specifically augmenting CXCR4 function in vascular cells are crucial if this receptor is to be useful as a therapeutic target in treating atherosclerosis and other vascular disorders. Here, we identified miR-206-3p as a vascular-specific CXCR4 repressor and exploited a target-site blocker (CXCR4-TSB) that disrupted the interaction of miR-206-3p with CXCR4 in vitro and in vivo. In vitro, CXCR4-TSB enhanced CXCR4 expression in human and murine ECs and VSMCs to modulate cell viability, proliferation, and migration. Systemic administration of CXCR4-TSB in Apoe -deficient mice enhanced Cxcr4 expression in ECs and VSMCs in the walls of blood vessels, reduced vascular permeability and monocyte adhesion to endothelium, and attenuated the development of diet-induced atherosclerosis. CXCR4-TSB also increased CXCR4 expression in B cells, corroborating its atheroprotective role in this cell type. Analyses of human atherosclerotic plaque specimens revealed a decrease in CXCR4 and an increase in miR-206-3p expression in advanced compared with early lesions, supporting a role for the miR-206-3p-CXCR4 interaction in human disease. Disrupting the miR-206-3p-CXCR4 interaction in a cell-specific manner with target-site blockers is a potential therapeutic approach that could be used to treat atherosclerosis and other vascular diseases.
- Published
- 2023
- Full Text
- View/download PDF
6. Short-Term Western Diet Causes Rapid and Lasting Alterations of Bone Marrow Physiology.
- Author
-
Bianchini M, Möller-Ramon Z, Weber C, Megens RTA, and Duchêne J
- Subjects
- Humans, Bone Marrow Cells, Bone Marrow, Diet, Western adverse effects
- Abstract
Competing Interests: None declared.
- Published
- 2023
- Full Text
- View/download PDF
7. Properties and fate of human mesenchymal stem cells upon miRNA let-7f-promoted recruitment to atherosclerotic plaques.
- Author
-
Egea V, Megens RTA, Santovito D, Wantha S, Brandl R, Siess W, Khani S, Soehnlein O, Bartelt A, Weber C, and Ries C
- Subjects
- Mice, Animals, Humans, Cytokines, Immunologic Factors, MicroRNAs genetics, Plaque, Atherosclerotic, Atherosclerosis genetics, Mesenchymal Stem Cells
- Abstract
Aims: Atherosclerosis is a chronic inflammatory disease of the arteries leading to the formation of atheromatous plaques. Human mesenchymal stem cells (hMSCs) are recruited from the circulation into plaques where in response to their environment they adopt a phenotype with immunomodulatory properties. However, the mechanisms underlying hMSC function in these processes are unclear. Recently, we described that miRNA let-7f controls hMSC invasion guided by inflammatory cytokines and chemokines. Here, we investigated the role of let-7f in hMSC tropism to human atheromas and the effects of the plaque microenvironment on cell fate and release of soluble factors., Methods and Results: Incubation of hMSCs with LL-37, an antimicrobial peptide abundantly found in plaques, increased biosynthesis of let-7f and N-formyl peptide receptor 2 (FPR2), enabling chemotactic invasion of the cells towards LL-37, as determined by qRT-PCR, flow cytometry, and cell invasion assay analysis. In an Apoe-/- mouse model of atherosclerosis, circulating hMSCs preferentially adhered to athero-prone endothelium. This property was facilitated by elevated levels of let-7f in the hMSCs, as assayed by ex vivo artery perfusion and two-photon laser scanning microscopy. Exposure of hMSCs to homogenized human atheromatous plaque material considerably induced the production of various cytokines, chemokines, matrix metalloproteinases, and tissue inhibitors of metalloproteinases, as studied by PCR array and western blot analysis. Moreover, exposure to human plaque extracts elicited differentiation of hMSCs into cells of the myogenic lineage, suggesting a potentially plaque-stabilizing effect., Conclusions: Our findings indicate that let-7f promotes hMSC tropism towards atheromas through the LL-37/FPR2 axis and demonstrate that hMSCs upon contact with human plaque environment develop a potentially athero-protective signature impacting the pathophysiology of atherosclerosis., Competing Interests: Conflict of interest: none declared., (© The Author(s) 2022. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2023
- Full Text
- View/download PDF
8. Ex Vivo Perfusion System to Analyze Chemokine-Driven Leukocyte Adhesion.
- Author
-
Maas SL, Megens RTA, and van der Vorst EPC
- Subjects
- Mice, Animals, Cell Adhesion physiology, Carotid Arteries, Perfusion, Endothelium, Vascular physiology, Leukocytes physiology, Chemokines
- Abstract
At the moment, many researchers are using in vitro techniques to investigate chemokine-driven leukocyte adhesion/recruitment, for example, by using a transwell or flow chamber system. Here we describe a more physiologically relevant, sophisticated, and highly flexible method to study leukocyte adhesion ex vivo in fresh murine carotid arteries under arterial flow conditions. This model mimics an in vivo situation and allows the combination of leukocytes and arteries isolated from different donors in one experiment, generating information on both vascular and leukocyte adhesive properties of both donors. This method provides a versatile, highly physiologically relevant model to investigate leukocyte adhesion., (© 2023. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
9. The Amino Acid Homoarginine Inhibits Atherogenesis by Modulating T-Cell Function.
- Author
-
Nitz K, Lacy M, Bianchini M, Wichapong K, Kücükgöze IA, Bonfiglio CA, Migheli R, Wu Y, Burger C, Li Y, Forné I, Ammar C, Janjic A, Mohanta S, Duchene J, Heemskerk JWM, Megens RTA, Schwedhelm E, Huveneers S, Lygate CA, Santovito D, Zimmer R, Imhof A, Weber C, Lutgens E, and Atzler D
- Subjects
- Amino Acids, Animals, Apolipoproteins E, Female, Homoarginine pharmacology, Mice, Myosin Heavy Chains, T-Lymphocytes metabolism, Atherosclerosis drug therapy, Atherosclerosis metabolism, Atherosclerosis prevention & control, Drinking Water, Plaque, Atherosclerotic
- Abstract
Background: Amino acid metabolism is crucial for inflammatory processes during atherogenesis. The endogenous amino acid homoarginine is a robust biomarker for cardiovascular outcome and mortality with high levels being protective. However, the underlying mechanisms remain elusive. We investigated the effect of homoarginine supplementation on atherosclerotic plaque development with a particular focus on inflammation., Methods: Female ApoE-deficient mice were supplemented with homoarginine (14 mg/L) in drinking water starting 2 weeks before and continuing throughout a 6-week period of Western-type diet feeding. Control mice received normal drinking water. Immunohistochemistry and flow cytometry were used for plaque- and immunological phenotyping. T cells were characterized using mass spectrometry-based proteomics, by functional in vitro approaches, for example, proliferation and migration/chemotaxis assays as well as by super-resolution microscopy., Results: Homoarginine supplementation led to a 2-fold increase in circulating homoarginine concentrations. Homoarginine-treated mice exhibited reduced atherosclerosis in the aortic root and brachiocephalic trunk. A substantial decrease in CD3
+ T cells in the atherosclerotic lesions suggested a T-cell-related effect of homoarginine supplementation, which was mainly attributed to CD4+ T cells. Macrophages, dendritic cells, and B cells were not affected. CD4+ T-cell proteomics and subsequent pathway analysis together with in vitro studies demonstrated that homoarginine profoundly modulated the spatial organization of the T-cell actin cytoskeleton and increased filopodia formation via inhibition of Myh9 (myosin heavy chain 9). Further mechanistic studies revealed an inhibition of T-cell proliferation as well as a striking impairment of the migratory capacities of T cells in response to relevant chemokines by homoarginine, all of which likely contribute to its atheroprotective effects., Conclusions: Our study unravels a novel mechanism by which the amino acid homoarginine reduces atherosclerosis, establishing that homoarginine modulates the T-cell cytoskeleton and thereby mitigates T-cell functions important during atherogenesis. These findings provide a molecular explanation for the beneficial effects of homoarginine in atherosclerotic cardiovascular disease.- Published
- 2022
- Full Text
- View/download PDF
10. Heterocomplexes between the atypical chemokine MIF and the CXC-motif chemokine CXCL4L1 regulate inflammation and thrombus formation.
- Author
-
Brandhofer M, Hoffmann A, Blanchet X, Siminkovitch E, Rohlfing AK, El Bounkari O, Nestele JA, Bild A, Kontos C, Hille K, Rohde V, Fröhlich A, Golemi J, Gokce O, Krammer C, Scheiermann P, Tsilimparis N, Sachs N, Kempf WE, Maegdefessel L, Otabil MK, Megens RTA, Ippel H, Koenen RR, Luo J, Engelmann B, Mayo KH, Gawaz M, Kapurniotu A, Weber C, von Hundelshausen P, and Bernhagen J
- Subjects
- HEK293 Cells, Humans, Inflammation metabolism, Intramolecular Oxidoreductases, Platelet Factor 4, Receptors, Interleukin-8B chemistry, Receptors, Interleukin-8B genetics, Receptors, Interleukin-8B metabolism, Atherosclerosis metabolism, Macrophage Migration-Inhibitory Factors metabolism, Thrombosis
- Abstract
To fulfil its orchestration of immune cell trafficking, a network of chemokines and receptors developed that capitalizes on specificity, redundancy, and functional selectivity. The discovery of heteromeric interactions in the chemokine interactome has expanded the complexity within this network. Moreover, some inflammatory mediators, not structurally linked to classical chemokines, bind to chemokine receptors and behave as atypical chemokines (ACKs). We identified macrophage migration inhibitory factor (MIF) as an ACK that binds to chemokine receptors CXCR2 and CXCR4 to promote atherogenic leukocyte recruitment. Here, we hypothesized that chemokine-chemokine interactions extend to ACKs and that MIF forms heterocomplexes with classical chemokines. We tested this hypothesis by using an unbiased chemokine protein array. Platelet chemokine CXCL4L1 (but not its variant CXCL4 or the CXCR2/CXCR4 ligands CXCL8 or CXCL12) was identified as a candidate interactor. MIF/CXCL4L1 complexation was verified by co-immunoprecipitation, surface plasmon-resonance analysis, and microscale thermophoresis, also establishing high-affinity binding. We next determined whether heterocomplex formation modulates inflammatory/atherogenic activities of MIF. Complex formation was observed to inhibit MIF-elicited T-cell chemotaxis as assessed by transwell migration assay and in a 3D-matrix-based live cell-imaging set-up. Heterocomplexation also blocked MIF-triggered migration of microglia in cortical cultures in situ, as well as MIF-mediated monocyte adhesion on aortic endothelial cell monolayers under flow stress conditions. Of note, CXCL4L1 blocked binding of Alexa-MIF to a soluble surrogate of CXCR4 and co-incubation with CXCL4L1 attenuated MIF responses in HEK293-CXCR4 transfectants, indicating that complex formation interferes with MIF/CXCR4 pathways. Because MIF and CXCL4L1 are platelet-derived products, we finally tested their role in platelet activation. Multi-photon microscopy, FLIM-FRET, and proximity-ligation assay visualized heterocomplexes in platelet aggregates and in clinical human thrombus sections obtained from peripheral artery disease (PAD) in patients undergoing thrombectomy. Moreover, heterocomplexes inhibited MIF-stimulated thrombus formation under flow and skewed the lamellipodia phenotype of adhering platelets. Our study establishes a novel molecular interaction that adds to the complexity of the chemokine interactome and chemokine/receptor-network. MIF/CXCL4L1, or more generally, ACK/CXC-motif chemokine heterocomplexes may be target structures that can be exploited to modulate inflammation and thrombosis., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
11. Designed peptides as nanomolar cross-amyloid inhibitors acting via supramolecular nanofiber co-assembly.
- Author
-
Taş K, Volta BD, Lindner C, El Bounkari O, Hille K, Tian Y, Puig-Bosch X, Ballmann M, Hornung S, Ortner M, Prem S, Meier L, Rammes G, Haslbeck M, Weber C, Megens RTA, Bernhagen J, and Kapurniotu A
- Subjects
- Amyloid pharmacology, Amyloid beta-Peptides chemistry, Amyloidogenic Proteins, Humans, Islet Amyloid Polypeptide chemistry, Alzheimer Disease drug therapy, Amyloidosis, Diabetes Mellitus, Type 2 drug therapy, Nanofibers
- Abstract
Amyloid self-assembly is linked to numerous devastating cell-degenerative diseases. However, designing inhibitors of this pathogenic process remains a major challenge. Cross-interactions between amyloid-β peptide (Aβ) and islet amyloid polypeptide (IAPP), key polypeptides of Alzheimer's disease (AD) and type 2 diabetes (T2D), have been suggested to link AD with T2D pathogenesis. Here, we show that constrained peptides designed to mimic the Aβ amyloid core (ACMs) are nanomolar cross-amyloid inhibitors of both IAPP and Aβ42 and effectively suppress reciprocal cross-seeding. Remarkably, ACMs act by co-assembling with IAPP or Aβ42 into amyloid fibril-resembling but non-toxic nanofibers and their highly ordered superstructures. Co-assembled nanofibers exhibit various potentially beneficial features including thermolability, proteolytic degradability, and effective cellular clearance which are reminiscent of labile/reversible functional amyloids. ACMs are thus promising leads for potent anti-amyloid drugs in both T2D and AD while the supramolecular nanofiber co-assemblies should inform the design of novel functional (hetero-)amyloid-based nanomaterials for biomedical/biotechnological applications., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
12. Endothelial ENPP2 (Ectonucleotide Pyrophosphatase/Phosphodiesterase 2) Increases Atherosclerosis in Female and Male Mice.
- Author
-
Karshovska E, Mohibullah R, Zhu M, Zahedi F, Thomas D, Magkrioti C, Geissler C, Megens RTA, Bianchini M, Nazari-Jahantigh M, Ferreirós N, Aidinis V, and Schober A
- Subjects
- Animals, Female, Lysophospholipids, Male, Mice, Mice, Knockout, ApoE, Pyrophosphatases metabolism, Tamoxifen, Atherosclerosis metabolism, Endothelial Cells metabolism, Phosphoric Diester Hydrolases genetics, Phosphoric Diester Hydrolases metabolism
- Abstract
Background: Maladapted endothelial cells (ECs) secrete ENPP2 (ectonucleotide pyrophosphatase/phosphodiesterase 2; autotaxin)-a lysophospholipase D that generates lysophosphatidic acids (LPAs). ENPP2 derived from the arterial wall promotes atherogenic monocyte adhesion induced by generating LPAs, such as arachidonoyl-LPA (LPA20:4), from oxidized lipoproteins. Here, we aimed to determine the role of endothelial ENPP2 in the production of LPAs and atherosclerosis., Methods: We quantified atherosclerosis in mice harboring loxP-flanked Enpp2 alleles crossed with Apoe
-/- mice expressing tamoxifen-inducible Cre recombinase under the control of the EC-specific bone marrow X kinase promoter after 12 weeks of high-fat diet feeding., Results: A tamoxifen-induced EC-specific Enpp2 knockout decreased atherosclerosis, accumulation of lesional macrophages, monocyte adhesion, and expression of endothelial CXCL (C-X-C motif chemokine ligand) 1 in male and female Apoe-/- mice. In vitro, ENPP2 mediated the mildly oxidized LDL (low-density lipoprotein)-induced expression of CXCL1 in aortic ECs by generating LPA20:4, palmitoyl-LPA (LPA16:0), and oleoyl-LPA (LPA18:1). ENPP2 and its activity were detected on the endothelial surface by confocal imaging. The expression of endothelial Enpp2 established a strong correlation between the plasma levels of LPA16:0, stearoyl-LPA (LPA18:0), and LPA18:1 and plaque size and a strong negative correlation between the LPA levels and ENPP2 activity in the plasma. Moreover, endothelial Enpp2 knockout increased the weight of high-fat diet-fed male Apoe-/- mice., Conclusions: We demonstrated that the expression of ENPP2 in ECs promotes atherosclerosis and endothelial inflammation in a sex-independent manner. This might be due to the generation of LPA20:4, LPA16:0, and LPA18:1 from mildly oxidized lipoproteins on the endothelial surface.- Published
- 2022
- Full Text
- View/download PDF
13. Murine bone marrow macrophages and human monocytes do not express atypical chemokine receptor 1.
- Author
-
Rot A, Gutjahr JC, Biswas A, Aslani M, Hub E, Thiriot A, von Andrian UH, Megens RTA, Weber C, and Duchene J
- Subjects
- Animals, Bone Marrow Cells, Humans, Macrophages, Mice, Receptors, Chemokine, Bone Marrow, Monocytes
- Abstract
Competing Interests: Declaration of interests The authors declare no competing interests.
- Published
- 2022
- Full Text
- View/download PDF
14. Endothelial ACKR3 drives atherosclerosis by promoting immune cell adhesion to vascular endothelium.
- Author
-
Gencer S, Döring Y, Jansen Y, Bayasgalan S, Yan Y, Bianchini M, Cimen I, Müller M, Peters LJF, Megens RTA, von Hundelshausen P, Duchene J, Lemnitzer P, Soehnlein O, Weber C, and van der Vorst EPC
- Subjects
- Animals, Cell Adhesion, Endothelial Cells metabolism, Endothelium, Vascular metabolism, Inflammation metabolism, Mice, Mice, Knockout, ApoE, Transcription Factor RelA metabolism, Atherosclerosis metabolism, Plaque, Atherosclerotic metabolism, Plaque, Atherosclerotic pathology, Receptors, CXCR metabolism
- Abstract
Atherosclerosis is the foundation of potentially fatal cardiovascular diseases and it is characterized by plaque formation in large arteries. Current treatments aimed at reducing atherosclerotic risk factors still allow room for a large residual risk; therefore, novel therapeutic candidates targeting inflammation are needed. The endothelium is the starting point of vascular inflammation underlying atherosclerosis and we could previously demonstrate that the chemokine axis CXCL12-CXCR4 plays an important role in disease development. However, the role of ACKR3, the alternative and higher affinity receptor for CXCL12 remained to be elucidated. We studied the role of arterial ACKR3 in atherosclerosis using western diet-fed Apoe
-/- mice lacking Ackr3 in arterial endothelial as well as smooth muscle cells. We show for the first time that arterial endothelial deficiency of ACKR3 attenuates atherosclerosis as a result of diminished arterial adhesion as well as invasion of immune cells. ACKR3 silencing in inflamed human coronary artery endothelial cells decreased adhesion molecule expression, establishing an initial human validation of ACKR3's role in endothelial adhesion. Concomitantly, ACKR3 silencing downregulated key mediators in the MAPK pathway, such as ERK1/2, as well as the phosphorylation of the NF-kB p65 subunit. Endothelial cells in atherosclerotic lesions also revealed decreased phospho-NF-kB p65 expression in ACKR3-deficient mice. Lack of smooth muscle cell-specific as well as hematopoietic ACKR3 did not impact atherosclerosis in mice. Collectively, our findings indicate that arterial endothelial ACKR3 fuels atherosclerosis by mediating endothelium-immune cell adhesion, most likely through inflammatory MAPK and NF-kB pathways., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
15. Neuroimmune cardiovascular interfaces control atherosclerosis.
- Author
-
Mohanta SK, Peng L, Li Y, Lu S, Sun T, Carnevale L, Perrotta M, Ma Z, Förstera B, Stanic K, Zhang C, Zhang X, Szczepaniak P, Bianchini M, Saeed BR, Carnevale R, Hu D, Nosalski R, Pallante F, Beer M, Santovito D, Ertürk A, Mettenleiter TC, Klupp BG, Megens RTA, Steffens S, Pelisek J, Eckstein HH, Kleemann R, Habenicht L, Mallat Z, Michel JB, Bernhagen J, Dichgans M, D'Agostino G, Guzik TJ, Olofsson PS, Yin C, Weber C, Lembo G, Carnevale D, and Habenicht AJR
- Subjects
- Animals, Disease Progression, Ganglia, Spinal, Ganglia, Sympathetic, Mice, Neurons physiology, Atherosclerosis prevention & control, Plaque, Atherosclerotic prevention & control
- Abstract
Atherosclerotic plaques develop in the inner intimal layer of arteries and can cause heart attacks and strokes
1 . As plaques lack innervation, the effects of neuronal control on atherosclerosis remain unclear. However, the immune system responds to plaques by forming leukocyte infiltrates in the outer connective tissue coat of arteries (the adventitia)2-6 . Here, because the peripheral nervous system uses the adventitia as its principal conduit to reach distant targets7-9 , we postulated that the peripheral nervous system may directly interact with diseased arteries. Unexpectedly, widespread neuroimmune cardiovascular interfaces (NICIs) arose in mouse and human atherosclerosis-diseased adventitia segments showed expanded axon networks, including growth cones at axon endings near immune cells and media smooth muscle cells. Mouse NICIs established a structural artery-brain circuit (ABC): abdominal adventitia nociceptive afferents10-14 entered the central nervous system through spinal cord T6 -T13 dorsal root ganglia and were traced to higher brain regions, including the parabrachial and central amygdala neurons; and sympathetic efferent neurons projected from medullary and hypothalamic neurons to the adventitia through spinal intermediolateral neurons and both coeliac and sympathetic chain ganglia. Moreover, ABC peripheral nervous system components were activated: splenic sympathetic and coeliac vagus nerve activities increased in parallel to disease progression, whereas coeliac ganglionectomy led to the disintegration of adventitial NICIs, reduced disease progression and enhanced plaque stability. Thus, the peripheral nervous system uses NICIs to assemble a structural ABC, and therapeutic intervention in the ABC attenuates atherosclerosis., (© 2022. The Author(s), under exclusive licence to Springer Nature Limited.)- Published
- 2022
- Full Text
- View/download PDF
16. Targeting platelet-derived CXCL12 impedes arterial thrombosis.
- Author
-
Leberzammer J, Agten SM, Blanchet X, Duan R, Ippel H, Megens RTA, Schulz C, Aslani M, Duchene J, Döring Y, Jooss NJ, Zhang P, Brandl R, Stark K, Siess W, Jurk K, Heemskerk JWM, Hackeng TM, Mayo KH, Weber C, and von Hundelshausen P
- Subjects
- Agammaglobulinaemia Tyrosine Kinase metabolism, Animals, Chemokine CXCL12 metabolism, Collagen metabolism, Mice, Platelet Activation, Platelet Aggregation, Platelet Glycoprotein GPIIb-IIIa Complex metabolism, Blood Platelets metabolism, Thrombosis metabolism
- Abstract
The prevention and treatment of arterial thrombosis continue to be clinically challenging, and understanding the relevant molecular mechanisms in detail may facilitate the quest to identify novel targets and therapeutic approaches that improve protection from ischemic and bleeding events. The chemokine CXCL12 augments collagen-induced platelet aggregation by activating its receptor CXCR4. Here we show that inhibition of CXCR4 attenuates platelet aggregation induced by collagen or human plaque homogenate under static and arterial flow conditions by antagonizing the action of platelet-secreted CXCL12. We further show that platelet-specific CXCL12 deficiency in mice limits arterial thrombosis by affecting thrombus growth and stability without increasing tail bleeding time. Accordingly, neointimal lesion formation after carotid artery injury was attenuated in these mice. Mechanistically, CXCL12 activated via CXCR4 a signaling cascade involving Bruton's tyrosine kinase (Btk) that led to integrin αIIbβ3 activation, platelet aggregation, and granule release. The heterodimeric interaction between CXCL12 and CCL5 can inhibit CXCL12-mediated effects as mimicked by CCL5-derived peptides such as [VREY]4. An improved variant of this peptide, i[VREY]4, binds to CXCL12 in a complex with CXCR4 on the surface of activated platelets, thereby inhibiting Btk activation and preventing platelet CXCL12-dependent arterial thrombosis. In contrast to standard antiplatelet therapies such as aspirin or P2Y12 inhibition, i[VREY]4 reduced CXCL12-induced platelet aggregation and yet did not prolong in vitro bleeding time. We provide evidence that platelet-derived CXCL12 is involved in arterial thrombosis and can be specifically targeted by peptides that harbor potential therapeutic value against atherothrombosis., (© 2022 by The American Society of Hematology.)
- Published
- 2022
- Full Text
- View/download PDF
17. Improved Quantification of Cell Density in the Arterial Wall-A Novel Nucleus Splitting Approach Applied to 3D Two-Photon Laser-Scanning Microscopy.
- Author
-
van der Laan KWF, Reesink KD, van der Bruggen MM, Jaminon AMG, Schurgers LJ, Megens RTA, Huberts W, Delhaas T, and Spronck B
- Abstract
Accurate information on vascular smooth muscle cell (VSMC) content, orientation, and distribution in blood vessels is indispensable to increase understanding of arterial remodeling and to improve modeling of vascular biomechanics. We have previously proposed an analysis method to automatically characterize VSMC orientation and transmural distribution in murine carotid arteries under well-controlled biomechanical conditions. However, coincident nuclei, erroneously detected as one large nucleus, were excluded from the analysis, hampering accurate VSMC content characterization and distorting transmural distributions. In the present study, therefore, we aim to (1) improve the previous method by adding a "nucleus splitting" procedure to split coinciding nuclei, (2) evaluate the accuracy of this novel method, and (3) test this method in a mouse model of VSMC apoptosis. After euthanasia, carotid arteries from SM22α-hDTR Apoe
-/- and control Apoe-/- mice were bluntly dissected, excised, mounted in a biaxial biomechanical tester and brought to in vivo axial stretch and a pressure of 100 mmHg. Nuclei and elastin fibers were then stained using Syto-41 and Eosin-Y, respectively, and imaged using 3D two-photon laser scanning microscopy. Nuclei were segmented from images and coincident nuclei were split. The nucleus splitting procedure determines the likelihood that voxel pairs within coincident nuclei belong to the same nucleus and utilizes these likelihoods to identify individual nuclei using spectral clustering. Manual nucleus counts were used as a reference to assess the performance of our splitting procedure. Before and after splitting, automatic nucleus counts differed -26.6 ± 9.90% ( p < 0.001) and -1.44 ± 7.05% ( p = 0.467) from the manual reference, respectively. Whereas the slope of the relative difference between the manual and automated counts as a function of the manual count was significantly negative before splitting ( p = 0.008), this slope became insignificant after splitting ( p = 0.653). Smooth muscle apoptosis led to a 33.7% decrease in VSMC density ( p = 0.008). Nucleus splitting improves the accuracy of automated cell content quantification in murine carotid arteries and overcomes the progressively worsening problem of coincident nuclei with increasing cell content in vessels. The presented image analysis framework provides a robust tool to quantify cell content, orientation, shape, and distribution in vessels to inform experimental and advanced computational studies on vascular structure and function., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 van der Laan, Reesink, van der Bruggen, Jaminon, Schurgers, Megens, Huberts, Delhaas and Spronck.)- Published
- 2022
- Full Text
- View/download PDF
18. Tissue Clearing Approaches in Atherosclerosis.
- Author
-
Sun T, Li Y, Förstera B, Stanic K, Lu S, Steffens S, Yin C, Ertürk A, Megens RTA, Weber C, Habenicht A, and Mohanta SK
- Subjects
- Arteries, Humans, Microscopy, Atherosclerosis, Imaging, Three-Dimensional methods
- Abstract
Recent advances in cardiovascular research have led to a more comprehensive understanding of molecular mechanisms of atherosclerosis. It has become apparent that the disease involves three layers of the arterial wall: the intima, the media, and a connective tissue coat termed the adventitia. It is also now appreciated that arteries are surrounded by adipose and neuronal tissues. In addition, adjacent to and within the adventitia, arteries are embedded in a loose connective tissue containing blood vessels (vasa vasora) and lymph vessels, artery-draining lymph nodes and components of the peripheral nervous system, including periarterial nerves and ganglia. During atherogenesis, each of these tissues undergoes marked structural and cellular alterations. We propose that a better understanding of these cell-cell and cell-tissue interactions may considerably advance our understanding of cardiovascular disease pathogenesis. Methods to acquire subcellular optical access to the intact tissues surrounding healthy and diseased arteries are urgently needed to achieve these aims. Tissue clearing is a landmark next-generation, three-dimensional (3D) microscopy technique that allows to image large-scale hitherto inaccessible intact deep tissue compartments. It allows for detailed reconstructions of arteries by a combination of labelling, clearing, advanced microscopies and other imaging and data-analysis tools. Here, we describe two distinct tissue clearing protocols; solvent-based modified three-dimensional imaging of solvent-cleared organs (3DISCO) clearing and another using aqueous-based 2,2'-thiodiethanol (TDE) clearing, both of which complement each other., (© 2022. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2022
- Full Text
- View/download PDF
19. T cells armed with C-X-C chemokine receptor type 6 enhance adoptive cell therapy for pancreatic tumours.
- Author
-
Lesch S, Blumenberg V, Stoiber S, Gottschlich A, Ogonek J, Cadilha BL, Dantes Z, Rataj F, Dorman K, Lutz J, Karches CH, Heise C, Kurzay M, Larimer BM, Grassmann S, Rapp M, Nottebrock A, Kruger S, Tokarew N, Metzger P, Hoerth C, Benmebarek MR, Dhoqina D, Grünmeier R, Seifert M, Oener A, Umut Ö, Joaquina S, Vimeux L, Tran T, Hank T, Baba T, Huynh D, Megens RTA, Janssen KP, Jastroch M, Lamp D, Ruehland S, Di Pilato M, Pruessmann JN, Thomas M, Marr C, Ormanns S, Reischer A, Hristov M, Tartour E, Donnadieu E, Rothenfusser S, Duewell P, König LM, Schnurr M, Subklewe M, Liss AS, Halama N, Reichert M, Mempel TR, Endres S, and Kobold S
- Subjects
- Animals, Cell- and Tissue-Based Therapy, Mesothelin, Mice, Receptors, Chemokine genetics, Immunotherapy, Adoptive, Pancreatic Neoplasms therapy, Receptors, CXCR6 metabolism, T-Lymphocytes
- Abstract
The efficacy of adoptive cell therapy for solid tumours is hampered by the poor accumulation of the transferred T cells in tumour tissue. Here, we show that forced expression of C-X-C chemokine receptor type 6 (whose ligand is highly expressed by human and murine pancreatic cancer cells and tumour-infiltrating immune cells) in antigen-specific T cells enhanced the recognition and lysis of pancreatic cancer cells and the efficacy of adoptive cell therapy for pancreatic cancer. In mice with subcutaneous pancreatic tumours treated with T cells with either a transgenic T-cell receptor or a murine chimeric antigen receptor targeting the tumour-associated antigen epithelial cell adhesion molecule, and in mice with orthotopic pancreatic tumours or patient-derived xenografts treated with T cells expressing a chimeric antigen receptor targeting mesothelin, the T cells exhibited enhanced intratumoral accumulation, exerted sustained anti-tumoral activity and prolonged animal survival only when co-expressing C-X-C chemokine receptor type 6. Arming tumour-specific T cells with tumour-specific chemokine receptors may represent a promising strategy for the realization of adoptive cell therapy for solid tumours., (© 2021. The Author(s), under exclusive licence to Springer Nature Limited.)
- Published
- 2021
- Full Text
- View/download PDF
20. MicroRNA-21 Controls Circadian Regulation of Apoptosis in Atherosclerotic Lesions.
- Author
-
Schober A, Blay RM, Saboor Maleki S, Zahedi F, Winklmaier AE, Kakar MY, Baatsch IM, Zhu M, Geißler C, Fusco AE, Eberlein A, Li N, Megens RTA, Banafsche R, Kumbrink J, Weber C, and Nazari-Jahantigh M
- Subjects
- Animals, Apoptosis physiology, Atherosclerosis genetics, Atherosclerosis pathology, Disease Models, Animal, Humans, Mice, Mice, Inbred C57BL, Atherosclerosis metabolism, MicroRNAs metabolism
- Abstract
Background: The necrotic core partly formed by ineffective efferocytosis increases the risk of an atherosclerotic plaque rupture. Microribonucleic acids contribute to necrotic core formation by regulating efferocytosis and macrophage apoptosis. Atherosclerotic plaque rupture occurs at increased frequency in the early morning, indicating diurnal changes in plaque vulnerability. Although circadian rhythms play a role in atherosclerosis, the molecular clock output pathways that control plaque composition and rupture susceptibility are unclear., Methods: Circadian gene expression, necrotic core size, apoptosis, and efferocytosis in aortic lesions were investigated at different times of the day in Apoe
-/- Mir21+/+ mice and Apoe-/- Mir21-/- mice after consumption of a high-fat diet for 12 weeks. Genome-wide gene expression and lesion formation were analyzed in bone marrow-transplanted mice. Diurnal changes in apoptosis and clock gene expression were determined in human atherosclerotic lesions., Results: The expression of molecular clock genes, lesional apoptosis, and necrotic core size were diurnally regulated in Apoe-/- mice. Efferocytosis did not match the diurnal increase in apoptosis at the beginning of the active phase. However, in parallel with apoptosis, expression levels of oscillating Mir21 strands decreased in the mouse atherosclerotic aorta. Mir21 knockout abolished circadian regulation of apoptosis and reduced necrotic core size but did not affect core clock gene expression. Further, Mir21 knockout upregulated expression of proapoptotic Xaf1 (XIAP-associated factor 1) in the atherosclerotic aorta, which abolished circadian expression of Xaf1. The antiapoptotic effect of Mir21 was mediated by noncanonical targeting of Xaf1 through both Mir21 strands. Mir21 knockout in bone marrow cells also reduced atherosclerosis and necrotic core size. Circadian regulation of clock gene expression was confirmed in human atherosclerotic lesions. Apoptosis oscillated diurnally in phase with XAF1 expression, demonstrating an early morning peak antiphase to that of the Mir21 strands., Conclusions: Our findings suggest that the molecular clock in atherosclerotic lesions induces a diurnal rhythm of apoptosis regulated by circadian Mir21 expression in macrophages that is not matched by efferocytosis, thus increasing the size of the necrotic core.- Published
- 2021
- Full Text
- View/download PDF
21. Cell-specific and divergent roles of the CD40L-CD40 axis in atherosclerotic vascular disease.
- Author
-
Lacy M, Bürger C, Shami A, Ahmadsei M, Winkels H, Nitz K, van Tiel CM, Seijkens TTP, Kusters PJH, Karshovka E, Prange KHM, Wu Y, Brouns SLN, Unterlugauer S, Kuijpers MJE, Reiche ME, Steffens S, Edsfeldt A, Megens RTA, Heemskerk JWM, Goncalves I, Weber C, Gerdes N, Atzler D, and Lutgens E
- Subjects
- Animals, Blood Platelets metabolism, CD4-Positive T-Lymphocytes cytology, Cardiovascular Diseases pathology, Dendritic Cells immunology, Mice, Mice, Knockout, Myocytes, Smooth Muscle cytology, Signal Transduction physiology, Thrombosis pathology, Atherosclerosis pathology, CD4-Positive T-Lymphocytes metabolism, CD40 Antigens metabolism, CD40 Ligand metabolism, Interferon-gamma metabolism, Plaque, Atherosclerotic pathology
- Abstract
Atherosclerosis is a major underlying cause of cardiovascular disease. Previous studies showed that inhibition of the co-stimulatory CD40 ligand (CD40L)-CD40 signaling axis profoundly attenuates atherosclerosis. As CD40L exerts multiple functions depending on the cell-cell interactions involved, we sought to investigate the function of the most relevant CD40L-expressing cell types in atherosclerosis: T cells and platelets. Atherosclerosis-prone mice with a CD40L-deficiency in CD4
+ T cells display impaired Th1 polarization, as reflected by reduced interferon-γ production, and smaller atherosclerotic plaques containing fewer T-cells, smaller necrotic cores, an increased number of smooth muscle cells and thicker fibrous caps. Mice with a corresponding CD40-deficiency in CD11c+ dendritic cells phenocopy these findings, suggesting that the T cell-dendritic cell CD40L-CD40 axis is crucial in atherogenesis. Accordingly, sCD40L/sCD40 and interferon-γ concentrations in carotid plaques and plasma are positively correlated in patients with cerebrovascular disease. Platelet-specific deficiency of CD40L does not affect atherogenesis but ameliorates atherothrombosis. Our results establish divergent and cell-specific roles of CD40L-CD40 in atherosclerosis, which has implications for therapeutic strategies targeting this pathway.- Published
- 2021
- Full Text
- View/download PDF
22. A MIF-Derived Cyclopeptide that Inhibits MIF Binding and Atherogenic Signaling via the Chemokine Receptor CXCR2.
- Author
-
Krammer C, Kontos C, Dewor M, Hille K, Dalla Volta B, El Bounkari O, Taş K, Sinitski D, Brandhofer M, Megens RTA, Weber C, Schultz JR, Bernhagen J, and Kapurniotu A
- Subjects
- Amino Acid Sequence, Animals, Cell Adhesion, Fluoresceins chemistry, HEK293 Cells, Humans, Leukocytes chemistry, Leukocytes cytology, Leukocytes metabolism, Mice, Mice, Inbred C57BL, Peptides, Cyclic blood, Peptides, Cyclic chemistry, Protein Binding, Protein Stability, Receptors, Interleukin-8B antagonists & inhibitors, Spectrometry, Fluorescence, Sulfonic Acids chemistry, Macrophage Migration-Inhibitory Factors chemistry, Peptides, Cyclic metabolism, Receptors, Interleukin-8B metabolism
- Abstract
Macrophage migration inhibitory factor (MIF) is an inflammatory cytokine and atypical chemokine with a key role in inflammatory diseases including atherosclerosis. Key atherogenic functions of MIF are mediated by noncognate interaction with the chemokine receptor CXCR2. The MIF N-like loop comprising the sequence 47-56 is an important structural determinant of the MIF/CXCR2 interface and MIF(47-56) blocks atherogenic MIF activities. However, the mechanism and critical structure-activity information within this sequence have remained elusive. Here, we show that MIF(47-56) directly binds to CXCR2 to compete with MIF receptor activation. By using alanine scanning, essential and dispensable residues were identified. Moreover, MIF(cyclo10), a designed cyclized variant of MIF(47-56), inhibited key inflammatory and atherogenic MIF activities in vitro and in vivo/ex vivo, and exhibited strongly improved resistance to proteolytic degradation in human plasma in vitro, thus suggesting that it could serve as a promising basis for MIF-derived anti-atherosclerotic peptides., (© 2020 The Authors. ChemBioChem published by Wiley-VCH GmbH.)
- Published
- 2021
- Full Text
- View/download PDF
23. An integrated set-up for ex vivo characterisation of biaxial murine artery biomechanics under pulsatile conditions.
- Author
-
van der Bruggen MM, Reesink KD, Spronck PJM, Bitsch N, Hameleers J, Megens RTA, Schalkwijk CG, Delhaas T, and Spronck B
- Subjects
- Animals, Biomechanical Phenomena, Blood Flow Velocity, Mice, Pulse Wave Analysis, Vascular Stiffness, Blood Pressure, Carotid Arteries, Models, Cardiovascular
- Abstract
Ex vivo characterisation of arterial biomechanics enables detailed discrimination of the various cellular and extracellular contributions to arterial stiffness. However, ex vivo biomechanical studies are commonly performed under quasi-static conditions, whereas dynamic biomechanical behaviour (as relevant in vivo) may differ substantially. Hence, we aim to (1) develop an integrated set-up for quasi-static and dynamic biaxial biomechanical testing, (2) quantify set-up reproducibility, and (3) illustrate the differences in measured arterial stiffness between quasi-static and dynamic conditions. Twenty-two mouse carotid arteries were mounted between glass micropipettes and kept fully vasodilated. While recording pressure, axial force (F), and inner diameter, arteries were exposed to (1) quasi-static pressure inflation from 0 to 200 mmHg; (2) 300 bpm dynamic pressure inflation (peaking at 80/120/160 mmHg); and (3) axial stretch (λ
z ) variation at constant pressures of 10/60/100/140/200 mmHg. Measurements were performed in duplicate. Single-point pulse wave velocities (PWV; Bramwell-Hill) and axial stiffness coefficients (cax = dF/dλz ) were calculated at the in vivo value of λz . Within-subject coefficients of variation were ~ 20%. Dynamic PWVs were consistently higher than quasi-static PWVs (p < 0.001); cax increased with increasing pressure. We demonstrated the feasibility of ex vivo biomechanical characterisation of biaxially-loaded murine carotid arteries under pulsatile conditions, and quantified reproducibility allowing for well-powered future study design.- Published
- 2021
- Full Text
- View/download PDF
24. Autophagy unleashes noncanonical microRNA functions.
- Author
-
Santovito D, Egea V, Bidzhekov K, Natarelli L, Mourão A, Blanchet X, Wichapong K, Aslani M, Brunßen C, Horckmans M, Hristov M, Geerlof A, Lutgens E, Daemen MJAP, Hackeng T, Ries C, Chavakis T, Morawietz H, Naumann R, Hundelshausen PV, Steffens S, Duchêne J, Megens RTA, Sattler M, and Weber C
- Subjects
- Autophagy genetics, Caspase 3, Endothelial Cells, Humans, Atherosclerosis, MicroRNAs genetics
- Abstract
MicroRNAs (miRNAs) are post-transcriptional regulators of gene expression which act by guiding AGO (argonaute) proteins to target RNA transcripts in the RNA-induced silencing complex (RISC). This macromolecular complex includes multiple additional components ( e.g ., TNRC6A) that allow for interaction with enzymes mediating inhibition of translation or RNA decay. However, miRNAs also reside in low-molecular weight complexes without being engaged in target repression, and their function in this context is largely unknown. Our recent findings show that endothelial cells exposed to protective high-shear stress or MTORC inhibition activate the macroautophagy/autophagy machinery to sustain viability by promoting differential trafficking of MIR126 strands and by enabling unconventional features of MIR126-5p . Whereas MIR126-3p is degraded upon autophagy activation, MIR126-5p interacts with the RNA-binding protein MEX3A to form a ternary complex with AGO2. This complex forms on the autophagosomal surface and facilitates its nuclear localization. Once in the nucleus, MIR126-5p dissociates from AGO2 and establishes aptamer-like interactions with the effector CASP3 (caspase 3). The binding to MIR126-5p prevents dimerization and proper active site formation of CASP3, thus inhibiting proteolytic activity and limiting apoptosis. Disrupting this pathway in vivo by genetic deletion of Mex3a or by specific deficiency of endothelial autophagy aggravates endothelial apoptosis and exacerbates the progression of atherosclerosis. The direct inhibition of CASP3 by MIR126-5p reveals a non-canonical mechanism by which miRNAs can modulate protein function and mediate the autophagy-apoptosis crosstalk.
- Published
- 2020
- Full Text
- View/download PDF
25. Designed CXCR4 mimic acts as a soluble chemokine receptor that blocks atherogenic inflammation by agonist-specific targeting.
- Author
-
Kontos C, El Bounkari O, Krammer C, Sinitski D, Hille K, Zan C, Yan G, Wang S, Gao Y, Brandhofer M, Megens RTA, Hoffmann A, Pauli J, Asare Y, Gerra S, Bourilhon P, Leng L, Eckstein HH, Kempf WE, Pelisek J, Gokce O, Maegdefessel L, Bucala R, Dichgans M, Weber C, Kapurniotu A, and Bernhagen J
- Subjects
- Aged, Animals, Antigens, CD metabolism, Atherosclerosis genetics, Atherosclerosis pathology, Atherosclerosis surgery, Binding Sites, Carotid Artery, Common pathology, Carotid Artery, Common surgery, Chemokine CXCL12 metabolism, Crystallography, X-Ray, Disease Models, Animal, Drug Design, Drug Evaluation, Preclinical, Endarterectomy, Carotid, Female, Humans, Intramolecular Oxidoreductases metabolism, Macrophage Migration-Inhibitory Factors metabolism, Male, Mice, Mice, Knockout, ApoE, Middle Aged, Peptide Fragments therapeutic use, Receptors, CXCR4 chemistry, Receptors, CXCR4 ultrastructure, Sialyltransferases metabolism, Signal Transduction drug effects, Atherosclerosis drug therapy, Intramolecular Oxidoreductases antagonists & inhibitors, Macrophage Migration-Inhibitory Factors antagonists & inhibitors, Peptide Fragments pharmacology, Receptors, CXCR4 metabolism
- Abstract
Targeting a specific chemokine/receptor axis in atherosclerosis remains challenging. Soluble receptor-based strategies are not established for chemokine receptors due to their discontinuous architecture. Macrophage migration-inhibitory factor (MIF) is an atypical chemokine that promotes atherosclerosis through CXC-motif chemokine receptor-4 (CXCR4). However, CXCR4/CXCL12 interactions also mediate atheroprotection. Here, we show that constrained 31-residue-peptides ('msR4Ms') designed to mimic the CXCR4-binding site to MIF, selectively bind MIF with nanomolar affinity and block MIF/CXCR4 without affecting CXCL12/CXCR4. We identify msR4M-L1, which blocks MIF- but not CXCL12-elicited CXCR4 vascular cell activities. Its potency compares well with established MIF inhibitors, whereas msR4M-L1 does not interfere with cardioprotective MIF/CD74 signaling. In vivo-administered msR4M-L1 enriches in atherosclerotic plaques, blocks arterial leukocyte adhesion, and inhibits atherosclerosis and inflammation in hyperlipidemic Apoe
-/- mice in vivo. Finally, msR4M-L1 binds to MIF in plaques from human carotid-endarterectomy specimens. Together, we establish an engineered GPCR-ectodomain-based mimicry principle that differentiates between disease-exacerbating and -protective pathways and chemokine-selectively interferes with atherosclerosis.- Published
- 2020
- Full Text
- View/download PDF
26. Histone Deacetylase 9 Activates IKK to Regulate Atherosclerotic Plaque Vulnerability.
- Author
-
Asare Y, Campbell-James TA, Bokov Y, Yu LL, Prestel M, El Bounkari O, Roth S, Megens RTA, Straub T, Thomas K, Yan G, Schneider M, Ziesch N, Tiedt S, Silvestre-Roig C, Braster Q, Huang Y, Schneider M, Malik R, Haffner C, Liesz A, Soehnlein O, Bernhagen J, and Dichgans M
- Subjects
- Acetylation, Aged, Aged, 80 and over, Animals, Arteries drug effects, Arteries pathology, Atherosclerosis drug therapy, Atherosclerosis genetics, Atherosclerosis pathology, CX3C Chemokine Receptor 1 genetics, CX3C Chemokine Receptor 1 metabolism, Cytokines metabolism, Disease Models, Animal, Endothelial Cells enzymology, Endothelial Cells pathology, Enzyme Activation, Female, Fibrosis, Histone Deacetylase Inhibitors pharmacology, Histone Deacetylases genetics, Humans, I-kappa B Kinase genetics, Inflammation Mediators metabolism, Leukocyte Rolling, Macrophages enzymology, Macrophages pathology, Male, Mice, Knockout, ApoE, Middle Aged, Monocytes enzymology, Monocytes pathology, Protein Binding, Repressor Proteins antagonists & inhibitors, Repressor Proteins genetics, Signal Transduction, Arteries enzymology, Atherosclerosis enzymology, Histone Deacetylases metabolism, I-kappa B Kinase metabolism, Plaque, Atherosclerotic, Repressor Proteins metabolism
- Abstract
Rationale: Arterial inflammation manifested as atherosclerosis is the leading cause of mortality worldwide. Genome-wide association studies have identified a prominent role of HDAC (histone deacetylase)-9 in atherosclerosis and its clinical complications including stroke and myocardial infarction., Objective: To determine the mechanisms linking HDAC9 to these vascular pathologies and explore its therapeutic potential for atheroprotection., Methods and Results: We studied the effects of Hdac9 on features of plaque vulnerability using bone marrow reconstitution experiments and pharmacological targeting with a small molecule inhibitor in hyperlipidemic mice. We further used 2-photon and intravital microscopy to study endothelial activation and leukocyte-endothelial interactions. We show that hematopoietic Hdac9 deficiency reduces lesional macrophage content while increasing fibrous cap thickness thus conferring plaque stability. We demonstrate that HDAC9 binds to IKK (inhibitory kappa B kinase)-α and β, resulting in their deacetylation and subsequent activation, which drives inflammatory responses in both macrophages and endothelial cells. Pharmacological inhibition of HDAC9 with the class IIa HDAC inhibitor TMP195 attenuates lesion formation by reducing endothelial activation and leukocyte recruitment along with limiting proinflammatory responses in macrophages. Transcriptional profiling using RNA sequencing revealed that TMP195 downregulates key inflammatory pathways consistent with inhibitory effects on IKKβ. TMP195 mitigates the progression of established lesions and inhibits the infiltration of inflammatory cells. Moreover, TMP195 diminishes features of plaque vulnerability and thereby enhances plaque stability in advanced lesions. Ex vivo treatment of monocytes from patients with established atherosclerosis reduced the production of inflammatory cytokines including IL (interleukin)-1β and IL-6., Conclusions: Our findings identify HDAC9 as a regulator of atherosclerotic plaque stability and IKK activation thus providing a mechanistic explanation for the prominence of HDAC9 as a vascular risk locus in genome-wide association studies. Its therapeutic inhibition may provide a potent lever to alleviate vascular inflammation. Graphical Abstract: A graphical abstract is available for this article.
- Published
- 2020
- Full Text
- View/download PDF
27. Noncanonical inhibition of caspase-3 by a nuclear microRNA confers endothelial protection by autophagy in atherosclerosis.
- Author
-
Santovito D, Egea V, Bidzhekov K, Natarelli L, Mourão A, Blanchet X, Wichapong K, Aslani M, Brunßen C, Horckmans M, Hristov M, Geerlof A, Lutgens E, Daemen MJAP, Hackeng T, Ries C, Chavakis T, Morawietz H, Naumann R, von Hundelshausen P, Steffens S, Duchêne J, Megens RTA, Sattler M, and Weber C
- Subjects
- Apoptosis, Autophagy, Caspase 3, Humans, Atherosclerosis genetics, MicroRNAs genetics
- Abstract
MicroRNAs (miRNAs) are versatile regulators of gene expression with profound implications for human disease including atherosclerosis, but whether they can exert posttranslational functions to control cell adaptation and whether such noncanonical features harbor pathophysiological relevance is unknown. Here, we show that miR-126-5p sustains endothelial integrity in the context of high shear stress and autophagy. Bound to argonaute-2 (Ago2), miR-126-5p forms a complex with Mex3a, which occurs on the surface of autophagic vesicles and guides its transport into the nucleus. Mutational studies and biophysical measurements demonstrate that Mex3a binds to the central U- and G-rich regions of miR-126-5p with nanomolar affinity via its two K homology domains. In the nucleus, miR-126-5p dissociates from Ago2 and binds to caspase-3 in an aptamer-like fashion with its seed sequence, preventing dimerization of the caspase and inhibiting its activity to limit apoptosis. The antiapoptotic effect of miR-126-5p outside of the RNA-induced silencing complex is important for endothelial integrity under conditions of high shear stress promoting autophagy: ablation of Mex3a or ATG5 in vivo attenuates nuclear import of miR-126-5p, aggravates endothelial apoptosis, and exacerbates atherosclerosis. In human plaques, we found reduced nuclear miR-126-5p and active caspase-3 in areas of disturbed flow. The direct inhibition of caspase-3 by nuclear miR-126-5p reveals a noncanonical mechanism by which miRNAs can modulate protein function., (Copyright © 2020 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)
- Published
- 2020
- Full Text
- View/download PDF
28. Glycoprotein VI is not a Functional Platelet Receptor for Fibrin Formed in Plasma or Blood.
- Author
-
Zhang D, Ebrahim M, Adler K, Blanchet X, Jamasbi J, Megens RTA, Uhland K, Ungerer M, Münch G, Deckmyn H, Weber C, Elia N, Lorenz R, and Siess W
- Subjects
- Agammaglobulinaemia Tyrosine Kinase blood, Agammaglobulinaemia Tyrosine Kinase physiology, Enzyme Activation, Fibrinogen metabolism, Hemorheology, Humans, Microscopy, Confocal methods, Plasma, Platelet Adhesiveness, Platelet Aggregation, Platelet Glycoprotein GPIb-IX Complex metabolism, Platelet Membrane Glycoproteins antagonists & inhibitors, Platelet Membrane Glycoproteins immunology, Protein Binding, Recombinant Proteins metabolism, Syk Kinase antagonists & inhibitors, Syk Kinase blood, Syk Kinase physiology, Thromboplastin metabolism, Blood Platelets metabolism, Fibrin metabolism, Platelet Membrane Glycoproteins physiology, Receptors, Peptide metabolism
- Abstract
Glycoprotein VI (GPVI), a platelet collagen receptor, is crucial in mediating atherothrombosis. Besides collagen, injured plaques expose tissue factor (TF) that triggers fibrin formation. Previous studies reported that GPVI also is a platelet receptor for fibrinogen and fibrin. We studied the effect of anti-GPVI antibodies and inhibitors of GPVI signaling kinases (Syk and Btk) on platelet adhesion and aggregate formation onto immobilized fibrinogen and different types of fibrin under arterial flow conditions. Fibrin was prepared from isolated fibrinogen ("pure fibrin"), recombinant fibrinogen ("recombinant fibrin"), or generated more physiologically from endogenous fibrinogen in plasma ("plasma fibrin") or by exposing TF-coated surfaces to flowing blood ("blood fibrin"). Inhibition of GPVI and Syk did not inhibit platelet adhesion and aggregate formation onto fibrinogen. In contrast anti-GPVI antibodies, inhibitors of Syk and Btk and the anti-GPIb antibody 6B4 inhibited platelet aggregate formation onto pure and recombinant fibrin. However, inhibition of GPVI and GPVI signaling did not significantly reduce platelet coverage of plasma fibrin and blood fibrin. Plasma fibrin contained many proteins incorporated during clot formation. Advanced optical imaging revealed plasma fibrin as a spongiform cushion with thicker, knotty, and long fibers and little activation of adhering platelets. Albumin intercalated in plasma fibrin fibers left only little space for platelet attachment. Pure fibrin was different showing a dense mesh of thin fibers with strongly activated platelets. We conclude that fibrin formed in plasma and blood contains plasma proteins shielding GPVI-activating epitopes. Our findings do not support a role of GPVI for platelet activation by physiologic fibrin., Competing Interests: K.A. and K.U. are employees of advanceCOR GmbH which produces the anti-GPVI antibodies. G.M. and M.U. are managing directors of advanceCOR GmbH and own shares of the company. The other authors report no conflict of interest., (Georg Thieme Verlag KG Stuttgart · New York.)
- Published
- 2020
- Full Text
- View/download PDF
29. Tick Saliva Protein Evasin-3 Allows for Visualization of Inflammation in Arteries through Interactions with CXC-Type Chemokines Deposited on Activated Endothelium.
- Author
-
Denisov SS, Heinzmann ACA, Vajen T, Vries MHM, Megens RTA, Suylen D, Koenen RR, Post MJ, Ippel JH, Hackeng TM, and Dijkgraaf I
- Subjects
- Animals, Carotid Artery Diseases metabolism, Glycosaminoglycans metabolism, Human Umbilical Vein Endothelial Cells metabolism, Humans, Inflammation diagnostic imaging, Inflammation metabolism, Mice, Arthropod Proteins metabolism, Carotid Artery Diseases diagnostic imaging, Chemokines, CXC metabolism, Endothelium, Vascular metabolism, Ticks
- Abstract
Atherosclerosis is one of the leading causes of mortality in developed and developing countries. The onset of atherosclerosis development is accompanied by overexpression of several inflammatory chemokines. Neutralization of these chemokines by chemokine-binding agents attenuates atherosclerosis progression. Here, we studied structural binding features of the tick protein Evasin-3 to chemokine (C-X-C motif) ligand 1 (CXCL1). We showed that Evasin-3-bound CXCL1 is unable to activate the CXCR2 receptor, but retains affinity to glycosaminoglycans. This observation was exploited to detect inflammation by visualizing a group of closely related CXC-type chemokines deposited on cell walls in human endothelial cells and murine carotid arteries by a fluorescent Evasin-3 conjugate. This work highlights the applicability of tick-derived chemokine-binding conjugates as a platform for the development of new agents for inflammation imaging.
- Published
- 2020
- Full Text
- View/download PDF
30. PD-L1 expression on nonclassical monocytes reveals their origin and immunoregulatory function.
- Author
-
Bianchini M, Duchêne J, Santovito D, Schloss MJ, Evrard M, Winkels H, Aslani M, Mohanta SK, Horckmans M, Blanchet X, Lacy M, von Hundelshausen P, Atzler D, Habenicht A, Gerdes N, Pelisek J, Ng LG, Steffens S, Weber C, and Megens RTA
- Subjects
- Abdominal Muscles immunology, Animals, Antibodies pharmacology, Bone Marrow immunology, Female, Femur, Immunoglobulin G immunology, Male, Mice, Inbred C57BL, Mice, Transgenic, T-Lymphocytes immunology, B7-H1 Antigen immunology, Monocytes immunology
- Abstract
The role of nonclassical monocytes (NCMs) in health and disease is emerging, but their location and function within tissues remain poorly explored. Imaging of NCMs has been limited by the lack of an established single NCM marker. Here, we characterize the immune checkpoint molecule PD-L1 (CD274) as an unequivocal marker for tracking NCMs in circulation and pinpoint their compartmentalized distribution in tissues by two-photon microscopy. Visualization of PD-L1
+ NCMs in relation to bone marrow vasculature reveals that conversion of classical monocytes into NCMs requires contact with endosteal vessels. Furthermore, PD-L1+ NCMs are present in tertiary lymphoid organs (TLOs) under inflammatory conditions in both mice and humans, and NCMs exhibit a PD-L1-dependent immunomodulatory function that promotes T cell apoptosis within TLOs. Our findings establish an unambiguous tool for the investigation of NCMs and shed light on their origin and function., (Copyright © 2019 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works.)- Published
- 2019
- Full Text
- View/download PDF
31. Externalized histone H4 orchestrates chronic inflammation by inducing lytic cell death.
- Author
-
Silvestre-Roig C, Braster Q, Wichapong K, Lee EY, Teulon JM, Berrebeh N, Winter J, Adrover JM, Santos GS, Froese A, Lemnitzer P, Ortega-Gómez A, Chevre R, Marschner J, Schumski A, Winter C, Perez-Olivares L, Pan C, Paulin N, Schoufour T, Hartwig H, González-Ramos S, Kamp F, Megens RTA, Mowen KA, Gunzer M, Maegdefessel L, Hackeng T, Lutgens E, Daemen M, von Blume J, Anders HJ, Nikolaev VO, Pellequer JL, Weber C, Hidalgo A, Nicolaes GAF, Wong GCL, and Soehnlein O
- Subjects
- Animals, Arteries pathology, Cell Membrane drug effects, Disease Models, Animal, Female, Histones antagonists & inhibitors, Mice, Mice, Inbred C57BL, Myocytes, Smooth Muscle pathology, Neutrophils cytology, Protein Binding drug effects, Atherosclerosis pathology, Cell Death, Cell Membrane metabolism, Histones metabolism, Inflammation metabolism, Inflammation pathology, Porosity
- Abstract
The perpetuation of inflammation is an important pathophysiological contributor to the global medical burden. Chronic inflammation is promoted by non-programmed cell death
1,2 ; however, how inflammation is instigated, its cellular and molecular mediators, and its therapeutic value are poorly defined. Here we use mouse models of atherosclerosis-a major underlying cause of mortality worldwide-to demonstrate that extracellular histone H4-mediated membrane lysis of smooth muscle cells (SMCs) triggers arterial tissue damage and inflammation. We show that activated lesional SMCs attract neutrophils, triggering the ejection of neutrophil extracellular traps that contain nuclear proteins. Among them, histone H4 binds to and lyses SMCs, leading to the destabilization of plaques; conversely, the neutralization of histone H4 prevents cell death of SMCs and stabilizes atherosclerotic lesions. Our data identify a form of cell death found at the core of chronic vascular disease that is instigated by leukocytes and can be targeted therapeutically.- Published
- 2019
- Full Text
- View/download PDF
32. Publisher Correction: ApoE attenuates unresolvable inflammation by complex formation with activated C1q.
- Author
-
Yin C, Ackermann S, Ma Z, Mohanta SK, Zhang C, Li Y, Nietzsche S, Westermann M, Peng L, Hu D, Bontha SV, Srikakulapu P, Beer M, Megens RTA, Steffens S, Hildner M, Halder LD, Eckstein HH, Pelisek J, Herms J, Roeber S, Arzberger T, Borodovsky A, Habenicht L, Binder CJ, Weber C, Zipfel PF, Skerka C, and Habenicht AJR
- Abstract
In the version of this article originally published, a sentence was erroneously included in the author contributions, and information regarding second shared authorship was missing from the author contributions. The following should not have been included in the author contributions: "C.W. and A.J.R.H. supervised the work presented in Figs. 1, 2, 5, 6; P.Z. and C.S. supervised the work presented in Figs. 3, 4." Additionally, this sentence should have appeared at the beginning of the author contributions: "These authors contributed equally: C.W., P.F.Z., C.S., and A.J.R.H." The errors have been corrected in the print, PDF and HTML versions of the article.
- Published
- 2019
- Full Text
- View/download PDF
33. ApoE attenuates unresolvable inflammation by complex formation with activated C1q.
- Author
-
Yin C, Ackermann S, Ma Z, Mohanta SK, Zhang C, Li Y, Nietzsche S, Westermann M, Peng L, Hu D, Bontha SV, Srikakulapu P, Beer M, Megens RTA, Steffens S, Hildner M, Halder LD, Eckstein HH, Pelisek J, Herms J, Roeber S, Arzberger T, Borodovsky A, Habenicht L, Binder CJ, Weber C, Zipfel PF, Skerka C, and Habenicht AJR
- Subjects
- Aged, Aged, 80 and over, Amyloid beta-Peptides immunology, Animals, Aorta immunology, Aorta pathology, Atherosclerosis immunology, Atherosclerosis pathology, Brain immunology, Brain pathology, Carotid Arteries immunology, Carotid Arteries pathology, Carotid Artery Diseases pathology, Choroid Plexus pathology, Cognitive Dysfunction pathology, Complement C5, Female, Humans, Leukocytes, Male, Mice, Knockout, ApoE, Microscopy, Fluorescence, Middle Aged, Plaque, Amyloid immunology, Plaque, Amyloid pathology, Protein Isoforms immunology, RNA, Small Interfering, Antigen-Antibody Complex immunology, Apolipoproteins E immunology, Carotid Artery Diseases immunology, Choroid Plexus immunology, Cognitive Dysfunction immunology, Complement C1q immunology, Complement Pathway, Classical immunology
- Abstract
Apolipoprotein-E (ApoE) has been implicated in Alzheimer's disease, atherosclerosis, and other unresolvable inflammatory conditions but a common mechanism of action remains elusive. We found in ApoE-deficient mice that oxidized lipids activated the classical complement cascade (CCC), resulting in leukocyte infiltration of the choroid plexus (ChP). All human ApoE isoforms attenuated CCC activity via high-affinity binding to the activated CCC-initiating C1q protein (K
D ~140-580 pM) in vitro, and C1q-ApoE complexes emerged as markers for ongoing complement activity of diseased ChPs, Aβ plaques, and atherosclerosis in vivo. C1q-ApoE complexes in human ChPs, Aβ plaques, and arteries correlated with cognitive decline and atherosclerosis, respectively. Treatment with small interfering RNA (siRNA) against C5, which is formed by all complement pathways, attenuated murine ChP inflammation, Aβ-associated microglia accumulation, and atherosclerosis. Thus, ApoE is a direct checkpoint inhibitor of unresolvable inflammation, and reducing C5 attenuates disease burden.- Published
- 2019
- Full Text
- View/download PDF
34. CD36-triggered cell invasion and persistent tissue colonization by tumor microvesicles during metastasis.
- Author
-
Pfeiler S, Thakur M, Grünauer P, Megens RTA, Joshi U, Coletti R, Samara V, Müller-Stoy G, Ishikawa-Ankerhold H, Stark K, Klingl A, Fröhlich T, Arnold GJ, Wörmann S, Bruns CJ, Algül H, Weber C, Massberg S, and Engelmann B
- Subjects
- Cell-Derived Microparticles pathology, Humans, Lysosomes pathology, Macrophages pathology, Neoplasm Invasiveness, Neoplasm Metastasis, Pancreatic Neoplasms pathology, THP-1 Cells, CD36 Antigens immunology, Cell-Derived Microparticles immunology, Lysosomes immunology, Macrophages immunology, Pancreatic Neoplasms immunology
- Abstract
Tumor microvesicles are a peculiar type of extracellular vesicles that circulate in the blood of patients with metastatic cancer. The itineraries and immune cell interactions of tumor microvesicles during the intravascular and extravascular stages of metastasis are largely unknown. We found that the lipid receptor CD36 is a major mediator of the engulfment of pancreatic tumor microvesicles by myeloid immune cells in vitro and critically samples circulating tumor microvesicles by resident liver macrophages in mice in vivo. Direct nanoscopic imaging of individual tumor microvesicles shows that the microvesicles rapidly decay during engulfment whereby their cargo is targeted concomitantly to the plasma membrane and the cytoplasm excluding lysosomal compartments. CD36 also promotes internalization of blood cell (nontumor) microvesicles, which involves endolysosomal pathways. A portion of tumor microvesicles circulating in the liver microcirculation traverses the vessel wall in a CD36-dependent way. Extravasated microvesicles colonize distinct perivascular Ly6C
- macrophages for at least 2 wk. Thus, the microvesicles are increasingly integrated into CD36-induced premetastatic cell clusters and enhance development of liver metastasis. Hence, promotion of metastasis by pancreatic tumor microvesicles is associated with CD36-regulated immune cell invasion and extravasation of microvesicles and persistent infiltration of specific tissue macrophages by microvesicle cargo.-Pfeiler, S., Thakur, M., Grünauer, P., Megens, R. T. A., Joshi, U., Coletti, R., Samara, V., Müller-Stoy, G., Ishikawa-Ankerhold, H., Stark, K., Klingl, A., Fröhlich, T., Arnold, G. J., Wörmann, S., Bruns, C. J., Algül, H., Weber, C., Massberg, S., Engelmann, B. CD36-triggered cell invasion and persistent tissue colonization by tumor microvesicles during metastasis.- Published
- 2019
- Full Text
- View/download PDF
35. The Actin Regulator Coronin-1A Modulates Platelet Shape Change and Consolidates Arterial Thrombosis.
- Author
-
Stocker TJ, Pircher J, Skenderi A, Ehrlich A, Eberle C, Megens RTA, Petzold T, Zhang Z, Walzog B, Müller-Taubenberger A, Weber C, Massberg S, Ishikawa-Ankerhold H, and Schulz C
- Subjects
- Actin Cytoskeleton metabolism, Animals, Cell Shape, Cells, Cultured, Chlorides metabolism, Cofilin 1 metabolism, Collagen metabolism, Ferric Compounds metabolism, Humans, Mice, Mice, Inbred C57BL, Mice, Knockout, Microfilament Proteins genetics, Protein Multimerization, Blood Platelets physiology, Infarction, Middle Cerebral Artery metabolism, Microfilament Proteins metabolism
- Abstract
Coronin-1A (Coro1A) belongs to a family of highly conserved actin-binding proteins that regulate cytoskeletal re-arrangement. In mammalians, Coro1A expression is most abundant in the haematopoietic lineage, where it regulates various cellular processes. The role of Coro1A in platelets has been previously unknown. Here, we identified Coro1A in human and mouse platelets. Genetic absence of Coro1A in mouse platelets inhibited agonist-induced actin polymerization and altered cofilin phosphoregulation, leading to a reduction in spreading and low-dose collagen induced aggregation. Furthermore, Coro1A-deficient mice displayed a defect in ferric chloride-induced arterial thrombosis with prolonged thrombus formation and reduced thrombus size. Immunofluorescence analysis revealed a less compact thrombus structure with reduced density of platelets and fibrinogen. In summary, Coro1A has a role in platelet biology with impact on spreading, aggregation and thrombosis., Competing Interests: None., (Georg Thieme Verlag KG Stuttgart · New York.)
- Published
- 2018
- Full Text
- View/download PDF
36. Mif-deficiency favors an atheroprotective autoantibody phenotype in atherosclerosis.
- Author
-
Schmitz C, Noels H, El Bounkari O, Straussfeld E, Megens RTA, Sternkopf M, Alampour-Rajabi S, Krammer C, Tilstam PV, Gerdes N, Bürger C, Kapurniotu A, Bucala R, Jankowski J, Weber C, and Bernhagen J
- Subjects
- Animals, Aorta metabolism, Apolipoproteins E metabolism, B-Lymphocytes metabolism, Cell Differentiation physiology, Female, Lipoproteins, LDL metabolism, Male, Mice, Mice, Inbred C57BL, Monocytes metabolism, Phenotype, Plaque, Atherosclerotic metabolism, Atherosclerosis metabolism, Autoantibodies metabolism, Intramolecular Oxidoreductases metabolism, Macrophage Migration-Inhibitory Factors metabolism
- Abstract
The inflammatory cytokine macrophage migration-inhibitory factor (MIF) promotes atherosclerosis via lesional monocyte and T-cell recruitment. B cells have emerged as important components in atherogenesis, but the interaction between MIF and B cells in atherogenesis is unknown. Here, we investigated the atherosclerotic phenotype of Mif-gene deletion in Apoe
-/- mice. Apoe-/- Mif-/- mice fed a Western diet exhibited strongly reduced atherosclerotic lesions in brachiocephalic artery (BC) and abdominal aorta compared with controls. This phenotype was accompanied by reduced circulating B cells. Flow cytometry revealed a B-cell developmental defect with increased premature and immature B-cell counts in bone marrow (BM) of Apoe-/- Mif-/- mice and diminished B-cell numbers in spleen. This finding was linked with a decreased expression of Baff-R and differentiation-driving transcription factors at the immature B-cell stage, whereas peritoneal B cells exhibited unchanged CD80 and CD86 expression but vastly decreased CD9 and elevated CD23 levels, indicating that the developmental block favors the generation of immature, egressing, and reactive B cells. Mif deficiency did not affect absolute B-cell numbers in the vessel wall but favored a relative increase of B cells in the atheroprone BC region and the appearance of periadventitial B-cell-rich clusters. Of note, Mif-/- mice exhibited a significant increase in oxidized low-density lipoprotein (oxLDL)-specific antibodies after the injection of oxLDL, indicating that Mif deficiency is associated with higher sensitivity of B cells against natural-occurring antigens such as oxLDL. Importantly, Apoe-/- mice adoptively transplanted with Apoe-/- Mif-/- BM showed reduced peripheral B cells compared with Apoe-/- BM transplantation but no atheroprotection in the BC; also, whereas there was a selective increase in atheroprotective IgM-anti-oxLDL-antibodies in global Mif deficiency, BM-specific Mif deficiency also led to elevated proatherogenic anti-oxLDL-IgG. Together, these findings reveal a novel link between MIF and B cells in atherogenesis. Protection from atherosclerosis by Mif deficiency is associated with enhanced B-cell hypersensitivity, which in global but not BM-restricted Mif deficiency favors an atheroprotective autoantibody profile in atherosclerotic mice. Targeting MIF may induce protective B-cell responses in atherosclerosis.-Schmitz, C., Noels, H., El Bounkari, O., Straussfeld, E., Megens, R. T. A., Sternkopf, M., Alampour-Rajabi, S., Krammer, C., Tilstam, P. V., Gerdes, N., Bürger, C., Kapurniotu, A., Bucala, R., Jankowski, J., Weber, C., Bernhagen, J. Mif-deficiency favors an atheroprotective autoantibody phenotype in atherosclerosis.- Published
- 2018
- Full Text
- View/download PDF
37. Chronic Intake of the Selective Serotonin Reuptake Inhibitor Fluoxetine Enhances Atherosclerosis.
- Author
-
Rami M, Guillamat-Prats R, Rinne P, Salvermoser M, Ring L, Bianchini M, Blanchet X, Megens RTA, Döring Y, Walzog B, Soehnlein O, Weber C, Faussner A, and Steffens S
- Subjects
- Animals, Aorta metabolism, Aorta pathology, Aortic Diseases blood, Aortic Diseases genetics, Aortic Diseases pathology, Atherosclerosis blood, Atherosclerosis genetics, Atherosclerosis pathology, Blood Platelets drug effects, Blood Platelets metabolism, CD18 Antigens blood, Capillary Permeability drug effects, Carotid Arteries metabolism, Carotid Arteries pathology, Carotid Artery Diseases blood, Carotid Artery Diseases genetics, Carotid Artery Diseases pathology, Cell Adhesion drug effects, Chemokine CCL5 blood, Disease Models, Animal, Disease Progression, Drug Administration Schedule, Fluoxetine administration & dosage, HEK293 Cells, HL-60 Cells, Humans, Integrin beta1 blood, Male, Mice, Inbred C57BL, Mice, Knockout, ApoE, Myeloid Cells drug effects, Myeloid Cells metabolism, Serotonin blood, Selective Serotonin Reuptake Inhibitors administration & dosage, Signal Transduction, Time Factors, Aorta drug effects, Aortic Diseases chemically induced, Atherosclerosis chemically induced, Carotid Arteries drug effects, Carotid Artery Diseases chemically induced, Fluoxetine toxicity, Plaque, Atherosclerotic, Selective Serotonin Reuptake Inhibitors toxicity
- Abstract
Objective: Cardiovascular diseases and depression are the leading causes of disability in Western countries. Clinical data on potential cardiovascular effects of serotonin reuptake inhibitors (SSRIs), the most commonly used antidepressant drugs, are controversial. In addition to blocking serotonin reuptake transporter in the brain, SSRIs deplete the major peripheral serotonin (5-hydroxytryptamine [5-HT]) storage by inhibiting serotonin reuptake transporter-mediated uptake in platelets. In this study, we aimed to investigate the effect of chronic SSRI intake on the development of atherosclerosis., Approach and Results: Treatment of apolipoprotein E-deficient mice with the SSRI fluoxetine for 2, 4, or 16 weeks increased atherosclerotic lesion formation, with most pronounced effect during early plaque development. Intravital microscopy of carotid arteries revealed enhanced myeloid cell adhesion on fluoxetine treatment. Mechanistically, we found that fluoxetine augmented vascular permeability and increased chemokine-induced integrin-binding activity of circulating leukocytes. In vitro stimulation of murine blood demonstrated that fluoxetine, but not 5-HT, could directly promote β1 and β2 integrin activation provided C-C motif chemokine ligand 5 was also present. Similar effects were observed with the SSRI escitalopram. Enhanced C-C motif chemokine ligand 5-induced integrin activation by fluoxetine was also confirmed in a human neutrophil-like cell line. In contrast to the proatherogenic properties of fluoxetine, pharmacological inhibition of the peripheral 5-HT synthesizing enzyme tryptophan hydroxylase 1 did not promote atherosclerosis, suggesting that the proatherogenic effect of fluoxetine occurs independent of peripheral 5-HT depletion., Conclusions: SSRI intake may promote atherosclerosis and therefore potentially increase the risk for acute cardiovascular events by a mechanism that is independent of 5-HT depletion., (© 2018 American Heart Association, Inc.)
- Published
- 2018
- Full Text
- View/download PDF
38. Pericardial Adipose Tissue Regulates Granulopoiesis, Fibrosis, and Cardiac Function After Myocardial Infarction.
- Author
-
Horckmans M, Bianchini M, Santovito D, Megens RTA, Springael JY, Negri I, Vacca M, Di Eusanio M, Moschetta A, Weber C, Duchene J, and Steffens S
- Subjects
- Animals, Cell Differentiation, Disease Models, Animal, Female, Fibrosis, Humans, Immunity, Innate, Mice, Mice, Inbred C57BL, Mice, Knockout, Myocardial Infarction metabolism, Receptor, Cannabinoid, CB2 genetics, Receptors, CCR7 genetics, Wound Healing, Adipose Tissue physiology, Granulocytes immunology, Myocardial Infarction pathology, Myocardium pathology, Pericardium pathology
- Abstract
Background: The pericardial adipose tissue (AT) contains a high density of lymphoid clusters. It is unknown whether these clusters play a role in post-myocardial infarction (MI) inflammatory responses and cardiac outcome., Methods: Lymphoid clusters were examined in epicardial AT of humans with or without coronary artery disease. Murine pericardial lymphoid clusters were visualized in mice subjected to coronary artery ligation. To study the relevance of pericardial clusters during inflammatory responses after MI, we surgically removed the pericardial AT and performed B-cell depletion and granulocyte-macrophage colony-stimulating factor blockade. Leukocytes in murine hearts, pericardial AT, spleen, mediastinal lymph nodes, and bone marrow were quantified by flow cytometry. Cannabinoid receptor CB2 (CB2
-/- ) mice were used as a model for enhanced B-cell responses. The effect of impaired dendritic cell (DC) trafficking on pericardial AT inflammatory responses was tested in CCR7-/- mice subjected to MI. Cardiac fibrosis and ventricular function were assessed by histology and echocardiography., Results: We identified larger B-cell clusters in epicardial AT of human patients with coronary artery disease in comparison with controls without coronary artery disease. Infarcted mice also had larger pericardial clusters and 3-fold upregulated numbers of granulocyte-macrophage colony-stimulating factor-producing B cells within pericardial AT, but not spleen or lymph nodes. This was associated with higher DC and T-cell counts in pericardial AT, which outnumbered DCs and T cells in lymph nodes. Analysis of DC maturation markers, tracking experiments with fluorescently labeled cells, and use of CCR7-deficient mice suggested that activated DCs migrate from infarcts into pericardial AT via CCR7. B-cell depletion or granulocyte-macrophage colony-stimulating factor neutralization inhibited DC and T-cell expansion within pericardial AT, and translated into reduced bone marrow granulopoiesis and cardiac neutrophil infiltration 3 days after MI. The relevance of the pericardial AT in mediating all these effects was confirmed by removal of pericardial AT and ex vivo coculture with pericardial AT and granulocyte progenitors. Finally, enhanced fibrosis and worsened ejection fraction in CB2-/- mice were limited by pericardial AT removal., Conclusions: Our findings unveil a new mechanism by which the pericardial AT coordinates immune cell activation, granulopoiesis, and outcome after MI., (© 2017 American Heart Association, Inc.)- Published
- 2018
- Full Text
- View/download PDF
39. Targeting CD40-Induced TRAF6 Signaling in Macrophages Reduces Atherosclerosis.
- Author
-
Seijkens TTP, van Tiel CM, Kusters PJH, Atzler D, Soehnlein O, Zarzycka B, Aarts SABM, Lameijer M, Gijbels MJ, Beckers L, den Toom M, Slütter B, Kuiper J, Duchene J, Aslani M, Megens RTA, van 't Veer C, Kooij G, Schrijver R, Hoeksema MA, Boon L, Fay F, Tang J, Baxter S, Jongejan A, Moerland PD, Vriend G, Bleijlevens B, Fisher EA, Duivenvoorden R, Gerdes N, de Winther MPJ, Nicolaes GA, Mulder WJM, Weber C, and Lutgens E
- Subjects
- Aniline Compounds pharmacology, Animals, Cell Culture Techniques, Cell Movement drug effects, Disease Models, Animal, Humans, Mice, Mice, Inbred C57BL, Monocytes drug effects, Propiophenones pharmacology, Atherosclerosis pathology, Atherosclerosis prevention & control, CD40 Ligand antagonists & inhibitors, Macrophages drug effects, Signal Transduction drug effects, TNF Receptor-Associated Factor 6 antagonists & inhibitors
- Abstract
Background: Disrupting the costimulatory CD40-CD40L dyad reduces atherosclerosis, but can result in immune suppression. The authors recently identified small molecule inhibitors that block the interaction between CD40 and tumor necrosis factor receptor-associated factor (TRAF) 6 (TRAF-STOPs), while leaving CD40-TRAF2/3/5 interactions intact, thereby preserving CD40-mediated immunity., Objectives: This study evaluates the potential of TRAF-STOP treatment in atherosclerosis., Methods: The effects of TRAF-STOPs on atherosclerosis were investigated in apolipoprotein E deficient (Apoe
-/- ) mice. Recombinant high-density lipoprotein (rHDL) nanoparticles were used to target TRAF-STOPs to macrophages., Results: TRAF-STOP treatment of young Apoe-/- mice reduced atherosclerosis by reducing CD40 and integrin expression in classical monocytes, thereby hampering monocyte recruitment. When Apoe-/- mice with established atherosclerosis were treated with TRAF-STOPs, plaque progression was halted, and plaques contained an increase in collagen, developed small necrotic cores, and contained only a few immune cells. TRAF-STOP treatment did not impair "classical" immune pathways of CD40, including T-cell proliferation and costimulation, Ig isotype switching, or germinal center formation, but reduced CD40 and β2-integrin expression in inflammatory monocytes. In vitro testing and transcriptional profiling showed that TRAF-STOPs are effective in reducing macrophage migration and activation, which could be attributed to reduced phosphorylation of signaling intermediates of the canonical NF-κB pathway. To target TRAF-STOPs specifically to macrophages, TRAF-STOP 6877002 was incorporated into rHDL nanoparticles. Six weeks of rHDL-6877002 treatment attenuated the initiation of atherosclerosis in Apoe-/- mice., Conclusions: TRAF-STOPs can overcome the current limitations of long-term CD40 inhibition in atherosclerosis and have the potential to become a future therapeutic for atherosclerosis., (Published by Elsevier Inc.)- Published
- 2018
- Full Text
- View/download PDF
40. Dimeric Glycoprotein VI Binds to Collagen but Not to Fibrin.
- Author
-
Ebrahim M, Jamasbi J, Adler K, Megens RTA, M'Bengue Y, Blanchet X, Uhland K, Ungerer M, Brandl R, Weber C, Elia N, Lorenz R, Münch G, and Siess W
- Subjects
- Atherosclerosis metabolism, Blood Coagulation, Blood Platelets metabolism, Humans, Microscopy, Fluorescence, Plaque, Atherosclerotic metabolism, Platelet Activation, Platelet Adhesiveness, Platelet Aggregation, Protein Binding, Protein Multimerization, Recombinant Proteins, Collagen metabolism, Fibrin metabolism, Fibrinogen metabolism, Platelet Membrane Glycoproteins metabolism, Thrombin metabolism
- Abstract
Platelet glycoprotein VI (GPVI) acts as a decisive collagen receptor in atherothrombosis. Besides collagen, injured atherosclerotic plaques expose tissue factor (TF) that triggers fibrin formation. Two recent studies reported that platelet GPVI also functions as fibrin receptor, which would importantly widen the mode of action of GPVI-targeted antithrombotic drugs. We studied the binding of two GPVI fusion proteins to fibrin under static and arterial flow conditions. Fibrin was prepared from purified fibrinogen or generated more physiologically from endogenous fibrinogen by coagulating plasma with thrombin. Fibrin formation was also triggered by exposing TF-coated surfaces or human atherosclerotic plaque slices to arterially flowing blood. By binding studies and advanced optical imaging, we found that recombinant dimeric GPVI-Fc fusion proteins with Fc from either IgG1 (GPVI-Fc1) or IgG2 (GPVI-Fc2) bound to collagen fibres, but neither to fibrin prepared from purified fibrinogen obtained from three suppliers, nor to physiological fibrin formed by thrombin in plasma or triggered by exposing TF or atherosclerotic plaque slices to arterially flowing blood. Our findings do not support a role of dimeric platelet GPVI as receptor for fibrin. This is important for the understanding of plaque-triggered platelet thrombus formation and is clinically relevant for future GPVI-targeting therapies with recombinant GPVI-Fc and anti-GPVI antibodies., Competing Interests: Kristin Adler, Kerstin Uhland, Götz Münch and Martin Ungerer report a possible conflict of interest. Kristin Adler and Kerstin Uhland are employees of advanceCor GmbH which produces Revacept; Götz Münch and Martin Ungerer are managing directors of advanceCor GmbH and own shares of the company. The other authors report no conflict of interest., (Schattauer GmbH Stuttgart.)
- Published
- 2018
- Full Text
- View/download PDF
41. CD27 co-stimulation increases the abundance of regulatory T cells and reduces atherosclerosis in hyperlipidaemic mice.
- Author
-
Winkels H, Meiler S, Lievens D, Engel D, Spitz C, Bürger C, Beckers L, Dandl A, Reim S, Ahmadsei M, Hartwig H, Holdt LM, Hristov M, Megens RTA, Schmitt MM, Biessen EA, Borst J, Faussner A, Weber C, Lutgens E, and Gerdes N
- Subjects
- Animals, Apoptosis, Atherosclerosis etiology, Atherosclerosis prevention & control, Cells, Cultured, Cytokines metabolism, Disease Models, Animal, Humans, Hyperlipidemias immunology, Hyperlipidemias metabolism, Macrophages immunology, Mice, Mice, Inbred C57BL, Mice, Knockout, T-Lymphocytes, Regulatory immunology, Atherosclerosis immunology, Hyperlipidemias complications, Tumor Necrosis Factor Receptor Superfamily, Member 7 immunology
- Abstract
Aims: The co-stimulatory receptor CD27 modulates responses of T cells, B cells, and NK cells. Various T cell subsets participate in atherogenesis. However, the role of CD27 in atherosclerosis remains unexplored., Methods and Results: Here we investigated the effect of bone marrow-derived and systemic CD27 deficiency in Apolipoprotein E-deficient (Apoe-/-) mice in early and advanced stages of atherosclerosis. Lethally-irradiated Apoe-/- mice reconstituted with Cd27-/-Apoe-/- bone marrow and consuming an atherogenic diet displayed a markedly increased plaque size and lesional inflammation compared to mice receiving Cd27+/+Apoe-/- bone marrow. Accordingly, chow diet-fed Cd27-/-Apoe-/- mice showed exacerbated lesion development and increased inflammation at the age of 18 weeks. At a more advanced stage of atherosclerosis (28 weeks), lesion size and phenotype did not differ between the two groups. Systemic and bone marrow-derived CD27 deficiency reduced the abundance of regulatory T cells (Treg) in blood, lymphoid organs, and the aorta. Numbers of other immune cells were not affected while expression of inflammatory cytokine genes (e.g. IL-1β and IL-6) was increased in the aorta when haematopoietic CD27 was lacking. In vitro, Tregs of CD27-deficient mice showed similar suppressive capacity compared with their wild-type controls and migrated equally towards CCL19 and CCL21. However, thymic Cd27-/- Tregs underwent increased apoptosis and expressed fewer markers of proliferation in vivo. Reconstitution of Cd27-/-Apoe-/- mice with Cd27+/+Apoe-/- Tregs reversed the increase in atherosclerosis., Conclusion: We demonstrate that CD27 co-stimulation increases the number of Tregs and limits lesion development and inflammation in experimental atherosclerosis, particularly during early stages of disease. Thus, our study suggests that promotion of CD27 function may mitigate atherosclerosis., (Published on behalf of the European Society of Cardiology. All rights reserved. © The Author 2017. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2017
- Full Text
- View/download PDF
42. Multi-photon microscopy in cardiovascular research.
- Author
-
Wu Z, Rademakers T, Kiessling F, Vogt M, Westein E, Weber C, Megens RTA, and van Zandvoort M
- Subjects
- Animals, Biomedical Research trends, Humans, Microscopy, Fluorescence, Multiphoton trends, Biomedical Research methods, Cardiovascular Diseases diagnostic imaging, Microscopy, Fluorescence, Multiphoton methods
- Abstract
Multiphoton laser scanning microscopy has proven profound value for ex vivo 3D histology and in vivo imaging of motionless tissue. The development of triggering systems and fast imaging methods, combined with advanced preparation procedures solved the challenging task of intravital imaging of the fast pulsating heart and major arteries in animals and further increased the popularity of intravital multiphoton imaging in cardiovascular research. This review article will highlight the potential of multiphoton microscopy for the visualization and characterization of dynamical and structural processes involved in cardiac and vascular diseases, both in an ex vivo and an intravital animal setting. Examples will be given how multiphoton microscopy can be applied to imaging of atherosclerotic plaque development and progression at subcellular level as well as to intravital imaging of inflammatory processes in the heart. In addition to highlighting the potential of multiphoton microscopy in preclinical cardiovascular research, we will discuss how this tool and its applications may be clinically translated to support disease diagnosis and therapy in patients., (Copyright © 2017 Elsevier Inc. All rights reserved.)
- Published
- 2017
- Full Text
- View/download PDF
43. Deletion of junctional adhesion molecule A from platelets increases early-stage neointima formation after wire injury in hyperlipidemic mice.
- Author
-
Zhao Z, Vajen T, Karshovska E, Dickhout A, Schmitt MM, Megens RTA, von Hundelshausen P, Koeppel TA, Hackeng TM, Weber C, and Koenen RR
- Subjects
- Animals, Apolipoproteins E deficiency, Atherosclerosis complications, Atherosclerosis metabolism, Atherosclerosis pathology, Blood Platelets metabolism, Blood Platelets pathology, Carotid Arteries metabolism, Carotid Arteries pathology, Carotid Artery Injuries complications, Carotid Artery Injuries metabolism, Carotid Artery Injuries pathology, Cell Adhesion, Cell Adhesion Molecules deficiency, Female, Gene Expression Regulation, Hyperlipidemias complications, Hyperlipidemias metabolism, Hyperlipidemias pathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Monocytes metabolism, Monocytes pathology, Myocytes, Smooth Muscle metabolism, Myocytes, Smooth Muscle pathology, Neointima complications, Neointima metabolism, Neointima pathology, Receptors, Cell Surface deficiency, Signal Transduction, Vascular Remodeling genetics, Apolipoproteins E genetics, Atherosclerosis genetics, Carotid Artery Injuries genetics, Cell Adhesion Molecules genetics, Hyperlipidemias genetics, Neointima genetics, Receptors, Cell Surface genetics
- Abstract
Platelets play an important role in the pathogenesis of vascular remodelling after injury. Junctional adhesion molecule A (JAM-A) was recently described to regulate platelet activation. Specific deletion of JAM-A from platelets resulted in increased reactivity and in accelerated progression of atherosclerosis. The aim of this study was to investigate the specific contribution of platelet-derived JAM-A to neointima formation after vascular injury. Mice with or without platelet-specific (tr)JAM-A-deficiency in an apolipoprotein e (apoe
-/- ) background underwent wire-induced injury of the common carotid artery. Ex vivo imaging by two-photon microscopy revealed increased platelet coverage at the site of injury in trJAM-A-deficient mice. Cell recruitment assays showed increased adhesion of monocytic cells to activated JAM-A-deficient platelets than to control platelets. Inhibition of αM β2 or GPIbα, but not of CD62P, suppressed those differences. Up to 4 weeks after wire injury, intimal neoplasia and neointimal cellular content were analysed. Neointimal lesion area was increased in trJAM-A-/- apoe-/- mice and the lesions showed an increased macrophage accumulation and proliferating smooth muscle cells compared with trJAM-A+/+ apoe-/- littermates 2 weeks, but not 4 weeks after injury. Re-endothelialization was decreased in trJAM-A-/- apoe-/- mice compared with controls 2 weeks after injury, yet it was complete in both groups after 4 weeks. A platelet gain of function by deletion of JAM-A accelerates neointima formation only during earlier phases after vascular injury, through an increased recruitment of mononuclear cells. Thus, the contribution of platelets might become less important when neointima formation progresses to later stages., (© 2017 The Authors. Journal of Cellular and Molecular Medicine published by John Wiley & Sons Ltd and Foundation for Cellular and Molecular Medicine.)- Published
- 2017
- Full Text
- View/download PDF
44. Vascular CXCR4 Limits Atherosclerosis by Maintaining Arterial Integrity: Evidence From Mouse and Human Studies.
- Author
-
Döring Y, Noels H, van der Vorst EPC, Neideck C, Egea V, Drechsler M, Mandl M, Pawig L, Jansen Y, Schröder K, Bidzhekov K, Megens RTA, Theelen W, Klinkhammer BM, Boor P, Schurgers L, van Gorp R, Ries C, Kusters PJH, van der Wal A, Hackeng TM, Gäbel G, Brandes RP, Soehnlein O, Lutgens E, Vestweber D, Teupser D, Holdt LM, Rader DJ, Saleheen D, and Weber C
- Subjects
- Animals, Atherosclerosis genetics, Capillary Permeability physiology, Female, Humans, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Receptors, CXCR4 genetics, Atherosclerosis metabolism, Atherosclerosis prevention & control, Endothelial Cells metabolism, Receptors, CXCR4 biosynthesis
- Abstract
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 (BmxCreER
T2 -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/β-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/β-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., (© 2017 American Heart Association, Inc.)- Published
- 2017
- Full Text
- View/download PDF
45. Atypical chemokine receptor 1 on nucleated erythroid cells regulates hematopoiesis.
- Author
-
Duchene J, Novitzky-Basso I, Thiriot A, Casanova-Acebes M, Bianchini M, Etheridge SL, Hub E, Nitz K, Artinger K, Eller K, Caamaño J, Rülicke T, Moss P, Megens RTA, von Andrian UH, Hidalgo A, Weber C, and Rot A
- Subjects
- Animals, Humans, Mice, Black People genetics, Bone Marrow pathology, Bone Marrow Cells metabolism, Cell Proliferation, Flow Cytometry, Fluorescent Antibody Technique, Microscopy, Confocal, Receptors, Chemokine genetics, Receptors, Chemokine metabolism, Duffy Blood-Group System genetics, Duffy Blood-Group System metabolism, Erythroblasts metabolism, Hematopoiesis genetics, Hematopoietic Stem Cells cytology, Hematopoietic Stem Cells metabolism, Neutropenia genetics, Neutrophils cytology, Neutrophils metabolism, Receptors, Cell Surface genetics, Receptors, Cell Surface metabolism
- Abstract
Healthy individuals of African ancestry have neutropenia that has been linked with the variant rs2814778(G) of the gene encoding atypical chemokine receptor 1 (ACKR1). This polymorphism selectively abolishes the expression of ACKR1 in erythroid cells, causing a Duffy-negative phenotype. Here we describe an unexpected fundamental role for ACKR1 in hematopoiesis and provide the mechanism that links its absence with neutropenia. Nucleated erythroid cells had high expression of ACKR1, which facilitated their direct contact with hematopoietic stem cells. The absence of erythroid ACKR1 altered mouse hematopoiesis including stem and progenitor cells, which ultimately gave rise to phenotypically distinct neutrophils that readily left the circulation, causing neutropenia. Individuals with a Duffy-negative phenotype developed a distinct profile of neutrophil effector molecules that closely reflected the one observed in the ACKR1-deficient mice. Thus, alternative physiological patterns of hematopoiesis and bone marrow cell outputs depend on the expression of ACKR1 in the erythroid lineage, findings with major implications for the selection advantages that have resulted in the paramount fixation of the ACKR1 rs2814778(G) polymorphism in Africa.
- Published
- 2017
- Full Text
- View/download PDF
46. Functional ex-vivo Imaging of Arterial Cellular Recruitment and Lipid Extravasation.
- Author
-
van der Vorst EPC, Maas SL, Ortega-Gomez A, Hameleers JMM, Bianchini M, Asare Y, Soehnlein O, Döring Y, Weber C, and Megens RTA
- Abstract
The main purpose of this sophisticated and highly versatile method is to visualize and quantify structural vessel wall properties, cellular recruitment, and lipid/dextran extravasation under physiological conditions in living arteries. This will be of interest for a broad range of researchers within the field of inflammation, hypertension, atherosclerosis, and even the pharmaceutical industry. Currently, many researchers are using in vitro techniques to evaluate cellular recruitment, like transwell or flow chamber systems with cultured cells, with unclear physiological comparability. The here introduced method describes in detail the use of a sophisticated and flexible method to study arterial wall properties and leukocyte recruitment in fresh and viable murine carotid arteries ex vivo under arterial flow conditions. This model mimics the in vivo situation and allows the use of cells and arteries isolated from two different donors (for example, wildtype vs. specific knockouts) to be combined into one experiments, thereby providing information on both leukocyte and/or endothelial cell properties of both donors. As such, this model can be considered an alternative for the complicated and invasive in vivo studies, such as parabiotic experiments.
- Published
- 2017
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.