Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Analyst ; 147(6): 1222-1235, 2022 Mar 14.
Article in English | MEDLINE | ID: mdl-35212697

ABSTRACT

Microvascular thrombosis and inflammation (thromboinflammation) are major causes of morbidity and mortality in critically ill patients with limited therapeutic options. Platelets are central to thromboinflammation, and microvascular platelet thrombi are highly effective at recruiting and activating leukocytes at sites of endothelial injury. Whilst parallel-plate flow chambers, microslides and straight microchannel assays have been widely used to recapitulate leukocyte adhesive behavior on 2-dimensional (2D) surfaces, none of these methods achieve high fidelity 3-dimensional (3D) geometries emulating microvascular platelet thrombi. As a result, the role of hydrodynamic factors in regulating leukocyte interactions with platelet thrombi remains ill-defined. Here, we report a microfluidic post model that allows visualization and analysis of neutrophil-platelet interactions in a 3D flow field. We have utilized the unique mechanosensitive features of platelets to enable selective micropatterning of the 3D posts with human or mouse platelets. By modulating the activation status of platelets, our method enables precise control of platelet surface reactivity and neutrophil recruitment. In addition, our microfluidic post assay accurately recapitulated the rolling versus stationary adhesion behavior of single neutrophils and demonstrated the efficacy of the P-selectin and Mac-1 blocking antibodies to reduce neutrophil recruitment and stationary adhesion, respectively. Moreover, the geometry of posts had a major influence on the efficiency of neutrophil recruitment and adhesion stability. This new post method highlights the importance of platelet 3D geometries in facilitating efficient, localized neutrophil recruitment. These findings have potentially important implications for the potent proinflammatory function of microvascular platelet thrombi.


Subject(s)
Blood Platelets , Thrombosis , Animals , Cell Adhesion , Humans , Inflammation , Leukocytes , Mice , Microfluidics , Neutrophils
2.
Eur Heart J ; 43(6): 488-500, 2022 02 10.
Article in English | MEDLINE | ID: mdl-34132336

ABSTRACT

AIMS: Assessment of endothelial function in humans by measuring flow-mediated dilation (FMD) risk-stratifies individuals with established cardiovascular disease, whereas its predictive value is limited in primary prevention. We therefore aimed to establish and evaluate novel markers of FMD at the population level. METHODS AND RESULTS: In order to identify novel targets that were negatively correlated with FMD and investigate their contribution to vascular function, we performed a genome-wide association study (GWAS) of 4175 participants of the population based Gutenberg Health Study. Subsequently, conditional knockout mouse models deleting the gene of interest were generated and characterized. GWAS analysis revealed that single-nucleotide polymorphisms (SNPs) in the tubulin-folding cofactor E (TBCE) gene were negatively correlated with endothelial function and TBCE expression. Vascular smooth muscle cell (VSMC)-targeted TBCE deficiency was associated with endothelial dysfunction, aortic wall hypertrophy, and endoplasmic reticulum (ER) stress-mediated VSMC hyperproliferation in mice, paralleled by calnexin up-regulation and exacerbated by the blood pressure hormone angiotensin II. Treating SMMHC-ERT2-Cre+/-TBCEfl/fl mice with the ER stress modulator tauroursodeoxycholic acid amplified Raptor/Beclin-1-dependent autophagy and reversed vascular dysfunction. CONCLUSION: TBCE and tubulin homeostasis seem to be novel predictors of vascular function and offer a new drug target to ameliorate ER stress-dependent vascular dysfunction.


Subject(s)
Endoplasmic Reticulum Stress , Tubulin , Animals , Aorta , Endothelium, Vascular/metabolism , Genome-Wide Association Study , Humans , Mice , Mice, Knockout , Tubulin/metabolism
3.
Acta Physiol (Oxf) ; 232(1): e13628, 2021 05.
Article in English | MEDLINE | ID: mdl-33590724

ABSTRACT

AIM: Recent evidence suggests that arterial hypertension could be alternatively explained as a physiological adaptation response to water shortage, termed aestivation, which relies on complex multi-organ metabolic adjustments to prevent dehydration. Here, we tested the hypothesis that chronic water loss across diseased skin leads to similar adaptive water conservation responses as observed in experimental renal failure or high salt diet. METHODS: We studied mice with keratinocyte-specific overexpression of IL-17A which develop severe psoriasis-like skin disease. We measured transepidermal water loss and solute and water excretion in the urine. We quantified glomerular filtration rate (GFR) by intravital microscopy, and energy and nitrogen pathways by metabolomics. We measured skin blood flow and transepidermal water loss (TEWL) in conjunction with renal resistive indices and arterial blood pressure. RESULTS: Psoriatic animals lost large amounts of water across their defective cutaneous epithelial barrier. Metabolic adaptive water conservation included mobilization of nitrogen and energy from muscle to increase organic osmolyte production, solute-driven maximal anti-diuresis at normal GFR, increased metanephrine and angiotensin 2 levels, and cutaneous vasoconstriction to limit TEWL. Heat exposure led to cutaneous vasodilation and blood pressure normalization without parallel changes in renal resistive index, albeit at the expense of further increased TEWL. CONCLUSION: Severe cutaneous water loss predisposes psoriatic mice to lethal dehydration. In response to this dehydration stress, the mice activate aestivation-like water conservation motifs to maintain their body hydration status. The circulatory water conservation response explains their arterial hypertension. The nitrogen-dependency of the metabolic water conservation response explains their catabolic muscle wasting.


Subject(s)
Hypertension , Water Loss, Insensible , Animals , Estivation , Mice , Muscles , Skin
4.
Cardiovasc Res ; 117(1): 162-177, 2021 01 01.
Article in English | MEDLINE | ID: mdl-32077922

ABSTRACT

AIMS: Heart failure (HF) ensuing myocardial infarction (MI) is characterized by the initiation of a systemic inflammatory response. We aimed to elucidate the impact of myelomonocytic cells and their activation by angiotensin II on vascular endothelial function in a mouse model of HF after MI. METHODS AND RESULTS: HF was induced in male C57BL/6J mice by permanent ligation of the left anterior descending coronary artery. Compared to sham, HF mice had significantly impaired endothelial function accompanied by enhanced mobilization of Sca-1+c-Kit+ haematopoietic stem cells and Sca-1-c-Kit+ common myeloid and granulocyte-macrophage progenitors in the bone marrow as well as increased vascular infiltration of CD11b+Ly6G-Ly6Chigh monocytes and accumulation of CD11b+ F4/80+ macrophages, assessed by flow cytometry. Using mice with Cre-inducible expression of diphtheria toxin receptor in myeloid cells, we selectively depleted lysozyme M+ myelomonocytic cells for 10 days starting 28 days after MI. While the cardiac phenotype remained unaltered until 38 days post-MI, myeloid cell depletion attenuated vascular accumulation of Nox2+CD45+ cells, endothelial dysfunction, oxidative stress, and vascular expression of adhesion molecules and angiotensin II receptor type 1 (AT1R). Pharmacological blockade of this receptor for 4 weeks did not significantly alter cardiac function, but mimicked the effects of myeloid cell depletion: telmisartan (20 mg/kg/day, fed to C57BL/6J mice) diminished bone marrow myelopoesis and myeloid reactive oxygen species production, attenuated endothelial leucocyte rolling and vascular accumulation of CD11b+Ly6G-Ly6Chigh monocytes and macrophages, resulting in improved vascular function with less abundance of Nox2+CD45+ cells. CONCLUSION: Endothelial dysfunction in HF ensuing MI is mediated by inflammatory Nox2+ myeloid cells infiltrating the vessel wall that can be targeted by AT1R blockade.


Subject(s)
Angiotensin II/metabolism , Endothelial Cells/metabolism , Heart Failure/etiology , Myeloid Cells/enzymology , Myocardial Infarction/complications , NADPH Oxidase 2/metabolism , Receptor, Angiotensin, Type 1/metabolism , Vasculitis/etiology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Animals, Genetically Modified , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/immunology , Heart Failure/drug therapy , Heart Failure/enzymology , Heart Failure/immunology , Leukocyte Rolling , Macrophages/immunology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Monocytes/immunology , Monocytes/metabolism , Muramidase/genetics , Muramidase/metabolism , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myocardial Infarction/enzymology , Myocardial Infarction/immunology , Oxidative Stress , Signal Transduction , Telmisartan/pharmacology , Vasculitis/drug therapy , Vasculitis/enzymology , Vasculitis/immunology
5.
Int J Mol Sci ; 21(14)2020 Jul 12.
Article in English | MEDLINE | ID: mdl-32664652

ABSTRACT

Myeloid cells are crucial for the development of vascular inflammation. Low-density lipoprotein receptor-related protein 8 (LRP8) or Apolipoprotein E receptor 2 (ApoER2), is expressed by macrophages, endothelial cells and platelets and has been implicated in the development of cardiovascular diseases. Our aim was to evaluate the role of LRP8, in particular from immune cells, in the development of vascular inflammation. METHODS: LRP8+/+ and LRP8-/- mice (on B6;129S background) were infused with angiotensin II (AngII, 1 mg/kg/day for 7 to 28 day) using osmotic minipumps. Blood pressure was recorded using tail cuff measurements. Vascular reactivity was assessed in isolated aortic segments. Leukocyte activation and infiltration were assessed by flow cytometry of aortic tissue and intravital videomicroscopy imaging. Histological analysis of aortic sections was conducted using sirius red staining. RESULTS: AngII infusion worsened endothelial-dependent vascular relaxation and immune cells rolling and adherence to the carotid artery in both LRP8+/+ as well as LRP8-/- mice. However, only LRP8-/- mice demonstrated a drastically increased mortality rate in response to AngII due to aortic dissection. Bone marrow transplantation revealed that chimeras with LRP8 deficient myeloid cells phenocopied LRP8-/- mice. CONCLUSION: AngII-infused LRP8 deficient mice could be a useful animal model to study aortic dissection reflecting the lethality of this disease in humans.


Subject(s)
Angiotensin II/toxicity , Aortic Dissection/chemically induced , LDL-Receptor Related Proteins/deficiency , Acetylcholine/pharmacology , Animals , Blood Pressure , Bone Marrow Transplantation , Cell Adhesion/drug effects , Chemotaxis, Leukocyte/drug effects , Disease Models, Animal , Gene Expression Regulation/drug effects , Infusion Pumps , LDL-Receptor Related Proteins/physiology , Male , Mice , Microscopy, Video , Myeloid Cells/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Radiation Chimera , Vasodilation/drug effects
6.
Int J Mol Sci ; 20(17)2019 Aug 30.
Article in English | MEDLINE | ID: mdl-31480330

ABSTRACT

BACKGROUND: Psoriasis is hallmarked by vascular dysfunction, arterial hypertension, and an increased risk for cardiovascular diseases. We have shown recently that skin-driven interleukin (IL)-17A expression promotes psoriasis-like disease in mice, and this is associated with vascular inflammation, vascular dysfunction, and hypertension. As an intensive risk-factor reduction is recommended for psoriasis patients, we aimed to elucidate the impact of the angiotensin II receptor type 1 (AT1) antagonist telmisartan in a mouse model of severe psoriasis-like skin disease. METHODS AND RESULTS: Elevated blood pressure measured by tail-cuff plethysmography in mice with keratinocyte-specific IL-17A overexpression (K14-IL-17Aind/+ mice) was significantly reduced in response to telmisartan. Importantly, vascular dysfunction, as assessed by isometric tension studies of isolated aortic rings, vascular inflammation measured by flow cytometry analysis of CD45+CD11b+ immune cells, as well as the increased peripheral oxidative stress levels assessed by L-012-enhanced chemiluminescence were not attenuated by telmisartan treatment of K14-IL-17Aind/+ mice, nor was the persisting skin inflammation. CONCLUSION: We provide first evidence for an effective antihypertensive treatment in experimental psoriasis by AT1 blockade, but without any impact on vascular inflammation and dysfunction in our mouse model of severe psoriasis-like skin disease. This suggests that vascular function and inflammation in psoriasis might not be attenuated as long as skin inflammation persists.


Subject(s)
Blood Pressure/drug effects , Endothelium, Vascular/physiopathology , Inflammation/pathology , Psoriasis/drug therapy , Psoriasis/physiopathology , Telmisartan/therapeutic use , Animals , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Mice , Oxidative Stress/drug effects , Skin/drug effects , Skin/pathology , Telmisartan/pharmacology
7.
Oxid Med Cell Longev ; 2019: 6721531, 2019.
Article in English | MEDLINE | ID: mdl-31396305

ABSTRACT

AIMS: The neutrophil recruiting cytokine Interleukin-17A (IL-17A) is a key component in vascular dysfunction and arterial hypertension. Moreover, IL-17A has a central role for the vascular infiltration of myeloid cells into the arterial wall in Angiotensin II-induced vascular inflammation. The intention of our study was to analyze the impact of T cell-derived IL-17A on hypertension, vascular function, and inflammation. METHODS AND RESULTS: Chronic IL-17A overexpression in T cells (CD4-IL-17Aind/+ mice) resulted in elevated reactive oxygen species in the peripheral blood and a significant vascular dysfunction compared to control mice. The vascular dysfunction seen in the CD4-IL-17Aind/+ mice was only accompanied by a modest and nonsignificant accumulation of inflammatory cells within the vessel wall. Therefore, infiltrating myeloid cells did not serve as an explanation of the vascular dysfunction seen in a chronic IL-17A-driven mouse model. In addition to vascular dysfunction, CD4-IL-17Aind/+ mice displayed vascular fibrosis with highly proliferative fibroblasts. This fibroblast proliferation was induced by exposure to IL-17A as confirmed by in vitro experiments with primary murine fibroblastic cells. We also found that the ·NO/cGMP pathway was downregulated in the vasculature of the CD4-IL-17Aind/+ mice, while levels of protein tyrosine kinase 2 (PYK2), an oxidative stress-triggered process associated with T cell activation, were upregulated in the perivascular fat tissue (PVAT). CONCLUSIONS: Our data demonstrate that T cell-derived IL-17A elicits vascular dysfunction by mediating proliferation of fibroblasts and subsequent vascular fibrosis associated with PYK2 upregulation.


Subject(s)
Cyclic GMP/metabolism , Endothelium, Vascular/physiopathology , Interleukin-17/metabolism , Nitric Oxide/metabolism , T-Lymphocytes/metabolism , Animals , Aorta/metabolism , Aorta/pathology , Cell Proliferation , Down-Regulation , Fibroblasts/cytology , Fibroblasts/metabolism , Fibrosis , Focal Adhesion Kinase 2/metabolism , Interleukin-17/genetics , Male , Mice , Mice, Inbred C57BL , Protein Subunits/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Soluble Guanylyl Cyclase/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Up-Regulation
8.
Cardiovasc Res ; 115(13): 1907-1917, 2019 Nov 01.
Article in English | MEDLINE | ID: mdl-30949687

ABSTRACT

AIMS: Myelomonocytic cells are critical in injury and healing post-myocardial infarction (MI). Mechanisms of regulation, however, are incompletely understood. The aim of the study was to elucidate the role of interferon gamma (IFN-γ) in the orchestrated inflammatory response in a murine model of MI. METHODS AND RESULTS: MI was induced in 8- to 12-week-old male mice (C57BL/6 background) by permanent ligation of the left anterior descending (LAD) coronary artery. Lysozyme M (LysM)+ cell-depleted LysMiDTR transgenic mice displayed a reduced influx of CD45.2+/CD3-/CD11b+/Gr-1high neutrophils into infarcted myocardium 1 day post-MI compared with infarcted controls, paralleled by decreased cardiac mRNA levels of IFN-γ and tumour necrosis factor alpha (TNF-α). Mortality after MI was significantly increased in LysM+ cell-depleted mice within 28 days post-MI. To more specifically address the role of neutrophils, we depleted C57BL/6 mice with a monoclonal anti-Gr-1 antibody and found increased mortality, deteriorated cardiac function as well as decreased cardiac IFN-γ mRNA expression early after MI. Ccl2, Cxcl1, Cx3cl1, and Il12b mRNA were reduced 3 days after MI, as was the amount of CD11b+/Ly-6G-/Ly-6Chigh inflammatory monocytes. LAD-ligated Cramp-/- mice lacking cathelicidin important in neutrophil-dependent monocyte chemotaxis as well as IFNγ-/- and TNFα-/- mice phenocopied Gr-1+ cell-depleted mice, supporting a regulatory role of IFN-γ impacting on both the sequence of inflammatory cell invasion and cardiac outcome early after MI. The use of conditional IFN-γ receptor deficient mice indicated a direct effect of IFN-γ on LysM+ cells in cardiac injury post-MI. Using IFN-γ reporter mice and flow cytometry, we identified cardiac lymphoid cells (CD4+ and CD8+ T cells and natural killer cells) as primary source of this cytokine in the cardiac inflammatory response post-MI. CONCLUSION: IFN-γ directs a sequential chemotactic cellular immune response and determines survival and cardiac function post-MI.


Subject(s)
Chemotaxis, Leukocyte , Immunity, Cellular , Interferon-gamma/metabolism , Lymphocytes/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Animals , Antimicrobial Cationic Peptides/genetics , Antimicrobial Cationic Peptides/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Interferon-gamma/genetics , Interferon-gamma/immunology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Lymphocytes/immunology , Male , Mice, Inbred C57BL , Mice, Knockout , Muramidase/genetics , Muramidase/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/immunology , Myocardial Infarction/physiopathology , Myocardium/immunology , Myocardium/pathology , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Signal Transduction , Time Factors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Cathelicidins , Interferon gamma Receptor
9.
Basic Res Cardiol ; 114(2): 8, 2019 01 14.
Article in English | MEDLINE | ID: mdl-30643968

ABSTRACT

Mice with a global deletion of α1AMPK are characterized by endothelial dysfunction and NADPH oxidase subunit 2 (NOX-2)-mediated vascular oxidative stress. However, the underlying mechanisms are incompletely understood and may involve endothelial NOX-2 upregulation or facilitated vascular infiltration of phagocytic cells. Therefore, the current study was designed to investigate the vascular effects of chronic angiotensin II (AngII) infusion in mice with an endothelial-specific α1AMPK deletion. A mouse strain with endothelial-specific α1AMPK deletion was generated by breeding α1AMPKflox/flox mice with TekCre+ or Cadh5Cre+ mice. Chronic AngII infusion (0.5 mg/kg/day for 7day) caused mild endothelial dysfunction in wild-type mice that was significantly aggravated in endothelial α1AMPK knockout mice. Aortic NOX-2 and CD68 expression were increased, indicating that infiltrating leukocytes may significantly contribute to enhanced vascular oxidative stress. Flow cytometry revealed a higher abundance of aortic CD90.2+ T-cells, CD11b+F4/80+ macrophages and Ly6G-Ly6C+ monocytes. Vascular mRNA expression of monocyte chemoattractant protein 1, CCL5 and vascular cell adhesion molecule 1 was enhanced in AngII-infused mice lacking endothelial α1AMPK, facilitating the recruitment of inflammatory cells to the vessel wall. In addition, AngII-induced upregulation of cytoprotective heme oxygenase 1 (HO-1) was blunted in mice with endothelial α1AMPK deletion, compatible with an impaired antioxidant defense in these animals. In summary, endothelial expressed α1AMPK limits the recruitment of inflammatory cells to the vessel wall and maintains HO-1 mediated antioxidant defense. Both mechanisms reduce vascular oxidative damage and preserve endothelial function during chronic AngII treatment.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Endothelium, Vascular/metabolism , Angiotensin II/toxicity , Animals , Antioxidants/metabolism , Endothelium, Vascular/drug effects , Heme Oxygenase-1/metabolism , Inflammation/metabolism , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress/drug effects , Oxidative Stress/physiology
10.
J Invest Dermatol ; 139(3): 638-647, 2019 03.
Article in English | MEDLINE | ID: mdl-30367871

ABSTRACT

Besides skin inflammation, patients with severe psoriasis suffer from an increased risk of cardiovascular mortality. IL-17A plays a central role in the development of psoriasis and might connect skin and vascular disease. The aim of this study was to clarify whether anti-IL-17A therapy could also ameliorate the vascular dysfunction associated with severe psoriasis. We analyzed three murine models with varying severities of psoriasis-like skin disease concerning their vascular function and inflammation: (i) K14-IL-17Aind/+ mice with keratinocyte-specific IL-17A overexpression and an early-onset severe psoriasis-like phenotype; (ii) homozygous CD11c-IL-17Aind/ind and heterozygous CD11c-IL-17Aind/+ mice overexpressing IL-17A in CD11c+ cells, leading to a delayed onset of moderate psoriasis-like skin disease; and (iii) the acute model of imiquimod-induced psoriasis-like skin inflammation. Similar to the severity of skin disease, vascular dysfunction correlated with peripheral IL-17A levels and neutrophil infiltration into the aortic vessel wall. Successful anti-IL-17A treatment of psoriatic skin lesions diminished peripheral oxidative stress levels, proinflammatory cytokines, and vascular inflammation. These data highlight the pivotal role of IL-17A linking the development of skin lesions and vascular disease in psoriasis. Anti-IL-17A therapy might thus represent a useful approach to attenuate and prevent vascular disease in psoriasis patients.


Subject(s)
Aorta/immunology , Immunotherapy/methods , Inflammation/immunology , Interleukin-17/metabolism , Psoriasis/immunology , Skin/immunology , Vascular Diseases/immunology , Animals , Antibodies, Blocking/administration & dosage , Disease Models, Animal , Disease Progression , Humans , Imiquimod , Interleukin-17/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Neutrophil Infiltration , Oxidative Stress , Psoriasis/therapy , Reactive Oxygen Species/metabolism
11.
Oxid Med Cell Longev ; 2018: 1860513, 2018.
Article in English | MEDLINE | ID: mdl-29983855

ABSTRACT

Pulmonary embolism (PE) results from deep vein thrombosis (DVT) and can lead to chronic thromboembolic pulmonary hypertension (CTEPH) involving vascular dysfunction. Mechanisms are incompletely understood, in part due to lack of mouse models. We induced PE in C57BL/6 mice by intravenous injection of thrombin (166 U/kg BW), confirmed by a sudden bradycardia, bradypnea, and an increase in pulmonary artery (PA) pressure observed by high-frequency ultrasound. While symptoms resolved rapidly after single thrombin application, repeated PEs resulted in sustained PA-pressure increase, increased PA superoxide formation assessed by oxidative fluorescent microtopography, increased PA gp91phox expression, and endothelial dysfunction assessed by isometric tension studies of isolated PA segments after 24 hours. DVT was modeled in C57BL/6 mice by ligation of the inferior vena cava (IVC). Importantly, small pulmonary emboli could be detected along with a mild phenotype of PA endothelial dysfunction and oxidative stress in the absence of PA-pressure elevation. mRNA expression of plasminogen activator inhibitor-1 was increased in PAs of mice with recurrent PE after repetitive thrombin injections and to a lesser extent in DVT mice. In summary, our data suggest that PA endothelial dysfunction, induced by gp91phox-derived ROS, is an early event upon repetitive PE. This phenomenon might help to elucidate the mechanisms of PA dysfunction in the pathogenesis of CTEPH.


Subject(s)
Hypertension, Pulmonary/metabolism , NADPH Oxidases/metabolism , Pulmonary Embolism/metabolism , Superoxides/metabolism , Venous Thrombosis/metabolism , Animals , Cell Line , Echocardiography , Humans , Immunohistochemistry , Male , Mice , Reverse Transcriptase Polymerase Chain Reaction , Vena Cava, Inferior/metabolism
12.
Sci Rep ; 8(1): 3013, 2018 02 14.
Article in English | MEDLINE | ID: mdl-29445199

ABSTRACT

The role of leukocytes in deep vein thrombosis (DVT) resolution is incompletely understood. We determined how depletion of lysozyme positive (LysM+) cells and a switched-off type 1 immune response influences thrombus resolution. DVT was induced in 12-week-old male mice by inferior vena cava (IVC) stenosis. Toxin mediated depletion of myeloid cells improved thrombus resolution in mice with Cre-inducible expression of the diphtheria toxin receptor in LysM+ cells. This correlated with decreased CD45+ cells, a population shift of Gr-1+ to Gr-1- CD11b+ myelomonocytic cells (flow cytometry) and an increase in CC-chemokine ligand 2, interleukin-4 and interleukin-10 mRNA expressions. Tbx21-/- mice (lacking transcription factor T-bet and marked by an attenuated type 1 immune response) with DVT had faster thrombus resolution, a reduction of pro-inflammatory Ly6Chi monocytes in thrombi and decreased interleukin-12p40 mRNA expression than control mice resulting in increased vascular endothelial growth factor mRNA expression and improved neovascularization of thrombotic veins. Transfer of Tbx21-/- bone marrow into irradiated Tbx21+/+ recipients lead to accelerated thrombus resolution with lower T-bet-dependent interleukin-12p40 mRNA levels following IVC-stenosis. We conclude that inhibition of Tbet+ interleukin-12 forming myelomonocytic cells accelerated thrombus resolution. Modulating the inflammatory immune response might be an approach to improve therapy of DVT.


Subject(s)
Interleukin-12 Subunit p40/metabolism , Monocytes/physiology , T-Box Domain Proteins/metabolism , Venous Thrombosis/immunology , Animals , Antigens, Ly/metabolism , Diphtheria Toxin/genetics , Disease Models, Animal , Humans , Interleukin-12 Subunit p40/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Neovascularization, Physiologic , T-Box Domain Proteins/genetics , Transplantation Chimera , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vena Cava, Inferior/surgery
13.
J Vis Exp ; (131)2018 01 04.
Article in English | MEDLINE | ID: mdl-29364261

ABSTRACT

Epifluorescence intravital video microscopy (IVM) of blood vessels is an established method to evaluate the activation of immune cells and their ability to role and adhere to the endothelial layer. Visualization of circulating cells by injection of fluorescent dyes or fluorophore-coupled antibodies is commonly used. Alternatively, fluorescent reporter mice can be used. Interactions of leukocytes, in particular lysozyme M+ (LysM+) monocytes, with the vessel wall play pivotal roles in promoting vascular dysfunction and arterial hypertension. We here present the technique to visualize and quantify leukocyte rolling and adhesion in carotid arteries in angiotensin II (AngII)-induced hypertension in mice by IVM. The implantation of a catheter damages the vascular wall and leads to altered blood cell responses. We compared different injection techniques and administration routes to visualize leukocytes in a LysMCre+IRG+ mouse with widespread expression of red fluorescent protein and conditional expression of green fluorescent protein in LysM+ cells. To study LysM+ cell activation, we used AngII infused mice in which rolling and adhesion of leukocytes to the endothelium is increased. We either injected acridine orange using a jugular catheter or directly though the tail vein and compared the amount of rolling and adhering cells. We found that jugular catheter implantation per se increased the number of rolling and adhering LysM+ cells in sham-infused LysMCre+IRG+ mice compared to controls. This activation was augmented in AngII-infused mice. Interestingly, injecting acridine orange directly through the tail vein did not increase LysM+ cell adhesion or rolling in sham-infused mice. We thereby demonstrated the importance of transgenic reporter mice expressing fluorescent proteins to not interfere with in vivo processes during experimentation. Furthermore, tail vein injection of fluorescent tracers might be a possible alternative to jugular catheter injections.


Subject(s)
Angiotensin II/administration & dosage , Cell Adhesion/physiology , Leukocyte Rolling/physiology , Animals , Carotid Arteries/drug effects , Carotid Arteries/pathology , Cell Adhesion/drug effects , Hypertension/blood , Hypertension/chemically induced , Hypertension/pathology , Leukocyte Rolling/drug effects , Leukocytes/drug effects , Leukocytes/pathology , Male , Mice , Mice, Transgenic
15.
Eur Heart J ; 38(37): 2838-2849, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28329261

ABSTRACT

Aims: Epidemiological studies indicate that traffic noise increases the incidence of coronary artery disease, hypertension and stroke. The underlying mechanisms remain largely unknown. Field studies with nighttime noise exposure demonstrate that aircraft noise leads to vascular dysfunction, which is markedly improved by vitamin C, suggesting a key role of oxidative stress in causing this phenomenon. Methods and results: We developed a novel animal model to study the vascular consequences of aircraft noise exposure. Peak sound levels of 85 and mean sound level of 72 dBA applied by loudspeakers for 4 days caused an increase in systolic blood pressure, plasma noradrenaline and angiotensin II levels and induced endothelial dysfunction. Noise increased eNOS expression but reduced vascular NO levels because of eNOS uncoupling. Noise increased circulating levels of nitrotyrosine, interleukine-6 and vascular expression of the NADPH oxidase subunit Nox2, nitrotyrosine-positive proteins and of endothelin-1. FACS analysis demonstrated an increase in infiltrated natural killer-cells and neutrophils into the vasculature. Equal mean sound pressure levels of white noise for 4 days did not induce these changes. Comparative Illumina sequencing of transcriptomes of aortic tissues from aircraft noise-treated animals displayed significant changes of genes in part responsible for the regulation of vascular function, vascular remodelling, and cell death. Conclusion: We established a novel and unique aircraft noise stress model with increased blood pressure and vascular dysfunction associated with oxidative stress. This animal model enables future studies of molecular mechanisms, mitigation strategies, and pharmacological interventions to protect from noise-induced vascular damage.


Subject(s)
Aircraft , Noise, Transportation/adverse effects , Oxidative Stress/physiology , Animals , Aorta/physiology , Blood Pressure/physiology , Disease Models, Animal , Endothelium, Vascular/physiology , Hormones/metabolism , Mice, Inbred C57BL , Myocardium/metabolism , NADPH Oxidases/metabolism , Nitric Oxide Synthase Type III/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction , Vasculitis/physiopathology , Vasoconstriction/physiology , Vasodilation/physiology
16.
Circulation ; 135(22): 2163-2177, 2017 May 30.
Article in English | MEDLINE | ID: mdl-28298457

ABSTRACT

BACKGROUND: Hypertension caused by increased renin-angiotensin system activation is associated with elevated reactive oxygen species production. Previous studies implicate NADPH oxidase (Nox) proteins as important reactive oxygen species sources during renin-angiotensin system activation, with different Nox isoforms being potentially involved. Among these, Nox2 is expressed in multiple cell types, including endothelial cells, fibroblasts, immune cells, and microglia. Blood pressure (BP) is regulated at the central nervous system, renal, and vascular levels, but the cell-specific role of Nox2 in BP regulation is unknown. METHODS: We generated a novel mouse model with a floxed Nox2 gene and used Tie2-Cre, LysM Cre, or Cdh5-CreERT2 driver lines to develop cell-specific models of Nox2 perturbation to investigate its role in BP regulation. RESULTS: Unexpectedly, Nox2 deletion in myeloid but not endothelial cells resulted in a significant reduction in basal BP. Both Tie2-CreNox2 knockout (KO) mice (in which Nox2 was deficient in both endothelial cells and myeloid cells) and LysM CreNox2KO mice (in which Nox2 was deficient in myeloid cells) had significantly lower BP than littermate controls, whereas basal BP was unaltered in Cdh5-CreERT2 Nox2KO mice (in which Nox2 is deficient only in endothelial cells). The lower BP was attributable to an increased NO bioavailability that dynamically dilated resistance vessels in vivo under basal conditions without a change in renal function. Myeloid-specific Nox2 deletion had no effect on angiotensin II-induced hypertension, which, however, was blunted in Tie2-CreNox2KO mice, along with preservation of endothelium-dependent relaxation during angiotensin II stimulation. CONCLUSIONS: We identify a hitherto unrecognized modulation of basal BP by myeloid cell Nox2, whereas endothelial cell Nox2 regulates angiotensin II-induced hypertension. These results identify distinct cell-specific roles for Nox2 in BP regulation.


Subject(s)
Blood Pressure/physiology , Endothelial Cells/enzymology , Hypertension/enzymology , Membrane Glycoproteins/deficiency , Myeloid Cells/enzymology , NADPH Oxidases/deficiency , Angiotensin II/toxicity , Animals , Blood Pressure/drug effects , Electron Spin Resonance Spectroscopy/methods , Endothelial Cells/drug effects , Hypertension/chemically induced , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/drug effects , NADPH Oxidase 2
17.
Sci Transl Med ; 9(375)2017 02 01.
Article in English | MEDLINE | ID: mdl-28148841

ABSTRACT

Multicellular interactions of platelets, leukocytes, and the blood vessel wall support coagulation and precipitate arterial and venous thrombosis. High levels of angiotensin II cause arterial hypertension by a complex vascular inflammatory pathway that requires leukocyte recruitment and reactive oxygen species production and is followed by vascular dysfunction. We delineate a previously undescribed, proinflammatory coagulation-vascular circuit that is a major regulator of vascular tone, blood pressure, and endothelial function. In mice with angiotensin II-induced hypertension, tissue factor was up-regulated, as was thrombin-dependent endothelial cell vascular cellular adhesion molecule 1 expression and integrin αMß2- and platelet-dependent leukocyte adhesion to arterial vessels. The resulting vascular inflammation and dysfunction was mediated by activation of thrombin-driven factor XI (FXI) feedback, independent of factor XII. The FXI receptor glycoprotein Ibα on platelets was required for this thrombin feedback activation in angiotensin II-infused mice. Inhibition of FXI synthesis with an antisense oligonucleotide was sufficient to prevent thrombin propagation on platelets, vascular leukocyte infiltration, angiotensin II-induced endothelial dysfunction, and arterial hypertension in mice and rats. Antisense oligonucleotide against FXI also reduced the increased blood pressure and attenuated vascular and kidney dysfunction in rats with established arterial hypertension. Further, platelet-localized thrombin generation was amplified in an FXI-dependent manner in patients with uncontrolled arterial hypertension, suggesting that platelet-localized thrombin generation may serve as an inflammatory marker of high blood pressure. Our results outline a coagulation-inflammation circuit that promotes vascular dysfunction, and highlight the possible utility of FXI-targeted anticoagulants in treating hypertension, beyond their application as antithrombotic agents in cardiovascular disease.


Subject(s)
Blood Coagulation , Blood Platelets/cytology , Factor XI/physiology , Hypertension/physiopathology , Thrombin/physiology , Aged , Angiotensin II , Animals , Blood Pressure , Factor XI/antagonists & inhibitors , Female , Humans , Hypertension/chemically induced , Macrophage-1 Antigen/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Oligonucleotides, Antisense/pharmacology , Platelet Glycoprotein GPIb-IX Complex/metabolism , Rats, Wistar , Thromboplastin/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
18.
Free Radic Biol Med ; 109: 48-60, 2017 08.
Article in English | MEDLINE | ID: mdl-28108279

ABSTRACT

Oxidative stress is a major hallmark of cardiovascular diseases although a causal link was so far not proven by large clinical trials. However, there is a close association between oxidative stress and inflammation and increasing evidence for a causal role of (low-grade) inflammation for the onset and progression of cardiovascular diseases, which may serve as the missing link between oxidative stress and cardiovascular morbidity and mortality. With the present review we would like to highlight the multiple redox regulated pathways in inflammation, discuss the sources of reactive oxygen and nitrogen species that are of interest for these processes and finally discuss the importance of angiotensin II (AT-II) as a trigger of cardiovascular inflammation and the initiation and progression of cardiovascular diseases.


Subject(s)
Cardiovascular Diseases/metabolism , Cardiovascular System/metabolism , Mitochondria/metabolism , Reactive Nitrogen Species/metabolism , Reactive Oxygen Species/metabolism , Angiotensin II/genetics , Angiotensin II/immunology , Cardiovascular Diseases/genetics , Cardiovascular Diseases/immunology , Cardiovascular Diseases/pathology , Cardiovascular System/immunology , Cardiovascular System/pathology , Endothelial Cells/immunology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Extracellular Traps/immunology , Extracellular Traps/metabolism , Gene Expression Regulation , Humans , Inflammation , Mitochondria/immunology , NADPH Oxidases/genetics , NADPH Oxidases/immunology , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Oxidation-Reduction , Oxidative Stress , Reactive Nitrogen Species/immunology , Reactive Oxygen Species/immunology , Signal Transduction
19.
Methods Mol Biol ; 1559: 439-453, 2017.
Article in English | MEDLINE | ID: mdl-28063062

ABSTRACT

Vascular inflammation in cardiovascular diseases is recognized to be linked with immune cell activation. Recruitment of immune cells into the vessel wall is an early step in angiotensin II-induced vascular dysfunction and arterial hypertension. Exploring the role of monocytes and macrophages in angiotensin II-induced hypertension and vascular inflammation in mouse models highlights the importance of these pathophysiological processes. Here we describe our routinely used protocols concerning angiotensin II-induced hypertension, assessment of blood pressure, vascular function, and immune cell infiltration.


Subject(s)
Angiotensin II/administration & dosage , Aorta/physiopathology , Endothelium, Vascular/physiopathology , Hypertension/physiopathology , Vasculitis/physiopathology , Acetylcholine/pharmacology , Animals , Aorta/drug effects , Aorta/immunology , Blood Pressure , Blood Pressure Monitors , Cell Separation/methods , Drug Administration Schedule , Endothelium, Vascular/drug effects , Endothelium, Vascular/immunology , Hypertension/chemically induced , Hypertension/immunology , Infusion Pumps, Implantable , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Mice , Monocytes/drug effects , Monocytes/immunology , Monocytes/pathology , Organ Culture Techniques , Osmotic Pressure , Vasculitis/chemically induced , Vasculitis/immunology
20.
Sci Rep ; 6: 32554, 2016 09 14.
Article in English | MEDLINE | ID: mdl-27624556

ABSTRACT

Alcoholic cardiomyopathy (ACM) resulting from excess alcohol consumption is an important cause of heart failure (HF). Although it is assumed that the cardiotoxicity of the ethanol (EtOH)-metabolite acetaldehyde (ACA) is central for its development and progression, the exact mechanisms remain obscure. Murine cardiomyocytes (CMs) exposed to ACA or EtOH showed increased superoxide (O2(•-)) levels and decreased mitochondrial polarization, both being normalized by NADPH oxidase (NOX) inhibition. C57BL/6 mice and mice deficient for the ACA-degrading enzyme mitochondrial aldehyde dehydrogenase (ALDH-2(-/-)) were fed a 2% EtOH diet for 5 weeks creating an ACA-overload. 2% EtOH-fed ALDH-2(-/-) mice exhibited a decreased cardiac function, increased heart-to-body and lung-to-body weight ratios, increased cardiac levels of the lipid peroxidation product malondialdehyde (MDA) as well as increased NOX activity and NOX2/glycoprotein 91(phox) (NOX2/gp91(phox)) subunit expression compared to 2% EtOH-fed C57BL/6 mice. Echocardiography revealed that ALDH-2(-/-)/gp91(phox-/-) mice were protected from ACA-overload-induced HF after 5 weeks of 2% EtOH-diet, demonstrating that NOX2-derived O2(•-) contributes to the development of ACM. Translated to human pathophysiology, we found increased gp91(phox) expression in endomyocardial biopsies of ACM patients. In conclusion, ACM is promoted by ACA-driven mitochondrial dysfunction and can be improved by ablation of NOX2/gp91(phox). NOX2/gp91(phox) therefore might be a potential pharmacological target to treat ACM.


Subject(s)
Cardiomyopathy, Alcoholic/genetics , Heart Failure/genetics , NADPH Oxidase 2/genetics , Acetaldehyde/toxicity , Aldehyde Dehydrogenase, Mitochondrial/genetics , Animals , Cardiomyopathy, Alcoholic/pathology , Disease Models, Animal , Ethanol/toxicity , Gene Expression Regulation/drug effects , Heart Failure/chemically induced , Heart Failure/pathology , Humans , Mice , Mitochondria/drug effects , Mitochondria/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Reactive Oxygen Species/metabolism , Superoxides/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...