Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
J Am Heart Assoc ; 11(16): e024581, 2022 08 16.
Article in English | MEDLINE | ID: mdl-35929448

ABSTRACT

Background Arteriovenous fistula (AVF) maturation failure is a main limitation of vascular access. Maturation is determined by the intricate balance between outward remodeling and intimal hyperplasia, whereby endothelial cell dysfunction, platelet aggregation, and vascular smooth muscle cell (VSMC) proliferation play a crucial role. von Willebrand Factor (vWF) is an endothelial cell-derived protein involved in platelet aggregation and VSMC proliferation. We investigated AVF vascular remodeling in vWF-deficient mice and vWF expression in failed and matured human AVFs. Methods and Results Jugular-carotid AVFs were created in wild-type and vWF-/- mice. AVF flow was determined longitudinally using ultrasonography, whereupon AVFs were harvested 14 days after surgery. VSMCs were isolated from vena cavae to study the effect of vWF on VSMC proliferation. Patient-matched samples of the basilic vein were obtained before brachio-basilic AVF construction and during superficialization or salvage procedure 6 weeks after AVF creation. vWF deficiency reduced VSMC proliferation and macrophage infiltration in the intimal hyperplasia. vWF-/- mice showed reduced outward remodeling (1.5-fold, P=0.002) and intimal hyperplasia (10.2-fold, P<0.0001). AVF flow in wild-type mice was incremental over 2 weeks, whereas flow in vWF-/- mice did not increase, resulting in a two-fold lower flow at 14 days compared with wild-type mice (P=0.016). Outward remodeling in matured patient AVFs coincided with increased local vWF expression in the media of the venous outflow tract. Absence of vWF in the intimal layer correlated with an increase in the intima-media ratio. Conclusions vWF enhances AVF maturation because its positive effect on outward remodeling outweighs its stimulating effect on intimal hyperplasia.


Subject(s)
Arteriovenous Fistula , Arteriovenous Shunt, Surgical , Myocytes, Smooth Muscle , von Willebrand Factor , Animals , Arteriovenous Shunt, Surgical/methods , Cell Proliferation , Humans , Hyperplasia , Mice , Mice, Knockout , Myocytes, Smooth Muscle/cytology , von Willebrand Factor/metabolism
2.
J Thromb Haemost ; 20(4): 996-1007, 2022 04.
Article in English | MEDLINE | ID: mdl-35037739

ABSTRACT

BACKGROUND: Atrial fibrillation (AF) can lead to the loss of microvascular integrity thereby enhancing AF progression. Mechanistically, the pro-coagulant state that drives the risk of stroke in patients with AF may also play a causal role in microvascular loss. Direct oral anticoagulants (DOACs), the preferred anticoagulants for AF, can target factors upstream (factor Xa [FXa]) or downstream (thrombin) in the coagulation cascade and mediate differential vascular effects through interaction with protease-activated receptors (PARs). OBJECTIVE: To investigate the potential effect of different DOACs on vascular integrity. METHODS: To model the impact of DOACs on vascular integrity, we utilized platelet-free plasma in thrombin generation assays and endothelial barrier assays under identical experimental conditions. These multifactorial systems provide all coagulation factors and their respective natural inhibitors in physiological ratios in combination with the pro-coagulant endothelial surface on which coagulation is initiated. Furthermore, the system provides pro- and anti-barrier factors and monitoring both assays simultaneously permits coupling of thrombin kinetics to endothelial barrier dynamics. RESULTS: We provide evidence that the anti-FXa DOAC rivaroxaban and the anti-thrombin DOAC dabigatran are efficient in blocking their target proteases. However, while rivaroxaban could preserve endothelial barrier function, dabigatran failed to protect endothelial integrity over time, which could be prevented in the presence of a custom-made peptide that blocks thrombin's exosite-I. CONCLUSIONS: Proteolytically inactive thrombin in complex with dabigatran evokes loss of barrier function that can be prevented by a protease-activated receptor-1 mimicking peptide blocking thrombin's exosite-I.


Subject(s)
Atrial Fibrillation , Dabigatran , Administration, Oral , Anticoagulants/therapeutic use , Atrial Fibrillation/drug therapy , Dabigatran/adverse effects , Factor Xa/therapeutic use , Factor Xa Inhibitors/adverse effects , Humans , Receptor, PAR-1 , Rivaroxaban/adverse effects , Thrombin/therapeutic use
3.
Int J Mol Sci ; 21(17)2020 Sep 02.
Article in English | MEDLINE | ID: mdl-32887275

ABSTRACT

Atherosclerosis is the underlying pathology in a major part of cardiovascular disease, the leading cause of mortality in developed countries. The infiltration of monocytes into the vessel walls of large arteries is a key denominator of atherogenesis, making monocytes accountable for the development of atherosclerosis. With the development of high-throughput transcriptome profiling platforms and cytometric methods for circulating cells, it is now feasible to study in-depth the predicted functional change of circulating monocytes reflected by changes of gene expression in certain pathways and correlate the changes to disease outcome. Neuroimmune guidance cues comprise a group of circulating- and cell membrane-associated signaling proteins that are progressively involved in monocyte functions. Here, we employed the CIRCULATING CELLS study cohort to classify cardiovascular disease patients and healthy individuals in relation to their expression of neuroimmune guidance cues in circulating monocytes. To cope with the complexity of human datasets featured by noisy data, nonlinearity and multidimensionality, we assessed various machine-learning methods. Of these, the linear discriminant analysis, Naïve Bayesian model and stochastic gradient boost model yielded perfect or near-perfect sensibility and specificity and revealed that expression levels of the neuroimmune guidance cues SEMA6B, SEMA6D and EPHA2 in circulating monocytes were of predictive values for cardiovascular disease outcome.


Subject(s)
Biomarkers/blood , Cardiovascular Diseases/diagnosis , Ephrins/blood , Machine Learning , Monocytes/metabolism , Netrin-1/blood , Semaphorins/blood , Adult , Cardiovascular Diseases/blood , Cardiovascular Diseases/genetics , Case-Control Studies , Cohort Studies , Ephrins/genetics , Female , Humans , Male , Middle Aged , Netrin-1/genetics , Semaphorins/genetics , Transcriptome
4.
J Thromb Haemost ; 18(7): 1714-1727, 2020 07.
Article in English | MEDLINE | ID: mdl-32297475

ABSTRACT

BACKGROUND: Genome wide association studies (GWAS) identified SLC44A2 as a novel susceptibility gene for venous thrombosis (VT) and previous work established that SLC44A2 contributed to clot formation upon vascular injury. OBJECTIVE: To further investigate the role of SLC44A2 in VT by utilizing SLC44A2 deficient mice (Slc44a2-/- ) in two representative disease models. METHODS: Mice were included in a hypercoagulability model driven by siRNA-mediated hepatic gene silencing of anticoagulants Serpinc1 (antithrombin) and Proc (protein C) and a flow restriction (stenosis) model induced by partial ligation of the inferior vena cava. RESULTS: In the hypercoagulability model, no effect in onset was observed in Slc44a2-/- animals; however, a drop in plasma fibrinogen and von Willebrand factor coinciding with an increase in blood neutrophils was recorded. In the neutrophil dependent stenosis model after 48 hours, Slc44a2-/- mice had significantly smaller thrombi both in length and weight with less platelet accumulation as a percentage of the total thrombus area. During the initiation of thrombosis at 6 hours post-stenosis, Slc44a2-/- mice also had smaller thrombi both in length and weight, with circulating platelets remaining elevated in Slc44a2-/- animals. Platelet activation and aggregation under both static- and venous and arterial shear conditions were normal for blood from Slc44a2-/- mice. CONCLUSIONS: These studies corroborate the original GWAS findings and establish a contributing role for SLC44A2 during the initiation of VT, with indications that this may be related to platelet-neutrophil interaction. The precise mechanism however remains elusive and warrants further investigation.


Subject(s)
Thrombophilia , Venous Thrombosis , Animals , Blood Platelets , Constriction, Pathologic , Disease Models, Animal , Genome-Wide Association Study , Membrane Transport Proteins/genetics , Mice , Platelet Activation , Thrombophilia/genetics , Venous Thrombosis/genetics
5.
Cells ; 9(4)2020 03 29.
Article in English | MEDLINE | ID: mdl-32235347

ABSTRACT

We recently reported that loss of hyaluronan (HA) from the endothelial glycocalyx leads to loss of vessel stability in specific microcirculatory vascular beds. Here we hypothesized that such derangements in the glycocalyx may also impair the adaptive response to vascular ischemia. Endothelial specific conditional hyaluronan synthase 2-KO (Has2-cKO) mice revealed reduced endothelial HA expression and lower hindlimb perfusion at baseline compared to control mice. After a single ligation of the common femoral artery in these mice, we observed dysregulated angiogenesis in the gastrocnemius muscle which did not restore capillary perfusion. Mechanistically, decreased endothelial binding of the pericyte-derived molecule angiopoietin1 (Ang1) could be observed in the Has2-cKO mouse. In vitro angiogenesis assays with an endothelial cell-pericyte coculture confirmed such disturbed Ang1-TIE2 signaling resulting in excessive angiogenesis upon loss of HA. These data could be of relevance to diabetes patients, where we confirm loss of endothelial HA in the microcirculation of muscle tissue, indicating that this may contribute to the known disturbed adaptation to ischemia in these patients. In summary, loss of endothelial HA results in impaired microvascular perfusion and endothelial stability in ischemic gastrocnemius muscle. Endothelial HA is a potential target to improve angiogenic therapy in diabetic patients with critical limb ischemia.


Subject(s)
Endothelial Cells/metabolism , Femoral Artery/pathology , Femoral Artery/physiopathology , Glycocalyx/metabolism , Ischemia/pathology , Ischemia/physiopathology , Vascular Remodeling , Angiopoietin-1/metabolism , Animals , Diabetes Mellitus, Experimental/pathology , Hindlimb/pathology , Humans , Hyaluronic Acid/metabolism , Ligation , Mice, Inbred C57BL , Mice, Knockout , Muscles/pathology , Neovascularization, Physiologic , Perfusion
6.
Epigenomes ; 4(1)2020 Feb 13.
Article in English | MEDLINE | ID: mdl-34968236

ABSTRACT

In the pathophysiologic setting of acute and chronic kidney injury, the excessive activation and recruitment of blood-borne monocytes prompts their differentiation into inflammatory macrophages, a process that leads to progressive glomerulosclerosis and interstitial fibrosis. Importantly, this differentiation of monocytes into macrophages requires the meticulous coordination of gene expression at both the transcriptional and post-transcriptional level. The transcriptomes of these cells are ultimately determined by RNA-binding proteins such as QUAKING (QKI), that define their pre-mRNA splicing and mRNA transcript patterns. Using two mouse models, namely (1) quaking viable mice (qkv) and (2) the conditional deletion in the myeloid cell lineage using the lysozyme 2-Cre (QKIFL/FL;LysM-Cre mice), we demonstrate that the abrogation of QKI expression in the myeloid cell lineage reduces macrophage infiltration following kidney injury induced by unilateral urethral obstruction (UUO). The qkv and QKIFL/FL;LysM-Cre mice both showed significant diminished interstitial collagen deposition and fibrosis in the UUO-damaged kidney, as compared to wild-type littermates. We show that macrophages isolated from QKIFL/FL;LysM-Cre mice are associated with defects in pre-mRNA splicing. Our findings demonstrate that reduced expression of the alternative splice regulator QKI in the cells of myeloid lineage attenuates renal interstitial fibrosis, suggesting that inhibition of this splice regulator may be of therapeutic value for certain kidney diseases.

7.
JBMR Plus ; 3(11): e10230, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31768489

ABSTRACT

Fibrodysplasia ossificans progressiva (FOP) is an extremely rare congenital form of heterotopic ossification (HO), caused by heterozygous mutations in the activin A type I receptor (ACVR1), that encodes the bone morphogenetic protein (BMP) type I receptor ALK2. These mutations enable ALK2 to induce downstream signaling in response to activins, thereby turning them into bone-inducing agents. To date, there is no cure for FOP. The further development of FOP patient-derived models may contribute to the discovery of novel biomarkers and therapeutic approaches. Nevertheless, this has traditionally been a challenge, as biopsy sampling often triggers HO. We have characterized peripheral blood-derived endothelial colony-forming cells (ECFCs) from three independent FOP donors as a new model for FOP. FOP ECFCs are prone to undergo endothelial-to-mesenchymal transition and exhibit increased ALK2 downstream signaling and subsequent osteogenic differentiation upon stimulation with activin A. Moreover, we have identified a new class of small molecule macrocycles with potential activity against ALK2 kinase. Finally, using FOP ECFCs, we have selected OD36 and OD52 as potent inhibitors with excellent kinase selectivity profiles that potently antagonize mutant ALK2 signaling and osteogenic differentiation. We expect that these results will contribute to the development of novel ALK2 clinical candidates for the treatment of FOP. © 2019 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.

8.
PLoS One ; 14(6): e0217582, 2019.
Article in English | MEDLINE | ID: mdl-31199833

ABSTRACT

BACKGROUND: Hypercholesterolemia is a major risk factor for ischemic heart disease including acute myocardial infarction. However, long-term effects of hypercholesterolemia in a rodent myocardial ischemia-reperfusion injury model are unknown. Therefore, the effects of diet-induced hypercholesterolemia on cardiac function and remodeling were investigated up to eight weeks after myocardial ischemia-reperfusion (MI-R) injury which was induced in either normocholesterolemic (NC-MI) or hypercholesterolemic (HC-MI) APOE*3-Leiden mice. METHODS: Left ventricular (LV) dimensions were serially assessed using parasternal long-axis echocardiography followed by LV pressure-volume measurements. Subsequently, infarct size and the inflammatory response were analyzed by histology and fluorescence-activated cell sorting (FACS) analysis. RESULTS: Intrinsic LV function eight weeks after MI-R was significantly impaired in HC-MI compared to NC-MI mice as assessed by end-systolic pressure, dP/dtMAX, and -dP/dtMIN. Paradoxically, infarct size was significantly decreased in HC-MI compared to NC-MI mice, accompanied by an increased wall thickness. Hypercholesterolemia caused a pre-ischemic peripheral monocytosis, in particular of Ly-6Chi monocytes whereas accumulation of macrophages in the ischemic-reperfused myocardium of HC-MI mice was decreased. CONCLUSION: Diet-induced hypercholesterolemia caused impaired LV function eight weeks after MI-R injury despite a reduced post-ischemic infarct size. This was preceded by a pre-ischemic peripheral monocytosis, while there was a suppressed accumulation of inflammatory cells in the ischemic-reperfused myocardium after eight weeks. This experimental model using hypercholesterolemic APOE*3-Leiden mice exposed to MI-R seems suitable to study novel cardioprotective therapies in a more clinically relevant animal model.


Subject(s)
Apolipoprotein E3 , Heart Ventricles , Hypercholesterolemia , Myocardial Reperfusion Injury , Ventricular Function, Left , Animals , Apolipoprotein E3/genetics , Apolipoprotein E3/metabolism , Female , Heart Ventricles/metabolism , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Hypercholesterolemia/genetics , Hypercholesterolemia/metabolism , Hypercholesterolemia/pathology , Hypercholesterolemia/physiopathology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Mice , Mice, Transgenic , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Time Factors
9.
Circulation ; 138(23): 2698-2712, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30571259

ABSTRACT

BACKGROUND: Hereditary Hemorrhagic Telangiectasia type 2 (HHT2) is an inherited genetic disorder characterized by vascular malformations and hemorrhage. HHT2 results from ACVRL1 haploinsufficiency, the remaining wild-type allele being unable to contribute sufficient protein to sustain endothelial cell function. Blood vessels function normally but are prone to respond to angiogenic stimuli, leading to the development of telangiectasic lesions that can bleed. How ACVRL1 haploinsufficiency leads to pathological angiogenesis is unknown. METHODS: We took advantage of Acvrl1+/- mutant mice that exhibit HHT2 vascular lesions and focused on the neonatal retina and the airway system after Mycoplasma pulmonis infection, as physiological and pathological models of angiogenesis, respectively. We elucidated underlying disease mechanisms in vitro by generating Acvrl1+/- mouse embryonic stem cell lines that underwent sprouting angiogenesis and performed genetic complementation experiments. Finally, HHT2 plasma samples and skin biopsies were analyzed to determine whether the mechanisms evident in mice are conserved in humans. RESULTS: Acvrl1+/- retinas at postnatal day 7 showed excessive angiogenesis and numerous endothelial "tip cells" at the vascular front that displayed migratory defects. Vascular endothelial growth factor receptor 1 (VEGFR1; Flt-1) levels were reduced in Acvrl1+/- mice and HHT2 patients, suggesting similar mechanisms in humans. In sprouting angiogenesis, VEGFR1 is expressed in stalk cells to inhibit VEGFR2 (Flk-1, KDR) signaling and thus limit tip cell formation. Soluble VEGFR1 (sVEGFR1) is also secreted, creating a VEGF gradient that promotes orientated sprout migration. Acvrl1+/- embryonic stem cell lines recapitulated the vascular anomalies in Acvrl1+/- (HHT2) mice. Genetic insertion of either the membrane or soluble form of VEGFR1 into the ROSA26 locus of Acvrl1+/- embryonic stem cell lines prevented the vascular anomalies, suggesting that high VEGFR2 activity in Acvrl1+/- endothelial cells induces HHT2 vascular anomalies. To confirm our hypothesis, Acvrl1+/- mice were infected by Mycoplasma pulmonis to induce sustained airway inflammation. Infected Acvrl1+/- tracheas showed excessive angiogenesis with the formation of multiple telangiectases, vascular defects that were prevented by VEGFR2 blocking antibodies. CONCLUSIONS: Our findings demonstrate a key role of VEGFR1 in HHT2 pathogenesis and provide mechanisms explaining why HHT2 blood vessels respond abnormally to angiogenic signals. This supports the case for using anti-VEGF therapy in HHT2.


Subject(s)
Telangiectasia, Hereditary Hemorrhagic/pathology , Vascular Endothelial Growth Factor Receptor-1/metabolism , Activin Receptors, Type I/genetics , Activin Receptors, Type II , Adult , Animals , Antibodies/administration & dosage , Antibodies/immunology , Arteriovenous Malformations/etiology , Disease Models, Animal , Female , Heterozygote , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Mouse Embryonic Stem Cells/cytology , Mouse Embryonic Stem Cells/metabolism , Mycoplasma pulmonis/physiology , Neovascularization, Physiologic , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Retinal Vessels/physiology , Signal Transduction , Skin/pathology , Telangiectasia, Hereditary Hemorrhagic/metabolism , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/immunology
10.
PLoS One ; 12(12): e0187118, 2017.
Article in English | MEDLINE | ID: mdl-29206835

ABSTRACT

We recently demonstrated that the adult human kidney cortex contains a perivascular stromal cell (kPSC) that shows organotypic properties and is important for repair and stabilisation of kidney function. Not only the kidney cortex but also the kidney capsule contains stromal cells that are important for the three dimensional organisation of the kidney during nephrogenesis. They provide the barrier function of the capsule which is critical for homeostatic processes such as pressure natriuresis. We postulated that stromal cells derived from the kidney capsule may therefore also have specific properties and functions. To this end, we isolated these capsule mesenchymal stromal cells (cMSC) from human cadaveric kidneys that were not suitable for transplantation. There were several similarities between cMSCs and kPSCs including support of vascular plexus formation, phenotypic marker expression and resistance against myofibroblast transformation. However, compared to kPSCs, cMSCs showed distinct mRNA and miRNA expression profiles, showed increased immunosuppressive capacity, and displayed strongly reduced HGF production, contributing to the inability to enhance kidney epithelial repair. Therefore cMSCs are a distinct, novel human kidney-derived MSC-population and these data underpin the large functional diversity of phenotypic similar stromal cells in relation to their anatomic site, even within one organ.


Subject(s)
Kidney/cytology , Mesenchymal Stem Cells/cytology , Cell Differentiation , Cell Lineage , Cells, Cultured , Coculture Techniques , Gene Expression Profiling , Humans , Kidney/metabolism , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , RNA, Messenger/genetics , Transforming Growth Factor beta/metabolism
11.
PLoS One ; 12(9): e0183741, 2017.
Article in English | MEDLINE | ID: mdl-28910300

ABSTRACT

BACKGROUND: COPD is a pulmonary disorder often accompanied by cardiovascular disease (CVD), and current treatment of this comorbidity is suboptimal. Systemic inflammation in COPD triggered by smoke and microbial exposure is suggested to link COPD and CVD. Mesenchymal stromal cells (MSC) possess anti-inflammatory capacities and MSC treatment is considered an attractive treatment option for various chronic inflammatory diseases. Therefore, we investigated the immunomodulatory properties of MSC in an acute and chronic model of lipopolysaccharide (LPS)-induced inflammation, emphysema and atherosclerosis development in APOE*3-Leiden (E3L) mice. METHODS: Hyperlipidemic E3L mice were intranasally instilled with 10 µg LPS or vehicle twice in an acute 4-day study, or twice weekly during 20 weeks Western-type diet feeding in a chronic study. Mice received 0.5x106 MSC or vehicle intravenously twice after the first LPS instillation (acute study) or in week 14, 16, 18 and 20 (chronic study). Inflammatory parameters were measured in bronchoalveolar lavage (BAL) and lung tissue. Emphysema, pulmonary inflammation and atherosclerosis were assessed in the chronic study. RESULTS: In the acute study, intranasal LPS administration induced a marked systemic IL-6 response on day 3, which was inhibited after MSC treatment. Furthermore, MSC treatment reduced LPS-induced total cell count in BAL due to reduced neutrophil numbers. In the chronic study, LPS increased emphysema but did not aggravate atherosclerosis. Emphysema and atherosclerosis development were unaffected after MSC treatment. CONCLUSION: These data show that MSC inhibit LPS-induced pulmonary and systemic inflammation in the acute study, whereas MSC treatment had no effect on inflammation, emphysema and atherosclerosis development in the chronic study.


Subject(s)
Atherosclerosis/therapy , Interleukin-6/metabolism , Lipopolysaccharides/adverse effects , Pneumonia/therapy , Pulmonary Emphysema/therapy , Administration, Intranasal , Animals , Atherosclerosis/chemically induced , Atherosclerosis/immunology , Bronchoalveolar Lavage Fluid/immunology , Disease Models, Animal , Female , Lipopolysaccharides/administration & dosage , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Mice , Pneumonia/chemically induced , Pneumonia/immunology , Pulmonary Emphysema/chemically induced , Pulmonary Emphysema/immunology , Treatment Outcome
12.
Am J Pathol ; 187(11): 2430-2440, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28837800

ABSTRACT

Inhibition of monocyte chemotactic protein-1 (MCP-1) with the Spiegelmer emapticap pegol (NOX-E36) shows long-lasting albuminuria-reducing effects in diabetic nephropathy. MCP-1 regulates inflammatory cell recruitment and differentiation of macrophages. Because the endothelial glycocalyx is also reduced in diabetic nephropathy, we hypothesized that MCP-1 inhibition restores glomerular barrier function through influencing macrophage cathepsin L secretion, thus reducing activation of the glycocalyx-degrading enzyme heparanase. Four weeks of treatment of diabetic Apoe knockout mice with the mouse-specific NOX-E36 attenuated albuminuria without any change in systemic hemodynamics, despite persistent loss of podocyte function. MCP-1 inhibition, however, increased glomerular endothelial glycocalyx coverage, with preservation of heparan sulfate. Mechanistically, both glomerular cathepsin L and heparanase expression were reduced. MCP-1 inhibition resulted in reduced CCR2-expressing Ly6Chi monocytes in the peripheral blood, without affecting overall number of kidney macrophages at the tissue level. However, the CD206+/Mac3+ cell ratio, as an index of presence of anti-inflammatory macrophages, increased in diabetic mice after treatment. Functional analysis of isolated renal macrophages showed increased release of IL-10, whereas tumor necrosis factor and cathepsin L release was reduced, further confirming polarization of tissue macrophages toward an anti-inflammatory phenotype during mouse-specific NOX-E36 treatment. We show that MCP-1 inhibition restores glomerular endothelial glycocalyx and barrier function and reduces tissue inflammation in the presence of ongoing diabetic injury, suggesting a therapeutic potential for NOX-E36 in diabetic nephropathy.


Subject(s)
Chemokine CCL2/metabolism , Diabetic Nephropathies/metabolism , Glycocalyx/metabolism , Macrophages/metabolism , Podocytes/metabolism , Animals , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/pathology , Kidney/pathology , Male , Mice, Knockout , Monocytes/metabolism
13.
Stem Cells Transl Med ; 6(2): 405-418, 2017 02.
Article in English | MEDLINE | ID: mdl-28191776

ABSTRACT

Mesenchymal stromal cells (MSCs) are immunomodulatory and tissue homeostatic cells that have shown beneficial effects in kidney diseases and transplantation. Perivascular stromal cells (PSCs) identified within several different organs share characteristics of bone marrow-derived MSCs (BM-MSCs). These PSCs may also possess tissue-specific properties and play a role in local tissue homeostasis. We hypothesized that human kidney-derived PSCs (hkPSCs) would elicit improved kidney repair in comparison with BM-MSCs. Here we introduce a novel, clinical-grade isolation method of hkPSCs from cadaveric kidneys by enriching for the perivascular marker, NG2. hkPSCs show strong transcriptional similarities to BM-MSCs but also show organotypic expression signatures, including the HoxD10 and HoxD11 nephrogenic transcription factors. Comparable to BM-MSCs, hkPSCs showed immunosuppressive potential and, when cocultured with endothelial cells, vascular plexus formation was supported, which was specifically in the hkPSCs accompanied by an increased NG2 expression. hkPSCs did not undergo myofibroblast transformation after exposure to transforming growth factor-ß, further corroborating their potential regulatory role in tissue homeostasis. This was further supported by the observation that hkPSCs induced accelerated repair in a tubular epithelial wound scratch assay, which was mediated through hepatocyte growth factor release. In vivo, in a neonatal kidney injection model, hkPSCs reintegrated and survived in the interstitial compartment, whereas BM-MSCs did not show this potential. Moreover, hkPSCs gave protection against the development of acute kidney injury in vivo in a model of rhabdomyolysis-mediated nephrotoxicity. Overall, this suggests a superior therapeutic potential for the use of hkPSCs and their secretome in the treatment of kidney diseases. Stem Cells Translational Medicine 2017;6:405-418.


Subject(s)
Cell Separation/methods , Kidney Diseases/surgery , Kidney Transplantation/methods , Kidney/blood supply , Kidney/cytology , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Regeneration , Regenerative Medicine/methods , Animals , Antigens/metabolism , Biomarkers/metabolism , Cell Differentiation , Cell Lineage , Cell Movement , Cell Proliferation , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Genotype , Homeodomain Proteins/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells/physiology , Humans , Kidney Diseases/metabolism , Kidney Diseases/pathology , Kidney Diseases/physiopathology , Male , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Neovascularization, Physiologic , Phenotype , Proteoglycans/metabolism , Transcription Factors/metabolism
14.
Eur Heart J ; 38(1): 38-50, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27071821

ABSTRACT

AIMS: Atrial fibrillation (AF) produces a hypercoagulable state. Stimulation of protease-activated receptors by coagulation factors provokes pro-fibrotic, pro-hypertrophic, and pro-inflammatory responses in a variety of tissues. We studied the effects of thrombin on atrial fibroblasts and tested the hypothesis that hypercoagulability contributes to the development of a substrate for AF. METHODS AND RESULTS: In isolated rat atrial fibroblasts, thrombin enhanced the phosphorylation of the pro-fibrotic signalling molecules Akt and Erk and increased the expression of transforming growth factor ß1 (2.7-fold) and the pro-inflammatory factor monocyte chemoattractant protein-1 (6.1-fold). Thrombin also increased the incorporation of 3H-proline, suggesting enhanced collagen synthesis by fibroblasts (2.5-fold). All effects could be attenuated by the thrombin inhibitor dabigatran. In transgenic mice with a pro-coagulant phenotype (TMpro/pro), the inducibility of AF episodes lasting >1 s was higher (7 out of 12 vs. 1 out of 10 in wild type) and duration of AF episodes was longer compared with wild type mice (maximum episode duration 42.8 ± 68.4 vs. 0.23 ± 0.39 s). In six goats with persistent AF treated with nadroparin, targeting Factor Xa-mediated thrombin generation, the complexity of the AF substrate was less pronounced than in control animals (LA maximal activation time differences 23.3 ± 3.1 ms in control vs. 15.7 ± 2.1 ms in nadroparin, P < 0.05). In the treated animals, AF-induced α-smooth muscle actin expression was lower and endomysial fibrosis was less pronounced. CONCLUSION: The hypercoagulable state during AF causes pro-fibrotic and pro-inflammatory responses in adult atrial fibroblasts. Hypercoagulability promotes the development of a substrate for AF in transgenic mice and in goats with persistent AF. In AF goats, nadroparin attenuates atrial fibrosis and the complexity of the AF substrate. Inhibition of coagulation may not only prevent strokes but also inhibit the development of a substrate for AF.


Subject(s)
Atrial Fibrillation/etiology , Receptors, Thrombin/drug effects , Thrombin/pharmacology , Thrombophilia/physiopathology , Analysis of Variance , Animals , Antithrombins/pharmacology , Cell Proliferation/drug effects , Dabigatran/pharmacology , Female , Fibrinolytic Agents/pharmacology , Fibroblasts/drug effects , Fibrosis/etiology , Goats , Heart Atria/pathology , Indazoles/pharmacology , Mice, Transgenic , Nadroparin/pharmacology , Peptide Hydrolases/drug effects , Pyrroles/pharmacokinetics , Quinazolines/pharmacokinetics , Rats , Urea/analogs & derivatives , Urea/pharmacology
15.
Tissue Eng Part C Methods ; 22(10): 923-931, 2016 10.
Article in English | MEDLINE | ID: mdl-27604583

ABSTRACT

Elastin, critical for its structural and regulatory functions, is a missing link in vascular tissue engineering. Several elastin-inducting compounds have previously been reported, but their relative efficiency in promoting elastogenesis by adult arterial and venous vascular smooth muscle cells (VSMCs) and fibroblasts, four main vascular and elastogenic cells, has not been described. In addition to elasto-inductive substances, microRNA-29a was recently established as a potent post-transcriptional inhibitor of elastogenesis. Here, we explored if stimulating positive regulators or blocking inhibitors of elastogenesis could maximize elastin production. We tested whether the elasto-inducing compounds IGF-1, TGF-ß1, and minoxidil could indeed augment elastin production, and whether microRNA-29a antagonism could block elastin production in adult arterial and venous fibroblasts and VSMCs. The effects on elastin, lysyl oxidase, and fibrillin-1 mRNA expression levels and tropoelastin protein were determined. IGF-1 and minoxidil exerted little effect on tropoelastin mRNA expression levels in all cell types, while TGF-ß1 predominantly enhanced mRNA tropoelastin levels, but this mRNA increase did not impact tropoelastin protein abundance. In contrast, microRNA29a inhibition resulted in the upregulation of tropoelastin mRNA in all cell types, but most pronounced in venous VSMCs. Importantly, microRNA-29a-antagonism also enhanced lysyl oxidase and fibrillin-1 mRNA expression, and revealed a dose-dependent increase in tropoelastin protein expression in venous VSMCs. Our studies suggest that the elastogenic potential of microRNA-29a inhibition in vascular cells is superior to that of established elastin-stimulating compounds IGF-1, TGF-ß1, and minoxidil. Thus, microRNA-29a antagonism could serve as an attractive means of enhancing elastin synthesis in tissue-engineered blood vessels.


Subject(s)
Arteries/metabolism , Fibroblasts/metabolism , Gene Expression Regulation/drug effects , Muscle, Smooth, Vascular/metabolism , Tissue Engineering/methods , Tropoelastin/metabolism , Veins/metabolism , Animals , Arteries/cytology , Cells, Cultured , Female , Fibroblasts/cytology , Insulin-Like Growth Factor I/pharmacology , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Minoxidil/pharmacology , Muscle, Smooth, Vascular/cytology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Swine , Transforming Growth Factor beta1/pharmacology , Tropoelastin/genetics , Vasodilator Agents/pharmacology , Veins/cytology
16.
Nat Commun ; 7: 10846, 2016 Mar 31.
Article in English | MEDLINE | ID: mdl-27029405

ABSTRACT

A hallmark of inflammatory diseases is the excessive recruitment and influx of monocytes to sites of tissue damage and their ensuing differentiation into macrophages. Numerous stimuli are known to induce transcriptional changes associated with macrophage phenotype, but posttranscriptional control of human macrophage differentiation is less well understood. Here we show that expression levels of the RNA-binding protein Quaking (QKI) are low in monocytes and early human atherosclerotic lesions, but are abundant in macrophages of advanced plaques. Depletion of QKI protein impairs monocyte adhesion, migration, differentiation into macrophages and foam cell formation in vitro and in vivo. RNA-seq and microarray analysis of human monocyte and macrophage transcriptomes, including those of a unique QKI haploinsufficient patient, reveal striking changes in QKI-dependent messenger RNA levels and splicing of RNA transcripts. The biological importance of these transcripts and requirement for QKI during differentiation illustrates a central role for QKI in posttranscriptionally guiding macrophage identity and function.


Subject(s)
Macrophages/physiology , Monocytes/physiology , RNA Splicing , RNA-Binding Proteins/physiology , Animals , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Differentiation , Foam Cells/cytology , Foam Cells/metabolism , Gene Expression Regulation , Humans , Macrophages/cytology , Macrophages/metabolism , Mice , Models, Biological , Models, Genetic , Monocytes/cytology , Monocytes/metabolism , RNA, Messenger/metabolism , RNA, Messenger/physiology , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
17.
Sci Rep ; 6: 21643, 2016 Feb 24.
Article in English | MEDLINE | ID: mdl-26905650

ABSTRACT

Proper regulation of endothelial cell-cell contacts is essential for physiological functioning of the endothelium. Interendothelial junctions are actively involved in the control of vascular leakage, leukocyte diapedesis, and the initiation and progression of angiogenesis. We found that the RNA-binding protein quaking is highly expressed by endothelial cells, and that its expression was augmented by prolonged culture under laminar flow and the transcription factor KLF2 binding to the promoter. Moreover, we demonstrated that quaking directly binds to the mRNA of VE-cadherin and ß-catenin and can induce mRNA translation mediated by the 3'UTR of these genes. Reduced quaking levels attenuated VE-cadherin and ß-catenin expression and endothelial barrier function in vitro and resulted in increased bradykinin-induced vascular leakage in vivo. Taken together, we report that quaking is essential in maintaining endothelial barrier function. Our results provide novel insight into the importance of post-transcriptional regulation in controlling vascular integrity.


Subject(s)
Antigens, CD/genetics , Cadherins/genetics , Human Umbilical Vein Endothelial Cells/physiology , RNA-Binding Proteins/physiology , beta Catenin/genetics , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Capillary Permeability , Female , Gene Expression , HEK293 Cells , Humans , Kruppel-Like Transcription Factors/physiology , Mice, Inbred C57BL , Mice, Transgenic , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , Transcriptional Activation , beta Catenin/metabolism
19.
Curr Vasc Pharmacol ; 13(3): 341-51, 2015.
Article in English | MEDLINE | ID: mdl-23713864

ABSTRACT

MicroRNAs are negative regulators of gene expression that have been shown to be essential elements in the coordination of complex regulatory pathways. One of these short non-coding RNAs, microRNA-126, is highly enriched in the vascular endothelium and was shown to play distinct roles in angiogenesis, vasculogenesis and endothelial inflammation. Abrogation of this microRNA leads to severe complications in the response in vascular development as well as vital repair mechanisms carried out by endothelial cells. Interestingly, recent data suggest that the homeostatic role of microRNA-126 may reach far beyond its endothelial functions as this microRNA was also found to be present in cells of the hematopoietic system and in microvesicles or 'free-form' in the periphery. MicroRNA-126 is controlling the fate and/or function of a variety of cells differentiating from the hematopoietic lineage, including megakaryocytes and erythrocytes. Recent studies identified circulating microRNA-126 as a biomarker for myocardial injury and vascular damage in diabetes. Furthermore, reports have suggested a protective role of circulating microRNA-126 in murine models of organ ischemia. Here, we review current insights in the role of microRNA-126 in vascular homeostasis and conclude that this microRNA may serve to integrate and facilitate both local as well as systemic functions in vascular maintenance and repair.


Subject(s)
Blood Vessels/physiology , Homeostasis/genetics , Homeostasis/physiology , MicroRNAs/physiology , Animals , Humans , Mice , MicroRNAs/genetics , Vascular Diseases/genetics , Vascular Diseases/pathology
20.
Microrna ; 3(3): 144-9, 2014.
Article in English | MEDLINE | ID: mdl-25541911

ABSTRACT

Integrity of the capillary network in the kidney is essential in the recovery from ischemia/ reperfusion injury (IRI), a phenomenon central to kidney transplantation and acute kidney injury. MicroRNA- 126 (miR-126) is known to be important in maintaining vascular homeostasis by facilitating vascular regeneration and modulating the mobilization of vascular progenitor cells. Stromal cell-derived factor 1 (SDF-1), important in the mobilization of vascular progenitor cells, is a direct target of miR-126 and modulation of miR-126 was previously shown to affect the number of circulating Sca-1(+)/Lin(-) vascular progenitor cells in a mouse model for hind limb ischemia. Here, we assessed the in vivo contribution of miR-126 to progenitor cell mobilization and kidney function following IRI in mice. A three day follow up of blood urea levels following kidney IRI demonstrated that systemic antagomir silencing of miR-126 did not impact the loss or subsequent restoration of kidney function. However, whole kidney lysates displayed elevated gene expression levels of Sdf-1, Vegf-A and eNOS after IRI as a result of systemic silencing of miR-126. Furthermore, FACS-analysis on whole blood three days after surgery revealed a marked up regulation of the number of circulating Sca-1(+)/Lin(-) progenitor cells in the antagomir-126 treated mice, in an ischemia dependent manner. Our data indicate that silencing of miR-126 can enhance renal expression of Sdf-1 after IRI, leading to the mobilization of vascular progenitor cells into the circulation.


Subject(s)
Antigens, Ly/metabolism , Chemokine CXCL12/metabolism , Endothelial Progenitor Cells/metabolism , Kidney/metabolism , Membrane Proteins/metabolism , MicroRNAs/metabolism , Reperfusion Injury/metabolism , Animals , Antigens, Ly/genetics , Chemokine CXCL12/genetics , Endothelial Progenitor Cells/cytology , Gene Silencing , Kidney/blood supply , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...