20 research outputs found

    Systemic Monocyte Chemotactic Protein-1 Inhibition Modifies Renal Macrophages and Restores Glomerular Endothelial Glycocalyx and Barrier Function in Diabetic Nephropathy

    No full text
    Contains fulltext : 182393.pdf (Publisher’s version ) (Open Access)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 Ly6C(hi) 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

    Protease-activated receptor (PAR)2, but not PAR1, is involved in collateral formation and anti-inflammatory monocyte polarization in a mouse hind limb ischemia model

    Get PDF
    In collateral development (i.e. arteriogenesis), mononuclear cells are important and exist as a heterogeneous population consisting of pro-inflammatory and anti-inflammatory/repair-associated cells. Protease-activated receptor (PAR)1 and PAR2 are G-protein-coupled receptors that are both expressed by mononuclear cells and are involved in pro-inflammatory reactions, while PAR2 also plays a role in repair-associated responses. Here, we investigated the physiological role of PAR1 and PAR2 in arteriogenesis in a murine hind limb ischemia model. PAR1-deficient (PAR1-/-), PAR2-deficient (PAR2-/-) and wild-type (WT) mice underwent femoral artery ligation. Laser Doppler measurements revealed reduced post-ischemic blood flow recovery in PAR2-/- hind limbs when compared to WT, while PAR1-/- mice were not affected. Upon ischemia, reduced numbers of smooth muscle actin (SMA)-positive collaterals and CD31-positive capillaries were found in PAR2-/- mice when compared to WT mice, whereas these parameters in PAR1-/- mice did not differ from WT mice. The pool of circulating repair-associated (Ly6C-low) monocytes and the number of repair-associated (CD206-positive) macrophages surrounding collaterals in the hind limbs were increased in WT and PAR1-/- mice, but unaffected in PAR2-/- mice. The number of repair-associated macrophages in PAR2-/- hind limbs correlated with CD11b- and CD115-expression on the circulating monocytes in these animals, suggesting that monocyte extravasation and M-CSF-dependent differentiation into repair-associated cells are hampered. PAR2, but not PAR1, is involved in arteriogenesis and promotes the repair-associated response in ischemic tissues. Therefore, PAR2 potentially forms a new pro-arteriogenic target in coronary artery disease (CAD) patient

    SLC44A2 deficient mice have a reduced response in stenosis but not in hypercoagulability driven venous thrombosis

    Get PDF
    International audienceBackground 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

    TLR4 Accessory Molecule RP105 (CD180) Regulates Monocyte-Driven Arteriogenesis in a Murine Hind Limb Ischemia Model

    No full text
    <div><p>Aims</p><p>We investigated the role of the TLR4-accessory molecule RP105 (CD180) in post-ischemic neovascularization, i.e. arteriogenesis and angiogenesis. TLR4-mediated activation of pro-inflammatory Ly6C<sup>hi</sup> monocytes is crucial for effective neovascularization. Immunohistochemical analyses revealed that RP105<sup>+</sup> monocytes are present in the perivascular space of remodeling collateral arterioles. As RP105 inhibits TLR4 signaling, we hypothesized that RP105 deficiency would lead to an unrestrained TLR4-mediated inflammatory response and hence to enhanced blood flow recovery after ischemia.</p><p>Methods and Results</p><p>RP105<sup>−/−</sup> and wild type (WT) mice were subjected to hind limb ischemia and blood flow recovery was followed by Laser Doppler Perfusion Imaging. Surprisingly, we found that blood flow recovery was severely impaired in RP105<sup>−/−</sup> mice. Immunohistochemistry showed that arteriogenesis was reduced in these mice compared to the WT. However, both in vivo and ex vivo analyses showed that circulatory pro-arteriogenic Ly6C<sup>hi</sup> monocytes were more readily activated in RP105<sup>−/−</sup> mice. FACS analyses showed that Ly6C<sup>hi</sup> monocytes became activated and migrated to the affected muscle tissues in WT mice following induction of hind limb ischemia. Although Ly6C<sup>hi</sup> monocytes were readily activated in RP105<sup>−/−</sup> mice, migration into the ischemic tissues was hampered and instead, Ly6C<sup>hi</sup> monocytes accumulated in their storage compartments, bone marrow and spleen, in RP105<sup>−/−</sup> mice.</p><p>Conclusions</p><p>RP105 deficiency results in an unrestrained inflammatory response and monocyte over-activation, most likely due to the lack of TLR4 regulation. Inappropriate, premature systemic activation of pro-inflammatory Ly6C<sup>hi</sup> monocytes results in reduced infiltration of Ly6C<sup>hi</sup> monocytes in ischemic tissues and in impaired blood flow recovery.</p></div

    Blood flow recovery in RP105<sup>−/−</sup> mice.

    Get PDF
    <p>(<b>A</b>) Representative Laser Doppler Perfusion Imaging (LDPI) images of paws from WT and RP105<sup>−/−</sup> mice after induction of HLI in the left limb. High blood flow is displayed in red. (<b>B</b>) Quantification of LDPI measurements of RP105<sup>−/−</sup> (n = 10) and WT (n = 9) mice over time. Data are calculated as the ratio of ligated over non-ligated paw. (<b>C</b>) Quantification of LDPI measurements of WT and RP105<sup>−/−</sup> mice directly after induction of HLI. (<b>D</b>) Quantification of LDPI measurements 10 days after induction of HLI. (<b>E</b>) Immunohistochemical staining of paraffin-embedded adductor muscle group of WT (n = 6) and RP105<sup>−/−</sup> (n = 6) mice, 10 days after HLI, using anti-αSMA (red) antibodies. Smallest lumen diameter of αSMA<sup>+</sup> vessels is indicated by black bars. (<b>F</b>) Immunohistochemical staining on fresh frozen sections of gastrocnemius muscles of WT (n = 6) and RP105<sup>−/−</sup> (n = 6) mice, 10 days after HLI, using anti-CD31 (brown) antibodies. Number (<b>G</b>) and lumen area (µm<sup>2</sup>) (<b>H</b>) of αSMA<sup>+</sup> vessels, measured at the center of the adductor muscle group in ligated and non-ligated limbs of RP105<sup>−/−</sup> and WT mice. (<b>I</b>) Capillary density in gastrocnemius muscles, defined as the number of CD31<sup>+</sup> vessels per section. pt = pre-treatment. ns = non-significant. All values are presented as the mean ± SEM. *P<0.05, **P<0.01, ***P<0.001.</p

    Revascularization upon ischemia is PAR2-dependent.

    No full text
    <p>(A) SMA staining was performed to detect collaterals in the ischemic adductor muscles and subsequently quantified. Mean collateral number in WT mice was compared to PAR1-/- mice (B) and PAR2-/- mice. (C) Capillary density was quantified after CD31 staining of ischemic calf muscles. Mean capillary number of WT was compared to PAR1-/- mice (D) and PAR2-/- mice. (E) Images represent the CD31 and SMA stainings of ischemic hind limbs of WT, PAR1-/- and PAR2-/- mice. Arrowheads indicate vessels. Bar, 100 µm. * p<0.05.</p

    Increased accumulation of repair-associated macrophages surrounding collaterals in ischemic hind limbs is PAR2-dependent.

    No full text
    <p>(A) Stainings of CD206-positive macrophages (green) and SMA-positive vessels (red) in non-ischemic (control) and ischemic (ligated) hind limbs of WT, PAR1-/- and PAR2-/- mice are shown. Nuclei were visualized with DAPI (blue). Arrows indicate single macrophages in the non-ischemic adductor. Quantification of the average number of repair-associated macrophages per vessel is indicated on the right. (B) Correlation between the number of CD206-positive macrophages in the ischemic tissues and the expression of CD11b and (C) CD115 on monocytes. ** p<0.01, *** p<0.001.</p
    corecore