Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Int J Mol Sci ; 25(1)2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38203193

ABSTRACT

Improved human-relevant preclinical models of coronary artery disease (CAD) are needed to improve translational research and drug discovery. Mitochondrial dysfunction and associated oxidative stress contribute to endothelial dysfunction and are a significant factor in the development and progression of CAD. Endothelial colony-forming cells (ECFCs) can be derived from peripheral blood mononuclear cells (PBMCs) and offer a unique potentially personalised means for investigating new potential therapies targeting important components of vascular function. We describe the application of the high-throughput and confocal Opera Phenix® High-Content Screening System to examine mitochondrial superoxide (mROS) levels, mitochondrial membrane potential, and mitochondrial area in both established cell lines and patient-derived ECFCs simultaneously. Unlike traditional plate readers, the Opera Phenix® is an imaging system that integrates automated confocal microscopy, precise fluorescent detection, and multi-parameter algorithms to visualize and precisely quantify targeted biological processes at a cellular level. In this study, we measured mROS production in human umbilical vein endothelial cells (HUVECs) and patient-derived ECFCs using the mROS production probe, MitoSOXTM Red. HUVECs exposed to oxidized low-density lipoprotein (oxLDL) increased mROS levels by 47.7% (p < 0.0001). A pooled group of patient-derived ECFCs from participants with CAD (n = 14) exhibited 30.9% higher mROS levels compared to patients with no CAD when stimulated with oxLDL (n = 14; p < 0.05). When tested against a small group of candidate compounds, this signal was attenuated by PKT-100 (36.22% reduction, p = 0.03), a novel P2X7 receptor antagonist. This suggests the P2X7 receptor as a valid target against excess mROS levels. As such, these findings highlight the potential of the MitoSOX-Opera Phenix technique to be used for drug discovery efforts in CAD.


Subject(s)
Coronary Artery Disease , Humans , Coronary Artery Disease/drug therapy , Superoxides , Leukocytes, Mononuclear , Mitochondria , Human Umbilical Vein Endothelial Cells
2.
Antioxidants (Basel) ; 10(10)2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34679682

ABSTRACT

Mechanisms involved in the individual susceptibility to atherosclerotic coronary artery disease (CAD) beyond traditional risk factors are poorly understood. Here, we describe the utility of cultured patient-derived endothelial colony-forming cells (ECFCs) in examining novel mechanisms of CAD susceptibility, particularly the role of dysregulated redox signalling. ECFCs were selectively cultured from peripheral blood mononuclear cells from 828 patients from the BioHEART-CT cohort, each with corresponding demographic, clinical and CT coronary angiographic imaging data. Spontaneous growth occurred in 178 (21.5%) patients and was more common in patients with hypertension (OR 1.45 (95% CI 1.03-2.02), p = 0.031), and less likely in patients with obesity (OR 0.62 [95% CI 0.40-0.95], p = 0.027) or obstructive CAD (stenosis > 50%) (OR 0.60 [95% CI 0.38-0.95], p = 0.027). ECFCs from patients with CAD had higher mitochondrial production of superoxide (O2--MitoSOX assay). The latter was strongly correlated with the severity of CAD as measured by either coronary artery calcium score (R2 = 0.46; p = 0.0051) or Gensini Score (R2 = 0.67; p = 0.0002). Patient-derived ECFCs were successfully cultured in 3D culture pulsatile mini-vessels. Patient-derived ECFCs can provide a novel resource for discovering mechanisms of CAD disease susceptibility, particularly in relation to mitochondrial redox signalling.

4.
Am J Physiol Heart Circ Physiol ; 319(1): H183-H191, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32469637

ABSTRACT

In pulmonary hypertension (PH) a proinflammatory milieu drives pulmonary vascular remodeling, maladaptive right ventricular (RV) remodeling, and right-sided heart failure. There is an unmet need for RV-targeted pharmaco-therapies to improve mortality. Targeting of the P2X7 receptor (P2X7R) reduces pulmonary pressures; however, its effects on the RV are presently unknown. We investigated the effect of P2X7 receptor (P2X7R) inhibition on the pulmonary vasculature and RV remodeling using the novel P2X7R antagonist PKT100. C57BL/6 mice were administered intratracheal bleomycin or saline and treated with PKT100 (0.2 mg·kg-1·day-1) or DMSO vehicle. RV was assessed by right heart catheterization and echocardiography, 21 days posttreatment. Cytokines in serum and bronchoalveolar lavage fluid (BALF) were analyzed by ELISA and flow cytometry. Lungs and hearts were analyzed histologically for pulmonary vascular and RV remodeling. Focused-PCR using genes involved in RV remodeling was performed. Right ventricular systolic pressure (RVSP) was elevated in bleomycin-treated mice (30.2 ± 1.1; n = 7) compared with control mice (23.5 ± 1.0; n = 10; P = 0.008). PKT100 treatment did not alter RVSP (32.4 ± 1.8; n = 9), but it substantially improved survival (93% vs. 57% DMSO). There were no differences between DMSO and PKT100 bleomycin mice in pulmonary inflammation or remodeling. However, RV hypertrophy was reduced in PKT100 mice. Bleomycin decreased echocardiographic surrogates of RV systolic performance, which were significantly improved with PKT100. Four genes involved in RV remodeling (RPSA, Rplp0, Add2, and Scn7a) were differentially expressed between DMSO and PKT100-treated groups. The novel P2X7R inhibitor, PKT100, attenuates RV hypertrophy and improves RV contractile function and survival in a mouse model of PH independently of effects on the pulmonary vasculature. PKT100 may improve ventricular response to increased afterload and merits further investigation into the potential role of P2X7R antagonists as direct RV-focused therapies in PH.NEW & NOTEWORTHY This study demonstrates the therapeutic potential for right-sided heart failure of a novel inhibitor of the P2X7 receptor (P2X7R). Inflammatory signaling and right ventricular function were improved in a mouse model of pulmonary fibrosis with secondary pulmonary hypertension when treated with this inhibitor. Importantly, survival was also improved, suggesting that this inhibitor, and other P2X7R antagonists, could be uniquely effective in right ventricle (RV)-targeted therapy in pulmonary hypertension. This addresses a major limitation of current treatment options, where the significant improvements in pulmonary pressures ultimately do not prevent mortality due to RV failure.


Subject(s)
Heart Ventricles/drug effects , Hypertension, Pulmonary/drug therapy , Purinergic P2X Receptor Antagonists/therapeutic use , Animals , Blood Pressure , Bronchoalveolar Lavage Fluid/cytology , Cytokines/blood , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Heart Ventricles/metabolism , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Lung/blood supply , Lung/drug effects , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Purinergic P2X Receptor Antagonists/pharmacology , Receptors, Laminin/genetics , Receptors, Laminin/metabolism , Receptors, Purinergic P2X7/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Ventricular Remodeling , Voltage-Gated Sodium Channels/genetics , Voltage-Gated Sodium Channels/metabolism
5.
Mol Ther Nucleic Acids ; 17: 49-62, 2019 Sep 06.
Article in English | MEDLINE | ID: mdl-31220779

ABSTRACT

MicroRNA-15a (miR-15a) and miR-16, which are transcribed from the miR-15a/miR-16-1 cluster, inhibit post-ischemic angiogenesis. MicroRNA (miRNA) binding to mRNA coding sequences (CDSs) is a newly emerging mechanism of gene expression regulation. We aimed to (1) identify new mediators of the anti-angiogenic action of miR-15a and -16, (2) develop an adenovirus (Ad)-based miR-15a/16 decoy system carrying a luciferase reporter (Luc) to both sense and inhibit miR-15a/16 activity, and (3) investigate Ad.Luc-Decoy-15a/16 therapeutic potential in a mouse limb ischemia (LI) model. LI increased miR-15a and -16 expression in mouse muscular endothelial cells (ECs). The miRNAs also increased in cultured human umbilical vein ECs (HUVECs) exposed to serum starvation, but not hypoxia. Using bioinformatic tools and luciferase activity assays, we characterized miR-15a and -16 binding to Tie2 CDS. In HUVECs, miR-15a or -16 overexpression reduced Tie2 at the protein, but not the mRNA, level. Conversely, miR-15a or -16 inhibition improved angiogenesis in a Tie2-dependent manner. Local Ad.Luc-Decoy-15a/16 delivery increased Tie2 levels in ischemic skeletal muscle and improved post-LI angiogenesis and perfusion recovery, with reduced toe necrosis. Bioluminescent imaging (in vivo imaging system [IVIS]) provided evidence that the Ad.Luc-Decoy-15a/16 system responds to miR-15a/16 increases. In conclusion, we have provided novel mechanistic evidence of the therapeutic potential of local miR-15a/16 inhibition in LI.

6.
Mol Ther ; 26(7): 1694-1705, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29908843

ABSTRACT

Therapies based on circulating proangiogenic cells (PACs) have shown promise in ischemic disease models but require further optimization to reach the bedside. Ischemia-associated hypoxia robustly increases microRNA-210 (miR-210) expression in several cell types, including endothelial cells (ECs). In ECs, miR-210 represses EphrinA3 (EFNA3), inducing proangiogenic responses. This study provides new mechanistic evidences for a role of miR-210 in PACs. PACs were obtained from either adult peripheral blood or cord blood. miR-210 expression was modulated with either an inhibitory complementary oligonucleotide (anti-miR-210) or a miRNA mimic (pre-miR-210). Scramble and absence of transfection served as controls. As expected, hypoxia increased miR-210 in PACs. In vivo, migration toward and adhesion to the ischemic endothelium facilitate the proangiogenic actions of transplanted PACs. In vitro, PAC migration toward SDF-1α/CXCL12 was impaired by anti-miR-210 and enhanced by pre-miR-210. Moreover, pre-miR-210 increased PAC adhesion to ECs and supported angiogenic responses in co-cultured ECs. These responses were not associated with changes in extracellular miR-210 and were abrogated by lentivirus-mediated EFNA3 overexpression. Finally, ex-vivo pre-miR-210 transfection predisposed PACs to induce post-ischemic therapeutic neovascularization and blood flow recovery in an immunodeficient mouse limb ischemia model. In conclusion, miR-210 modulates PAC functions and improves their therapeutic potential in limb ischemia.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow/physiology , Hindlimb/cytology , Ischemia/genetics , Ischemia/therapy , MicroRNAs/genetics , Neovascularization, Physiologic/physiology , Adult , Animals , Cell Line , Chemokine CXCL12/genetics , Endothelial Cells/cytology , Female , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Inbred C57BL , Neovascularization, Pathologic/genetics , Neovascularization, Physiologic/genetics , Transfection/methods
7.
Mol Ther ; 26(7): 1669-1684, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29703701

ABSTRACT

Pluripotent stem cell-derived differentiated endothelial cells offer high potential in regenerative medicine in the cardiovascular system. With the aim of translating the use of a human stem cell-derived endothelial cell product (hESC-ECP) for treatment of critical limb ischemia (CLI) in man, we report a good manufacturing practice (GMP)-compatible protocol and detailed cell tracking and efficacy data in multiple preclinical models. The clinical-grade cell line RC11 was used to generate hESC-ECP, which was identified as mostly endothelial (60% CD31+/CD144+), with the remainder of the subset expressing various pericyte/mesenchymal stem cell markers. Cell tracking using MRI, PET, and qPCR in a murine model of limb ischemia demonstrated that hESC-ECP was detectable up to day 7 following injection. Efficacy in several murine models of limb ischemia (immunocompromised/immunocompetent mice and mice with either type I/II diabetes mellitus) demonstrated significantly increased blood perfusion and capillary density. Overall, we demonstrate a GMP-compatible hESC-ECP that improved ischemic limb perfusion and increased local angiogenesis without engraftment, paving the way for translation of this therapy.


Subject(s)
Endothelial Cells/cytology , Hindlimb/cytology , Ischemia/therapy , Neovascularization, Physiologic/physiology , Animals , Biomarkers/metabolism , Cell Differentiation/physiology , Cell Line , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endothelial Cells/metabolism , Hindlimb/metabolism , Humans , Ischemia/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Pericytes/cytology , Pericytes/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Stem Cell Transplantation/methods
8.
Am J Physiol Heart Circ Physiol ; 314(6): H1279-H1288, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29569957

ABSTRACT

We have previously shown that protein tyrosine phosphatase 1B (PTP1B) inactivation in mice [PTP1B-deficient (PTP1B-/-) mice] improves left ventricular (LV) angiogenesis, perfusion, remodeling, and function and limits endothelial dysfunction after myocardial infarction. However, whether PTP1B inactivation slows aging-associated cardiovascular dysfunction remains unknown. Wild-type (WT) and PTP1B-/- mice were allowed to age until 18 mo. Compared with old WT mice, in which aging increased the LV mRNA expression of PTP1B, old PTP1B-/- mice had 1) reduced cardiac hypertrophy with decreased LV mRNA levels of hypertrophic markers and atrial and brain natriuretic peptides, 2) lower LV fibrosis (collagen: 16 ± 3% in WT mice and 5 ± 3% in PTP1B-/- mice, P < 0.001) with decreased mRNA levels of transforming growth-factor-ß1 and matrix metalloproteinase-2, and 3) higher LV capillary density and lower LV mRNA level of hypoxic inducible factor-1α, which was associated over time with a higher rate of proangiogenic M2 type macrophages and a stable LV mRNA level of VEGF receptor-2. Echocardiography revealed an age-dependent LV increase in end-diastolic volume in WT mice together with alterations of fractional shortening and diastole (transmitral Doppler E-to-A wave ratio). Invasive hemodynamics showed better LV systolic contractility and better diastolic compliance in old PTP1B-/- mice (LV end-systolic pressure-volume relation: 13.9 ± 0.9 in WT mice and 18.4 ± 1.6 in PTP1B-/- mice; LV end-diastolic pressure-volume relation: 5.1 ± 0.8 mmHg/relative volume unit in WT mice and 1.2 ± 0.3 mmHg/relative volume unit in PTP1B-/- mice, P < 0.05). In addition, old PTP1B-/- mice displayed a reduced amount of LV reactive oxygen species. Finally, in isolated resistance mesenteric arteries, PTP1B inactivation reduced aging-associated endothelial dysfunction (flow-mediated dilatation: -0.4 ± 2.1% in WT mice and 8.2 ± 2.8% in PTP1B-/- mice, P < 0.05). We conclude that PTP1B inactivation slows aging-associated LV remodeling and dysfunction and reduces endothelial dysfunction in mesenteric arteries. NEW & NOTEWORTHY The present study shows that protein tyrosine phosphatase 1B inactivation in aged mice improves left ventricular systolic and diastolic function associated with reduced adverse cardiac remodeling (hypertrophy, fibrosis, and capillary rarefaction) and limits vascular endothelial dysfunction. This suggests that protein tyrosine phosphatase 1B inhibition could be an interesting treatment approach in age-related cardiovascular dysfunction.


Subject(s)
Heart Failure/prevention & control , Heart Ventricles/enzymology , Hypertrophy, Left Ventricular/prevention & control , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Ventricular Dysfunction, Left/prevention & control , Ventricular Function, Left , Ventricular Remodeling , Age Factors , Aging/genetics , Aging/metabolism , Animals , Disease Models, Animal , Fibrosis , Gene Expression Regulation, Enzymologic , Heart Failure/enzymology , Heart Failure/genetics , Heart Failure/physiopathology , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Hemodynamics , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/physiopathology , Male , Mesenteric Arteries/enzymology , Mesenteric Arteries/physiopathology , Mice, Inbred BALB C , Mice, Knockout , Neovascularization, Physiologic , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/physiopathology
9.
Nat Commun ; 8(1): 747, 2017 09 29.
Article in English | MEDLINE | ID: mdl-28963481

ABSTRACT

Several cell-based therapies are under pre-clinical and clinical evaluation for the treatment of ischemic diseases. Poor survival and vascular engraftment rates of transplanted cells force them to work mainly via time-limited paracrine actions. Although several approaches, including the use of soluble vascular endothelial growth factor (sVEGF)-VEGF165, have been developed in the last 10 years to enhance cell survival, they showed limited efficacy. Here, we report a pro-survival approach based on VEGF-immobilized microparticles (VEGF-MPs). VEGF-MPs prolong VEGFR-2 and Akt phosphorylation in cord blood-derived late outgrowth endothelial progenitor cells (OEPCs). In vivo, OEPC aggregates containing VEGF-MPs show higher survival than those treated with sVEGF. Additionally, VEGF-MPs decrease miR-17 expression in OEPCs, thus increasing the expression of its target genes CDKN1A and ZNF652. The therapeutic effect of OEPCs is improved in vivo by inhibiting miR-17. Overall, our data show an experimental approach to improve therapeutic efficacy of proangiogenic cells for the treatment of ischemic diseases.Soluble vascular endothelial growth factor (VEGF) enhances vascular engraftment of transplanted cells but the efficacy is low. Here, the authors show that VEGF-immobilized microparticles prolong survival of endothelial progenitors in vitro and in vivo by downregulating miR17 and upregulating CDKN1A and ZNF652.


Subject(s)
Cell Survival , Endothelial Progenitor Cells/metabolism , MicroRNAs/metabolism , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Cell- and Tissue-Based Therapy , Cell-Derived Microparticles , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA-Binding Proteins/genetics , Endothelial Progenitor Cells/cytology , Fetal Blood/cytology , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells , Humans , Ischemia/metabolism , Ischemia/therapy , Neovascularization, Physiologic , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism
10.
Mol Ther ; 25(3): 679-693, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28159509

ABSTRACT

The pericardial fluid (PF) is contained in the pericardial sac surrounding the heart. MicroRNA (miRNA) exchange via exosomes (endogenous nanoparticles) contributes to cell-to-cell communication. We investigated the hypotheses that the PF is enriched with miRNAs secreted by the heart and that it mediates vascular responses through exosome exchange of miRNAs. The study was developed using leftover material from aortic valve surgery. We found that in comparison with peripheral plasma, the PF contains exosomes enriched with miRNAs co-expressed in patients' myocardium and vasculature. At a functional level, PF exosomes improved survival, proliferation, and networking of cultured endothelial cells (ECs) and restored the angiogenic capacity of ECs depleted (via Dicer silencing) of their endogenous miRNA content. Moreover, PF exosomes improved post-ischemic blood flow recovery and angiogenesis in mice. Mechanistically, (1) let-7b-5p is proangiogenic and inhibits its target gene, TGFBR1, in ECs; (2) PF exosomes transfer a functional let-7b-5p to ECs, thus reducing their TGFBR1 expression; and (3) let-7b-5p depletion in PF exosomes impairs the angiogenic response to these nanoparticles. Collectively, our data support the concept that PF exosomes orchestrate vascular repair via miRNA transfer.


Subject(s)
Cardiovascular System/metabolism , Exosomes/metabolism , MicroRNAs/genetics , Neovascularization, Pathologic , Neovascularization, Physiologic , Pericardial Fluid/metabolism , Animals , Argonaute Proteins/genetics , Endothelial Cells/metabolism , Extracellular Vesicles/metabolism , Gene Expression Profiling , Humans , Male , Mice , Ribonuclease III/genetics
11.
J Mol Cell Cardiol ; 101: 50-57, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27596049

ABSTRACT

Protein Tyrosine Phosphatase 1B (PTP1B) is mostly involved in negative regulation of signaling mediated by Tyrosine Kinase Receptors, especially the insulin and leptin receptors. This enzyme thus plays a major role in the development of diseases associated with insulin resistance, such as obesity and diabetes. PTP1B inhibition is currently considered as an attractive treatment of insulin resistance and associated metabolic disorders. In parallel, emerging evidence also suggests that PTP1B is widely expressed in cardiovascular tissues, notably in the heart and the endothelium, and that it could also be a potential treatment of several cardiovascular diseases. PTP1B is especially present in endothelial cells, and appears to contribute to endothelial dysfunction. Indeed, preclinical evidence shows that pharmacological inhibition of gene deletion of PTP1B reduces endothelial dysfunction in various cardiovascular diseases associated or not with insulin resistance. In parallel, because PTP1B also negatively modulates VEGF signaling, inhibition of this enzyme also tends to favor cardiac angiogenesis. Importantly, blocking PTP1B also results in beneficial effects on cardiac dysfunction and remodeling not only in metabolic diseases but also in the context of heart failure, thus this enzyme represents an attractive new target for the treatment of this disease. This beneficial effect in heart failure may to a large extent result from the endothelial protective and/or proangiogenic effects of PTP1B blockade. Finally, PTP1B inhibition also reduces cardiac dysfunction, but also systemic inflammation and mortality in experimental models of septic shock, and thus may also constitute a new treatment of this disease. Altogether, accumulating preclinical evidence suggests that PTP1B represents an interesting molecular target to treat both cardiovascular and metabolic diseases, which often share the same risk factors. This concept now deserves to be tested in clinical studies that should soon be possible with the current development of selective PTP1B inhibitors.


Subject(s)
Cardiovascular Diseases/etiology , Cardiovascular Diseases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Animals , Cardiovascular Diseases/pathology , Cardiovascular Diseases/therapy , Cardiovascular Physiological Phenomena , Cardiovascular System/metabolism , Endothelial Cells/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Gene Expression Regulation , Gene Targeting , Humans , Multigene Family , Myocytes, Cardiac/metabolism , Myocytes, Smooth Muscle/metabolism , Protein Processing, Post-Translational , Protein Transport , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors
12.
Circ Heart Fail ; 9(4): e002895, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27059805

ABSTRACT

BACKGROUND: Chronic heart failure (CHF) induces endothelial dysfunction in part because of decreased nitric oxide (NO(·)) production, but the direct link between endothelial dysfunction and aggravation of CHF is not directly established. We previously reported that increased NO production via inhibition of protein tyrosine phosphatase 1B (PTP1B) is associated with reduced cardiac dysfunction in CHF. Investigation of the role of endothelial PTP1B in these effects may provide direct evidence of the link between endothelial dysfunction and CHF. METHODS AND RESULTS: Endothelial deletion of PTP1B was obtained by crossing LoxP-PTP1B with Tie2-Cre mice. CHF was assessed 4 months after myocardial infarction. In some experiments, to exclude gene extinction in hematopoietic cells, Tie2-Cre/LoxP-PTP1B mice were lethally irradiated and reconstituted with bone marrow from wild-type mice, to obtain mouse with endothelial-specific deletion of PTP1B. Vascular function evaluated ex vivo in mesenteric arteries showed that in wild-type mice, CHF markedly impaired NO-dependent flow-mediated dilatation. CHF-induced endothelial dysfunction was less marked in endoPTP1B(-/-) mice, suggesting restored NO production. Echocardiographic, hemodynamic, and histological evaluations demonstrated that the selectively improved endothelial function was associated with reduced left ventricular dysfunction and remodeling, as well as increased survival, in the absence of signs of stimulated angiogenesis or increased cardiac perfusion. CONCLUSIONS: Prevention of endothelial dysfunction, by endothelial PTP1B deficiency, is sufficient to reduce cardiac dysfunction post myocardial infarction. Our results provide for the first time a direct demonstration that endothelial protection per se reduces CHF and further suggest a causal role for endothelial dysfunction in CHF development.


Subject(s)
Endothelium, Vascular/enzymology , Heart Failure/prevention & control , Mesenteric Arteries/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Vasodilation , Ventricular Dysfunction, Left/prevention & control , Animals , Bone Marrow Transplantation , Chronic Disease , Disease Models, Animal , Endothelium, Vascular/physiopathology , Heart Failure/enzymology , Heart Failure/genetics , Heart Failure/physiopathology , Mesenteric Arteries/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/enzymology , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Nitric Oxide/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Signal Transduction , Time Factors , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left , Ventricular Remodeling
13.
Hypertension ; 67(4): 717-23, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26902493

ABSTRACT

Mineralocorticoid receptor (MR) antagonists slow down the progression of heart failure after myocardial infarction (MI), but the cell-specific role of MR in these benefits is unclear. In this study, the role of MR expressed in vascular smooth muscle cells (VSMCs) was investigated. Two months after coronary artery ligation causing MI, mice with VSMC-specific MR deletion (MI-MR(SMKO)) and mice treated with the MR antagonist finerenone (MI-fine) had improved left ventricular compliance and elastance when compared with infarcted control mice (MI-CTL), as well as reduced interstitial fibrosis. Importantly, the coronary reserve assessed by magnetic resonance imaging was preserved (difference in myocardial perfusion before and after induction of vasodilatation, mL mg(-1) min(-1): MI-CTL: 1.1 ± 0.5, nonsignificant; MI-MR(SMKO): 4.6 ± 1.6 [P<0.05]; MI-fine: 3.6 ± 0.7 [P<0.01]). The endothelial function, tested on isolated septal coronary arteries by analyzing the acetylcholine-induced nitric oxide-dependent relaxation, was also improved by MR deletion in VSMCs or by finerenone treatment (relaxation %: MI-CTL: 36 ± 5, MI-MR(SMKO): 54 ± 3, and MI-fine: 76 ± 4; P<0.05). Such impairment of the coronary endothelial function on MI involved an oxidative stress that was reduced when MR was deleted in VSMCs or by finerenone treatment. Moreover, short-term incubation of coronary arteries isolated from noninfarcted animals with low-dose angiotensin-II (10(-9) mol/L) induced oxidative stress and impaired acetylcholine-induced relaxation in CTL but neither in MR(SMKO) nor in mice pretreated with finerenone. In conclusion, deletion of MR in VSMCs improved left ventricular dysfunction after MI, likely through maintenance of the coronary reserve and improvement of coronary endothelial function. MR blockage by finerenone had similar effects.


Subject(s)
Mineralocorticoid Receptor Antagonists/pharmacology , Muscle, Smooth, Vascular/drug effects , Myocardial Infarction/complications , Naphthyridines/pharmacology , Ventricular Dysfunction, Left/drug therapy , Ventricular Dysfunction, Left/etiology , Animals , Disease Models, Animal , Disease Progression , Heart Function Tests , Magnetic Resonance Imaging/methods , Male , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/metabolism , Myocardial Infarction/diagnosis , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Random Allocation , Receptors, Mineralocorticoid/drug effects , Reference Values , Risk Assessment , Risk Factors , Treatment Outcome , Ventricular Dysfunction, Left/diagnosis , Ventricular Remodeling/drug effects , Ventricular Remodeling/physiology
14.
Nat Commun ; 6: 8024, 2015 Aug 13.
Article in English | MEDLINE | ID: mdl-26268439

ABSTRACT

The communication between vascular endothelial cells (ECs) and pericytes in the microvasculature is fundamental for vascular growth and homeostasis; however, these processes are disrupted by diabetes. Here we show that modulation of p75(NTR) expression in ECs exposed to high glucose activates transcription of miR-503, which negatively affects pericyte function. p75(NTR) activates NF-κB to bind the miR-503 promoter and upregulate miR-503 expression in ECs. NF-κB further induces activation of Rho kinase and shedding of endothelial microparticles carrying miR-503, which transfer miR-503 from ECs to vascular pericytes. The integrin-mediated uptake of miR-503 in the recipient pericytes reduces expression of EFNB2 and VEGFA, resulting in impaired migration and proliferation. We confirm operation of the above mechanisms in mouse models of diabetes, in which EC-derived miR-503 reduces pericyte coverage of capillaries, increased permeability and impaired post-ischaemic angiogenesis in limb muscles. Collectively, our data demonstrate that miR-503 regulates pericyte-endothelial crosstalk in microvascular diabetic complications.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Endothelial Cells/metabolism , MicroRNAs/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/genetics , Gene Expression Regulation/physiology , Hindlimb/blood supply , Humans , Ischemia , Male , Mice , Mice, Knockout , MicroRNAs/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Nerve Tissue Proteins/genetics , Oligonucleotide Array Sequence Analysis , Pericytes/physiology , Receptors, Nerve Growth Factor/genetics , Transcription, Genetic/physiology
15.
Prostaglandins Other Lipid Mediat ; 120: 148-54, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26022136

ABSTRACT

This study addressed the hypothesis that inhibition of the EETs degrading enzyme soluble epoxide hydrolase affords renal protection in the early stage of diabetic nephropathy. The renal effects of the sEH inhibitor t-AUCB (10mg/l in drinking water) were compared to those of the sulfonylurea glibenclamide (80mg/l), both administered for 8 weeks in FVB mice subjected to a high-fat diet (HFD, 60% fat) for 16 weeks. Mice on control chow diet (10% fat) and non-treated HFD mice served as controls. Compared with non-treated HFD mice, HFD mice treated with t-AUCB had a decreased EET degradation, as shown by their higher plasma EETs-to-DHETs ratio, and an increased EET production, as shown by the increase in EETs+DHETs levels, which was associated with induction of CYP450 epoxygenase expression. Both agents similarly reduced fasting glycemia but only t-AUCB prevented the increase in the urinary albumine-to-creatinine ratio in HFD mice. Histopathological analysis showed that t-AUCB reduced renal inflammation, which was associated with an increased mRNA expression of the NFκB inhibitor Iκ≡ and related decrease in MCP-1, COX2 and VCAM-1 expressions. Finally, there was a marginally significant increase in reactive oxygen species production in HFD mice, together with an enhanced NOX2 expression. Both agents did not modify these parameters but t-AUCB increased the expression of the antioxidant enzyme superoxide dismutase 1. These results demonstrate that, independently from its glucose-lowering effect, sEH inhibition prevents microalbuminuria and renal inflammation in overweight hyperglycemic mice, suggesting that this pharmacological strategy could be useful in the management of diabetic nephropathy.


Subject(s)
Blood Glucose/metabolism , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/antagonists & inhibitors , Epoxide Hydrolases/chemistry , Kidney/drug effects , Overweight/blood , Animals , Benzoates/pharmacology , Body Weight/drug effects , Diet, High-Fat/adverse effects , Eicosanoids/metabolism , Fasting/blood , Glyburide/pharmacology , Kidney/metabolism , Kidney/pathology , Kidney/physiopathology , Mice , Mice, Obese , Organ Size/drug effects , Overweight/metabolism , Overweight/pathology , Overweight/physiopathology , Oxidative Stress/drug effects , Solubility , Urea/analogs & derivatives , Urea/pharmacology
16.
Am J Physiol Heart Circ Physiol ; 308(9): H1020-9, 2015 May 01.
Article in English | MEDLINE | ID: mdl-25724490

ABSTRACT

This study addressed the hypothesis that inhibiting the soluble epoxide hydrolase (sEH)-mediated degradation of epoxy-fatty acids, notably epoxyeicosatrienoic acids, has an additional impact against cardiovascular damage in insulin resistance, beyond its previously demonstrated beneficial effect on glucose homeostasis. The cardiovascular and metabolic effects of the sEH inhibitor trans-4-[4-(3-adamantan-1-yl-ureido)-cyclohexyloxy]-benzoic acid (t-AUCB; 10 mg/l in drinking water) were compared with those of the sulfonylurea glibenclamide (80 mg/l), both administered for 8 wk in FVB mice subjected to a high-fat diet (HFD; 60% fat) for 16 wk. Mice on control chow diet (10% fat) and nontreated HFD mice served as controls. Glibenclamide and t-AUCB similarly prevented the increased fasting glycemia in HFD mice, but only t-AUCB improved glucose tolerance and decreased gluconeogenesis, without modifying weight gain. Moreover, t-AUCB reduced adipose tissue inflammation, plasma free fatty acids, and LDL cholesterol and prevented hepatic steatosis. Furthermore, only the sEH inhibitor improved endothelium-dependent relaxations to acetylcholine, assessed by myography in isolated coronary arteries. This improvement was related to a restoration of epoxyeicosatrienoic acid and nitric oxide pathways, as shown by the increased inhibitory effects of the nitric oxide synthase and cytochrome P-450 epoxygenase inhibitors l-NA and MSPPOH on these relaxations. Moreover, t-AUCB decreased cardiac hypertrophy, fibrosis, and inflammation and improved diastolic function, as demonstrated by the increased E/A ratio (echocardiography) and decreased slope of the end-diastolic pressure-volume relation (invasive hemodynamics). These results demonstrate that sEH inhibition improves coronary endothelial function and prevents cardiac remodeling and diastolic dysfunction in obese insulin-resistant mice.


Subject(s)
Benzoates/pharmacology , Coronary Vessels/drug effects , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Epoxide Hydrolases/antagonists & inhibitors , Heart Diseases/prevention & control , Insulin Resistance , Obesity/drug therapy , Urea/analogs & derivatives , Vasodilation/drug effects , Animals , Blood Glucose/drug effects , Blood Glucose/metabolism , Coronary Vessels/enzymology , Coronary Vessels/physiopathology , Disease Models, Animal , Dose-Response Relationship, Drug , Eicosanoids/metabolism , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiopathology , Epoxide Hydrolases/metabolism , Glyburide/pharmacology , Heart Diseases/enzymology , Heart Diseases/etiology , Heart Diseases/physiopathology , Hypoglycemic Agents/pharmacology , Inflammation Mediators/metabolism , Lipids/blood , Male , Mice , Nitric Oxide/metabolism , Obesity/blood , Obesity/complications , Obesity/enzymology , Obesity/physiopathology , Time Factors , Urea/pharmacology , Vasodilator Agents/pharmacology , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
17.
FASEB J ; 28(8): 3351-61, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24760754

ABSTRACT

The protein tyrosine phosphatase 1B (PTP1B) modulates tyrosine kinase receptors, among which is the vascular endothelial growth factor receptor type 2 (VEGFR2), a key component of angiogenesis. Because PTP1B deficiency in mice improves left ventricular (LV) function 2 mo after myocardial infarction (MI), we hypothesized that enhanced angiogenesis early after MI via activated VEGFR2 contributes to this improvement. At 3 d after MI, capillary density was increased at the infarct border of PTP1B(-/-) mice [+7±2% vs. wild-type (WT), P = 0.05]. This was associated with increased extracellular signal-regulated kinase 2 phosphorylation and VEGFR2 activation (i.e., phosphorylated-Src/Src/VEGFR2 and dissociation of endothelial VEGFR2/VE-cadherin), together with higher infiltration of proangiogenic M2 macrophages within unchanged overall infiltration. In vitro, we showed that PTP1B inhibition or silencing using RNA interference increased VEGF-induced migration and proliferation of mouse heart microvascular endothelial cells as well as fibroblast growth factor (FGF)-induced proliferation of rat aortic smooth muscle cells. At 8 d after MI in PTP1B(-/-) mice, increased LV capillary density (+21±3% vs. WT; P<0.05) and an increased number of small diameter arteries (15-50 µm) were likely to participate in increased LV perfusion assessed by magnetic resonance imaging and improved LV compliance, indicating reduced diastolic dysfunction. In conclusion, PTP1B deficiency reduces MI-induced heart failure promptly after ischemia by enhancing angiogenesis, myocardial perfusion, and diastolic function.


Subject(s)
Coronary Circulation/physiology , Myocardial Infarction/physiopathology , Neovascularization, Physiologic/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Animals , Aorta , Arterioles/physiopathology , Capillaries/physiopathology , Cardiotonic Agents/pharmacology , Cell Division , Cell Movement , Cells, Cultured , Diastole , Endothelial Cells/pathology , Heart Failure/etiology , Heart Failure/physiopathology , Heart Failure/prevention & control , Mice , Mice, Inbred BALB C , Mice, Knockout , Molecular Targeted Therapy , Myocardial Infarction/complications , Myocardial Infarction/enzymology , Myocytes, Smooth Muscle/cytology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/physiology , RNA Interference , Rats , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/physiology , Ventricular Dysfunction, Left/etiology , Ventricular Dysfunction, Left/physiopathology , Ventricular Remodeling
18.
J Mol Cell Cardiol ; 52(6): 1257-64, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22446161

ABSTRACT

Protein tyrosine phosphatase 1B (PTP1B) regulates tyrosine kinase receptor-mediated responses, and especially negatively influences insulin sensitivity, thus PTP1B inhibitors (PTP1Bi) are currently evaluated in the context of diabetes. We recently revealed another important target for PTP1Bi, consisting in endothelial protection. The present study was designed to test whether reduction of PTP1B activity may be beneficial in chronic heart failure (CHF). We evaluated the impact of either a 2 month pharmacological inhibition, or a gene deletion of PTP1B (PTP1B(-/-)) in CHF mice (2 months post-myocardial infarction). PTP1Bi and PTP1B deficiency reduced adverse LV remodeling, and improved LV function, as shown by the increased LV fractional shortening and cardiac output (measured by echocardiography), the increased LV end systolic pressure, and the decreased LV end diastolic pressure, at identical infarct sizes. This was accompanied by reduced cardiac fibrosis, myocyte hypertrophy and cardiac expression of ANP. In vitro vascular studies performed in small mesenteric artery segments showed a restored endothelial function (i.e. improved NO-dependent, flow-mediated dilatation, increased eNOS phosphorylation) after either pharmacological inhibition or gene deletion. PTP1B(-/-) CHF also displayed an improved insulin sensitivity (assessed by euglycemic-hyperinsulinemic clamp studies), when compared to wild-type CHF associated with an increased insulin mediated mesenteric artery dilation. Thus, chronic pharmacological inhibition or gene deletion of PTP1B improves cardiac dysfunction and cardiac remodeling in the absence of changes in infarct size. Thus this enzyme may be a new therapeutic target in CHF. Diabetic patients with cardiac complications may potentially benefit from PTP1B inhibition via two different mechanisms, reduced diabetic complications, and reduced heart failure.


Subject(s)
Gene Deletion , Heart Failure/therapy , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Animals , Disease Models, Animal , Echocardiography , Enzyme Inhibitors/pharmacology , Gene Expression , Heart Failure/drug therapy , Heart Failure/physiopathology , Hemodynamics , Insulin Resistance , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myocardial Infarction/drug therapy , Myocardial Infarction/genetics , Myocardium/metabolism , Myocardium/pathology , Nitric Oxide Synthase/genetics , Ventricular Remodeling
SELECTION OF CITATIONS
SEARCH DETAIL
...