Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
2.
Am J Physiol Heart Circ Physiol ; 321(5): H920-H932, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34533398

ABSTRACT

Bone marrow-derived endothelial progenitor cells (EPCs) contribute to endothelial repair and angiogenesis. Reduced number of circulating EPCs is associated with future cardiovascular events. We tested whether dysregulated glucose and/or triglyceride (TG) metabolism has an impact on EPC homeostasis. The analysis of metabolic factors associated with circulating EPC number in humans revealed that postprandial hyperglycemia is negatively correlated with circulating EPC number, and this correlation appears to be further enhanced in the presence of postprandial hypertriglyceridemia (hTG). We therefore examined the effect of glucose/TG spikes on bone marrow lineage-sca-1+ c-kit+ (LSK) cells in mice, because primitive EPCs reside in bone marrow LSK fraction. Repetitive glucose + lipid (GL) spikes, but not glucose (G) or lipid (L) spikes alone, induced senescence-like phenotypes of LSK cells, and this phenomenon was reversible after cessation of GL spikes. G spikes and GL spikes differentially affected transcriptional program of LSK cell metabolism and differentiation. GL spikes upregulated a histone H3K27 demethylase JMJD3, and inhibition of JMJD3 eliminated GL spikes-induced LSK cell senescence-like phenotypes. These observations suggest that postprandial glucose/TG dysmetabolism modulate transcriptional regulation in LSK cells through H3K27 demethylase-mediated epigenetic regulation, leading to senescence-like phenotypes of LSK cells, reduced number of circulating EPCs, and development of atherosclerotic cardiovascular disease.NEW & NOTEWORTHY Combination of hyperglycemia and hypertriglyceridemia is associated with increased risk of atherosclerotic cardiovascular disease. We found that 1) hypertriglyceridemia may enhance the negative impact of hyperglycemia on circulating EPC number in humans and 2) metabolic stress induced by glucose + triglyceride spikes in mice results in senescence-like phenotypes of bone marrow stem/progenitor cells via H3K27me3 demethylase-mediated epigenetic regulation. These findings have important implications for understanding the pathogenesis of atherosclerotic cardiovascular disease in patients with T2DM.


Subject(s)
Blood Glucose/metabolism , Bone Marrow Cells/enzymology , Cellular Senescence , DNA Methylation , Diabetes Mellitus, Type 2/blood , Endothelial Progenitor Cells/enzymology , Epigenesis, Genetic , Hyperglycemia/blood , Hypertriglyceridemia/blood , Jumonji Domain-Containing Histone Demethylases/metabolism , Triglycerides/blood , Adult , Aged , Animals , Bone Marrow Cells/pathology , Case-Control Studies , Cell Lineage , Cells, Cultured , Diabetes Mellitus, Type 2/enzymology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Disease Models, Animal , Endothelial Progenitor Cells/pathology , Female , Glycated Hemoglobin , Humans , Hyperglycemia/enzymology , Hyperglycemia/genetics , Hyperglycemia/pathology , Hypertriglyceridemia/enzymology , Hypertriglyceridemia/genetics , Hypertriglyceridemia/pathology , Jumonji Domain-Containing Histone Demethylases/genetics , Male , Mice, Inbred C57BL , Middle Aged , Phenotype
3.
Microvasc Res ; 136: 104165, 2021 07.
Article in English | MEDLINE | ID: mdl-33845105

ABSTRACT

Phototherapy has been tried for treating cardiovascular diseases. In particular, ultraviolet and blue visible lights were suggested to be useful due to their nitric oxide (NO)-production ability in the skin. However, the effects of blue light on the arterial contractility are controversial. Here, we hypothesized that appropriate protocol of blue laser can induce selective vasorelaxation by activating vasodilating signaling molecules in arteries. Using organ chamber arterial mechanics, NO assay, Matrigel assay, and microarray, we showed that a 200-Hz, 300-µs, 445-nm pulsed-laser (total energy of 600 mJ; spot size 4 mm) induced selective vasorelaxation, without vasocontraction in rat mesenteric arteries. The laser stimulation increased NO production in the cord blood-endothelial progenitor cells (CB-EPCs). Both the laser-induced vasorelaxation and NO production were inhibited by a non-selective, pan-NO synthase inhibitor, L-NG-Nitro arginine methyl ester. Microarray study in CB-EPCs suggested up-regulation of cryptochrome (CRY)2 as well as NO synthase (NOS)1 and NOSTRIN (NOS trafficking) by the laser. In conclusion, this study suggests that the 445-nm blue puled-laser can induce vasorelaxation possibly via the CRY photoreceptors and NOSs activation. The blue laser-therapy would be useful for treating systemic hypertension as well as improving local blood flow depending on the area of irradiation.


Subject(s)
Endothelial Progenitor Cells/radiation effects , Lasers , Low-Level Light Therapy/instrumentation , Mesenteric Arteries/radiation effects , Nitric Oxide Synthase/metabolism , Nitric Oxide/metabolism , Vasodilation/radiation effects , Animals , Cells, Cultured , Endothelial Progenitor Cells/enzymology , Enzyme Activation , Fetal Blood/cytology , Gene Expression Regulation , Humans , Male , Mesenteric Arteries/enzymology , Nitric Oxide Synthase/genetics , Rats, Sprague-Dawley , Signal Transduction
4.
Microvasc Res ; 135: 104134, 2021 05.
Article in English | MEDLINE | ID: mdl-33428882

ABSTRACT

BACKGROUND: Clinical data show that aneurysm rupture causes high mortality in aged men. MicroRNAs (miRNAs) were reported to regulate endothelial progenitor cells (EPCs) which play a vital role in repairing endothelial damage and maintaining vascular integrity. This study identified a novel miRNA regulator for the functions of EPCs in aneurysm repair. METHODS: Abdominal aortic aneurysm (AAA) model was established on Sprague-Dawley rats which later underwent antagomiR-222 treatment. The histopathological changes of AAA rats were examined by hematoxylin-eosin staining. Flow cytometry was performed to quantify EPCs in peripheral blood and identify EPCs isolated from the rat femur. The potential target of miR-222-3p was predicted by TargetScan v7.2 and validated by Dual-luciferase reporter assay. The effects of miR-222-3p and ADIPOR1 on the migration, invasion and tube formation of EPCs were evaluated by wound healing, Transwell and tube formation assays. The expressions of miR-222-3p and ADIPOR1 in aortic aneurysm tissues and EPCs were assessed by qRT-PCR or Western blot. RESULTS: AAA exhibited histopathological abnormality, a decreased number of EPCs in the peripheral blood and an increased miR-222-3p expression. AntagomiR-222 injection reversed all these phenomena in AAA rats. Upregulating miR-222-3p expression inhibited the migration, invasion, and tube formation of EPCs, and the expressions of ADIPOR1 and phosphorylated-AMKP, while downregulating miR-222-3p expression exerted opposite effects in EPCs. ADIPOR1 was identified as a target gene of miR-222-3p. Overexpressing ADIPOR1 abrogated the effects of miR-222-3p upregulation on EPCs. CONCLUSION: Downregulated miR-222-3p prompted the migration, invasion and recruitment of EPCs by targeting ADIPOR1-induced AMKP activation.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Aorta, Abdominal/enzymology , Aortic Aneurysm, Abdominal/enzymology , Cell Movement , Endothelial Progenitor Cells/enzymology , MicroRNAs/metabolism , Neovascularization, Physiologic , Receptors, Adiponectin/metabolism , Animals , Antagomirs/genetics , Antagomirs/metabolism , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/genetics , Aortic Aneurysm, Abdominal/pathology , Cells, Cultured , Disease Models, Animal , Down-Regulation , Endothelial Progenitor Cells/pathology , Enzyme Activation , Humans , Male , MicroRNAs/genetics , Phosphorylation , Rats, Sprague-Dawley , Receptors, Adiponectin/genetics , Signal Transduction
5.
Arterioscler Thromb Vasc Biol ; 40(12): 2875-2890, 2020 12.
Article in English | MEDLINE | ID: mdl-33115267

ABSTRACT

OBJECTIVE: Endothelial progenitors migrate early during embryogenesis to form the primary vascular plexus. The regulatory mechanisms that govern their migration are not completely defined. Here, we describe a novel role for ETV2 (Ets variant transcription factor 2) in cell migration and provide evidence for an ETV2-Rhoj network as a mechanism responsible for this process. Approach and Results: Analysis of RNAseq datasets showed robust enrichment of migratory/motility pathways following overexpression of ETV2 during mesodermal differentiation. We then analyzed ETV2 chromatin immunoprecipitation-seq and assay for transposase accessible chromatin-seq datasets, which showed enrichment of chromatin immunoprecipitation-seq peaks with increased chromatin accessibility in migratory genes following overexpression of ETV2. Migratory assays showed that overexpression of ETV2 enhanced cell migration in mouse embryonic stem cells, embryoid bodies, and mouse embryonic fibroblasts. Knockout of Etv2 led to migratory defects of Etv2-EYFP+ angioblasts to their predefined regions of developing embryos relative to wild-type controls at embryonic day (E) 8.5, supporting its role during migration. Mechanistically, we showed that ETV2 binds the promoter region of Rhoj serving as an upstream regulator of cell migration. Single-cell RNAseq analysis of Etv2-EYFP+ sorted cells revealed coexpression of Etv2 and Rhoj in endothelial progenitors at E7.75 and E8.25. Overexpression of ETV2 led to a robust increase in Rhoj in both embryoid bodies and mouse embryonic fibroblasts, whereas, its expression was abolished in the Etv2 knockout embryoid bodies. Finally, shRNA-mediated knockdown of Rhoj resulted in migration defects, which were partially rescued by overexpression of ETV2. CONCLUSIONS: These results define an ETV2-Rhoj cascade, which is important for the regulation of endothelial progenitor cell migration.


Subject(s)
Cell Movement , Embryonic Stem Cells/enzymology , Endothelial Progenitor Cells/enzymology , Transcription Factors/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cells, Cultured , Embryonic Development , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Gestational Age , Mice, Transgenic , Signal Transduction , Transcription Factors/genetics , rho GTP-Binding Proteins/genetics
6.
Aging (Albany NY) ; 12(16): 16294-16303, 2020 08 25.
Article in English | MEDLINE | ID: mdl-32843584

ABSTRACT

BACKGROUND: The angiogenesis post myocardial infarction (MI) is compromised in diabetes. MiR-144-3p is reported to be highly expressed in circulating exosomes of diabetic patients, implying its role in diabetic complications. However, whether circulating exosomes and enriched miR-144-3p are involved in the impaired neovascularization in diabetes and the underlying mechanism is unclear. RESULTS: DMexo and miR-144-3p mimic-treated MSCs had elevated miR-144-3p levels and decreased MMP9, Ets1 and PLG expression. The percentage of EPCs were relatively lower in DMexo-treated or agomir-treated MI mice compared with MI mice. Finally, the luciferase assay confirmed the direct binding between miR-144-3p and Ets1. CONCLUSION: Exosomal miR-144-3p could impair the mobilization ability of EPCs, which was associated with impaired ischemia-induced neovascularization. METHODS: Circulating exosomes were isolated from Streptozotocin (STZ)-induced mice. In vitro, mesenchymal stem cells (MSCs) were incubated with exosomes from diabetic mice (DMexo), and miR-144-3p mimic or inhibitor. miR-144-3p, and MMP9 pathway were measured using qPCR and immunoblotting. In vivo, MI mice induced by left anterior descending ligation were treated with DMexo, as well as miR-144-3p agomir. Flow cytometry was used to profile endothelial progenitor cells (EPCs) in peripheral blood and bone marrow post 24 hours respectively.


Subject(s)
Cell Movement , Circulating MicroRNA/blood , Diabetes Mellitus, Experimental/blood , Endothelial Progenitor Cells/enzymology , Exosomes/metabolism , Matrix Metalloproteinase 9/metabolism , MicroRNAs/blood , Myocardial Infarction/enzymology , Neovascularization, Physiologic , 3' Untranslated Regions , Animals , Binding Sites , Cells, Cultured , Circulating MicroRNA/genetics , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/genetics , Endothelial Progenitor Cells/pathology , Exosomes/genetics , Male , Matrix Metalloproteinase 9/genetics , Mice, Inbred C57BL , MicroRNAs/genetics , Myocardial Infarction/blood , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Protein c-ets-1/metabolism , Signal Transduction
7.
Fertil Steril ; 113(5): 1067-1079.e5, 2020 05.
Article in English | MEDLINE | ID: mdl-32386617

ABSTRACT

OBJECTIVE: To explore the possible mechanism of protein kinase CK2, which participates in estrogen recruitment of endothelial progenitor cells (EPCs), and its role in the angiogenesis of endometriosis lesions. DESIGN: Laboratory study. SETTING: University. ANIMAL(S): BALB/c mice. INTERVENTION(S): Exposure of human endometrial stromal cells (HESCs) to estrogen and CK2 inhibitor CX-4945 and endometrial stromal cells transfected with the protein kinase CK2 vector (HESC-CK2). Endometriosis models were induced by allogeneic mice transplantation of the endometrium into dorsal skinfold chambers. The mice received an IP injection of 50 mg/kg emodin per day or were treated with 100 µg/kg estrogen by SC injection once a week. MAIN OUTCOME MEASURE(S): The concentration of cytokines in cells was measured with ELISA. The migration of EPCs was examined using the scratch assay method and Transwell, a capillary tube-formation assay to determine EPC tube-forming capacity, and protein and mRNA expression with Western blot and polymerase chain reaction analyses, respectively. RESULT(S): Protein kinase CK2 participates in estrogen-mediated EPC homing to endometriotic lesions through stromal cells in a stromal cell-derived factor-1 (SDF-1)-CXCR4-dependent manner. Conditioned medium from endometrial stromal cells that were stably transfected with the protein kinase CK2 vector (HESC-CK2) or pretreated with estrogen significantly enhanced the migration and recruitment of EPCs. In contrast, conditioned medium from HESCs that were treated with CX-4945, a selective inhibitor of CK2, inhibited the mobility and viability of EPCs. Furthermore, CK2 overexpression significantly upregulated SDF-1 expression and secretion in endometrial stromal cells by activating the AKT/mTOR pathway. Moreover, treatment with the SDF-1 receptor CXCR4-specific inhibitor AMD3100 completely reversed the CK2-enhanced migration of EPCs. CONCLUSION(S): This study demonstrates that CK2 participates in estrogen-mediated EPC homing to endometriotic lesions through stromal cells in an SDF-1-CXCR4-dependent manner and may be a therapeutic target.


Subject(s)
Casein Kinase II/metabolism , Chemokine CXCL12/metabolism , Endometriosis/enzymology , Endometrium/enzymology , Endothelial Progenitor Cells/enzymology , Receptors, CXCR4/metabolism , Stromal Cells/enzymology , Animals , Casein Kinase II/genetics , Cell Line , Cell Movement/drug effects , Coculture Techniques , Disease Models, Animal , Endometriosis/genetics , Endometriosis/pathology , Endometrium/pathology , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/pathology , Estrogens/pharmacology , Female , Humans , Mice, Inbred BALB C , Neovascularization, Pathologic , Paracrine Communication/drug effects , Signal Transduction , Stromal Cells/drug effects , Stromal Cells/pathology
8.
Cardiovasc Res ; 116(2): 393-405, 2020 02 01.
Article in English | MEDLINE | ID: mdl-30937452

ABSTRACT

AIMS: Cord blood-derived endothelial colony-forming cells (CB-ECFCs) are a defined progenitor population with established roles in vascular homeostasis and angiogenesis, which possess low immunogenicity and high potential for allogeneic therapy and are highly sensitive to regulation by reactive oxygen species (ROS). The aim of this study was to define the precise role of the major ROS-producing enzyme, NOX4 NADPH oxidase, in CB-ECFC vasoreparative function. METHODS AND RESULTS: In vitro CB-ECFC migration (scratch-wound assay) and tubulogenesis (tube length, branch number) was enhanced by phorbol 12-myristate 13-acetate (PMA)-induced superoxide in a NOX-dependent manner. CB-ECFCs highly-expressed NOX4, which was further induced by PMA, whilst NOX4 siRNA and plasmid overexpression reduced and potentiated in vitro function, respectively. Increased ROS generation in NOX4-overexpressing CB-ECFCs (DCF fluorescence, flow cytometry) was specifically reduced by superoxide dismutase, highlighting induction of ROS-specific signalling. Laser Doppler imaging of mouse ischaemic hindlimbs at 7 days indicated that NOX4-knockdown CB-ECFCs inhibited blood flow recovery, which was enhanced by NOX4-overexpressing CB-ECFCs. Tissue analysis at 14 days revealed consistent alterations in vascular density (lectin expression) and eNOS protein despite clearance of injected CB-ECFCs, suggesting NOX4-mediated modulation of host tissue. Indeed, proteome array analysis indicated that NOX4-knockdown CB-ECFCs largely suppressed tissue angiogenesis, whilst NOX4-overexpressing CB-ECFCs up-regulated a number of pro-angiogenic factors specifically-linked with eNOS signalling, in parallel with equivalent modulation of NOX-dependent ROS generation, suggesting that CB-ECFC NOX4 signalling may promote host vascular repair. CONCLUSION: Taken together, these findings indicate a key role for NOX4 in CB-ECFCs, thereby highlighting its potential as a target for enhancing their reparative function through therapeutic priming to support creation of a pro-reparative microenvironment and effective post-ischaemic revascularization.


Subject(s)
Endothelial Progenitor Cells/transplantation , Ischemia/surgery , Muscle, Skeletal/blood supply , NADPH Oxidase 4/metabolism , Neovascularization, Physiologic , Animals , Cell Movement , Cells, Cultured , Cellular Microenvironment , Disease Models, Animal , Endothelial Progenitor Cells/enzymology , Fetal Blood/cytology , Hindlimb , Humans , Ischemia/enzymology , Ischemia/genetics , Ischemia/physiopathology , Mice, Inbred NOD , NADPH Oxidase 4/genetics , Reactive Oxygen Species/metabolism , Recovery of Function , Signal Transduction
9.
Cardiovasc Res ; 116(2): 262-268, 2020 02 01.
Article in English | MEDLINE | ID: mdl-31393561

ABSTRACT

The differentiation of stem cells into endothelial cells involves the modulation of highly interconnected metabolic and epigenetic processes. Therefore, the differentiation of endothelial cells is a tightly controlled process, which is adjusted at multiple levels, meaning that even the smallest variation can result in major consequences. Reactive oxygen species (ROS) represent a group of second messengers that can interfere with both metabolic and epigenetic processes. Besides their generation by mitochondria, ROS are produced in a controlled manner by the family of NADPH oxidases. The different members of the NADPH oxidase family produce superoxide anions or hydrogen peroxide. Due to the specific sub-cellular localization of the different NADPH oxidases, ROS are produced at diverse sites in the cell, such as the plasma membrane or the endoplasmic reticulum. Once produced, ROS interfere with proteins, lipids, and DNA to modulate intracellular signal cascades. Accordingly, ROS represent a group of readily available and specifically localized modulators of the highly sophisticated signalling network that eventually leads to the differentiation of stem cells into endothelial cells. This review focuses on the role of NADPH oxidases in the differentiation of stem cells into endothelial cells.


Subject(s)
Cell Differentiation , Endothelial Progenitor Cells/enzymology , NADPH Oxidases/metabolism , Animals , Humans , Reactive Oxygen Species/metabolism , Signal Transduction
10.
Ann Vasc Surg ; 64: 339-346, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31639479

ABSTRACT

OBJECTIVE: Bone-marrow-derived endothelial progenitor cells (EPCs) can accelerate the dissolution of thrombi. However, EPC functions are weakened in deep vein thrombosis (DVT), and miR-130a-3p is downregulated in DVT. As little is known about the function of miR-130a-3p in EPCs, we aimed to explore the effects of miR-130a-3p on EPC functions and the mechanisms of miR-130a-3p regulation of EPCs in DVT. METHODS: The EPCs were transfected with miR-130a-3p mimics or miR-130a-3p inhibitor. Migration and angiogenesis of EPCs were detected by wound healing, Transwell, and tube formation assays. Dual luciferase assay was used to test the relation of miR-130a-3p and phosphatase and tensin homolog (PTEN). Protein and mRNA levels of associated genes were measured by western blotting (WB) and qRT-PCR. RESULTS: miR-103a-3p could promote EPC migration and angiogenesis, and it was also downregulated in EPCs isolated from DVT patients. Moreover, PTEN was a target of miR-130a-3p. Upregulation of PTEN rescued the auxoaction of miR-130a-3p in EPC function. CONCLUSIONS: Downregulation of miR-103a-3p contributes to EPC dysfunction in DVT via targeting PTEN. Thus, miR-130a-3p may be a potential target for DVT treatment.


Subject(s)
Endothelial Progenitor Cells/enzymology , MicroRNAs/metabolism , Neovascularization, Physiologic , PTEN Phosphohydrolase/metabolism , Venous Thrombosis/enzymology , Case-Control Studies , Cell Movement , Cells, Cultured , Down-Regulation , Endothelial Progenitor Cells/pathology , Humans , MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins c-akt , Signal Transduction , Venous Thrombosis/genetics , Venous Thrombosis/pathology
11.
J Cardiovasc Transl Res ; 13(2): 158-170, 2020 04.
Article in English | MEDLINE | ID: mdl-31701352

ABSTRACT

The enhanced activity of endothelial progenitor cells (EPCs) by AMP-activated protein kinase (AMPK) agonists might explain the reversal of chronic heart failure (CHF)-mediated endothelial dysfunction. We studied baseline circulating EPC numbers in patients with heart failure and clarified the effect of fenofibrate on both circulating angiogenic cell (CAC) and late EPC activity. The numbers of circulating EPCs in CHF patients were quantified by flow cytometry. Blood-derived mononuclear cells were cultured, and CAC and late EPC functions, including fibronectin adhesion, tube formation, and migration, were evaluated. We focused on the effect of fenofibrate, an AMPK agonist, on EPC function and Akt/eNOS cascade activation in vitro. The number of circulating EPCs (CD34+/KDR+) was significantly lower in CHF patients (ischemic cardiomyopathy (ICMP): 0.07%, dilated cardiomyopathy (DCMP): 0.068%; p < 0.05) than in healthy subjects (0.102% of the gating region). In CACs, fibronectin adhesion function was reversed by fenofibrate treatment (p < 0.05). Similar results were also found for tube formation and migration in late EPCs, which were significantly improved by fenofibrate in an AMPK-dependent manner (p < 0.05), suggesting that fenofibrate reversed CACs and late EPC dysfunction in CHF patients. The present findings reveal the potential application of the AMPK agonist fenofibrate to reverse endothelial dysfunction in CHF patients.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Endothelial Progenitor Cells/drug effects , Enzyme Activators/pharmacology , Fenofibrate/pharmacology , Heart Failure/drug therapy , Neovascularization, Physiologic/drug effects , Adult , Aged , Antigens, CD34/metabolism , Case-Control Studies , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chronic Disease , Endothelial Progenitor Cells/enzymology , Endothelial Progenitor Cells/pathology , Enzyme Activation , Female , Heart Failure/enzymology , Heart Failure/physiopathology , Humans , Male , Middle Aged , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism
12.
Circ J ; 83(12): 2537-2546, 2019 11 25.
Article in English | MEDLINE | ID: mdl-31645525

ABSTRACT

BACKGROUND: Given that cathepsin S (CatS) gained attention due to its enzymatic and non-enzymatic functions in signaling, the role of CatS in ischemia-induced angiogenesis of aged mice was explored.Methods and Results:To study the role of CatS in the decline in aging-related vascular regeneration capacity, a hindlimb ischemia model was applied to aged wild-type (CatS+/+) and CatS-deficient (CatS-/-) mice. CatS-/-mice exhibited impaired blood flow recovery and capillary formation and increased levels of p-insulin receptor substrate-1, Wnt5a, and SC35 proteins and decreased levels of phospho-endothelial nitric oxide synthase (p-eNOS), p-mTOR, p-Akt, p-ERK1/2, p-glycogen synthase kinase-3α/ß, and galatin-3 proteins, as well as decreased macrophage infiltration and matrix metalloproteinase-2/-9 activities in the ischemic muscles. In vitro, CatS knockdown altered the levels of these targeted essential molecules for angiogenesis. Together, the results suggested that CatS-/-leads to defective endothelial cell functions and that CatS-/-is associated with decreased circulating endothelial progenitor cell (EPC)-like CD31+/c-Kit+cells. This notion was reinforced by the study finding that pharmacological CatS inhibition led to a declined angiogenic capacity accompanied by increased Wnt5a and SC35 levels and decreased eNOS/Akt-ERK1/2 signaling in response to ischemia. CONCLUSIONS: These findings demonstrated that the impairment of ischemia-induced neovascularization in aged CatS-/-mice is due, at least in part, to the attenuation of endothelial cell/EPC functions and/or mobilization associated with Wnt5a/SC35 activation in advanced age.


Subject(s)
Cathepsins/metabolism , Endothelial Progenitor Cells/enzymology , Ischemia/enzymology , Muscle, Skeletal/blood supply , Serine-Arginine Splicing Factors/metabolism , Wnt-5a Protein/metabolism , Age Factors , Animals , Cathepsins/deficiency , Cathepsins/genetics , Cells, Cultured , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Hindlimb , Human Umbilical Vein Endothelial Cells/enzymology , Humans , Ischemia/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
13.
Am J Physiol Heart Circ Physiol ; 316(6): H1468-H1479, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30951365

ABSTRACT

20-Hydroxyeicosatetraenoic acid (20-HETE) was recently identified as a novel contributor of ischemia-induced neovascularization based on the key observation that pharmacological interferences of CYP4A/20-HETE decrease ischemic neovascularization. The objective of the present study is to examine whether the underlying cellular mechanisms involve endothelial progenitor cells (EPCs) and preexisting endothelial cells (ECs). We found that ischemia leads to a time-dependent increase of cyp4a12 expression and 20-HETE production, which are endothelial in origin, using immunofluorescent microscopy, Western blot analysis, and LC-MS/MS. This is accompanied by increases in the tissue stromal cell-derived factor-1α (SDF-1α) expressions as well as SDF-1α plasma levels, EPC mobilization from bone marrow, and subsequent homing to ischemic tissues. Pharmacological interferences of CYP4A/20-HETE with a 20-HETE synthesis inhibitor, dibromo-dodecenyl-methylsulfimide (DDMS), or a 20-HETE antagonist, N-(20-hydroxyeicosa-6(Z), 15(Z)-dienoyl) glycine (6, 15-20-HEDGE), significantly attenuated these increases. Importantly, we also determined that 20-HETE plays a novel role in maintaining EPC functions and increasing the expression of Oct4, Sox2, and Nanog, which are indicative of increased progenitor cell stemness. Flow cytometric analysis revealed that pharmacological interferences of CYP4A/20-HETE decrease the EPC population in culture, whereas 20-HETE increases the cultured EPC population. Furthermore, ischemia also markedly increased the proliferation, oxidative stress, and ICAM-1 expression in the preexisting EC in the hindlimb gracilis muscles. We found that these increases were markedly negated by DDMS and 6, 15-20-HEDGE. Taken together, CYP4A/20-HETE regulates ischemia-induced compensatory neovascularization via its combined actions on promoting EPC and local preexisting EC responses that are associated with increased neovascularization. NEW & NOTEWORTHY CYP4A/20-hydroxyeicosatetraenoic acid (20-HETE) was recently discovered as a novel contributor of ischemia-induced neovascularization. However, the underlying molecular and cellular mechanisms are completely unknown. Here, we show that CYP4A/20-HETE regulates the ischemic neovascularization process via its combined actions on both endothelial progenitor cells (EPCs) and preexisting endothelial cells. Moreover, this is the first study, to the best of our knowledge, that associates CYP4A/20-HETE with EPC differentiation and stemness.


Subject(s)
Cytochrome P-450 CYP4A/metabolism , Endothelial Cells/enzymology , Endothelial Progenitor Cells/enzymology , Hydroxyeicosatetraenoic Acids/metabolism , Ischemia/enzymology , Muscle, Skeletal/blood supply , Neovascularization, Physiologic , Animals , Cells, Cultured , Chemokine CXCL12/metabolism , Cytochrome P450 Family 4/metabolism , Disease Models, Animal , Hindlimb , Humans , Ischemia/physiopathology , Male , Mice, Inbred BALB C , Nanog Homeobox Protein/genetics , Nanog Homeobox Protein/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Signal Transduction , Time Factors
14.
J Vasc Res ; 56(1): 17-27, 2019.
Article in English | MEDLINE | ID: mdl-30879014

ABSTRACT

Vitamin D has an important protective effect on chronic inflammatory disease. Angiotensin II (AngII) triggers vascular damage and plays a key role in vascular diseases via several mechanisms, including inflammation. Conversely, vitamin D has been shown to have an important protective effect on chronic inflammation. There is evidence showing that vitamin D can reverse the effects of AngII, but the molecular mechanisms by which this occurs are not known. Our results demonstrate that vitamin D improved the viability, migration ability, and tube formation of AngII-pretreated endothelial progenitor cells (EPCs) and inhibited the apoptosis of EPCs induced by AngII. Vitamin D also reversed reactive oxygen species production, vascular inflammatory cytokine generation, and nuclear factor kappa-B activation in EPCs induced by AngII. Furthermore, EPC pretreatment with GW9662 (the antagonist for PPAR-γ) or siHO-1 decreased the protective effect of vitamin D on AngII-induced EPC injury. Overall, our data indicate that vitamin D ameliorated AngII-induced abnormal EPC injury by decreasing oxidative stress and inflammatory cytokine levels. These findings also suggest that vitamin D protected EPCs from AngII-induced vascular injury via the activation of the PPAR-γ/HO-1 signaling pathway.


Subject(s)
Angiotensin II/toxicity , Anti-Inflammatory Agents/pharmacology , Antioxidants/pharmacology , Endothelial Progenitor Cells/drug effects , Heme Oxygenase-1/metabolism , PPAR gamma/metabolism , Vitamin D/pharmacology , Apoptosis/drug effects , Cell Movement/drug effects , Cells, Cultured , Cytokines/metabolism , Cytoprotection , Endothelial Progenitor Cells/enzymology , Endothelial Progenitor Cells/pathology , Humans , Inflammation Mediators/metabolism , Neovascularization, Physiologic/drug effects , Oxidative Stress/drug effects , Signal Transduction
15.
Crit Care ; 23(1): 44, 2019 02 13.
Article in English | MEDLINE | ID: mdl-30760290

ABSTRACT

BACKGROUND: The acute respiratory distress syndrome (ARDS) is characterized by disruption of the alveolar-capillary barrier resulting in accumulation of proteinaceous edema and increased inflammatory cells in the alveolar space. We previously found that endothelial progenitor cell (EPC) exosomes prevent endothelial dysfunction and lung injury in sepsis in part due to their encapsulation of miRNA-126. However, the effects of EPC exosomes in acute lung injury (ALI) remain unknown. METHODS: To determine if EPC exosomes would have beneficial effects in ALI, intratracheal administration of lipopolysaccharide (LPS) was used to induce ALI in mice. Lung permeability, inflammation, and the role of miRNA-126 in the alveolar-epithelial barrier function were examined. RESULTS: The intratracheal administration of EPC exosomes reduced lung injury following LPS-induced ALI at 24 and 48 h. Compared to placebo, intratracheal administration of EPC exosomes significantly reduced the cell number, protein concentration, and cytokines/chemokines in the bronchoalveolar lavage fluid (BALF), indicating a reduction in permeability and inflammation. Further, EPC exosomes reduced myeloperoxidase (MPO) activity, lung injury score, and pulmonary edema, demonstrating protection against lung injury. Murine fibroblast (NIH3T3) exosomes, which do not contain abundant miRNA-126, did not provide these beneficial effects. In human small airway epithelial cells (SAECs), we found that overexpression of miRNA-126-3p can target phosphoinositide-3-kinase regulatory subunit 2 (PIK3R2), while overexpression of miRNA-126-5p inhibits the inflammatory alarmin HMGB1 and permeability factor VEGFα. Interestingly, both miR-126-3p and 5p increase the expression of tight junction proteins suggesting a potential mechanism by which miRNA-126 may mitigate LPS-induced lung injury. CONCLUSIONS: Our data demonstrated that human EPC exosomes are beneficial in LPS-induced ALI mice, in part through the delivery of miRNA-126 into the injured alveolus.


Subject(s)
Acute Lung Injury/drug therapy , Endothelial Progenitor Cells/enzymology , Inflammation/physiopathology , Acute Lung Injury/physiopathology , Animals , Blotting, Western/methods , Exosomes/metabolism , HMGB1 Protein/metabolism , Inflammation/metabolism , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/adverse effects , Mice , MicroRNAs/physiology , Peroxidase/metabolism , Peroxidase/physiology , Phosphatidylinositol 3-Kinases/metabolism , Real-Time Polymerase Chain Reaction/methods , Severity of Illness Index , Trachea/drug effects , Vascular Endothelial Growth Factor A/metabolism
16.
J Thorac Cardiovasc Surg ; 157(2): 803-814, 2019 02.
Article in English | MEDLINE | ID: mdl-30391008

ABSTRACT

OBJECTIVE: Endothelial progenitor cells (EPCs) can improve endothelial integrity. This study aimed to examine the effects and the mechanism of EPCs on lung ischemia-reperfusion injury (LIRI). METHODS: Wistar rats were randomized into the sham or the left lung transplantation group. The recipients were randomized and treated with vehicle as the LIRI group, with EPC as the EPC group, or with N5-(1-iminoethyl)-l-ornithine-pretreated EPC as the EPC/L group (n = 8 per group). The ratios of arterial oxygen partial pressure to fractional inspiratory oxygen were measured. The lung wet-to-dry weight ratios, protein levels, and injury, as well as the levels of plasma cytokines, were examined. The levels of endothelin (ET)-1, endothelial nitric oxide synthase (eNOS), phosphorylated eNOS, inducible NOS, phosphorylated myosin light chain, nuclear factor-κBp65, Bax, Bcl-2, cleaved caspase-3, and myeloperoxidase in the graft lungs were detected. RESULTS: Compared with the LIRI group, EPC treatment significantly increased the ratios of arterial oxygen partial pressure to fractional inspiratory oxygen and decreased the lung wet-to-dry weight ratios and protein levels in the grafts, accompanied by increasing eNOS expression and phosphorylation, but decreasing endothelin-1, inducible NOS, phosphorylated nuclear factor-kBp65, phosphorylated myosin light chain expression, and myeloperoxidase activity. EPCs reduced lung tissue damage and apoptosis associated with decreased levels of Bax and cleaved caspase-3 expression, but increased Bcl-2 expression. EPC treatment significantly reduced the levels of serum proinflammatory factors, but elevated levels of interleukin-10. In contrast, the protective effect of EPCs were mitigated and abrogated by N5-(1-iminoethyl)-l-ornithine pretreatment. CONCLUSIONS: Data indicated that EPC ameliorated LIRI by increasing eNOS expression.


Subject(s)
Endothelial Progenitor Cells/transplantation , Lung Transplantation/adverse effects , Nitric Oxide Synthase Type III/metabolism , Reperfusion Injury/prevention & control , Stem Cell Transplantation , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Cell Survival , Cells, Cultured , Disease Models, Animal , Endothelial Progenitor Cells/enzymology , Endothelial Progenitor Cells/pathology , Graft Survival , Inflammation Mediators/metabolism , Male , NF-kappa B/metabolism , Phosphorylation , Rats, Wistar , Reperfusion Injury/enzymology , Reperfusion Injury/pathology , Signal Transduction
17.
J Cell Physiol ; 234(5): 7090-7103, 2019 05.
Article in English | MEDLINE | ID: mdl-30552827

ABSTRACT

Ischemic stroke is a refractory disease caused by cerebral ischemic injury, which results in brain dysfunction. This study intends to investigate the effects of microRNA-212 (miR-212) on the recovery function and vascular regeneration of endothelial progenitor cells (EPCs) by inactivation of the Notch signaling pathway by binding to matrix metallopeptidase 9 (MMP9) in mice with ischemic stroke. According to the results of database retrieval systems and data analysis, MMP9 was predicted as a gene related to ischemic stroke and miR-212 is a potential regulating mRNA of MMP9. All 72 healthy adult C57BL6 mice were selected for middle cerebral artery occlusion (MCAO) establishment. Cerebral infarction was observed under triphenyltetrazolium chloride staining. A series of inhibitors, activators, and siRNAs were introduced to the verified regulatory functions for miR-212 governing MMP9 in ischemic stroke. Cell proliferation was detected by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and tube-forming ability by tubule formation test. Reverse transcription quantitative polymerase chain reaction and Western blot analysis were used to detect the expressions of miR-212, MMP9, Hes-1, and Notch-1. The corresponding results demonstrated that the area of cerebral infarction and the number of neuronal necrosis increased in the MCAO group in contrast to the sham group. Meanwhile, upregulation of miR-212 or downregulation of MMP9 decreases the expressions of MMP9, Hes-1 Notch-1, increases cell proliferation and tube-forming ability and improves the pathological conditions of EPCs. Our study suggests that miR-212 promotes recovery function and vascular regeneration of EPCs through negative regulation of the Notch signaling pathway via downregulating expression of MMP9, thus provides a clinical theoretical basis for ischemic stroke therapy.


Subject(s)
Brain/blood supply , Cell Proliferation , Endothelial Progenitor Cells/enzymology , Infarction, Middle Cerebral Artery/enzymology , Matrix Metalloproteinase 9/metabolism , MicroRNAs/metabolism , Neovascularization, Physiologic , Receptor, Notch1/metabolism , Animals , Case-Control Studies , Cells, Cultured , Databases, Genetic , Disease Models, Animal , Endothelial Progenitor Cells/pathology , Humans , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Infarction, Middle Cerebral Artery/physiopathology , Male , Matrix Metalloproteinase 9/genetics , Mice, Inbred C57BL , MicroRNAs/genetics , Receptor, Notch1/genetics , Signal Transduction , Transcription Factor HES-1/genetics , Transcription Factor HES-1/metabolism
18.
Sci Rep ; 8(1): 13433, 2018 09 07.
Article in English | MEDLINE | ID: mdl-30194334

ABSTRACT

Early damage to transplanted organs initiates excess inflammation that can cause ongoing injury, a leading cause for late graft loss. The endothelial glycocalyx modulates immune reactions and chemokine-mediated haptotaxis, potentially driving graft loss. In prior work, conditional deficiency of the glycocalyx-modifying enzyme N-deacetylase-N-sulfotransferase-1 (Ndst1f/f TekCre+) reduced aortic allograft inflammation. Here we investigated modification of heparan sulfate (HS) and chemokine interactions in whole-organ renal allografts. Conditional donor allograft Ndst1 deficiency (Ndst1-/-; C57Bl/6 background) was compared to systemic treatment with M-T7, a broad-spectrum chemokine-glycosaminoglycan (GAG) inhibitor. Early rejection was significantly reduced in Ndst1-/- kidneys engrafted into wildtype BALB/c mice (Ndst1+/+) and comparable to M-T7 treatment in C57Bl/6 allografts (P < 0.0081). M-T7 lost activity in Ndst1-/- allografts, while M-T7 point mutants with modified GAG-chemokine binding displayed a range of anti-rejection activity. CD3+ T cells (P < 0.0001), HS (P < 0.005) and CXC chemokine staining (P < 0.012), gene expression in NFκB and JAK/STAT pathways, and HS and CS disaccharide content were significantly altered with reduced rejection. Transplant of donor allografts with conditional Ndst1 deficiency exhibit significantly reduced acute rejection, comparable to systemic chemokine-GAG inhibition. Modified disaccharides in engrafted organs correlate with reduced rejection. Altered disaccharides in engrafted organs provide markers for rejection with potential to guide new therapeutic approaches in allograft rejection.


Subject(s)
Allogeneic Cells/enzymology , Aorta/transplantation , Endothelial Progenitor Cells/enzymology , Graft Rejection/enzymology , Myeloid Progenitor Cells/enzymology , Sulfotransferases , Allogeneic Cells/pathology , Animals , Aorta/pathology , Endothelial Progenitor Cells/pathology , Gene Deletion , Graft Rejection/genetics , Graft Rejection/pathology , Graft Rejection/prevention & control , Mice , Mice, Inbred BALB C , Myeloid Progenitor Cells/pathology , Sulfotransferases/genetics , Sulfotransferases/metabolism
19.
J Tissue Eng Regen Med ; 12(7): 1594-1607, 2018 07.
Article in English | MEDLINE | ID: mdl-29702753

ABSTRACT

Diabetes mellitus (DM) causes dysfunction of endothelial progenitor cells (EPCs), resulting in impaired wound healing. EPC therapy is a potential substitute to the current treatments of chronic wounds. Because EPCs isolated from diabetic patients are dysfunctional and therefore pose an obstacle in their efficacious employment in autologous cell therapy, a strategy to rescue them prior to transplantation would be expected to improve the efficacy of autologous cell therapy multifold. Compromised reactive oxygen species scavenging ability being the main cause of EPC dysfunction (EPCD), reactive oxygen species scavengers are likely to reverse or rescue EPCD. Therefore, in this study, we evaluated the potential of curcumin in reversing DM-induced EPCD. We found that in vitro treatment of bone marrow EPCs from diabetic mice (D-EPC) with curcumin restored their functionality, as judged by colony formation, tubule formation, and migration assays. Most importantly, autologous transplantation of curcumin-treated D-EPCs onto diabetic wounds also resulted in accelerated wound healing. Furthermore, curcumin-treated diabetic mice exhibited improved wound healing, as compared with their vehicle-treated diabetic counterparts, underscoring the efficacy of curcumin in vivo as well. The levels and activity of manganese superoxide dismutase (MnSOD) in D-EPCs treated in vitro with curcumin or those isolated from curcumin-treated diabetic mice were comparable with those in non-diabetic EPCs. Addition of methyl mercury chloride to inhibit MnSOD activity during curcumin treatment abolished the salutary effects of curcumin. Our data demonstrate that curcumin reverses DM-induced EPCD by boosting MnSOD expression and activity and emphasizes its potential for use in autologous cell therapy for diabetic wound management.


Subject(s)
Curcumin/pharmacology , Diabetes Mellitus, Experimental/drug therapy , Diabetic Angiopathies/drug therapy , Endothelial Progenitor Cells/enzymology , Gene Expression Regulation, Enzymologic/drug effects , Superoxide Dismutase/biosynthesis , Animals , Diabetes Mellitus, Experimental/enzymology , Diabetes Mellitus, Experimental/pathology , Diabetic Angiopathies/enzymology , Diabetic Angiopathies/pathology , Endothelial Progenitor Cells/pathology , Female , Male , Mice
20.
Med Sci Monit ; 24: 951-960, 2018 Feb 15.
Article in English | MEDLINE | ID: mdl-29447140

ABSTRACT

BACKGROUND Endothelial progenitor cells (EPCs) were found to be a potential therapeutic choice for low extremity deep vein thrombosis. The aim of our research was to investigate the effect of resveratrol (RSV) on EPCs that may promote thrombus resolution and its potential pathway. MATERIAL AND METHODS EPCs were pretreated with RSV and migration; angiogenesis were evaluated ex vivo. Expression of miR-138 and focal adhesion kinase (FAK) was also tested. A murine model of venous thrombosis was developed as an in vivo model. The effects of RSV treatment on mice with inferior venous thrombosis were evaluated. RESULTS We found that RSV increased EPCs migration and tube formation ex vivo. RSV significantly inhibited miR-138 expression. Moreover, we demonstrated that FAK was a target of miR-138 and revealed that FAK knockdown downregulated migration and angiogenesis of RSV-treated EPCs. In addition, RSV-induced EPCs promoted thrombus resolution in a murine model of venous thrombosis. CONCLUSIONS We found the first evidence that intravenous injection of RSV-treated EPCs enhanced thrombus resolution in vivo. RSV exerted its role by reducing miR-138 expression and therefore upregulated FAK.


Subject(s)
Endothelial Progenitor Cells/enzymology , Endothelial Progenitor Cells/pathology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , MicroRNAs/metabolism , Stilbenes/therapeutic use , Thrombosis/drug therapy , Thrombosis/enzymology , Adult , Animals , Cell Movement/drug effects , Endothelial Progenitor Cells/drug effects , Humans , Male , MicroRNAs/genetics , Neovascularization, Physiologic/drug effects , Rats, Nude , Resveratrol , Stilbenes/pharmacology , Thrombosis/genetics , Up-Regulation/drug effects , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL