Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Neuromolecular Med ; 22(4): 503-516, 2020 12.
Article in English | MEDLINE | ID: mdl-32820422

ABSTRACT

Repairing the damaged blood-CNS-barrier in amyotrophic lateral sclerosis (ALS) is necessary to prevent entry of detrimental blood-borne factors contributing to motor neuron dysfunction. Recently, we showed benefits of human bone marrow endothelial progenitor cell (hBM-EPC) transplantation into symptomatic ALS mice on barrier restoration by replacing damaged endothelial cells (ECs). Additionally, transplanted cells may endogenously repair ECs by secreting angiogenic factors as our subsequent in vitro study demonstrated. Based on these study results, hBM-EPCs may secrete extracellular vesicles, which may contain and transfer diverse vesicular biomolecules towards maintenance of EC functionality. The study aimed to characterize extracellular vesicles (EVs) derived from hBM-EPCs as potential cell-free therapeutics for endothelium repair in ALS. EVs were isolated from hBM-EPC media at different culture times and vesicle properties were evaluated. The protective effects of EVs on mouse brain endothelial cells (mBECs) exposed to ALS mouse plasma were investigated. Uptake and blockage of EVs from GFP-transfected hBM-EPCs in ECs were determined in vitro. Results showed that EVs isolated from hBM-EPCs as nanosized vesicles significantly reduced mBEC damage from the pathological environment and these EVs were taken up by cells. Blockage of ß1 integrin on EVs prevented internalization of vesicles in mBECs. Together, these results provide evidence for potential of hBM-EPC-derived EVs as novel cell-free therapeutics for repair of endothelium in ALS. Although determining translational potential of hBM-EPC-derived EVs will require evaluation in vivo, this in vitro study represents a step towards an extracellular vesicle-based approach for repair of the damaged microvascular endothelium in ALS.


Subject(s)
Amyotrophic Lateral Sclerosis/therapy , Endothelial Progenitor Cells/ultrastructure , Extracellular Vesicles/transplantation , Amyotrophic Lateral Sclerosis/blood , Animals , Blood-Brain Barrier , Bone Marrow Cells , Cells, Cultured , Culture Media, Conditioned/chemistry , Disease Models, Animal , Endothelium, Vascular/pathology , Extracellular Vesicles/ultrastructure , Genes, Reporter , Humans , Male , Mice , Superoxide Dismutase-1/genetics
2.
Cells ; 9(3)2020 03 23.
Article in English | MEDLINE | ID: mdl-32210018

ABSTRACT

Appropriate mechanical properties and fast endothelialization of synthetic grafts are key to ensure long-term functionality of implants. We used a newly developed biostable polyurethane elastomer (TPCU) to engineer electrospun vascular scaffolds with promising mechanical properties (E-modulus: 4.8 ± 0.6 MPa, burst pressure: 3326 ± 78 mmHg), which were biofunctionalized with fibronectin (FN) and decorin (DCN). Neither uncoated nor biofunctionalized TPCU scaffolds induced major adverse immune responses except for minor signs of polymorph nuclear cell activation. The in vivo endothelial progenitor cell homing potential of the biofunctionalized scaffolds was simulated in vitro by attracting endothelial colony-forming cells (ECFCs). Although DCN coating did attract ECFCs in combination with FN (FN + DCN), DCN-coated TPCU scaffolds showed a cell-repellent effect in the absence of FN. In a tissue-engineering approach, the electrospun and biofunctionalized tubular grafts were cultured with primary-isolated vascular endothelial cells in a custom-made bioreactor under dynamic conditions with the aim to engineer an advanced therapy medicinal product. Both FN and FN + DCN functionalization supported the formation of a confluent and functional endothelial layer.


Subject(s)
Blood Vessel Prosthesis , Endothelial Progenitor Cells/metabolism , Fibronectins/metabolism , Tissue Engineering , Adsorption , Bioreactors , Cells, Cultured , Decorin/metabolism , Endothelial Progenitor Cells/ultrastructure , Humans , Immunity , Male , Tissue Scaffolds/chemistry
3.
Vascul Pharmacol ; 124: 106601, 2020 01.
Article in English | MEDLINE | ID: mdl-31689530

ABSTRACT

Arterial hypertension is a condition associated with endothelial dysfunction, accompanied by an imbalance in the production of reactive oxygen species (ROS) and NO. The aim of this study was to investigate and elucidate the possible mechanisms of sildenafil, a selective phosphodiesterase-5 inhibitor, actions on endothelial function in aortas from spontaneously hypertensive rats (SHR). SHR treated with sildenafil (40 mg/kg/day, p.o., 3 weeks) were compared to untreated SHR and Wistar-Kyoto (WKY) rats. Systolic blood pressure (SBP) was measured by tail-cuff plethysmography and vascular reactivity was determined in isolated rat aortic rings. Circulating endothelial progenitor cells and systemic ROS were measured by flow cytometry. Plasmatic total antioxidant capacity, NO production and aorta lipid peroxidation were determined by spectrophotometry. Scanning electron microscopy was used for structural analysis of the endothelial surface. Sildenafil reduced high SBP and partially restored the vasodilator response to acetylcholine and sodium nitroprusside in SHR aortic rings. Using selective inhibitors, our experiments revealed an augmented participation of NO, with a simultaneous decrease of oxidative stress and of cyclooxygenase-1 (COX-1)-derived prostanoids contribution in the endothelium-dependent vasodilation in sildenafil-treated SHR compared to non-treated SHR. Also, the relaxant responses to sildenafil and 8-Br-cGMP were normalized in sildenafil-treated SHR and sildenafil restored the pro-oxidant/antioxidant balance and the endothelial architecture. In conclusion, sildenafil reverses endothelial dysfunction in SHR by improving vascular relaxation to acetylcholine with increased NO bioavailability, reducing the oxidative stress and COX-1 prostanoids, and improving cGMP/PKG signaling. Also, sildenafil reduces structural endothelial damage. Thus, sildenafil is a promising novel pharmacologic strategy to treat endothelial dysfunction in hypertensive states reinforcing its potential role as adjuvant in the pharmacotherapy of cardiovascular diseases.


Subject(s)
Antihypertensive Agents/pharmacology , Aorta/drug effects , Blood Pressure/drug effects , Cyclooxygenase 1/metabolism , Endothelium, Vascular/drug effects , Hypertension/drug therapy , Membrane Proteins/metabolism , NADP/metabolism , Nitric Oxide/metabolism , Sildenafil Citrate/pharmacology , Vasodilator Agents/pharmacology , Animals , Aorta/enzymology , Aorta/physiopathology , Aorta/ultrastructure , Cyclic GMP/metabolism , Disease Models, Animal , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/ultrastructure , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiopathology , Endothelium, Vascular/ultrastructure , Hypertension/enzymology , Hypertension/pathology , Hypertension/physiopathology , Lipid Peroxidation/drug effects , Male , Oxidative Stress/drug effects , Phosphodiesterase 5 Inhibitors/pharmacology , Rats, Inbred SHR , Rats, Inbred WKY , Signal Transduction , Vasodilation/drug effects
4.
Dev Cell ; 51(3): 374-386.e5, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31564611

ABSTRACT

The lymphatic system comprises blind-ended tubes that collect interstitial fluid and return it to the circulatory system. In mammals, unidirectional lymphatic flow is driven by muscle contraction working in conjunction with valves. Accordingly, defective lymphatic valve morphogenesis results in backflow leading to edema. In fish species, studies dating to the 18th century failed to identify lymphatic valves, a precedent that currently persists, raising the question of whether the zebrafish could be used to study the development of these structures. Here, we provide functional and morphological evidence of valves in the zebrafish lymphatic system. Electron microscopy revealed valve ultrastructure similar to mammals, while live imaging using transgenic lines identified the developmental origins of lymphatic valve progenitors. Zebrafish embryos bearing mutations in genes required for mammalian valve morphogenesis show defective lymphatic valve formation and edema. Together, our observations provide a foundation from which to further investigate lymphatic valve formation in zebrafish.


Subject(s)
Lymphatic Vessels/embryology , Zebrafish/embryology , Animals , Base Sequence , Embryo, Nonmammalian/metabolism , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/ultrastructure , Face/anatomy & histology , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/metabolism , Imaging, Three-Dimensional , Larva/anatomy & histology , Larva/metabolism , Lymphatic Vessels/anatomy & histology , Lymphatic Vessels/ultrastructure , Mice , Morphogenesis , Transcription Factors/metabolism , Zebrafish/genetics , Zebrafish Proteins/metabolism
5.
J Thromb Haemost ; 17(9): 1544-1554, 2019 09.
Article in English | MEDLINE | ID: mdl-31265169

ABSTRACT

Essentials Endothelial colony forming cells (ECFCs) are a powerful tool to study vascular diseases ex vivo. Separate ECFC lines show variations in morphology and von Willebrand factor-related parameters. Maximum cell density is correlated with von Willebrand factor expression in ECFCs. Variations in ECFC lines are dependent on the age and mesenchymal state of the cells. ABSTRACT: Background Endothelial colony forming cells (ECFCs) are cultured endothelial cells derived from peripheral blood. ECFCs are a powerful tool to study pathophysiological mechanisms underlying vascular diseases, including von Willebrand disease. In prior research, however, large variations between ECFC lines were observed in, among others, von Willebrand factor (VWF) expression. Objective Understand the relation between phenotypic characteristics and VWF-related parameters of healthy control ECFCs. Methods ECFC lines (n = 16) derived from six donors were studied at maximum cell density. Secreted and intracellular VWF antigen were measured by ELISA. The angiogenic capacity of ECFCs was investigated by the Matrigel tube formation assay. Differences in expression of genes involved in angiogenesis, aging, and endothelial to mesenchymal transition (EndoMT) were measured by quantitative PCR. Results Different ECFC lines show variable morphologies and cell density at maximum confluency and cell lines with a low maximum cell density show a mixed and more mesenchymal phenotype. We identified a significant positive correlation between maximum cell density and VWF production, both at protein and mRNA level. Also, significant correlations were observed between maximum cell density and several angiogenic, aging and EndoMT parameters. Conclusions We observed variations in morphology, maximum cell density, VWF production, and angiogenic potential between healthy control ECFCs. These variations seem to be attributable to differences in aging and EndoMT. Because variations correlate with cell density, we believe that ECFCs maintain a powerful tool to study vascular diseases. It is however important to compare cell lines with the same characteristics and perform experiments at maximum cell density.


Subject(s)
Endothelial Progenitor Cells/metabolism , von Willebrand Factor/metabolism , Cell Count , Cell Shape , Cell Transdifferentiation , Cells, Cultured , Cellular Senescence , Collagen , Culture Media, Conditioned/chemistry , Drug Combinations , Endothelial Progenitor Cells/ultrastructure , Gene Expression , Humans , Laminin , Mesoderm/cytology , Neovascularization, Physiologic , Proteoglycans , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Weibel-Palade Bodies/chemistry , von Willebrand Factor/analysis , von Willebrand Factor/genetics
6.
Hear Res ; 377: 179-188, 2019 06.
Article in English | MEDLINE | ID: mdl-30954884

ABSTRACT

Can damaged or degenerated vessels be regenerated in the ear? The question is clinically important, as disruption of cochlear blood flow is seen in a wide variety of hearing disorders, including in loud sound-induced hearing loss (endothelial injury), ageing-related hearing loss (lost vascular density), and genetic hearing loss (e.g., Norrie disease: strial avascularization). Progression in cochlear blood flow (CBF) pathology can parallel progression in hair cell and hearing loss. However, neither new vessel growth in the ear, nor the role of angiogenesis in hearing, have been investigated. In this study, we used an established ex vivo tissue explant model in conjunction with a matrigel matrix model to demonstrate for the first time that new vessels can be generated by activating a vascular endothelial growth factor (VEGF-A) signal. Most intriguingly, we found that the pattern of the newly formed vessels resembles the natural 'mesh pattern' of in situ strial vessels, with both lumen and expression of tight junctions. Sphigosine-1-phosphate (S1P) in synergy with VEGF-A control new vessel size and growth. Using transgenic neural/glial antigen 2 (NG2) fluorescent reporter mice, we have furthermore discovered that the progenitors of "de novo" strial vessels are NG2-derived cells. Taken together, our data demonstrates that damaged strial microvessels can be regenerated by reprogramming NG2-derived angiogenic cells. Restoration of the functional vasculature may be critical for recovery of vascular dysfunction related hearing loss.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Antigens/metabolism , Cochlea/blood supply , Endothelial Progenitor Cells/drug effects , Neovascularization, Physiologic/drug effects , Proteoglycans/metabolism , Stria Vascularis/drug effects , Vascular Endothelial Growth Factor A/pharmacology , Animals , Antigens/genetics , Cells, Cultured , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/ultrastructure , Lysophospholipids/pharmacology , Mice, Inbred C57BL , Mice, Transgenic , Proteoglycans/genetics , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Stria Vascularis/metabolism , Stria Vascularis/ultrastructure , Tight Junction Proteins/metabolism , Tight Junctions/drug effects , Tight Junctions/metabolism
7.
Cardiovasc Res ; 114(7): 1029-1040, 2018 06 01.
Article in English | MEDLINE | ID: mdl-29566124

ABSTRACT

Aims: Previous studies have demonstrated improved cardiac function following myocardial infarction (MI) after administration of endothelial progenitor cells (EPCs) into ischaemic myocardium. A growing body of literature supports paracrine effectors, including extracellular vesicles (EVs), as the main mediators of the therapeutic benefits of EPCs. The direct use of paracrine factors is an attractive strategy that harnesses the effects of cell therapy without concerns of cell engraftment or viability. We aim to reproduce the beneficial effects of EPC treatment through delivery of EPC-derived EVs within a shear-thinning gel (STG) for precise localization and sustained delivery. Methods and results: EVs were harvested from EPCs isolated from adult male Rattus norvegicus (Wistar) rats and characterized by electron microscopy, nanoparticle tracking analysis (NTA), and mass spectrometry. EVs were incorporated into the STG and injected at the border zone in rat models of MI. Haemodynamic function, angiogenesis, and myocardial remodelling were analyzed in five groups: phosphate buffered saline (PBS) control, STG control, EVs in PBS, EVs in STG, and EPCs in STG. Electron microscopy and NTA of EVs showed uniform particles of 50-200 nm. EV content analysis revealed several key angiogenic mediators. EV uptake by endothelial cells was confirmed and followed by robust therapeutic angiogenesis. In vivo animal experiments demonstrated that delivery of EVs within the STG resulted in increased peri-infarct vascular proliferation, preservation of ventricular geometry, and improved haemodynamic function post-MI. Conclusions: EPC-derived EVs delivered into ischaemic myocardium via an injectable hydrogel enhanced peri-infarct angiogenesis and myocardial haemodynamics in a rat model of MI. The STG greatly increased therapeutic efficiency and efficacy of EV-mediated myocardial preservation.


Subject(s)
Angiogenic Proteins/metabolism , Cell-Derived Microparticles/transplantation , Endothelial Progenitor Cells/transplantation , Hyaluronic Acid/chemistry , Myocardial Infarction/surgery , Neovascularization, Physiologic , Stem Cell Transplantation/methods , Ventricular Function, Left , Animals , Cell-Derived Microparticles/metabolism , Cell-Derived Microparticles/ultrastructure , Cells, Cultured , Disease Models, Animal , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/ultrastructure , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Hydrogels , Male , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocardium/pathology , Paracrine Communication , Rats, Wistar , Recovery of Function , Signal Transduction , Time Factors , Ventricular Pressure
8.
Cell Stem Cell ; 22(3): 384-397.e6, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29429943

ABSTRACT

The generation of new blood vessels via angiogenesis is critical for meeting tissue oxygen demands. A role for adult stem cells in this process remains unclear. Here, we identified CD157 (bst1, bone marrow stromal antigen 1) as a marker of tissue-resident vascular endothelial stem cells (VESCs) in large arteries and veins of numerous mouse organs. Single CD157+ VESCs form colonies in vitro and generate donor-derived portal vein, sinusoids, and central vein endothelial cells upon transplantation in the liver. In response to injury, VESCs expand and regenerate entire vasculature structures, supporting the existence of an endothelial hierarchy within blood vessels. Genetic lineage tracing revealed that VESCs maintain large vessels and sinusoids in the normal liver for more than a year, and transplantation of VESCs rescued bleeding phenotypes in a mouse model of hemophilia. Our findings show that tissue-resident VESCs display self-renewal capacity and that vascular regeneration potential exists in peripheral blood vessels.


Subject(s)
ADP-ribosyl Cyclase/metabolism , Antigens, CD/metabolism , Endothelial Progenitor Cells/metabolism , Homeostasis , Regeneration , Animals , Biomarkers/metabolism , Blood Vessels/metabolism , Cell Lineage , Colony-Forming Units Assay , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/transplantation , Endothelial Progenitor Cells/ultrastructure , Factor VIII/metabolism , GPI-Linked Proteins/metabolism , Liver/cytology , Liver/physiology , Mice, Inbred C57BL
9.
Sci Rep ; 8(1): 110, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29311692

ABSTRACT

Decorin (DCN) is an important small leucine-rich proteoglycan present in the extracellular matrix (ECM) of many organs and tissues. Endothelial progenitor cells (EPCs) are able to interact with the surrounding ECM and bind to molecules such as DCN. Here, we recombinantly produced full-length human DCN under good laboratory practice (GLP) conditions, and after detailed immunological characterization, we investigated its potential to attract murine and human EPCs (mEPCs and hECFCs). Electrospun polymeric scaffolds were coated with DCN or stromal cell-derived factor-1 (SDF-1α) and were then dynamically cultured with both cell types. Cell viability was assessed via imaging flow cytometry. The number of captured cells was counted and compared with the non-coated controls. To characterize cell-scaffold interactions, immunofluorescence staining and scanning electron microscopy analyses were performed. We identified that DCN reduced T cell responses and attracted innate immune cells, which are responsible for ECM remodeling. A significantly higher number of EPCs attached on DCN- and SDF-1α-coated scaffolds, when compared with the uncoated controls. Interestingly, DCN showed a higher attractant effect on hECFCs than SDF-1α. Here, we successfully demonstrated DCN as promising EPC-attracting coating, which is particularily interesting when aiming to generate off-the-shelf biomaterials with the potential of in vivo cell seeding.


Subject(s)
Decorin/metabolism , Endothelial Progenitor Cells/metabolism , Recombinant Proteins/metabolism , Animals , CHO Cells , Cell Survival/drug effects , Cells, Cultured , Cricetulus , Decorin/immunology , Decorin/pharmacology , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/ultrastructure , Extracellular Matrix/metabolism , Fluorescent Antibody Technique , Humans , Immunity , Monocytes/immunology , Monocytes/metabolism , Protein Binding , Recombinant Proteins/pharmacology , Tissue Scaffolds
10.
Biomed Pharmacother ; 92: 86-94, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28531804

ABSTRACT

Endothelial progenitor cells (EPCs), widely existing in bone marrow and peripheral blood, are involved in the repair of injured vascular endothelium and angiogenesis which are important to diabetic mellitus (DM) patients with vascular complications. The number and the function of EPCs are related to the advanced glycation end products (AGEs) generated in DM patients. Lycopene (Lyc) is an identified natural antioxidant that protects EPCs under the microenvironment of AGEs from damage. However, the underlying mechanism remains unclear. To investigate the effect of Lyc on EPCs, we isolated EPCs from DM rat bone marrow and determined cell proliferation, cell cycle,apoptosis and autophagy of EPCs. The present study showed that 10µg/mL Lyc improved cell proliferation and had low cytotoxicity in the presence of AGEs. In addition, Lyc rescued S phase of the cell cycle arrest, reduced apoptosis rate and decreased autophagic reaction including ROS and mitochondrial membrane potential (MMP) of EPCs. Moreover, Lyc combined use of autophagy inhibitors, 3-MA, had better protective effects. Taken together, our data suggests that Lyc promotes EPCs survival and protect EPCs from apoptosis and oxidative autophagy induced by AGEs, further remaining the number and function of EPCs. This study provides new insights into Lyc protective mechanism of AGEs-induced oxidative autophagy in EPCs from DM patients and offers a new therapy for DM vascular complications.


Subject(s)
Antioxidants/metabolism , Autophagy , Carotenoids/metabolism , Diabetes Mellitus, Type 2/metabolism , Endothelial Progenitor Cells/metabolism , Glycation End Products, Advanced/antagonists & inhibitors , Oxidative Stress , Animals , Antioxidants/adverse effects , Apoptosis , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Bone Marrow Cells/ultrastructure , Carotenoids/adverse effects , Cell Proliferation , Cells, Cultured , Diabetes Mellitus, Type 2/pathology , Dietary Supplements/adverse effects , Endothelial Progenitor Cells/pathology , Endothelial Progenitor Cells/ultrastructure , Glycation End Products, Advanced/adverse effects , Lycopene , Membrane Potential, Mitochondrial , Microscopy, Electron, Transmission , Rats, Sprague-Dawley , Reactive Oxygen Species/antagonists & inhibitors , Reactive Oxygen Species/metabolism , S Phase
11.
J Biomed Mater Res B Appl Biomater ; 105(8): 2443-2454, 2017 Nov.
Article in English | MEDLINE | ID: mdl-27654960

ABSTRACT

OBJECTIVE: The aim of this study was to explore an innovative cell-seeding technology applied on artificial vascular scaffolds. METHODS: Scaffolds were fabricated by electrospinning polycaprolactone (PCL) and seeded with rat endothelial progenitor cells differentiated from adipose-derived stem cells. Then, we modified the PCL scaffolds through the use of alginate hydrogel conglutinating cells (AHCC), a blank alginate hydrogel coating (BAHC), and natural sedimentation seeding cells (NSSC). The blank PCL (BP) scaffolds without any modifications were considered the blank control group. After modification, the scaffolds were implanted in a rat model. The implanted scaffolds were harvested and observed using histological and immunohistochemical methods and scanning electron microscopy (SEM) at 1, 2, and 4 weeks after implantation, respectively. RESULTS: The best regeneration and configuration of the endothelium tissue and the most similar morphology to that of natural endangium was observed qualitatively in the AHCC scaffolds. The BP scaffolds had qualitatively the worst regeneration and configuration and the most dissimilar morphology at the same time point. In the AHCC group, cells could adhere directly on the inner surface of the vascular scaffolds, eliminating the time delay via the NSSC method prior to cell adhesion. CONCLUSION: AHCC are an effective method for seeding cells on vascular scaffolds and can eliminate the time delay for cell adhesion. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater, 105B: 2443-2454, 2017.


Subject(s)
Alginates/chemistry , Blood Vessel Prosthesis , Endothelial Progenitor Cells/metabolism , Hydrogels/chemistry , Materials Testing , Polyesters/chemistry , Tissue Scaffolds/chemistry , Animals , Endothelial Progenitor Cells/ultrastructure , Glucuronic Acid/chemistry , Hexuronic Acids/chemistry , Male , Rats , Rats, Sprague-Dawley
12.
Mol Med Rep ; 14(6): 5551-5555, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27878275

ABSTRACT

Vascular development is a regulated process and is dependent on the participation and differentiation of many cell types including the proliferation and migration of vascular endothelial cells and differentiation of endothelial progenitor cells (EPCs) to mesodermal precursor cells. Thus, reconstitution of this process in vitro necessitates providing ambient conditions for generating and culturing EPCs in vitro and differentiating them to vascular endothelial cells. In the present study, we developed methods to differentiate bone marrow mesenchymal stem cells (MSC) into EPCs and to vascular endothelial cells. Bone marrow MSC from canines and human sources were differentiated in vitro in to EPCs. These EPCs were able to express a variety of endothelial markers following 7 days in culture. Further culturing led to the appearance of an increased number and proportion of endothelial cells. These cells were stable even after 30 generations in culture. There was a gradual loss of CD31 and increased expression of factor VIII, VEGFR and CD133. VEGF being highly angiogenic, helps in the vascular development. These results provide the basis for the possible development of vasculature in vitro conditions for biomedical applications and in vivo for organ/tissue reconstruction therapies.


Subject(s)
Cell Differentiation , Endothelial Cells/cytology , Endothelial Progenitor Cells/cytology , Mesenchymal Stem Cells/cytology , Aged , Animals , Biomarkers , Cell Culture Techniques , Cell Proliferation , Cells, Cultured , Dogs , Endothelial Cells/metabolism , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/ultrastructure , Female , Humans , Immunophenotyping , Male , Mesenchymal Stem Cells/metabolism , Middle Aged , Phenotype
13.
Mater Sci Eng C Mater Biol Appl ; 69: 780-8, 2016 Dec 01.
Article in English | MEDLINE | ID: mdl-27612772

ABSTRACT

Bone tissue engineering is sought to apply strategies for bone defects healing without limitations and short-comings of using either bone autografts or allografts and xenografts. The aim of this study was to fabricate a thin layer poly(lactic-co-glycolic) acid (PLGA) coated beta-tricalcium phosphate (ß-TCP) scaffold with sustained release of vascular endothelial growth factor (VEGF). PLGA coating increased compressive strength of the ß-TCP scaffolds significantly. For in vitro evaluations, canine mesenchymal stem cells (cMSCs) and canine endothelial progenitor cells (cEPCs) were isolated and characterized. Cell proliferation and attachment were demonstrated and the rate of cells proliferation on the VEGF released scaffold was significantly more than compared to the scaffolds with no VEGF loading. A significant increase in expression of COL1 and RUNX2 was indicated in the scaffolds loaded with VEGF and MSCs compared to the other groups. Consequently, PLGA coated ß-TCP scaffold with sustained and localized release of VEGF showed favourable results for bone regeneration in vitro, and this scaffold has the potential to use as a drug delivery device in the future.


Subject(s)
Bone and Bones/physiology , Calcium Phosphates/chemistry , Coated Materials, Biocompatible/chemistry , Lactic Acid/chemistry , Polyglycolic Acid/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Vascular Endothelial Growth Factor A/pharmacology , Animals , Bone and Bones/drug effects , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Compressive Strength , Dogs , Drug Liberation , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/ultrastructure , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/ultrastructure , Polylactic Acid-Polyglycolic Acid Copolymer , Porosity , Real-Time Polymerase Chain Reaction , X-Ray Diffraction
14.
Neurol Res ; 38(4): 333-41, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27125512

ABSTRACT

Recurrences of aneurysms remain the major drawback of detachable coils for the endovascular treatment of intracranial aneurysms. The aim of the present study is to develop new modified coils, coating the surface of platinum coils with silk fibroin (SF) consisting of stromal cell-derived factor-1α (SDF-1α), and evaluate its acceleration of organization of cavities and reduction of lumen size in a rat aneurysm model. The morphological characteristics of SDF-1α-coated coils were examined using scanning electron microscopy (SEM). Fifty experimental aneurysms were created and randomly divided into five groups: three groups were embolized with SDF-1α-coated coils (8 mm) and two of these groups need transplantation of mesenchymal stem cells (MSCs) or endothelial progenitor cells (EPCs); one group was embolized with bare coils (8 mm) and another group severed as control. After coil implantation for 14 or 28 days, the coils were harvested and histological analysis was performed. SEM photographs showed that SF/SDF-1α-coated coils have uniform size and a thin film compared with bare coils. In the group treated with SDF-1α-coated coils, tissue organization was accelerated and the proliferation of α-smooth muscle actin positive cells was promoted in the aneurysmal sac. Compared with unmodified coils, on day 28, tissue organization was significantly greater in the group treated with SDF-1α-coated coils and MSC or EPC transplantation. These results suggest that SDF-1α-coated coils with MSC or EPC transplantation may be beneficial in the aneurysm healing and endothelialization at the orifice of embolized aneurysm.


Subject(s)
Aneurysm/therapy , Chemokine CXCL12/therapeutic use , Coated Materials, Biocompatible/therapeutic use , Endothelial Progenitor Cells/transplantation , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/physiology , Animals , Chemokine CXCL12/metabolism , Disease Models, Animal , Embolization, Therapeutic , Endothelial Progenitor Cells/ultrastructure , Humans , Mesenchymal Stem Cells/ultrastructure , Microscopy, Electron, Scanning , Rats , Rats, Sprague-Dawley , Time Factors
15.
J Neurol Sci ; 362: 91-9, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26944125

ABSTRACT

BACKGROUND AND PURPOSE: Statins are active in reducing plasma lipids, suppressing inflammation and promoting angiogenesis. Because angiogenesis is critical for the absorbance of subdural hematoma (SDH), we hypothesize that atorvastatin promotes angiogenesis to enhance hematoma absorption. METHODS: SDH was induced in adult Wistar rats and treated with 3mg/kg, 8mg/kg of atorvastatin, or vehicle saline daily for 7days. The treated rats were examined for the level of CD34+/CD133+ endothelial progenitor cells (EPCs) in the circulation by flow cytometry, hematoma volumes by magnetic resonance imaging (MRI), and changes in cognitive functions. We also examined angiogenesis in the hematoma wall by transmission electronic microscopy and immunohistochemistry for the expression of vascular endothelial growth factor (VEGF), matrix metalloprotease 9 (MMP 9) and angiopoietin. RESULTS: SDH volume was significantly reduced and neurological deficits improved in rats receiving the low dose atorvastatin compared to those receiving either the high dose of atorvastatin or saline. Consistent with these outcome measures, the low dose atorvastatin increased the expression of angiopoient-1 and VEGF and reduced MMP9 expression in the connective tissue of the SDH wall, resulting in an increased vascular density and enhanced vascular maturation. CONCLUSIONS: The low-dose atorvastatin is effective in reducing SDH and improving neurological deficits in a rat model, primarily by promoting angiogenesis and vascular maturation.


Subject(s)
Anticholesteremic Agents/therapeutic use , Atorvastatin/therapeutic use , Hematoma, Subdural/drug therapy , Neovascularization, Physiologic/drug effects , AC133 Antigen/blood , Analysis of Variance , Angiopoietin-1/genetics , Angiopoietin-1/metabolism , Animals , Antigens, CD34/blood , Atorvastatin/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Progenitor Cells/drug effects , Endothelial Progenitor Cells/ultrastructure , Gene Expression Regulation/drug effects , Hematoma, Subdural/diagnostic imaging , Magnetic Resonance Imaging , Male , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Microscopy, Electron, Transmission , RNA, Messenger/metabolism , Rats , Rats, Wistar , Time Factors , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
16.
Sci Rep ; 5: 11594, 2015 Jun 25.
Article in English | MEDLINE | ID: mdl-26108983

ABSTRACT

Endothelial progenitor cells (EPCs) seeded on biomaterials can effectively promote diabetic ischemic wound healing. However, the function of transplanted EPCs is negatively affected by a high-glucose and ischemic microenvironment. Our experiments showed that EPC autophagy was inhibited and mitochondrial membrane potential (MMP) was increased in diabetic patients, while adenosine treatment decreased the energy requirements and increased the autophagy levels of EPCs. In animal experiments, we transplanted a biomaterial seeded with EPCs onto the surface of diabetic wounds and found that adenosine-stimulated EPCs effectively promoted wound healing. Increased microvascular genesis and survival of the transplanted cells were also observed in the adenosine-stimulated groups. Interestingly, our study showed that adenosine increased the autophagy of the transplanted EPCs seeded onto the biomaterial and maintained EPC survival at 48 and 96 hours. Moreover, we observed that adenosine induced EPC differentiation through increasing the level of autophagy. In conclusion, our study indicated that adenosine-stimulated EPCs seeded onto a biomaterial significantly improved wound healing in diabetic mice; mechanistically, adenosine might maintain EPC survival and differentiation by increasing high glucose-inhibited EPC autophagy and maintaining cellular energy metabolism.


Subject(s)
Adenosine/pharmacology , Autophagy/drug effects , Diabetes Mellitus, Experimental/physiopathology , Endothelial Progenitor Cells/drug effects , Ulcer/therapy , Wound Healing/drug effects , Animals , Apoptosis/drug effects , Biocompatible Materials , Blood Vessels/drug effects , Blood Vessels/physiopathology , Blotting, Western , Cell Differentiation/drug effects , Cell Survival/drug effects , Cells, Cultured , Endothelial Progenitor Cells/transplantation , Endothelial Progenitor Cells/ultrastructure , Energy Metabolism/drug effects , Humans , Ischemia/physiopathology , Male , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Stem Cell Transplantation/methods , Transplantation, Heterologous , Ulcer/physiopathology
17.
Histol Histopathol ; 30(11): 1333-40, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25972019

ABSTRACT

Development of an efficient vascular substitute by tissue engineering is strongly dependent on endothelial cell viability. The aim of this study was to evaluate cell viability of transdifferentiated endothelial-like cells (Tr-ELC) by using for the first time electron probe X-ray microanalysis (EPXMA), not only to accurately analyze cell viability by quantifying the intracellular ionic concentrations, but also to establish their possible use in vascular tissue engineering protocols. Human umbilical cord Wharton's jelly stem cells (HWJSC) and endothelial cells from the human umbilical vein (HUVEC) were isolated and cultured. Transdifferentiation from HWJSC to the endothelial phenotype was induced. EPXMA was carried out to analyze HUVEC, HWJSC and Tr-ELC cells by using a scanning electron microscope equipped with an EDAX DX-4 microanalytical system and a solid-state backscattered electron detector. To determine total ion content, the peak-to-local-background (P/B) ratio method was used with reference to standards composed of dextran containing known amounts of inorganic salts. Our results revealed a high K/Na ratio in Tr-ELC (9.41), in association with the maintenance of the intracellular levels of chlorine, phosphorous and magnesium and an increase of calcium (p=0.031) and sulfur (p=0.022) as compared to HWJSC. Calcium levels were similar for HUVEC and Tr-ELC. These results ensure that transdifferentiated cells are highly viable and resemble the phenotypic and microanalytical profile of endothelial cells. Tr-ELC induced from HWJSC may fulfill the requirements for use in tissue engineering protocols applied to the vascular system at the viability and microanalytical levels.


Subject(s)
Cell Transdifferentiation , Electron Probe Microanalysis , Endothelial Progenitor Cells/physiology , Human Umbilical Vein Endothelial Cells/physiology , Mesenchymal Stem Cells/physiology , Tissue Engineering/methods , Biomarkers/metabolism , Cell Separation , Cell Survival , Cells, Cultured , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/ultrastructure , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/ultrastructure , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/ultrastructure , Phenotype
18.
Mol Med Rep ; 11(5): 3814-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25529111

ABSTRACT

Endothelial progenitor cells (EPCs) have an essential role in counteracting risk factor­induced endothelial injury and protecting against the development of vascular injury, such as myocardial infarction. Magnetic resonance imaging (MRI) was reported to be effective in tracking transplanted stem cells following cell­labeling with superparamagnetic iron oxide (SPIO) nanoparticles. SPIO has previously been used to label and track EPCs; however, the safest concentration of SPIO for labeling EPCs on a cellular level has remained to be elucidated. In addition, the optimum number of SPIO­labeled cells required to produce the highest quality magnetic resonance images has not yet been determined. In the present study, EPCs were isolated from the bone marrow of minipigs using density gradient centrifugation. Their biological activity was then studied using flow cytometric analysis. Cells were incubated at different concentrations of SPIO for different durations and then the growth curve, apoptosis, morphology and labeling efficiency of the EPCs were detected using optical and electron microscopy. T2­weighted fast spin­echo (T2WITSE) MRI of the different numbers of SPIO­labeled EPCs (35 µg/ml) were then obtained in axial and sagittal planes. The results of the present study demonstrated that EPCs were efficiently labeled with SPIO, with a labeling efficiency in each group of ~100% following incubation for 24 h. SPIO was found to be localized in the endosomal vesicles of EPCs, which was confirmed by electron microscopy. When the concentration of SPIO was <70 µg/ml, no significant differences were observed in cell viability, proliferative capability (P>0.05) and morphology between labeled and unlabeled EPCs. Furthermore, the T2WITSE signal intensity was significantly decreased in the groups of 5.0x105/ml and 1.0x105/ml compared with that of the control (P<0.05). In conclusion, the results of the present study indicated that 35 µg/ml was the most effective concentration of SPIO to label EPCs in vitro and acquire a high quality MRI. These findings may therefore contribute to the development of a promising novel therapeutic method for the treatment of myocardial infarction following autograft with SPIO­labeled EPCs in vivo.


Subject(s)
Cell Tracking/methods , Endothelial Progenitor Cells/metabolism , Ferric Compounds , Magnetite Nanoparticles , Animals , Biomarkers , Cell Proliferation , Cell Survival , Endothelial Progenitor Cells/cytology , Endothelial Progenitor Cells/ultrastructure , Immunophenotyping , In Vitro Techniques , Magnetic Resonance Imaging/methods , Swine , Swine, Miniature
19.
Clin Exp Pharmacol Physiol ; 41(5): 351-7, 2014 May.
Article in English | MEDLINE | ID: mdl-24750349

ABSTRACT

Dysfunction of endothelial progenitor cells (EPC) contribute to diabetic vascular disease. MicroRNAs (miRNAs) are key regulators of diverse cellular processes, including angiogenesis. We recently reported that downregulated miR-130a in patients with Type 2 diabetes mellitus (DM) results in EPC dysfunction, including increased apoptosis, likely via its target runt-related transcription factor 3 (Runx3). However, whether miR-130a affects the autophagy of EPC is unknown. The aim of the present study was to explore the effects of miR-130a on the autophagy and cell death of EPC, as well as their expression of Beclin 1 (BECN1; an initiator of autophagosome formation) and the anti-apoptotic protein Bcl2 (which binds to and inactivates BECN1), and the role of Runx3 in mediating these effects. The EPC were cultured from peripheral blood mononuclear cells of diabetic patients and non-diabetic controls. Cells were transfected with an miR-130a inhibitor, or mimic-miR-130a or mimic-miR-130a plus lentiviral vector expressing Runx3 to manipulate miR-130a and/or Runx3 levels. The number of autophagosomes was counted under transmission electron microscopy and cell death was examined by flow cytometry. The mRNA expression of Beclin1 was measured by real-time polymerase chain reaction and the protein expression of Beclin1 and Bcl2 was determined by western blotting. Both the number of autophagosomes and Beclin1 expression were increased in EPC from patients with DM. Inhibition of miR-130a increased the number of autophagosomes and Beclin1 expression, but attenuated Bcl2 expression. Overexpression of miR-130a decreased the number of autophagosomes, cell death and Beclin1 expression, but promoted Bcl2 expression; these effects were mediated by Runx3. In conclusion, miR-130a is important for maintaining normal autophagy levels and promoting the survival of EPC via regulation of Bcl-2 and Beclin1 expression, via Runx3. MiR-130a may be a regulator linking apoptosis and the autophagy of EPC.


Subject(s)
Autophagy , Core Binding Factor Alpha 3 Subunit/metabolism , Diabetes Mellitus, Type 2 , Endothelial Progenitor Cells/pathology , MicroRNAs/genetics , Apoptosis Regulatory Proteins/genetics , Autophagy/genetics , Beclin-1 , Blotting, Western , Cells, Cultured , Core Binding Factor Alpha 3 Subunit/genetics , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Endothelial Progenitor Cells/metabolism , Endothelial Progenitor Cells/ultrastructure , Flow Cytometry , Humans , Lentivirus/genetics , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Leukocytes, Mononuclear/ultrastructure , Membrane Proteins/genetics , Microscopy, Electron, Transmission , Plasmids , Proto-Oncogene Proteins c-bcl-2/genetics , RNA, Small Interfering/genetics , Transfection , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...