Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
World J Stem Cells ; 11(1): 44-54, 2019 Jan 26.
Article in English | MEDLINE | ID: mdl-30705714

ABSTRACT

BACKGROUND: Pluripotent stem cell-derived cardiomyocytes (CMs) have become one of the most attractive cellular resources for cell-based therapy to rescue damaged cardiac tissue. AIM: We investigated the regenerative potential of mouse embryonic stem cell (ESC)-derived platelet-derived growth factor receptor-α (DGFRα)+ cardiac lineage-committed cells (CLCs), which have a proliferative capacity but are in a morphologically and functionally immature state compared with differentiated CMs. METHODS: We induced mouse ESCs into PDGFRα+ CLCs and αMHC+ CMs using a combination of the small molecule cyclosporin A, the rho-associated coiled-coil kinase inhibitor Y27632, the antioxidant Trolox, and the ALK5 inhibitor EW7197. We implanted PDGFRα+ CLCs and differentiated αMHC+ CMs into a myocardial infarction (MI) murine model and performed functional analysis using transthoracic echocardiography (TTE) and histologic analysis. RESULTS: Compared with the untreated MI hearts, the anterior and septal regional wall motion and systolic functional parameters were notably and similarly improved in the MI hearts implanted with PDGFRα+ CLCs and αMHC+ CMs based on TTE. In histologic analysis, the untreated MI hearts contained a thinner ventricular wall than did the controls, while the ventricular walls of MI hearts implanted with PDGFRα+ CLCs and αMHC+ CMs were similarly thicker compared with that of the untreated MI hearts. Furthermore, implanted PDGFRα+ CLCs aligned and integrated with host CMs and were mostly differentiated into α-actinin+ CMs, and they did not convert into CD31+ endothelial cells or αSMA+ mural cells. CONCLUSION: PDGFRα+ CLCs from mouse ESCs exhibiting proliferative capacity showed a regenerative effect in infarcted myocardium. Therefore, mouse ESC-derived PDGFRα+ CLCs may represent a potential cellular resource for cardiac regeneration.

2.
J Clin Invest ; 127(10): 3877-3896, 2017 Oct 02.
Article in English | MEDLINE | ID: mdl-28920924

ABSTRACT

Primary open-angle glaucoma (POAG) is often caused by elevated intraocular pressure (IOP), which arises due to increased resistance to aqueous humor outflow (AHO). Aqueous humor flows through Schlemm's canal (SC), a lymphatic-like vessel encircling the cornea, and via intercellular spaces of ciliary muscle cells. However, the mechanisms underlying increased AHO resistance are poorly understood. Here, we demonstrate that signaling between angiopoietin (Angpt) and the Angpt receptor Tie2, which is critical for SC formation, is also indispensable for maintaining SC integrity during adulthood. Deletion of Angpt1/Angpt2 or Tie2 in adult mice severely impaired SC integrity and transcytosis, leading to elevated IOP, retinal neuron damage, and impairment of retinal ganglion cell function, all hallmarks of POAG in humans. We found that SC integrity is maintained by interconnected and coordinated functions of Angpt-Tie2 signaling, AHO, and Prox1 activity. These functions diminish in the SC during aging, leading to impaired integrity and transcytosis. Intriguingly, Tie2 reactivation using a Tie2 agonistic antibody rescued the POAG phenotype in Angpt1/Angpt2-deficient mice and rejuvenated the SC in aged mice. These results indicate that the Angpt-Tie2 system is essential for SC integrity. The impairment of this system underlies POAG-associated pathogenesis, supporting the possibility that Tie2 agonists could be a therapeutic option for glaucoma.


Subject(s)
Angiopoietin-1/metabolism , Angiopoietin-2/metabolism , Cornea/metabolism , Glaucoma, Open-Angle/metabolism , Signal Transduction , Angiopoietin-1/genetics , Angiopoietin-2/genetics , Animals , Cornea/blood supply , Cornea/pathology , Female , Glaucoma, Open-Angle/genetics , Glaucoma, Open-Angle/pathology , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Male , Mice , Mice, Transgenic , Transcytosis/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
3.
Sci Rep ; 7: 41840, 2017 02 06.
Article in English | MEDLINE | ID: mdl-28165490

ABSTRACT

Isolating actively proliferating cardioblasts is the first crucial step for cardiac regeneration through cell implantation. However, the origin and identity of putative cardioblasts are still unclear. Here, we uncover a novel class of cardiac lineage cells, PDGFRα+Flk1- cardioblasts (PCBs), from mouse and human pluripotent stem cells induced using CsAYTE, a combination of the small molecules Cyclosporin A, the rho-associated coiled-coil kinase inhibitor Y27632, the antioxidant Trolox, and the ALK5 inhibitor EW7197. This novel population of actively proliferating cells is cardiac lineage-committed but in a morphologically and functionally immature state compared to mature cardiomyocytes. Most important, most of CsAYTE-induced PCBs spontaneously differentiated into functional αMHC+ cardiomyocytes (M+CMs) and could be a potential cellular resource for cardiac regeneration.


Subject(s)
Cell Differentiation , Myoblasts/cytology , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Receptor, Platelet-Derived Growth Factor alpha/metabolism , Amides/pharmacology , Aniline Compounds/pharmacology , Animals , Antioxidants/pharmacology , Cell Line , Cells, Cultured , Chromans/pharmacology , Cyclosporine/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Mice , Myoblasts/metabolism , Myocytes, Cardiac/metabolism , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Pyridines/pharmacology , Receptor, Platelet-Derived Growth Factor alpha/genetics , Triazoles/pharmacology
4.
Circ Res ; 119(7): 839-52, 2016 Sep 16.
Article in English | MEDLINE | ID: mdl-27528602

ABSTRACT

RATIONALE: Vascular endothelial growth factor (VEGF) signaling is a key pathway for angiogenesis and requires highly coordinated regulation. Although the Notch pathway-mediated suppression of excessive VEGF activity via negative feedback is well known, the positive feedback control for augmenting VEGF signaling remains poorly understood. Transcription factor Sox17 is indispensable for angiogenesis, but its association with VEGF signaling is largely unknown. The contribution of other Sox members to angiogenesis also remains to be determined. OBJECTIVE: To reveal the genetic interaction of Sox7, another Sox member, with Sox17 in developmental angiogenesis and their functional relationship with VEGF signaling. METHODS AND RESULTS: Sox7 is expressed specifically in endothelial cells and its global and endothelial-specific deletion resulted in embryonic lethality with severely impaired angiogenesis in mice, substantially overlapping with Sox17 in both expression and function. Interestingly, compound heterozygosity for Sox7 and Sox17 phenocopied vascular defects of Sox7 or Sox17 homozygous knockout, indicating that the genetic cooperation of Sox7 and Sox17 is sensitive to their combined gene dosage. VEGF signaling upregulated both Sox7 and Sox17 expression in angiogenesis via mTOR pathway. Furthermore, Sox7 and Sox17 promoted VEGFR2 (VEGF receptor 2) expression in angiogenic vessels, suggesting a positive feedback loop between VEGF signaling and SoxF. CONCLUSIONS: Our findings demonstrate that SoxF transcription factors are indispensable players in developmental angiogenesis by acting as positive feedback regulators of VEGF signaling.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic/physiology , SOXF Transcription Factors/physiology , Signal Transduction/physiology , Vascular Endothelial Growth Factor A/metabolism , Animals , Culture Techniques , Female , Humans , Mice , Mice, Knockout , Mice, Transgenic , Pregnancy
5.
Int J Cardiol ; 197: 33-43, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26113473

ABSTRACT

BACKGROUND: Endothelial colony forming cells (ECFCs), a subtype of endothelial progenitor cells, have been studied as a promising cellular source for therapeutic angiogenesis. Although ECFCs are very similar to mature endothelial cells, details regarding the role of ECFCs during angiogenesis are not known. We compared the cellular and angiogenic properties of ECFCs and mature endothelial cells (HUVECs). METHODS: HUVECs were used as control. Quantitative RT-PCR, western blotting, immunofluorescence staining, flow cytometric analyses and angiogenic cytokine array were performed. 3D-microfluidic angiogenesis assay system was adopted for in vitro angiogenic potential. In vivo angiogenic potential was assessed by Matrigel plug assay. RESULTS: ECFCs had higher expression of activated endothelial tip cell markers (Dll4, CXCR4, CD34, and VCAM1) and arterial genes (DLL4 and CX40), but lower expression of venous and lymphatic genes (COUP-TFII and PROX1). In 3D-microfluidic angiogenesis assay system, ECFCs induced robust sprouting vascular structures. Co-cultivation of both ECFCs and HUVECs gave rise to lumen-formed hybrid vascular structures, with the resulting ECFCs predominantly localized to the tip portion. This finding suggests that the ECFC has a role as a sprouting endothelial tip cell. Interestingly, VEGF-A phosphorylated VEGFR2 and its downstream signaling molecules more strongly in ECFCs than in HUVECs. Even small amount of VEGF-A successfully induced the sprouting angiogenesis of ECFCs. Finally, co-administration of ECFCs and human dermal fibroblasts successfully induced lumen-formed maturated neovessels in vivo. CONCLUSION: ECFCs derived from adult peripheral blood had enhanced sprouting angiogenic potential in vitro and in vivo through up-regulation of the VEGFR2 signaling pathway.


Subject(s)
Endothelial Progenitor Cells/physiology , Neovascularization, Physiologic/physiology , Signal Transduction/physiology , Up-Regulation/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Adult , Aged , Animals , Blotting, Western , Cells, Cultured , Coculture Techniques , Female , Flow Cytometry , Human Umbilical Vein Endothelial Cells/physiology , Humans , Male , Mice , Mice, SCID , Microcirculation/physiology , Middle Aged , Real-Time Polymerase Chain Reaction
6.
Circulation ; 131(11): 995-1005, 2015 Mar 17.
Article in English | MEDLINE | ID: mdl-25596186

ABSTRACT

BACKGROUND: Intracranial aneurysm (IA) is a common vascular disorder that frequently leads to fatal vascular rupture. Although various acquired risk factors associated with IA have been identified, the hereditary basis of IA remains poorly understood. As a result, genetically modified animals accurately modeling IA and related pathogenesis have been lacking, and subsequent drug development has been delayed. METHODS AND RESULTS: The transcription factor Sox17 is robustly expressed in endothelial cells of normal intracerebral arteries. The combination of Sox17 deficiency and angiotensin II infusion in mice induces vascular abnormalities closely resembling the cardinal features of IA such as luminal dilation, wall thinning, tortuosity, and subarachnoid hemorrhages. This combination impairs junctional assembly, cell-matrix adhesion, regeneration capacity, and paracrine secretion in endothelial cells of intracerebral arteries, highlighting key endothelial dysfunctions that lead to IA pathogenesis. Moreover, human IA samples showed reduced Sox17 expression and impaired endothelial integrity, further strengthening the applicability of this animal model to clinical settings. CONCLUSIONS: Our findings demonstrate that Sox17 deficiency in mouse can induce IA under hypertensive conditions, suggesting Sox17 deficiency as a potential genetic factor for IA formation. The Sox17-deficient mouse model provides a novel platform to develop therapeutics for incurable IA.


Subject(s)
Endothelium, Vascular/pathology , HMGB Proteins/deficiency , Intracranial Aneurysm/genetics , SOXF Transcription Factors/deficiency , SOXF Transcription Factors/physiology , Adult , Aged , Angiotensin II/toxicity , Animals , Aorta/pathology , Cells, Cultured , Cerebral Arteries/chemistry , Cerebral Arteries/pathology , Cyclin-Dependent Kinase Inhibitor Proteins/biosynthesis , Cyclin-Dependent Kinase Inhibitor Proteins/genetics , Dilatation, Pathologic/genetics , Dilatation, Pathologic/pathology , Disease Models, Animal , Endothelium, Vascular/metabolism , Female , HMGB Proteins/genetics , HMGB Proteins/physiology , Humans , Hypertension/complications , Intracranial Aneurysm/etiology , Intracranial Aneurysm/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Myocytes, Smooth Muscle/chemistry , Paracrine Communication , RNA Interference , SOXF Transcription Factors/analysis , SOXF Transcription Factors/genetics , Specific Pathogen-Free Organisms , Subarachnoid Hemorrhage/etiology , Transcription, Genetic , Up-Regulation , Veins/chemistry
7.
J Clin Invest ; 124(9): 3960-74, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25061877

ABSTRACT

Schlemm's canal (SC) is a specialized vascular structure in the eye that functions to drain aqueous humor from the intraocular chamber into systemic circulation. Dysfunction of SC has been proposed to underlie increased aqueous humor outflow (AHO) resistance, which leads to elevated ocular pressure, a factor for glaucoma development in humans. Here, using lymphatic and blood vasculature reporter mice, we determined that SC, which originates from blood vessels during the postnatal period, acquires lymphatic identity through upregulation of prospero homeobox protein 1 (PROX1), the master regulator of lymphatic development. SC expressed lymphatic valve markers FOXC2 and integrin α9 and exhibited continuous vascular endothelial-cadherin (VE-cadherin) junctions and basement membrane, similar to collecting lymphatics. SC notably lacked luminal valves and expression of the lymphatic endothelial cell markers podoplanin and lymphatic vessel endothelial hyaluronan receptor 1 (LYVE-1). Using an ocular puncture model, we determined that reduced AHO altered the fate of SC both during development and under pathologic conditions; however, alteration of VEGF-C/VEGFR3 signaling did not modulate SC integrity and identity. Intriguingly, PROX1 expression levels linearly correlated with SC functionality. For example, PROX1 expression was reduced or undetectable under pathogenic conditions and in deteriorated SCs. Collectively, our data indicate that PROX1 is an accurate and reliable biosensor of SC integrity and identity.


Subject(s)
Aqueous Humor/physiology , Cornea/blood supply , Homeodomain Proteins/physiology , Tumor Suppressor Proteins/physiology , Actins/analysis , Animals , Endothelial Cells/physiology , Epithelial-Mesenchymal Transition , Intraocular Pressure , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/physiology , Mice , Mice, Inbred C57BL , Morphogenesis , Vascular Endothelial Growth Factor C/physiology , Vascular Endothelial Growth Factor Receptor-3/physiology
8.
Circ Res ; 115(2): 215-26, 2014 Jul 07.
Article in English | MEDLINE | ID: mdl-24755984

ABSTRACT

RATIONALE: The Notch pathway stabilizes sprouting angiogenesis by favoring stalk cells over tip cells at the vascular front. Because tip and stalk cells have different properties in morphology and function, their transcriptional regulation remains to be distinguished. Transcription factor Sox17 is specifically expressed in endothelial cells, but its expression and role at the vascular front remain largely unknown. OBJECTIVE: To specify the role of Sox17 and its relationship with the Notch pathway in sprouting angiogenesis. METHODS AND RESULTS: Endothelial-specific Sox17 deletion reduces sprouting angiogenesis in mouse embryonic and postnatal vascular development, whereas Sox17 overexpression increases it. Sox17 promotes endothelial migration by destabilizing endothelial junctions and rearranging cytoskeletal structure and upregulates expression of several genes preferentially expressed in tip cells. Interestingly, Sox17 expression is suppressed in stalk cells in which Notch signaling is relatively high. Notch activation by overexpressing Notch intracellular domain reduces Sox17 expression both in primary endothelial cells and in retinal angiogenesis, whereas Notch inhibition by delta-like ligand 4 (Dll4) blockade increases it. The Notch pathway regulates Sox17 expression mainly at the post-transcriptional level. Furthermore, endothelial Sox17 ablation rescues vascular network from excessive tip cell formation and hyperbranching under Notch inhibition in developmental and tumor angiogenesis. CONCLUSIONS: Our findings demonstrate that the Notch pathway restricts sprouting angiogenesis by reducing the expression of proangiogenic regulator Sox17.


Subject(s)
Endothelial Cells/metabolism , HMGB Proteins/physiology , Neovascularization, Pathologic/physiopathology , Neovascularization, Physiologic/physiology , Receptors, Notch/physiology , SOXF Transcription Factors/physiology , Signal Transduction/physiology , Animals , Carcinoma, Lewis Lung/blood supply , Cell Differentiation , Cell Movement , Cytoskeleton/ultrastructure , Embryo, Mammalian/blood supply , Embryonic Stem Cells , Gene Expression Regulation , HMGB Proteins/biosynthesis , HMGB Proteins/genetics , Human Umbilical Vein Endothelial Cells , Humans , Intercellular Junctions/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Morphogenesis/genetics , Protein Structure, Tertiary , RNA, Small Interfering/pharmacology , Receptor, Notch1/genetics , Receptor, Notch1/physiology , Recombinant Fusion Proteins , Retinal Vessels/growth & development , SOXF Transcription Factors/biosynthesis , SOXF Transcription Factors/genetics , Specific Pathogen-Free Organisms , Transcription, Genetic
9.
J Am Heart Assoc ; 3(2): e000693, 2014 Mar 13.
Article in English | MEDLINE | ID: mdl-24627421

ABSTRACT

BACKGROUND: Cardiomyocytes that differentiate from pluripotent stem cells (PSCs) provide a crucial cellular resource for cardiac regeneration. The mechanisms of mitochondrial metabolic and redox regulation for efficient cardiomyocyte differentiation are, however, still poorly understood. Here, we show that inhibition of the mitochondrial permeability transition pore (mPTP) by Cyclosporin A (CsA) promotes cardiomyocyte differentiation from PSCs. METHODS AND RESULTS: We induced cardiomyocyte differentiation from mouse and human PSCs and examined the effect of CsA on the differentiation process. The cardiomyogenic effect of CsA mainly resulted from mPTP inhibition rather than from calcineurin inhibition. The mPTP inhibitor NIM811, which does not have an inhibitory effect on calcineurin, promoted cardiomyocyte differentiation as much as CsA did, but calcineurin inhibitor FK506 only slightly increased cardiomyocyte differentiation. CsA-treated cells showed an increase in mitochondrial calcium, mitochondrial membrane potential, oxygen consumption rate, ATP level, and expression of genes related to mitochondrial function. Furthermore, inhibition of mitochondrial oxidative metabolism reduced the cardiomyogenic effect of CsA while antioxidant treatment augmented the cardiomyogenic effect of CsA. CONCLUSIONS: Our data show that mPTP inhibition by CsA alters mitochondrial oxidative metabolism and redox signaling, which leads to differentiation of functional cardiomyocytes from PSCs.


Subject(s)
Cell Differentiation/drug effects , Cyclosporine/pharmacology , Energy Metabolism/drug effects , Mitochondria, Heart/drug effects , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Myocytes, Cardiac/drug effects , Pluripotent Stem Cells/drug effects , Signal Transduction/drug effects , Animals , Cell Line , Cell Proliferation/drug effects , Coculture Techniques , Dose-Response Relationship, Drug , Feeder Cells , Humans , Mice , Mitochondria, Heart/metabolism , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Permeability Transition Pore , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Pluripotent Stem Cells/metabolism , Time Factors
10.
Cell Mol Bioeng ; 6(2): 160-174, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23805169

ABSTRACT

The local hemodynamic shear stress waveforms present in an artery dictate the endothelial cell phenotype. The observed decrease of the apical glycocalyx layer on the endothelium in atheroprone regions of the circulation suggests that the glycocalyx may have a central role in determining atherosclerotic plaque formation. However, the kinetics for the cells' ability to adapt its glycocalyx to the environment have not been quantitatively resolved. Here we report that the heparan sulfate component of the glycocalyx of HUVECs increases by 1.4-fold following the onset of high shear stress, compared to static cultured cells, with a time constant of 19 h. Cell morphology experiments show that 12 h are required for the cells to elongate, but only after 36 h have the cells reached maximal alignment to the flow vector. Our findings demonstrate that following enzymatic degradation, heparan sulfate is restored to the cell surface within 12 h under flow whereas the time required is 20 h under static conditions. We also propose a model describing the contribution of endocytosis and exocytosis to apical heparan sulfate expression. The change in HS regrowth kinetics from static to high-shear EC phenotype implies a differential in the rate of endocytic and exocytic membrane turnover.

11.
Lab Chip ; 13(15): 3033-40, 2013 Aug 07.
Article in English | MEDLINE | ID: mdl-23743709

ABSTRACT

Although bacterial cancer targeting in animal models has been previously demonstrated and suggested as a possible therapeutic tool, a thorough understanding of the mechanisms responsible for cancer specificity would be required prior to clinical applications. To visualize bacterial preference for cancer cells over normal cells and to elucidate the cancer-targeting mechanism, a simple microfluidic platform has been developed for in vitro studies. This platform allows simultaneous cultures of multiple cell types in independent culture environments in isolated chambers, and creates a stable chemical gradient across a collagen-filled passage between each of these cell culture chambers and the central channel. The established chemical gradient induces chemotactic preferential migration of bacteria toward a particular cell type for quantitative analysis. As a demonstration, we tested differential bacterial behavior on a two-chamber device where we quantified bacterial preference based on the difference in fluorescence intensities of green fluorescence protein (GFP)-expressing bacteria at two exits of the collagen-filled passages. Analysis of the chemotactic behavior of Salmonella typhimurium toward normal versus cancer hepatocytes using the developed platform revealed an apparent preference for cancer hepatocytes. We also demonstrate that alpha-fetoprotein (AFP) is one of the key chemo-attractants for S. typhimurium in targeting liver cancer.


Subject(s)
Chemotaxis , Coculture Techniques/instrumentation , Hepatocytes/microbiology , Liver Neoplasms/therapy , Microfluidic Analytical Techniques/instrumentation , Salmonella typhimurium/physiology , Cell Line, Tumor , Equipment Design , Hepatocytes/metabolism , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/microbiology
12.
Lab Chip ; 13(8): 1602-11, 2013 Apr 21.
Article in English | MEDLINE | ID: mdl-23450300

ABSTRACT

Cells are inherently exposed to a number of different biophysical stimuli such as electric fields, shear stress, and tensile or compressive stress from the extracellular environment in vivo. Each of these biophysical cues can work simultaneously or independently to regulate cellular functions and tissue integrity in both physiological and pathological conditions. Thus, it is vital to understand the interaction of multiple stimuli on cells by decoupling and coupling the stimuli in simple combinations and by investigating cellular behaviors in response to these cues. Here, we report a novel microfluidic platform to apply the combinatorial stimulation of an electric field and fluid shear stress by controlling two directional cues independently. An integrated microfluidic platform was developed using soft lithography to monitor the cellular migration in real-time in response to an electric field and fluid shear stress in single, simultaneous, and sequential modes. When each of these stimulations is applied separately, normal human dermal fibroblasts migrate toward the anode and in the direction of fluid flow in a dose-dependent manner. Simultaneous stimulation with an electric field and shear stress, which mimics a wound in vivo, enhances the directional migration of fibroblasts by increasing both directedness and trajectory speed, suggesting the plausible scenario of cooperation between two physical cues to promote wound healing. When an electric field and shear stress are applied sequentially, migration behavior is affected by the applied stimulation as well as pre-existing stimulating conditions. This microfluidic platform can be utilized to understand other microenvironments such as embryogenesis, angiogenesis and tumor metastasis.


Subject(s)
Electricity , Fibroblasts/cytology , Microfluidic Analytical Techniques/methods , Shear Strength , Cell Movement , Cells, Cultured , Humans , Microfluidic Analytical Techniques/instrumentation , Time-Lapse Imaging
13.
Blood ; 120(13): 2733-44, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22896004

ABSTRACT

Successful differentiation and expansion of endothelial cells (ECs) from embryonic stem cell (ESC)-derived Flk1(+) mesodermal precursor cells (MPCs) requires supplementation of vascular endothelial growth factor-A (VEGF-A). While analyzing VEGF-A/VEGFR2 downstream signaling pathway that underlies the VEGF-A-induced differentiation and expansion of ECs, we fortuitously found that Rho-associated protein kinase (ROCK) inhibitor Y27632 profoundly promoted the differentiation and expansion of ECs from Flk1(+) MPCs while reducing the differentiation and expansion of mural cells. The ROCK suppression-induced expansion of ECs appears to have resulted from promotion of proliferation of ECs via activation of PI3-kinase-Akt signaling. The ECs obtained by the combination of ROCK suppression and VEGF-A supplementation faithfully expressed most pan-EC surface makers, and phenotypic analyses revealed that they were differentiated toward arterial EC. Further incubation of the ICAM2(+) ECs with Y27632 and VEGF-A for 2 days promoted expansion of ECs by 6.5-fold compared with those incubated with only VEGF-A. Importantly, the ROCK suppression-induced ECs displayed neovasculogenic abilities in vitro and in vivo. Thus, supplementation of ROCK inhibitor Y27632 along with VEGF-A in 2D Matrigel culture system provides a simple, efficient, and versatile method for obtaining ample amount of ESC-derived ECs at high purity suitable for use in therapeutic neovascularization.


Subject(s)
Cell Differentiation/drug effects , Embryonic Stem Cells/cytology , Endothelial Cells/cytology , Mesoderm/cytology , Neovascularization, Physiologic , Vascular Endothelial Growth Factor Receptor-2/metabolism , rho-Associated Kinases/antagonists & inhibitors , Amides/pharmacology , Animals , Blotting, Western , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Collagen/metabolism , Drug Combinations , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Enzyme Inhibitors/pharmacology , Flow Cytometry , Fluorescent Antibody Technique , Laminin/metabolism , Mesoderm/drug effects , Mesoderm/metabolism , Mice , Proteoglycans/metabolism , Pyridines/pharmacology , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/metabolism , rho-Associated Kinases/metabolism
14.
Biomaterials ; 33(7): 2007-15, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22189145

ABSTRACT

In this work, well-aligned endothelial cell (EC) layers were prepared by culturing ECs on surfaces containing nanoscale ridges/grooves fabricated by UV-assisted capillary force lithography. Then, the dynamics of T cells on well-aligned ECs were compared with that on randomly oriented ECs cultured on flat surfaces. With this experimental setting, we demonstrated for the first time that EC alignment is important for the regulation of transendothelial migration (TEM) of T cells, a critical step for leukocyte infiltration; T cells preferentially underwent TEM at the junctions surrounded by more than three ECs only if ECs surrounding those junctions were poorly aligned. As a result, TEM of T cells occurred more quickly and frequently on randomly oriented ECs cultured on flat surfaces than on well-aligned ECs cultured on nanostructured surfaces. This result will suggest a new strategy for the design of synthetic small diameter vascular grafts and extend our current knowledge of leukocyte dynamics on an inflamed endothelium.


Subject(s)
Cell Culture Techniques , Endothelial Cells/metabolism , Nanostructures , T-Lymphocytes/metabolism , Animals , Blood Vessel Prosthesis , Cell Movement/physiology , Cells, Cultured , Endothelial Cells/cytology , Inflammation/pathology , Leukocytes/cytology , Leukocytes/metabolism , Mice , Surface Properties , T-Lymphocytes/cytology
15.
Article in English | MEDLINE | ID: mdl-19964781

ABSTRACT

Vascular endothelial cell migration, which plays an important role in vascular remodeling, is known to be regulated by hemodynamic forces in the blood vessels. When shear stress is applied on mouse microvessel endothelial cells (bEnd.3) in vitro, cells exhibit upstream migration behavior with respect to the direction of the flow. To determine how shear stress magnitude influences mechanotaxis of the cells, endothelial cells were exposed to different magnitudes of unidirectional shear stress. While a higher flow rate reduces the speed of the motility, the horizontal component of the velocity parallel to the flow increases with the flow rate, indicating the higher alignment of cells in the direction parallel to the flow at a higher level of shear stress. In addition, cells seeded on softer substrate, whose elastic modulus is comparable to that of the blood vessels, show enhanced directional persistence when compared to those seeded on a stiffer substrate. The higher directionality accompanies increased stress fiber formation and focal adhesion turn-over, exhibiting higher mechanotaxis behavior. Therefore, the increased stiffness in the vessel may hinder the mechano-sensing mechanism of the endothelial cells, resulting in reduced mechanotaxis in response to hemodynamic shear stress. This substrate stiffness-dependent migration behavior can further elucidate the endothelial cell remodeling and wound healing in pathologically hardened vessels as well as re-endothelialization of vascular stents and grafted tissues.


Subject(s)
Cell Movement/physiology , Microcirculation/physiology , Actins/physiology , Animals , Blood Flow Velocity , Cell Adhesion/physiology , Cerebral Cortex/blood supply , Cerebral Cortex/cytology , Cerebral Cortex/physiology , Cerebrovascular Circulation/physiology , Endothelial Cells/physiology , Mice , Paxillin/physiology , Stress, Mechanical
SELECTION OF CITATIONS
SEARCH DETAIL
...