Literature
首页医源资料库在线期刊动脉硬化血栓血管生物学杂志2006年第26卷第10期

Sca-1 + Progenitors Derived From Embryonic Stem Cells Differentiate Into Endothelial Cells Capable of Vascular Repair After Arterial Injury

来源:《动脉硬化血栓血管生物学杂志》
摘要:【摘要】Background-Embryonicstemcellspossesstheabilitytodifferentiateintoendothelium。Theabilitytoproducelargevolumesofendotheliumfromembryonicstemcellscouldprovideapotentialtherapeuticmodalityforvascularinjury。Wedescribeanapproachthatselectsendothelia......

点击显示 收起

【摘要】  Background- Embryonic stem cells possess the ability to differentiate into endothelium. The ability to produce large volumes of endothelium from embryonic stem cells could provide a potential therapeutic modality for vascular injury. We describe an approach that selects endothelial cells using magnetic beads that may be used therapeutically to treat arterial injury.

Methods and Results- Large numbers of endothelial cells (ECs) with high purity were produced using Sca-1 + cells isolated with magnetic beads from predifferentiated embryonic stem cells (ESCs) cultured in -MEM containing 10 ng/mL VEGF 165 for a minimum of 21 days (esEC). The transcription regulator histone deacetylase (HDAC3) was essential for VEGF-induced EC differentiation. Immunofluorescence or fluorescence-activated cell sorter (FACS) analysis revealed that esECs expressed a full range of EC lineage-specific markers including CD31, CD106, CD144, Flk-1, Flt-1, and von Willebrand factor (vWF). FACS analysis confirmed that 99% of esECs were CD31-positive and 75% vWF-positive. Furthermore, almost all cells were positive for DiI-acLDL uptake. When matrigel containing esECs was subcutaneously implanted into mice, various vessel-like structures were observed indicating their endothelial cell like phenotype. In keeping with this, when esECs infected with adenovirus-LacZ were injected into denuded femoral arteries of mice, they were found to form a neo-endothelium that covered the injured areas (86%±13.6%), which resulted in a 73% decrease in neointimal area 2 weeks after injury.

Conclusions- We conclude that Sca-1 + cells can differentiate into functional ECs via activation of HDAC3, accelerating re-endothelialization of injured arteries and reducing neointima formation.

We have successfully established a method for producing large numbers of endothelial cells (esECs) with high purity from embryonic stem cells and evaluated the therapeutic effect of esECs on re-endothelialization and neointima formation in a mouse model of arterial injury. We also demonstrate for the first time to our knowledge that HDAC3 is involved in VEGF-induced EC differentiation in Sca-1 + progenitor cells.

【关键词】  endothelial cells histone deacetylase progenitor cells stem cells stem cell therapy vascular injury


Introduction


Balloon angioplasty and stenting is routinely used in clinical practice to treat patients with flow-limiting atherosclerosis. However, stenting is limited by restenosis, a homeostatic response to vascular injury, which leads to the recurrence of symptoms. Balloon inflation at high pressures and the foreign metallic stent struts lead to loss of the endothelium with subsequent smooth muscle cell (SMC) proliferation and matrix deposition leading to luminal narrowing after the procedure. It has been shown that accelerated re-endothelialization by mature endothelial cells (ECs) effectively inhibits SMC migration, proliferation, and neointima formation, and therefore, prevents the development of the early stages of restenosis after vascular injury. 1 In recent years, increasing evidence indicates a repairing capacity of endothelial progenitor cells (EPCs), 2,3 providing a novel cell therapeutic option for various vascular diseases.


Embryonic stem cells (ESCs) are a promising source of pluripotent stem cells. They have the capacity for unlimited growth and self-renewal and the ability to differentiate into all types of mature tissue cells. In the past several years, accumulating evidence indicates that ES cells can differentiate into ECs in vitro or in vivo. 4-8 Methods to produce large number of ECs with high purity from ES cells in vitro are lacking, and little is known about the mechanism of EC differentiation and the therapeutic potential of ESC-derived ECs in cardiovascular diseases.


ESCs undergo complex gene-specific and functionally important remodeling of chromatin structure necessary for differentiation. Histone acetyltransferases (HATs) and histone deacetylases (HDACs) are critical to these processes regulating the structure and function of chromatin. Inhibition of HDACs is reported to block tumor angiogenesis, 9-11 and prevent ESC differentiation, 12 indicating that some HDACs are involved in ESC differentiation. Modulation of HDACs to promote production of endothelial cells from ESCs is unexplored.


Our previous data showed that stem cell antigen-1 (Sca-1) can be used as a sorting marker for vascular progenitors to allow isolation of these cells from adult adventitial tissues. 13 We hypothesized that Sca-1 + progenitor cells derived from ESCs can serve as EC progenitors leading to ECs in vitro, and furthermore that HDAC3 is important in EC differentiation from Sca-1 + progenitor cells. Because endothelial dysfunction plays an essential role in vascular diseases, we also postulate that local transplantation of ESC-derived ECs (esECs) have therapeutic potential as a strategy to enhance the biological function of reconstituted endothelium. In the present study, we isolated Sca-1 + cells from mouse ES cells, differentiated these progenitors into ECs, and characterized these cells in detail. We have successfully prepared large numbers of ECs with high purity from ES cells, and evaluated the therapeutic effect of ECs on re-endothelialization and neointima formation in a mouse model of wire induced femoral artery injury.


Materials and Methods


The detailed materials and methodology for vascular endothelial growth factor (VEGF) treatment, trichostatin A (TSA) treatment, histone deacetylase activity assay, histology, chromosome detection, and X-gal staining for detection of LacZ, Western blot analysis, Uptake of DiI-Ac-LDL, double-immunofluorescence staining, flow cytometry analysis, esEC-induced angiogenesis in vivo, morphometric analysis, and quantification of lesion formation are available online (please see http://atvb.ahajournals.org).


ES Cell Culture, Cell Sorting, and EC Differentiation


Mouse ES cells (ES-D3 cell line; ATCC, Manassas, Va) were maintained as described previously. 14 ES-D3 cells were cultured on type IV mouse collagen-coated (Trevigen, Gaithersburg, Md) flasks in basic differentiation medium (DM) ( -minimal essential medium ; Gibco), supplemented with 10% fetal calf serum (Gibco) and 0.05 mmol/L 2-mercaptoethanol (Sigma) for 3 to 4 days. As described in previous studies, 13 3 to 10 x 10 5 of stem cell antigen-1-positive cells (Sca-1 + cells) were sorted from 1.0 x 10 7 of cultured ES cells by magnetic labeling cell sorting (MACS) with anti-Sca-1 immunomagnetic microbeads (Miltenyi Biotec, GmbH, Bergisch Gladbach, Germany). To obtain higher numbers of Sca-1 + 5 x 10 6 ), these cells were expanded in ES cell culture medium for 5 to 7 population doublings. For EC differentiation, Sca-1 + cells were plated on collagen IV-coated dishes or flasks, and cultured in EC-differentiated medium [basic DM plus 10 ng/mL VEGF 165 (Bender MedSystems, Vienna, Austria) for 3, 7, and 9 days, or passaged every 3 days to 21 days for longer EC differentiation.


Plasmids and Transient Transfection


Reporter systems for CD31, CD144, endothelial nitric oxide synthase (eNOS), Flk-1, Flt -1, and von Willebrand factor (vWF) were cloned into pGL3-Luc vectors, and confirmed by DNA sequencing. For transient transfection, Sca-1 + cells were cultured in collagen IV-coated 12-well plate for 4 days, then transfected with reporter gene (0.4 µg/well) alone or together with HDAC3-siRNA (0.067 nmol per well). pRL-TK vector (Promega; 50 ng per well) was included in all transfection assay as internal control, and control siRNA #4611 was used as mock control; 48 hours later, the transfected cells were subjected to 20 ng/mL VEGF treatment for 12 hours or 24 hours. Dual luciferase activity assay was performed at 72 hours after transfection, and detected with a commercial kit (Promega). Relative luciferase unit was defined as the ratio of luciferase activity of specific gene to Renilla luciferase activity with that of control set as 1.0.


Adenoviral HDAC3 Gene Transfer


HDAC3 cDNA was amplified from embryonic stem cell with a primer set of 5'-atgacc ggtaccgtggcgtatttctacgac-3 and 5'-cacagcaagcttgctgctctaaatctccac-3', and cloned into Kpn I/ Hind III sites of pShuttle2-Flag vector, creating the plasmid pShuttle2-Flag-HDAC3, which was verified by sequencing. Ad-Flag-HDAC3 was created from pShuttle2-Flag-HDAC3 with Adeno-XTM expression system (PT3414-1; Clontech, Mountain View, Calif) and virus was produced, amplified, and titered according to protocol provided. For adenoviral gene transfer, the Sca-1 + cells were cultured in DM for 4 days and infected with Ad-HDAC3 or Ad-TTA with 5 or 10 multiple of infection (MOI) for additional 3 days, followed by Western blot analysis. The Ad-TTA virus was used as control and to compensate the MOI.


siRNA Knockdown Experiment


The control siRNA (#4611) and the siRNA for HDAC3 (5'-ccucaucgccuggcauugatt-3' and 5'-ucaaugccaggcgaugaggtt-3') were purchased or synthesized from Ambion Ltd (Huntingdon, Cambridgeshire, UK). For siRNA knockdown experiments, Sca-1 + cells were cultured on collagen IV-coated plates for 3 days, and the medium was refreshed at 24 hours and 1 hour before transfection; 10 µL of 10 mmol/L siRNA per well (0.2 nmol per 10 cm 2 ) was introduced into the cells with siIMPORTER transfection reagents (Upstate) according to the protocol provided. The transfected cells were further cultured for 48 hours, subjected to VEGF treatment for 24 hours.


Mouse Femoral Artery Denudation Injury and Cell Delivery


ApoE-deficient mice were anesthetized and the surgical procedure was similar to that described previously. 15,16 Removal of the endothelium of the femoral arteries was achieved by 3 passages of a 0.25 mm angioplasty guide wire (Brivant Ltd, Ireland). After removal of the wire, the femoral artery was temporarily clamped at the level of the inguinal ligament, then 50 µL medium with or without esECs with passage number 5 to 10 (5 x 10 5 ), which means that esEC were cultured for 21 days or more in the presence of VEGF, was injected into the injured vessel and ligated. After 30 minutes of incubation, the clamp was removed, and the wound was closed. In some experiments, esECs were prelabeled with adenovirus-LacZ virus (MOI: 20) to trace these cells in vivo.


Statistical Analysis


Data expressed as the mean±SEM were analyzed with a 2-tailed Student t test for 2 groups or pair-wise comparisons. A value of P <0.05 was considered to be significant.


Results


Characterization of ESC-Derived ECs


The number and purity of differentiated ECs determine their potential as cell-based therapy in clinical medicine. To produce large numbers of ECs with high purity from ESCs, Sca-1 + cells were treated with 10 ng/mL VEGF in DM for 21 days or more, subcultured with a ratio of 1 to 3 every 2 to 3 days (esECs) (supplemental Figure IA, available online at http://atvb.ahajournals.org). Morphologically, ESCs displayed clusters in an undifferentiated 35 passages in our culture conditions, whereas Sca-1 + progenitors and esECs displayed a monolayer in culture. In addition, Sca-1 + cells grew as round cells, and esECs displayed typical "cobblestone" morphology (supplemental Figure IB). esECs, like ESCs, had a normal content of chromosomes, ie, 39.8±1.5, indicating cell fusion is not involved during cell differentiation (supplemental Figure IC). This technique provides the ability to produce large numbers of cells ( 3 to 6 x 10 8,9 ) because the population doubling time of esECs was 2.5±0.50 days and the number of population doublings of esEC reached 35±5.5 60 days after differentiation.


Previously we have shown that up to 35 passages ES cells are SSEA-1-positive but Sca-1-negative, whereas almost all Sca-1-positive cells are SSEA-1-negative. 17 When isolated Sca-1 + cells were treated with 10 ng/mL of VEGF 165 for 21 days or more, the majority of esECs were positive for EC linage-specific markers as demonstrated by immunofluorescence staining, with Flt-1 ( Figure 1 A; Flt-1), Flk-1 ( Figure 1 A; Flk-1), vWF ( Figure 1 A; vWF), CD31 ( Figure 1 A; CD31), CD106 ( Figure 1 A; CD106), and CD144 ( Figure 1 A; CD144). To validate the specificity of these antibodies, adult mature human umbilical vein endothelial cells (HUVECs) (positive control), adult mature aortic SMCs (negative control), and IgG control for individual primary antibodies were stained. Importantly CD31, vWF, and CD144 were expressed on HUVECs in the typical pattern, but not aortic SMCs (supplemental Figure IIA). FACS analysis demonstrated that 97.5% of ESCs were SSEA-1-positive and 32% CD31-positive (supplemental Figure IIB), yet Sca-1 and VE-cadherin-negative. After 3 or 4 days of culture in collagen-coated flasks, a small fraction of cells (8.6%) were Sca-1 +, whereas the majority of cells (94.7%) isolated with magnetic beads coupled with anti-Sca-1 antibodies were Sca-1 +. 17 Expanded Sca-1 + cells (passage 5 to 10) expressed some progenitor cell markers, eg, Flk-1 (32.6%), but not mature EC markers (CD106 and CD144). When Sca-1 + cells were treated with 10 ng/mL of VEGF 165 for 7 days, some mature EC linage-specific markers were expressed ( Figure 1 B; esEC-d7). As expected, the percentage of cells expressing mature EC markers further increased after 21 days or more of VEGF treatment, including CD31 (from 74.5% to 98.7%), CD106 (from 16.8% to 46.7%), CD144 (from 5.8% to 57.3%), Flt-1 (from 44.5% to 64.5%), and vWF (from 21.5% to 74.8%) ( P <0.05), whereas the percentage of progenitor markers decreased, ie, Flk-1 (from 39.8 to 16.8%) (table in Figure 1 B; supplemental Figure IIC). Lineage markers for nonendothelial cells, such as SMCs (smooth muscle a-actin ), T lymphocytes (CD3), common white blood cell antigen (CD45), and macrophages (Mac-1), did not or were very weakly expressed in esEC-d21 as demonstrated by FACS analysis (supplemental Figure IIC). To further characterize these esECs, they were incubated with DiI-acLDL. esECs rapidly took up DiI-acLDL and expressed high levels of CD31 ( Figure 1 C; supplemental Figure IID), whereas mouse SMCs did not uptake DiI-acLDL and were negative for CD31 (data not shown). To address the difference between esECs and mature adult ECs, which do not significantly contribute to neoangiogenesis and re-endothelialization in adult organisms, clonogenic potentials of Sca-1 + cells, esEC-p8, and HUVEC-p8 were assessed. Cells (2500) were re-plated into 6 well-plates coated with collagen IV and cultured for 6 days, followed by fixation and staining with hematoxylin and eosin. As shown in Figure 1 D, the clonogenic potential of esECs is much higher than that of HUVECs, but lower than Sca-1 + cells ( P <0.05).


Figure 1. Characteristics of esECs. A, Representative images showed that esECs are EC lineage-specific marker positive, including CD31, CD144, CD106, Flk-1, Flt-1, and vWF. Nuclear counterstaining was performed with DAPI (blue). Original magnification, x 400. B, FACS analysis of EC lineage-specific markers CD31, CD144, CD106, Flk-1, Flt-1, and vWF expression in esEC-d7 and esEC-d21. Left, Representative flow cytometeric graphs for vWF expression on esEC-d7 and esEC-d21 are presented. Right, Percentage of other EC linage-specific marker expressed on esEC-d7 and esEC-d21 are shown in the table (n=3). C, A representative image demonstrating CD31-positive staining and DiI-Ac-LDL uptake in esECs. Nuclear counterstaining was performed with DAPI (blue). Original magnification, x 400. D, Representative photographs demonstrating the clonogenic potential of Sca-1 +, esEC-p8, and HUVEC-p8 cells. * P <0.05. Each experiment was performed in triplicate and the mean presented (n=3).


Differentiaton of ECs From VEGF-Induced Sca-1 + Cells Is Dependent on HDAC


VEGF promoted Sca-1 + cells to differentiate into ECs, demonstrated by transfection assays using reporter genes that showed VEGF promoted EC lineage-specific gene expression of CD31, CD144, eNOS, vWF, and Flt-1 ( Figure 2 A) In keeping with these findings, protein levels of EC-linage-specific markers including CD31, eNOS, Flt-1, and Flk-1 were increased ( Figure 2 B), whereas Sca-1 protein levels decreased in a time-dependent manner. In the same experiment HDAC3 protein levels were increased in response to VEGF stimulation.


Figure 2. VEGF-induced expression of EC markers and HDAC3. A, Dual-luciferase activity showed that VEGF significantly increased EC lineage-specific gene expression. All values are mean±SEM, n=3 treatment/time point. B, VEGF increased EC-lineage specific markers and HDAC3 protein expression in Sca-1 + cells. Cultured Sca-1 + cells (d0) and -tubulin were included as controls. Upper panels show representative Western blots, and lower panels show means±SEM of densitometric analysis on day 7 and 9 (n=3), * P <0.05.


Published data showed that prevention of global histone deacetylation by treatment with TSA prevents ES cell differentiation. 12 To explore the role of HDACs in VEGF-induced EC differentiation, we used TSA in further VEGF treatment experiments. 50 nM TSA significantly abolished VEGF-induced HDAC activation (supplemental Figure IIIA). Furthermore, the increased protein levels of the EC-lineage markers CD31, Flk-1, and Flt-1 induced by VEGF were abolished by TSA ( Figure 3 A). To confirm the role of HDAC3 in EC differentiation, we overexpressed HDAC3 in Sca-1 + cells using adenoviral gene transfer, followed by Western blot analysis. Overexpression of HDAC3 increased protein levels of Flk-1, Flt-1, and eNOS protein level in a dose-dependent manner ( Figure 3 B). These results suggest that upregulation of HDAC3 could promote Sca-1 + cells differentiating into ECs.


Figure 3. HDAC3 is crucial in VEGF-induced EC differentiation. A, TSA abolished VEGF-induced EC linage-specific marker expression in Sca-1 + cells. Upper panel shows representative Western blots, and lower panels show means±SEM of densitometric analysis (n=2). B, Overexpression of HDAC3 promotes Sca-1 + cells differentiate into ECs. Adenovirus-TTA was used as control and to compensate the MOI. Upper panel shows representative Western blots and lower panels show means±SEM of densitometric analysis (n=3). C, HDAC3 siRNA abolished VEGF-induced EC-lineage specific marker expression in Sca-1 + cells. Left panel shows representative Western blot, and right panel shows means±SEM of densitometric analysis (n=3). D, HDAC3 siRNA ablated VEGF-induced EC lineage-specific genes expression in Sca-1 + cells. Values are mean±SEM, n=3 treatment/time point.


To determine whether HDAC3 is essential for VEGF-induced EC differentiation, we performed siRNA-mediated knockdown experiments. Knockdown of HDAC3 reduced protein levels of Flt-1, Flk-1, and eNOS ( Figure 3 C). In addition, luciferase reporter assays revealed that HDAC3 siRNA treatment inhibited CD144, eNOS, vWF, Flk-1, and Flt-1 gene expression under control conditions, and also reduced VEGF-induced CD31, CD144, eNOS, vWF, and Flt-1 gene expression ( Figure 3 D; supplemental Figure IIIB). These results indicate that HDAC3 is essential for VEGF-induced EC differentiation from Sca-1 + progenitor cells.


Angiogenesis In Vivo


To investigate whether esECs have angiogenic functions in vivo, esECs were mixed with Matrigel, injected into C57BL/6J mice and harvested at 1, 2, 3, or 4 weeks after the procedure. As shown in Figure 4A to 4 F, esECs formed various highly vascularized structures including capillary-like networks ( Figure 4 A), micro (<20 µm; Figure 4 B), small (25 to 50 µm; Figure 4 F), medium (50 to 100 µm; Figure 4 100 µm; Figure 4 D) vessels. To determine the origin of cells constituting the neovascularization within the implant, we prelabeled esECs with Adenovirus expressing LacZ before implantation. Most neovessels were LacZ-positive, indicating an esEC origin of these neovessels ( Figure 4E and 4 F). Quantitative analysis of X-gal staining for LacZ expression showed that (81.5±12.6%) of neovessels originated from esECs. Double staining for ß-gal and CD144 demonstrated that most cells lining the vessels were double-positive ( Figure 4G to 4 I). The EC-specific markers were expressed largely on the luminal surface of neovessels. Interestingly, double positive staining for ß-gal and SMC-specific markers, such as smooth muscle myosin heavy chain, were not observed in the present study (data not shown). These findings indicate that esECs do not contribute to the formation of perivascular smooth muscle, and that perivascular SMCs that participate in the formation of neovessels may be recruited from the host. Importantly, no tumor-like tissues formed from cell implantation at any time point (1 to 4 weeks). These results strongly suggest that these esECs have high purity and angiogenic function in vivo.


Figure 4. Angiogenesis of esECs in vivo. A to F, Representative images of angiogenesis of esECs in vivo. Matrigel plugs were harvested at 2 weeks after cell implantation. Frozen sections were prepared and stained with H&E (A to D) or X-gal (E and F). Various vascular-like structures including capillary networks (A and E), micro ( 100 µm) (D) were observed. Red arrow (E and F) indicates Lac-Z + cells or vessel (blue). G to I, Double immunofluorescence staining of Matrigel plugs. esECs (10 6 ) infected with adenovirus expressing LacZ gene were mixed with Matrigel at 4°C, and subcutaneously injected into C57BL/6J mouse. After 2 weeks, the implants were harvested and frozen sections were prepared and labeled with antibodies against ß-gal and EC specific marker CD144. Nuclear counterstaining was performed with DAPI (blue). Double-positive cells in merged images appear yellow.


Local Transfer of esECs Inhibits Neointima Formation


To evaluate the in vivo effect of esECs on neointima formation after arterial injury, mice were given sham-treatment (medium alone) or local esEC transfer after femoral artery wire induced injury. Figure 5 shows representative photographs of injured vessels, and quantitative morphometric measurements. Wire-induced arterial injury resulted in prominent neointima formation ( Figure 5 B), which was markedly inhibited by local injection of esECs ( Figure 5C and 5 D). Morphometric analysis revealed that local transfer of esECs significantly reduced neointima formation (8036 µm 2 ±866 µm 2 versus 30 026 µm 2 ±2500 µm 2, P <0.001) as well as the ratio of neointima/total vessel wall (0.198±0.06 versus 0.9±0.08, P <0.01) ( Figure 5 E).


Figure 5. esEC transfer inhibited neointimal formation in injured artery. A to D, Representative photographs of normal artery (A), wire-injured (B), and esEC-treated (C and D) femoral arteries. Two weeks after cell transfer, animals were euthanized, and injured arteries harvested, fixed, sectioned, and stained with H&E. Original magnification, x 100 (A to C); x 200 (D). E, Quantitative data of neointimal lesional area (left) and ratio of neointima/total vessel wall (right). Data are means±SEM, (n=15/group). Black arrow indicates neointima. *Significant difference from untreated control, P <0.01, ** P <0.001.


When adenovirus-LacZ-infected esECs were applied to injured arteries, ß-gal-positive cells were detected on the majority of the luminal surface ( Figure 6 B), and in some areas showed confluence ( Figure 6 C). No ß-gal-positive cells were observed in mice given medium alone ( Figure 6 A), indicating that transferred esECs were incorporated into the injured arterial site. Immunofluorescence staining confirmed double-positive cells for ß-gal and EC-specific markers, such as CD144 ( Figure 6D to 6 F) and vWF ( Figure 6G to 6 I). Luminal immunofluorescence intensity showed that local esEC transfer significantly increased re-endothelialization of injured arteries up to 86%±13.6%. These results further validate the finding that locally applied esECs contribute to re-endothelialization of injured arteries.


Figure 6. Transplanted esECs contribute to reendothelialization. esECs (5 x 10 5 ) infected with adenovirus encoding LacZ were injected into wire-injured femoral arteries. After 3 or 7 days, arterial samples were harvested, cryosectioned, and stained with X-gal to detect LacZ (A to C). Representative photographs of negative (A), partial (B), and confluent X-gal staining. Double-immunofluorescent staining (D through I) for ß-gal and EC specific markers CD144 and vWF were performed. Nuclear counterstaining was performed with DAPI (blue). Note that double-positive cells in merged images appear yellow. Original magnification, x 100 (A, D to I); x 200 (B and C).


Discussion


In the present study, we report a technique to produce large numbers of endothelial cells from embryonic stem cells ( 3 to 6 x 10 8,9 ECs from 10 7 ES cells), which may be used as therapy after vascular injury. We observed, first, that Sca-1 + progenitor cells derived from ES cells can differentiate into ECs with high purity, and the mechanism for this involves induction of HDAC3. Second, after incorporation into Matrigel esECs form vascular-like structures in vivo. Finally, local application of esECs to areas of vascular injury promotes re-endothelialization and retards neointima formation. These findings strongly suggest that Sca-1 + progenitor cells derived from embryonic stem cells may be used to form endothelial cells. These esECs have therapeutic implications for vascular diseases, and may be a promising cell source for cell-based vascular engineering and repair of injured vessels.


Several studies have reported that ES cells can spontaneously undergo vascular differentiation through first forming embryoid bodies in vitro. 14 These ES cells were differentiated into ECs by growth factor cocktails, including VEGF, basic fibroblast growth factor, insulin-like growth factor-1, and epidermal growth factor. 7 Another group reported that Flk-1 + cells derived from ES cells can differentiate into smooth muscle cells and ECs in vitro. 4-6 In most studies, ES cell-derived ECs are cell mixtures limiting their use for studying the functional role of stem cell-derived ECs. In the present study, we sorted Sca-1 progenitor cells from ES cell cultures, and demonstrate that Sca-1 may be used as a sorting marker for the isolation of vascular progenitor cells. These findings suggest that this novel technique is useful for obtaining large numbers of ECs with high purity.


A panel of genetic, antigenic and functional assays is required to provide optimal characterization of EC populations differentiated from primary ES cells. We demonstrated that esECs have EC functions based on the following observations. First, esECs display typical "cobblestone" endothelial cell morphology in culture. Second, esECs express high levels of mRNA of almost all the EC markers, including CD31, CD62E, CD105, CD106, CD109, CD141, CD144, CD146, Flk-1, Flt-1, Tie-1, Tie-2, and vWF (data not shown). Furthermore, immunofluorescence and FACS analysis show that esECs are positive for EC lineage-specific markers such as CD31, CD106, CD144, Flk-1, Flt-1, and vWF. We found that Flk-1 expression decreased with EC differentiation ( Figure 1 B) that may be attributed to higher expression of Flk-1 in progenitors. 4-6 Interestingly, high levels of CD31 mRNA or protein were unexpectedly detected on undifferentiated ES cells by reverse-transcription polymerase chain reaction, immunofluorescence staining, flow cytometry analysis, and Western blot analysis. Although CD31 is accepted as one of the main markers of ECs and platelets, this observation may reflect a possible expression of CD31 in the inner cell mass of blastocytes. Importantly, our results are consistent with some previous findings. Vittet et al found mouse ES cells are Tie-2 and CD31-positive regardless of culture conditions. 14 Third, almost all esECs are positive for CD31 and rapidly uptake Dil-acLDL, indicating CD31-positive esECs in our experiment show some EC characteristics. Fourth, in comparison to mature adult ECs, esECs have much higher clonogenic potential ( Figure 1 D), possibly providing improved ability to influence neoangiogenesis and re-endothelialization in the adult organism. Finally, vascular-like structures including capillary networks and vessels of various sizes were observed in the in vivo angiogenesis experiments. It is thus likely that ECs derived from ESCs share a similar phenotype and ability to function as ECs generated from adult progenitors and the vascular wall.


Previous studies have specified a number of molecules involved in vascular development. Among these molecules, VEGF, 18 fetal liver kinase-1 (also known VEGF-R2), 19 fms-like tyrosine kinase (Flt-1, also known VEGF-R1), 20 and VE-cadherin (also known CD144) 21 have been reported as essential for vasculogenesis. In particular, VEGF plays a critical role in endothelial proliferation, differentiation, and vascular development. 22,23 However, little is known about the mechanism by which VEGF promotes Sca-1 + cells to differentiate into ECs. Recently, Ajamian et al reported that mRNA of class II histone deacetylases increased during neural differentiation. 24 Inhibition of HDAC activity by Trichostatin A blocked tumor angiogenesis 9-11 and prevented ES cell differentiation. 12 In another study by Rossig et al, shear stress activated class I HDACs, which was essential in shear-induced EC differentiation in human EPCs. 25 In our study, we showed that VEGF increased HDAC3 protein levels and that inhibition using TSA and siRNA knockdown or upregulation with adenovirus modulated EC differentiation. These findings demonstrate for the first time to our knowledge that HDAC3 is critical for VEGF-induced EC differentiation from Sca-1 + cells.


The ultimate goal of understanding stem cell biology is the potential for using these cells therapeutically in clinical medicine. Accumulating evidence indicates that adult progenitor cells, such as bone marrow cells 26-28 and endothelial progenitor cells, 29-31 have beneficial effects on neovascularization following ischemia and re-endothelialization after vascular injury thereby providing a potentially novel therapeutic option for vascular disease states. In the present study, we found lesions in ApoE -/- mice were more severe and appeared earlier than in wild-type mice. This finding may explain why almost all injured arteries without cell treatment were occluded by neointimal lesions 2 weeks after injury. esEC transfer significantly enhanced re-endothelialization and reduced neointima formation in denuded vessels. Therefore, esEC-based therapeutic strategies may be a treatment option for prevention of vascular diseases such as atherosclerosis, restenosis, and graft failure.


Previous studies have shown that systemic transfusion of adult progenitor cells have beneficial effects on neovascularization after ischemia and re-endothelialization after endothelial injury. 26-31 We first used this strategy in our study but found esECs provided no significant benefit on re-endothelialization compared with sham treatment. Compared with systemic application, we found a striking beneficial effect of esECs on re-endothelialization after local application (supplemental Figure IV). These findings strongly suggest the use of different approaches in experimental and clinical cell therapy trials.


In conclusion, we have established a novel method for producing large numbers of ECs from ES cells, which regenerated the endothelium of denuded vessels and reduced neointimal lesion formation after arterial injury. HDAC3 was critical in VEGF-induced EC differentiation from Sca-1 + cells. These findings provide important details on targeting proteins that promote endothelial differentiation and activation, and provide direct evidence to support stem cell-based therapeutic approaches.


Acknowledgments


We thank Dr Neil Roberts and Evelyn Torsney for critical reading of the manuscript.


Sources of Funding


This work was supported by grants from the Oak Foundation and British Heart Foundation.


Disclosures


None

【参考文献】
  Nowak G, Karrar A, Holmen C, Nava S, Uzunel M, Hultenby K, Sumitran-Holgersson S. Expression of vascular endothelial growth factor receptor-2 or tie-2 on peripheral blood cells defines functionally competent cell populations capable of reendothelialization. Circulation. 2004; 110: 3699-3707.

Urbich C, Dimmeler S. Endothelial progenitor cells functional characterization. Trends Cardiovasc Med. 2004; 14: 318-322.

Xu Q. The impact of progenitor cells in atherosclerosis. Nat Clin Pract Cardiovasc Med. 2006; 3: 94-101.

Hirashima M, Ogawa M, Nishikawa S, Matsumura K, Kawasaki K, Shibuya M. A chemically defined culture of VEGFR2+ cells derived from embryonic stem cells reveals the role of VEGFR1 in tuning the threshold for VEGF in developing endothelial cells. Blood. 2003; 101: 2261-2267.

Sone M, Itoh H, Yamashita J, Yurugi-Kobayashi T, Suzuki Y, Kondo Y, Nonoguchi A, Sawada N, Yamahara K, Miyashita K, Park K, Shibuya M, Nito S, Nishikawa S, Nakao K. Different differentiation kinetics of vascular progenitor cells in primate and mouse embryonic stem cells. Circulation. 2003; 107: 2085-2088.

Yamashita J, Itoh H, Hirashima M, Ogawa M, Nishikawa S, Yurugi T, Naito M, Nakao K. Flk1-positive cells derived from embryonic stem cells serve as vascular progenitors. Nature. 2000; 408: 92-96.

Kaufman DS, Lewis RL, Hanson ET, Auerbach R, Plendl J, Thomson JA. Functional endothelial cells derived from rhesus monkey embryonic stem cells. Blood. 2004; 103: 1325-1332.

Levenberg S, Golub JS, Amit M, Itskovitz-Eldor J, Langer R. Endothelial cells derived from human embryonic stem cells. Proc Natl Acad Sci U S A. 2002; 99: 4391-4396.

Williams RJ. Trichostatin A, an inhibitor of histone deacetylase, inhibits hypoxia-induced angiogenesis. Expert Opin Investig Drugs. 2001; 10: 1571-1573.

Qian DZ, Wang X, Kachhap SK, Kato Y, Wei Y, Zhang L, Atadja P, Pili R. The histone deacetylase inhibitor NVP-LAQ824 inhibits angiogenesis and has a greater antitumor effect in combination with the vascular endothelial growth factor receptor tyrosine kinase inhibitor PTK787/ZK222584. Cancer Res. 2004; 64: 6626-6634.

Takimoto R, Kato J, Terui T, Takada K, Kuroiwa G, Wu J, Ohnuma H, Takahari D, Kobune M, Sato Y, Takayama T, Matsunaga T, Niitsu Y. Augmentation of antitumor effects of p53 gene therapy by combination with HDAC inhibitor. Cancer Biol Ther. 2005; 4: 421-428.

Lee JH, Hart SR, Skalnik DG. Histone deacetylase activity is required for embryonic stem cell differentiation. Genesis. 2004; 38: 323-328.

Hu Y, Zhang Z, Torsney E, Afzal AR, Davison F, Metzler B, Xu Q. Abundant progenitor cells in the adventitia contribute to atherosclerosis of vein grafts in ApoE-deficient mice. J Clin Invest. 2004; 113: 1258-1265.

Vittet D, Prandini MH, Berthier R, Schweitzer A, Martin-Sisteron H, Uzan G, Dejana E. Embryonic stem cells differentiate in vitro to endothelial cells through successive maturation steps. Blood. 1996; 88: 3424-3431.

Roque M, Fallon JT, Badimon JJ, Zhang WX, Taubman MB, Reis ED. Mouse model of femoral artery denudation injury associated with the rapid accumulation of adhesion molecules on the luminal surface and recruitment of neutrophils. Arterioscler Thromb Vasc Biol. 2000; 20: 335-342.

Lindner V, Fingerle J, Reidy MA. Mouse model of arterial injury. Circ Res. 1993; 73: 792-796.

Yin X, Mayr M, Xiao Q, Mayr U, Tarelli E, Wait R, Wang W, Xu Q. Proteomic dataset of Sca-1+ progenitor cells. Proteomics. 2005; 5: 4533-4545.

Leung DW, Cachianes G, Kuang WJ, Goeddel DV, Ferrara N. Vascular endothelial growth factor is a secreted angiogenic mitogen. Science. 1989; 246: 1306-1309.

Matthews W, Jordan CT, Gavin M, Jenkins NA, Copeland NG, Lemischka IR. A receptor tyrosine kinase cDNA isolated from a population of enriched primitive hematopoietic cells and exhibiting close genetic linkage to c-kit. Proc Natl Acad Sci U S A. 1991; 88: 9026-9030.

Shibuya M, Yamaguchi S, Yamane A, Ikeda T, Tojo A, Matsushime H, Sato M. Nucleotide sequence and expression of a novel human receptor-type tyrosine kinase gene (flt) closely related to the fms family. Oncogene. 1990; 5: 519-524.

Breier G, Breviario F, Caveda L, Berthier R, Schnurch H, Gotsch U, Vestweber D, Risau W, Dejana E. Molecular cloning and expression of murine vascular endothelial-cadherin in early stage development of cardiovascular system. Blood. 1996; 87: 630-641.

Coultas L, Chawengsaksophak K, Rossant J. Endothelial cells and VEGF in vascular development. Nature. 2005; 438: 937-945.

Ferrara N, Gerber HP, LeCouter J. The biology of VEGF and its receptors. Nat Med. 2003; 9: 669-676.

Ajamian F, Suuronen T, Salminen A, Reeben M. Upregulation of class II histone deacetylases mRNA during neural differentiation of cultured rat hippocampal progenitor cells. Neurosci Lett. 2003; 346: 57-60.

Rossig L, Urbich C, Bruhl T, Dernbach E, Heeschen C, Chavakis E, Sasaki K, Aicher D, Diehl F, Seeger F, Potente M, Aicher A, Zanetta L, Dejana E, Zeiher AM, Dimmeler S. Histone deacetylase activity is essential for the expression of HoxA9 and for endothelial commitment of progenitor cells. J Exp Med. 2005; 201: 1825-1835.

Tanaka K, Sata M, Hirata Y, Nagai R. Diverse contribution of bone marrow cells to neointimal hyperplasia after mechanical vascular injuries. Circ Res. 2003; 93: 783-790.

Xu Y, Arai H, Zhuge X, Sano H, Murayama T, Yoshimoto M, Heike T, Nakahata T, Nishikawa S, Kita T, Yokode M. Role of bone marrow-derived progenitor cells in cuff-induced vascular injury in mice. Arterioscler Thromb Vasc Biol. 2004; 24: 477-482.

Fujiyama S, Amano K, Uehira K, Yoshida M, Nishiwaki Y, Nozawa Y, Jin D, Takai S, Miyazaki M, Egashira K, Imada T, Iwasaka T, Matsubara H. Bone marrow monocyte lineage cells adhere on injured endothelium in a monocyte chemoattractant protein-1-dependent manner and accelerate reendothelialization as endothelial progenitor cells. Circ Res. 2003; 93: 980-989.

Werner N, Junk S, Laufs U, Link A, Walenta K, Bohm M, Nickenig G. Intravenous transfusion of endothelial progenitor cells reduces neointima formation after vascular injury. Circ Res. 2003; 93: e17-e24.

Kong D, Melo LG, Mangi AA, Zhang L, Lopez-Ilasaca M, Perrella MA, Liew CC, Pratt RE, Dzau VJ. Enhanced inhibition of neointimal hyperplasia by genetically engineered endothelial progenitor cells. Circulation. 2004; 109: 1769-1775.

Strehlow K, Werner N, Berweiler J, Link A, Dirnagl U, Priller J, Laufs K, Ghaeni L, Milosevic M, Bohm M, Nickenig G. Estrogen increases bone marrow-derived endothelial progenitor cell production and diminishes neointima formation. Circulation. 2003; 107: 3059-3065.


作者单位:Department of Cardiac and Vascular Sciences (Q.X., L.Z., Z.Z., A.M., Y.H., Q.X.), St George?s, University of London, London, United Kingdom; Department of Cardiovascular Medicine (Z.A., K.C), University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.

作者: Qingzhong Xiao; Lingfang Zeng; Zhongyi Zhang; Andr
医学百科App—中西医基础知识学习工具
  • 相关内容
  • 近期更新
  • 热文榜
  • 医学百科App—健康测试工具