CA2465173C - Endothelial cells derived from primate embryonic stem cells - Google Patents

Endothelial cells derived from primate embryonic stem cells Download PDF

Info

Publication number
CA2465173C
CA2465173C CA2465173A CA2465173A CA2465173C CA 2465173 C CA2465173 C CA 2465173C CA 2465173 A CA2465173 A CA 2465173A CA 2465173 A CA2465173 A CA 2465173A CA 2465173 C CA2465173 C CA 2465173C
Authority
CA
Canada
Prior art keywords
cells
endothelial
endothelial cells
cell
embryonic stem
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Lifetime
Application number
CA2465173A
Other languages
French (fr)
Other versions
CA2465173A1 (en
Inventor
Dan S. Kaufman
Rachel Lewis
Robert Auerbach
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Wisconsin Alumni Research Foundation
Original Assignee
Wisconsin Alumni Research Foundation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Wisconsin Alumni Research Foundation filed Critical Wisconsin Alumni Research Foundation
Publication of CA2465173A1 publication Critical patent/CA2465173A1/en
Application granted granted Critical
Publication of CA2465173C publication Critical patent/CA2465173C/en
Anticipated expiration legal-status Critical
Expired - Lifetime legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0603Embryonic cells ; Embryoid bodies
    • C12N5/0606Pluripotent embryonic cells, e.g. embryonic stem cells [ES]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/069Vascular Endothelial cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2500/00Specific components of cell culture medium
    • C12N2500/30Organic components
    • C12N2500/38Vitamins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/105Insulin-like growth factors [IGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/11Epidermal growth factor [EGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/165Vascular endothelial growth factor [VEGF]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2506/00Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells
    • C12N2506/02Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from embryonic cells

Abstract

A method is described to induce primate embryonic stem cells to differentiate into a relatively homogenous population of endothelial cells. The ES derived endothelial cells have the general morphological and cell surface marker characteristics of endothelial cells. The ES derived endothelial cells also are capable of inducing and participating in blood vessel formation (or vascularization) when transplanted into tissue in vivo.

Description

ENDOTHELIAL CELLS DERIVED FROM
PRIMATE EMBRYONIC STEM CELLS
BACKGROUND OF THE INVENTION
[0003] Stem cells are defined to be cells which are capable both of self-renewal and differentiation into one or more differentiated cell types. Human embryonic stem cells are a category of stem cells created from human pre-implantation blastocysts. Human embryonic stem cells are pluripotent and may be totipotent, meaning that they can certainly differentiate into many cell types evidenced in an adult human body and may be capable of differentiating into all cell types present in the human body.
[0004] Embryonic stem cells (ES cells) have also been derived in a number of animals other than humans. For example, much scientific work has been conducted with murine ES cells.
Once a method for the initiation of ES cell cultures for a particular species is worked out, it becomes possible to manipulate the ES cells, and animals which result therefrom, in a variety of ways to learn useful information about the genetics of the animal under study.
For example, it has become possible over the past decade to create cultures of murine ES cells in which one or another specific gene is knocked out in each murine stem cell culture. While some techniques that could be worked out in murine ES cell systems were transferable to other species, many were not. For example, the basic techniques which could be used to create murine ES cell cultures did not transfer well to many other animal species. For the development of techniques for the culture and manipulation of human ES cells, the murine cell may therefore not be the best model due to the phylogenic distance between humans and mice. However, in the course of the development of the science of human ES cell cultures and techniques, much of the preliminary work was conducted in non-human primates, such as the rhesus monkey. Other primate ES cell cultures have proven to be a relatively reliable model for systems that could be easily transferred to human cell culture. For an example, murine ES cell cultures require application of leukemia inhibitory factor (LIF) or another agonist of the gp130/STAT3 signaling pathway for maintenance of undifferentiated cell growth, whereas human and rhesus monkey ES cell cultures do not require LIF for undifferentiated cell growth. Prior work on hematopoiesis using rhesus monkey ES cells validates the utility of this system for doing pre-clinical investigations for tecluiiques that can be transferred to human ES cell cultures.
[0005] One of the exciting potential uses of stem cells is for human tissue transplantation.
It is hoped and expected that techniques can be developed to direct the differentiation of stem cells into specific lineages which can then be transferred into the human body to replace or enhance tissues of the body. In order to do that, first techniques must be developed to direct the differentiation of stem cells into the specific cell lineages desired.
Teclmiques have already been proposed to direct stem cell cultures into lineages of hematopoeic, neural, cardiomyocyte, pancreatic and other lineages. These techniques have proven to be quite different from each other and independent in the sense that a new and different technique is required for each new desired lineage.
[0006] Endothelial cells make up a network of interconnected cells in the human body that line blood vessels, lymphatic vessels, and form capillaries. Endothelial cells regulate the flow of nutrient substances and create and respond to diverse biologically active molecules.
While it has been demonstrate that human ES cells will differentiate into many progeny cells types, including endothelial cells, it has not been previously possible to create distinct cultures of derivatives of human ES cells directed into an endothelial lineage.

BRIEF SUMMARY OF THE INVENTION
[0007] The present invention is summarized in that a method has been developed which permits the direct differentiation of a culture of embryonic stem cells into a culture of endothelial cells. The method includes culturing the embryonic stem cells in a culture medium previously known to maintain endothelial cells and which, it now turns out, has the capability to support embryonic stem cells in the process of differentiation into endothelial cells.
[0008] The present invention is also summarized in that cultures of endothelial cells derived from embryonic stem cells which have morphology and cell surface marlcers characteristic of endothelial cells and which are capable of inducing vascularization of tissue in vivo=
[0009] It is a feature of the present invention in that it is relatively efficient to perfomi since its steps are simple, and the result is a culture that appears to be a relatively homogenous population of endothelial cells.
[00010] Other objects, advantages and features of the present invention will become apparent from the following specification when taken in conjunction with the accompanying drawings.

BRIEF DESCRIPTION OF THE SEVERAL VIEWS OF THE DRAWINGS
Not applicable.

DETAILED DESCRIPTION OF THE INVENTION
[00011] The present invention is directed both to a method to direct the differentiation of primate embryonic stem cells into endothelial cells and to the relatively pure population of endothelial cells so produced. The method is based on the cultivation of primate embryonic stem cells with a defined protein growth factor or factors which cause the cells so treated to change their morphology to become endothelial cells. In contrast to other techniques for the directed differentiation of cells of other lineages from embryonic stem cells, the culture of endothelial cells, derived from embryonic stem cells by the method described here, appears relatively uniform and is made up of primarily of endothelial cells having apparent angiogenic capability.
[00012] The culture method is based on the culture of undifferentiated primate embryonic stem (ES) cells in a medium containing vascular endothelial cell growth factors (VEGF), basic fibroblast growth factor (bFGF), insulin-like growth factor (IGF-1), and epidermal growth factor (EGF). These factors are all found in a commercially available medium known as endothelial cell basal medium (EBM-2, Clonetics/BioWhittaker). This medium was previously known and is used to sustain endothelial cells in culture. It was not previously known that this medium could be used to support the differentiation of ES cells into endothelial cells.
While this combination of growth factors has been found to be sufficient to support the differentiation of ES cells into endothelial cells, it may not be necessary to use all four factors in the culture medium, and whetller or not one of the factors can be omitted can readily be ascertained by empirical experimentation without departing from the concept of the present invention.
[00013] What separates this method from prior art derivation of heterogeneous mixtures including endothelial cells is the relative uniformity of the transition of the cell culture from ES
cells to endothelial cells. Other methods were tried, without success, to achieve this transition, such as application of phorbol esters, co-cultivation with stromal cells plus serum and isolation of endothelial cells from embryoid bodies. None of these efforts reproducibly yielded cultures of predominantly endothelial cells. In contrast, the method described here is simple and efficient and results in a cell culture of morphologically similar cells having the characteristics of endothelial cells.
[00014] The culture of endothelial cells made by the present invention will have certain characteristics. The cells have a characteristic morphology, similar to elongated or stellate shaped endothelial cells. In contrast, ES cells grown in other media differentiate into a heterogeneous population of cell types with no distinct endothelial-appearing cells. The endothelial cells rapidly form tubular structures when placed in Matrigel (TM) medium. The endothelial cells are positive for presence of the von Willebrand factor (vWF) and have liigh levels of tflex europaeus agglutinin 1(UEA-1) binding, as well as expression of the intergrin av(33 and the surface antigen CD 146. These cells also will take up acetylated LDL, another trait characteristic of endothelial cells. These cells do lack expression of CD31 and VE-cadherin, two antigens commonly, but not always, present on the surface of endothelial cells. These endothelial cells have the ability, when transferred into a SCID (severe combined immunodeficient) mouse together with tumor cells, to effect the vascularization of the resulting tumor, thus demonstrating the ability of the cells both to recruit and to participate in vascularization in vivo. The ability of these cell to participate in vascularization is particularly noteworthy, since that attribute makes in possible to transplant genetically altered endothelial cells into a tissue requiring vascularization with the altered cells surviving in vivo in the vascular matrix created to therefore express whatever gene was inserted into the cells.
[00015] In contrast to other cell types which can be induced to form from embryonic stem cells, the endothelial cell culture described and characterized here is relatively homogenous in cells coinmitted to the derivative lineage, i.e. to be endothelial cells. The ES derived endothelial cell culture is formed of cells having a uniform morphology and exhibiting the characteristics of endothelial cells. Given the limits of present cell culture technology, however, it cannot be said with certainty that the ES derived endothelial cell culture is entirely free of other cell types.
What can be said is that the ES derived endothelial cell culture is predominantly composed of endothelial cells and is a practical source of cells which will act as endothelial cells to promote and participate in vascularization of tissues when transplanted into a host in vivo. Using a common test for endothelial character, the ability to bind the Nex europaeus agglutinin 1(UEA-1) lectin, it has been found that reproducible over 90% of the cells in the derivative culture do bind the UEA-1 lectin. While in some variations of the method the percentage of cells which bind to UEA-1 might vary, in cultures of endothelial cells made by the method described here, at least 75%, and more preferably, over 90%, of the cells in the culture will test positive for the ability to bind the UEA-1 lectin.
t04016} While the examples below were conducted with rhesus monkey ES cells, the same processes and result can be obtained with human ES cells. Human endothelial cells derived from ES cells offer the possibility to develop tissues transplantable into human patients.
Transplantation of endothelial cells would be desirable for those applications in which vascularization of ischemic tissue is needed. In addition, the introduction of endothelial cells may be useful in any location on the body where improved vascularization is needed. Since the precursor ES cells can be grown in any number, this makes possible the generation of large numbers of endothelial cells for clinical experimentation or treatment.

EXAMPLES
[000171 METHODS
[00018] Cell Culture [00019] Undifferentiated rhesus monkey ES cells (R366.4 cell line) were cultured as previously described (Thomson et al. Proc. Natl. Acad. Sci. USA 92:7844-7848 (1996)). Briefly R366.4 cells were co-cultured with irradiated mouse embryonic fibroblast (MEF) cells in medium containing DMEM, 20% FBS (Hyclone, Ogden UT), 2mM L-glutamine (Sigma, St.
Louis, MO), 0.1 mM 2-mercaptoethanol (Sigma), and 1% Iv1EM non-essential amino acids (Invitrogen). Undifferentiated cells were fed daily with fresh medium and passaged onto new MEFs approximately every 5-7 days. To promote endothelial cell differentiation, the medium was removed from the ES cells 24 hours after plating and replaced with medium consisting of EGM2, 5% FBS, VEGF, bFGF, IGF-1, EGF, and ascorbic acid (EGM2) (EGM2-MV Bullet Kit, CloneticsBioWhittaker, Walkersville, MD). The ES cells were differentiated for 29 days in the EGM2 medium, which was changed every 3-5 days. Differentiated rhesus ES cells, were dissociated with 0.05% trypsin/0.53mM EDTA (GIBCO/BRL) for 5 minutes, centrifuged, and re-plated in EGM2 in 10 cm tissue culture dishes without irradiated MEF cells.
After 24 hours non-adherent cells were removed and adherent cells were fed fresh medium. The rhesus ES cell-derived endothelial cells (RESDECs) could be grown to confluence and serially passaged and expanded in the EBM2 medium.
[00020] Human umbilical vein cells (HUVECs) (Clonetics/Biowhittaker) were also grown and passaged in EGM2 by known methods.

[00021] Tube Formation on MatrigelTM
[000221 0.2m1 of MatrigelTM (Becton Dickinson) was added to each well of a 24 well tissue culture plate and allowed to solidify at 37 C for at least 30 minutes.
Following gelation, 0.2m1 of a cell suspension containing 5 x 104 - 1 x 105 RESDECs was placed on top of the MatrigelTM. The cultures were incubated at 37 C/5% CO2 and observed at 24, 48, and 72 hours for rearrangement of cells into tube-like capillary structures. Individual experiments were performed in triplicate and representative wells recorded by photomicrography.

[00023] VEGF and bFGF ELISA
[000241 RESDECs were cultured for 3 days in the absence of VEGF or bFGF in EGM2, EGM2 supplemented with 10% Knockout serum replacerTM (GIBCO) instead of FBS, or DMEM
supplemented with 10% FBS. After 72 hours the conditioned media (CM) was collected and centrifuged to remove dead cells. EGM2 medium alone served as a negative control. The amount of VEGF or bFGF in the CM was analyzed by colorimetric ELISA assay (R &
D
systems, Minneapolis, W.

[00025] Flow Cytometry [00026] RESDECs were washed with Ca2+ and Mg2+ free PBS and detached from the monolayer with 0.05% trypsin/0.53mM EDTA for 5 minutes. The dissociated cells were centrifuged and washed with FACS medium consisting of PBS supplemented with 2%
FBS and 0.1% sodium azide. After filtration through 80-micron nitex, the single-cell suspension was measured in aliquots and stained with either isotype control or antigen-specific antibodies diluted to appropriate concentrations in FACs media. Cell surface antigen expression was analyzed using antigen-specific primary antibody followed by fluorescent-tagged secondary antibodies (indirect staining), or fluorescently-conjugated antigen-specific antibodies (Direct staining).
Appropriate unconjugated mouse and goat IgGs (both Sigma) as well as FITC-conjugated mouse IgG (Pharrningen San Diego, CA)) were used as isotype controls. Unconjugated antigen-specific antibody against flk-I (Research Diagnostics) was detected with a FITC labeled anti-goat IgG
antibody (Sigma). Unconjugated antibodies against VEGF receptor 1(FIt-1) and VEGF receptor 2(flk-1) (both Sigma) were detected with a FITC labeled goat anti-mouse IgG
(Caltag).
Unconjugated P1H12 antibody (mouse IgGl, provided by Dr. Robert Hebbel, University of Minnesota) was detected with rat anti-mouse IgG-FITC conjugated secondary antibody (Caltag).
The cells were also tested for expression of the VEGF receptor using a biotinylated VEGF Kit (R&D Systems) and for their ability to bind the tqex europaeus agglutinin 1(UEA-1) (Vector labs). Direct conjugated antibodies used were I-iLA-A, B, C-FITC (Pharmingen) and aV(33/cl LM609-FITC (Chemicon). Human umbilical vein cells (HUVEC) (Clonetics) served as a positive control. Cells were analyzed without fixation on a FACScan or FACs Calibur (Becton Dickinson) using propidium iodide to exclude dead cells. Data analysis was carried out using CellQuestTM software (Becton Dickinson).

[00027] Immunostaining [00028] Analysis for the acetylated LDL receptor was performed by diluting diIAcLDL
(Molecular Probes) in serum-free EGM2. Cells were washed twice and incubated overnight in EGM2 medium containing difAcLDL. After washing, the cells were observed by fluorescence microscopy (UV, rhodamine filter). HiJVECs were used as a positive control.
[00029] Expression of vonWillebrand factor protein (vWF) (DAKO) was detected with a goat anti-rabbit IgG-FITC secondary antibody (Sigma). Cells were fixed and incubated at room temperature for one hour with vWF, washed, and incubated for 30 minutes in the secondary antibody. After a final wash, cells were observed by fluorescence microscopy (UV, FITC filter).
HUVECS again served as a positive control.

[000301 Matrigel Plugs [00031] In one experiment SCID niice (Baib/Scid, Harlan Sprague Dawley) were injected subcutaneously with 0.5m1 MatrigelTM (Becton Dickinson) containing 5 x 105 -1 x 106 RESDECs.
A second experiment was performed implanting a sponge containing the RESDECs into the solidified MatrigelTM. The MatrigelTM plugs were removed after 14, 21, 35, and 42 days. Vessels were observed by injecting high molecular weight FITC-dextran (Sigma) intravenously a few minutes before removing and fixing the plugs. Standard H/E slides were also prepared.

[00032] In Vivo Studies [00033] Adherent RESDECs were harvested by trypsinization and mixed with the mouse mammary carcinoma, C755 cell line. In two separate experiments Balb/c-SCID
mice were injected subcutaneously with either 1 x 106 C755 tumor cells, or 1 x 106 RESDECs, or with a mixture of 1 x 106 tumor cells and 1 x 106 RESDECs. After initial growth, tumors were measured by caliper every 3-5 days. Approximately tbree weeks after transplantation all mice were sacrificed for histochemical analysis of the grown tumors. Mice injected with only RESDECs failed to grow tumors.

1000341 Immunohistochemical Staining of Tumors [00035] In the first experiment tumors were isolated and fixed in 10% formalin for the preparation of paraffin sections. To prepare frozen sections tumors were fixed in 2%
paraformaldehyde. All sections were mounted onto Fisher Superfrost slides.
Using the standard ABC technique (VectaStain Elite ABC kit, Vector labs) sections were processed for expression of HLA -class I A,B, C (W6/32 antibody, IgG2a) and CD31 (IgGI) (Novacastra, Vector). Mouse IgG1 (Sigma) and IgG2a (Southern Bioteclmology) were used as isotype controls.
The peroxidase activity was visualized with a DAB substrate (Vector) and sections were counterstained with hematoxylin.

[00036] RESULTS
[00037] Derivation of endothelial cells from rhesus monkey ES cells:
[00038] Rhesus monkey ES cells were grown in EGM2 medium containing VEGF, bFGF, EGF, and IGF as described in materials and methods. After approximately 5-10 days, these ES
cells assumed a uniform morphology similar to elongated or stellate -shaped endothelial cells.
In contrast, ES cells grown in medium supplemented with FBS alone differentiated into a heterogeneous cell population with no distinct endothelial-appearing cells.
The potential endothelial cells were serially passaged and expanded for approximately 20 population doublings while grown in EGM2 with maintenance of a homogeneous appearance. As an initial test of endothelial cell characteristics, these cells were placed in Matrigel-based medium where they rapidly formed tube-like capillary structures similar those formed by HUVEC or other endothelial cell populations when placed in Matrigel-based medium. Cytogenetic studies showed all cells have a normal rhesus monkey 40 XY karyotype. These cytogenetic results were important to demonstrate that these cells were not transformed after prolonged culture, nor were they derived from potentially contaminating mouse embryonic fibroblast cells that are used for the growth of undifferentiated ES cells.
[000391 Electron micrographs of the rhesus ES cell-derived cells demonstrate typical endothelial cell features. These include multiple dense round or rod-shaped Weibel-Palade bodies, tight junctions between cells, and endocytic/exocytic vesicles.

[00040] Imnaunophenotyping [00041] Next, immunohistochemical staining of these rhesus-derived endothelial-like cells demonstrated the presence von Willebrand factor (vWF), the ability for these cells to rapidly take-up acetylated LDL and the ability to bind the UEA-1 lectin. Flow cytometric studies confirm.ed high levels of UEA-1 binding, as well as expression of the integrin av(33 and the surface antigen CD146 recognized by the P1H12 antibody. These proteins have been shown to be important in endothelial cell-cell interactions. These results led us to call these rhesus embryonic stem cell-derived endothelial cells (RESDECs). Surprisingly, antibodies against the VEGF receptors flk-1 and flt-1 did not bind these RESDECs, though binding to HUVEC cells was also weak. However, an assay using biotinylated VEGF and secondary streptavidin-FITC
showed binding to the RESDEC (and HUVEC) cells. Specificity of this, binding was demonstrated by blocking with an anti-VEGF antibody. This results suggests that either the anti-human flk-1 and flt-1 antibodies did not cross-react with the rhesus-derived cells, or these cells express a different VEGF receptor. RT-PCR studies showed expression of flk-1 mRNA in the RESDECs, suggesting that the antibodies may not cross-react. Of course, it is possible that protein derived from this mRNA is not properly expressed on the cell surface.
[00042] The RESDEC cells do differ from HUVEC cells by lack of expression of and VE-cadherin, two surface antigens commonly, but not uniformly, found on the surface of endothelial cells. RT-PCR studies confirm absence of mRNA expression of these genes.
However, some studies of mouse ES cell-derived endothelial cells also shows lack of CD31 and VE-cadherin in certain endothelial cell populations. While CD31 and VE-cadherin can serve as positive markers of endothelial cells, lack of expression does not preclude these being endothelial cells.

[00043] Angiogenesis from RESDEC cells:
[00044] In vivo function of the RESDECs was first assessed by a Matrigel plug assay.
Here, RESDECs were imbedded in a sponge that was suspended into solidified Matrigel. This Matrigel plug is- then implanted subcutaneously in a severe combined immunodeficient (SCID) mouse. After approximately 28 days, the mouse was injeceted intravenously with a FITC-dextran solution, followed by plug removal and imaging. This demonstrated intense vascular localization toward the RESDEC containing sponge, a chemotactic-like event typical of endothelial cells. From the appearance of this vascularization, it is likely that this represents an area of murine vessel angiogenesis in response to RESDEC-derived factors.
Indeed, subsequent analysis,of RESDEC supernatant by ELISA demonstrated a significant level of vascular endothelial growth factor (VEGF) produced by these cells. However, basic fibroblast growth factor (bFGF), another angiogenic protein often produced by endothelial cells, was not measured in RESDEC culture supernatant.
[00045] To demonstrate neo-vessels produced by the RESDECs, 0.5 -1.0 x 106 cells were evenly suspended in a Matrigel plug implanted subcutaneously in a SCID mouse.
Again, the animals were injected with FITC-dextran, followed by plug removal and imaging.
Here, larger vascular structures were seen and subsequent histological examination of the plug showed vascular formation by the RESDECs.
[00046] Next, to demonstrate the ability of RESDEC cells to contribute to active angiogenesis within tumors in vivo another SCID mouse model was used. Here, cells of the mouse mammary carcinoma line C755 were injected subcutaneously either alone or co-injected with an equivalent number of RESDECs. Tumor growth was measured at regular intervals and tumors grew significantly faster when co-injected with RESDECs. Importantly, 106 RESDECs injected alone did not lead to any measurable tumor growth, demonstrating these cells are not directly tumorigenic. These tumors were highly vascular and immunohistochemical staining of the tumors with anti-human specific antibodies (that cross-react to rhesus monkey but not mouse cells) clearly demonstrate a contribution to the endothelium from the RESDEC
cells. Staining of a substantial number of endothelial cells was positive using anti-MHC class I
or anti-vWF
antibodies in tumors co-injected with RESDEC cells, but endothelial cells in tumors derived from C755 alone were negative for these antigens.

Claims (4)

CLAIM OR CLAIMS

I/WE CLAIM:
1. A method for directing the differentiation of primate embryonic stem cells into endothelial cells comprising the steps of (a) culturing a culture of embryonic stem cells in a medium containing vascular endothelial cell growth factor, basic fibroblast growth factor, insulin-like growth factor and epidermal growth factor; and (b) sub-culturing cells which have the morphology of endothelial cells.
2. A method as claimed in claim 1 wherein the primate cells are rhesus monkey cells.
3. A method as claimed in claim 1 wherein the primate cells are human cells.
4. A method as claimed in claim 1 wherein the medium for the culture of embryonic stem cells into endothelial cells also includes mammalian serum.
CA2465173A 2001-11-02 2002-11-01 Endothelial cells derived from primate embryonic stem cells Expired - Lifetime CA2465173C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US33533201P 2001-11-02 2001-11-02
US60/335,332 2001-11-02
PCT/US2002/035278 WO2003040319A2 (en) 2001-11-02 2002-11-01 Endothelial cells derived from primate embryonic stem cells

Publications (2)

Publication Number Publication Date
CA2465173A1 CA2465173A1 (en) 2003-05-15
CA2465173C true CA2465173C (en) 2010-04-20

Family

ID=23311328

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2465173A Expired - Lifetime CA2465173C (en) 2001-11-02 2002-11-01 Endothelial cells derived from primate embryonic stem cells

Country Status (15)

Country Link
US (1) US7176023B2 (en)
EP (1) EP1446476A4 (en)
JP (1) JP4085062B2 (en)
KR (1) KR20050042215A (en)
CN (1) CN100540657C (en)
AU (1) AU2002356896B2 (en)
BR (1) BR0213815A (en)
CA (1) CA2465173C (en)
IL (2) IL161461A0 (en)
IS (1) IS7242A (en)
LU (1) LU91074B1 (en)
MX (1) MXPA04004067A (en)
NZ (1) NZ532170A (en)
SE (1) SE529427C8 (en)
WO (1) WO2003040319A2 (en)

Families Citing this family (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2003080801A2 (en) * 2002-03-19 2003-10-02 Advanced Research & Technology Transfer Adipose stromal stem cells for tissue and vascular modification
US20040009589A1 (en) * 2002-03-26 2004-01-15 Shulamit Levenberg Endothelial cells derived from human embryonic stem cells
EP1783207A4 (en) * 2004-06-22 2008-07-30 Mitsubishi Tanabe Pharma Corp Method of producing vascular endothelial cells from primate embryonic stem cells
KR20140146224A (en) 2006-04-14 2014-12-24 어드밴스드 셀 테크놀로지, 인코포레이티드 Hemangio-colony forming cells
US20080044847A1 (en) * 2006-06-23 2008-02-21 Shusta Eric V Blood-Brain Barrier Model
KR100986149B1 (en) * 2007-09-14 2010-10-07 차의과학대학교 산학협력단 A process for the differentiation of vascular endothelial progenitor cells from embryoid bodies derived from embryonic stem cells using hypoxic media condition
WO2009104825A1 (en) 2008-02-18 2009-08-27 Kaist Method for inducing the defferentiation of embryonic stem cells into hemangioblast
RU2359030C1 (en) * 2008-03-19 2009-06-20 Общество С Ограниченной Ответственностью "Лаборатория Клеточных Технологий" Method for obtaining endotheliocytes from human embryonic stem cells (versions)
US20110086424A1 (en) 2008-05-06 2011-04-14 Advanced Cell Technology, Inc. Methods for producing enucleated erythroid cells derived from pluripotent stem cells
WO2011159797A2 (en) 2010-06-15 2011-12-22 Cellular Dynamics International, Inc. A compendium of ready-built stem cell models for interrogation of biological response
JP5897002B2 (en) * 2010-07-07 2016-04-13 セルラー ダイナミクス インターナショナル, インコーポレイテッド Endothelial cell production by programming
US20120295347A1 (en) * 2011-05-20 2012-11-22 Steven Kessler Methods and Compositions for Producing Endothelial Progenitor Cells from Pluripotent Stem Cells
CN102719393B (en) * 2012-06-05 2014-02-12 中山大学 Serum-free medium capable of inducing tumor stem cells for differentiation towards lymphatic endothelial cells, and method therefor
CN114558032A (en) 2012-12-21 2022-05-31 安斯泰来再生医药协会 Method for preparing platelets from pluripotent stem cells and composition thereof
CN103194423B (en) * 2013-03-07 2015-02-25 中国人民解放军军事医学科学院野战输血研究所 Medium and purpose thereof
WO2014185151A1 (en) * 2013-05-13 2014-11-20 コニカミノルタ株式会社 Cell-staining method and specimen collection tube for use in method
EP3003290B1 (en) 2013-06-05 2021-03-10 AgeX Therapeutics, Inc. Compositions for use in the treatment of wounds in mammalian species
US11078462B2 (en) 2014-02-18 2021-08-03 ReCyte Therapeutics, Inc. Perivascular stromal cells from primate pluripotent stem cells
US10240127B2 (en) 2014-07-03 2019-03-26 ReCyte Therapeutics, Inc. Exosomes from clonal progenitor cells
KR101756337B1 (en) 2015-08-12 2017-07-10 고려대학교 산학협력단 Method for Inducing Endodermal and Mesodermal Differentiation from Human Pluripotent Stem Cells by CXCR2 Suppression
CA3173124A1 (en) * 2020-04-06 2021-10-14 Kyle M. LOH Generating populations of human blood and blood vessel progenitors from pluripotent stem cells
CN115316589A (en) * 2021-03-30 2022-11-11 云南师范大学 Method for removing agglutinin in stem and leaf of pseudo-ginseng

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5612211A (en) 1990-06-08 1997-03-18 New York University Stimulation, production and culturing of hematopoietic progenitor cells by fibroblast growth factors
NZ314644A (en) 1993-05-24 2000-11-24 Immunex Corp Use of flt3-ligands as a growth stimulator of stem cells in the transplantation of tissue
JPH078273A (en) * 1993-06-14 1995-01-13 Kurabo Ind Ltd Serumfree culture of adhesive animal cell
EP0757562A1 (en) * 1994-04-29 1997-02-12 W.L. Gore & Associates, Inc. Improved blood contact surfaces using endothelium on a subendothelial extracellular matrix
US5843780A (en) 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
WO1997047734A1 (en) 1996-06-14 1997-12-18 The Regents Of The University Of California In vitro derivation and culture of primate pluripotent stem cells and therapeutic uses thereof

Also Published As

Publication number Publication date
NZ532170A (en) 2008-05-30
CA2465173A1 (en) 2003-05-15
CN100540657C (en) 2009-09-16
LU91074B1 (en) 2004-04-30
WO2003040319A3 (en) 2003-10-23
WO2003040319A2 (en) 2003-05-15
SE0401132L (en) 2004-05-03
IL161461A0 (en) 2004-09-27
US20030166273A1 (en) 2003-09-04
IS7242A (en) 2004-04-30
JP2005508180A (en) 2005-03-31
US7176023B2 (en) 2007-02-13
MXPA04004067A (en) 2004-07-08
CN1582331A (en) 2005-02-16
SE529427C8 (en) 2007-09-25
BR0213815A (en) 2004-10-19
KR20050042215A (en) 2005-05-06
SE529427C2 (en) 2007-08-07
AU2002356896B2 (en) 2007-11-01
JP4085062B2 (en) 2008-04-30
IL161461A (en) 2010-11-30
EP1446476A4 (en) 2005-01-26
SE0401132D0 (en) 2004-05-03
EP1446476A2 (en) 2004-08-18

Similar Documents

Publication Publication Date Title
CA2465173C (en) Endothelial cells derived from primate embryonic stem cells
AU2002356896A1 (en) Endothelial cells derived from primate embryonic stem cells
Corbel et al. Hematopoietic potential of the pre-fusion allantois
KR101195838B1 (en) Isolated pluripotent adult stem cells and methods for isolating and cultivating the same
Chen et al. Stromal cell-derived factor-1/CXCR4 signaling modifies the capillary-like organization of human embryonic stem cell-derived endothelium in vitro
US20190367883A1 (en) Regulating stem cells
US20110312091A1 (en) Pluripotent stem cells, method for preparation thereof and uses thereof
MX2007009173A (en) Adipose derived adult stromal cells exhibiting characteristics of endothelial cells.
Domev et al. Efficient engineering of vascularized ectopic bone from human embryonic stem cell–derived mesenchymal stem cells
WO2002083864A2 (en) Methods and reagents for cell transplantation
CA2465908A1 (en) Adipose tissue-derived stromal cells for the repair of corneal and intra-orbital defects and uses thereof
Martini et al. Human placenta-derived mesenchymal stem cells acquire neural phenotype under the appropriate niche conditions
JP2003516141A (en) Long-term cell culture compositions and genetically modified animals derived therefrom
JPWO2005085425A1 (en) Methods for differentiating mesenchymal stem cells into steroidogenic cells
WO2014163206A1 (en) Use of functional melanocytes readily differentiated from multilineage-differentiating stress-enduring (Muse) cells, distinct stem cells in human fibroblasts
WO2002084281A1 (en) Encapsulated cell indicator system
Qiu et al. Skeletal myogenic potential of mouse skin-derived precursors
Chen et al. In vitro expansion and differentiation of rat pancreatic duct-derived stem cells into insulin secreting cells using a dynamic three-dimensional cell culture system
Vodicka et al. The minipig as an animal model in biomedical stem cell research
JP2005287478A (en) Human fat precursor cell line and method for using the same
IL193947A (en) Cultured cells that stain as cd31bright, method for stimulating the same to differentiate into a progenitor/precursor cell population (pcp), use of said pcp in the preparation of medicaments and apparatus for implantation comprising the same

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20221101