CA2390281C - Hematopoietic differentiation of human embryonic stem cells - Google Patents

Hematopoietic differentiation of human embryonic stem cells Download PDF

Info

Publication number
CA2390281C
CA2390281C CA002390281A CA2390281A CA2390281C CA 2390281 C CA2390281 C CA 2390281C CA 002390281 A CA002390281 A CA 002390281A CA 2390281 A CA2390281 A CA 2390281A CA 2390281 C CA2390281 C CA 2390281C
Authority
CA
Canada
Prior art keywords
cells
hematopoietic
human
embryonic stem
medium
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
CA002390281A
Other languages
French (fr)
Other versions
CA2390281A1 (en
Inventor
Dan S. Kaufman
James A. Thomson
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 CA2390281A1 publication Critical patent/CA2390281A1/en
Application granted granted Critical
Publication of CA2390281C publication Critical patent/CA2390281C/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
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0647Haematopoietic stem cells; Uncommitted or multipotent progenitors
    • 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
    • C12N5/0634Cells from the blood or the immune system
    • 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
    • A61K2035/122Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells for inducing tolerance or supression of immune responses
    • 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
    • A61K2035/124Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells the cells being hematopoietic, bone marrow derived or blood 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
    • 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
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/13Coculture with; Conditioned medium produced by connective tissue cells; generic mesenchyme cells, e.g. so-called "embryonic fibroblasts"
    • 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
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/13Coculture with; Conditioned medium produced by connective tissue cells; generic mesenchyme cells, e.g. so-called "embryonic fibroblasts"
    • C12N2502/1394Bone marrow stromal cells; whole marrow
    • 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

Disclosed herein are methods of obtaining human hematopoietic cells from human embryonic stem cells using mammalian stromal cells. Hematopoietic cells derived in this way are useful for creating cell cultures suitable for transplantation, transfusion, and other purposes.

Description

WO 01/34776 ~ ~ PCT/US00/23469 HEMATOPOIETIC DIFFERENTIATION OF HUMAN EMBRYONIC STEM CELLS
CROSS REFERENCES TO RELATED APPLICATIONS
Not applicable.
STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH
__ __ BACKGROUND OF THE INVENTION
The present invention relates to the use of human embryonic stem cells to create blood-related cells, and the use of those blood-related cells for various purposes.
Techniques for isolating stable cultures of human embryonic stem cells have recently been described by our laboratory. See U.S. patent 5,843,780 and J. Thomson et al., 282 Science 1145-1147 (1998).
We have deposited two of our human embryonic stem cell lines with the American Type Culture Collection, 10801 University Boulevard, Manassas, Virginia 20170-2209 U.S.A. on July 7, 1999 and July 15, 1999 respectively.
Taxonomic descriptions of these deposits are human embryonic stem cell lines H1 and H9 respectively. It has been proposed in these publications that such cell lines may be used for, among other things, providing a source of specified cell lines of various types for research, transplantation and other purposes.
Under the storage and culturing conditions described in these publications the cell lines are maintained long term without differentiation into specific cell types.
When the cell lines are subsequently injected into immunodeficient mice, they form teratomas demonstrating differentiation into multiple tissue types.
When ES cells are used to produce desired cells, it is often preferable to optimize differentiation towards specific cell types. In the case of hematopoietic cells it is desirable that this result in hematopoietic cells that can be isolated and used to form multiple hematopoietic lineages. These cells may include, but not be limited to, hematopoietic stem cells.
Hematopoietic stem cell populations have been isolated directly from bone marrow. See C. Baum et al.
89 PNAS USA 2804-2808 (1992). However, this relies on a supply of bone marrow to obtain the cells.
There have also been some attempts to direct murine embryonic cell populations towards hematopoietic cells.
See e'a. U.S. patent 5,914,268; G. Keller, 7 Current Opinion In Cell Biology, 862-869 (1995); and T. Nakano et al. 265 Science 1098-1101 (1994). See also M. Weiss, 11 Aplastic Anemia And Stem Cell Biology, 1185-1195 (1997);
and S. Morrison et al., 11 Annu. Rev. Cell Dev. Biol., 35-71 (1995).
However, applying these teachings to primates has proven difficult. For example, in F. Li et al., 92 Blood 368a (1998) there was a discussion of techniques for differentiation of rhesus embryonic stem cell lines using a stromal cell line and exogenous cytokines. However, that group has more recently reported that their techniques had inadequate formation of colonies.
The treatment of various diseases by tissue transplantation has become routine. However, there can be waiting lists to obtain natural donated organs, cells, or tissue. Even when the natural donor material becomes available there is often a problem with rejection.
Traditional approaches for suppressing an immune response of recipients have drawbacks. For example, immunosuppressive drugs are costly and often have side effects.
In WO 98/07841 there was discussed techniques of deriving embryonic stem cells that are MHC compatible with a selected donor (e. g. transplanting a donor nucleus into an enucleated oocyte, followed by derivation of the stem cells therefrom). The application suggested that the resulting cells could be used to obtain MHC
compatible hematopoietic stem cells for use in medical treatments requiring bone marrow transplantation.
However, some diseases such as type 1 diabetes mellitus or multiple sclerosis involve an autoimmune response. For example, merely transplanting pancreatic islets (which are MHC compatible to the diseased individual) to replace destroyed pancreatic islets will not provide sufficient long term reduction in type 1 diabetes mellitus, as the immune system of the host will still attack the transplanted islets.
It can therefore be seen that a need exists for techniques for causing human embryonic stem cell cultures to differentiate to desired hematopoietic colonies.
Further, it is desired to develop improved uses for hematopoietic cells.
BRIEF SUMMARY OF THE INVENTION
In one aspect the present invention provides a method for obtaining human hematopoietic cells. One exposes a human embryonic stem cell culture to mammalian hematopoietic stromal cells so as to thereby create human hematopoietic cells. At least some of the human hematopoietic cells that are so created are CD34+ and/or are capable of forming hematopoietic cell colony forming units in methylcellulose culture.
CD34 is a standard marker for hematopoietic stem cells, as described in C. Baum et al. 89 PNAS USA 2809-2808 (1992) and S. Morrison et al., 11 Annu. Rev. Cell Dev. Biol., 35-71 (1995). The property of capability of forming a colony forming unit is indicative that the cells have the desired characteristics to form more differentiated hematopoietic lineages.
The stromal cells are preferably derived from bone marrow cells or embryonic yolk sac cells. Murine stromal cells may be used for this purpose. However, primate WO 01/34776 CA 02390281 2002-05-03 pCT/US00/23469 stromal and other mammalian stromal cells should be suitable as well.
In another aspect the invention provides a human hematopoietic cell which was derived from a human embryonic stem cell culture in vitro, and is capable of forming hematopoietic cell colony forming units in methylcellulose culture. As used in this patent, the term "derived" is intended to mean obtained directly or indirectly (e.g. through one or more intermediates or passages).
In yet another aspect the invention provides a method of transplanting human cellular material into a human recipient host. One obtains human hematopoietic cells which have been derived in vitro from an embryonic stem cell culture. One then obtains a selected human cellular material other than hematopoietic cells, the selected non-hematopoietic material having major histocompatibility complex compatibility to the hematopoietic cells. One then transplants both the hematopoietic cells and selected human non-hematopoietic cellular material into the human host.
For example, one can obtain human hematopoietic cells which have been derived in vitro from an embryonic stem cell culture (e. g. using the techniques described below). One also obtains human pancreatic islets which have MHC compatibility to the hematopoietic cells. Both the hematopoietic cells and pancreatic islets are then transplanted into the human (preferably after the recipient's own bone marrow has been inactivated).
The pancreatic islets can be obtained directly from a donor whose cells were used to create the embryonic stem cell culture. Alternatively, a single embryonic stem cell culture can be differentiated along two different paths. In one process the above technique can be used to create hematopoietic stem cells. These cells should develop into multiple hematopoietic lineages when WO 01/34776 CA 02390281 2002-05-03 pCT/US00/23469 transplanted into appropriate hosts. These lineages should include lymphocytes which would be tolerant of other cells derived from the same parental embryonic stem cells. In another process the stem cells would be directed towards pancreatic islets.
In another example one could supply oligodendrocytes to a human who has a multiple sclerosis condition. One obtains human hematopoietic cells which have been derived in vitro from an embryonic stem cell culture (e. g. using a technique described below). One also obtains human oligodendrocytes which have MHC compatibility to the bone marrow cells and transplants both the bone marrow cells and oligodendrocytes into the human.
The same human whose genetic material was used to create the embryonic stem cell can be a donor for the oligodendrocytes. Alternatively, the same embryonic stem cell culture can be differentiated along two separate paths to provide the two transplantable materials.
With respect to either disease (and potentially other autoimmune diseases) the immune and autoimmune rejection problems should be reduced by this technique.
In this regard, the recipient's original bone marrow can be totally or partially inactivated by radiation or chemical means before the transplantation. Thereafter, it is replaced at least in part by the transplanted hematopoietic cells. The elimination/reduction of the original bone marrow reduces the body's ability to create an autoimmune response. The matching of the MHC of the replacement bone marrow and the second transplantable material insures that the second material won't be rejected by the transplanted bone marrow.
Moreover, co-transplantation of hematopoietic cells and other tissue can be done to promote acceptance of the second tissue (e. g. heart muscle plus hematopoietic cells for treating heart disease; hepatocytes plus hematopoietic cells for treating liver disease). By creating hematopoietic chimeras improved acceptance of tissues with similarly matched MHC type can be obtained.
The present invention should be suitable to obtain a wide variety of hematopoietic cells of interest, such as erythroid cells, granulocyte cells, macrophages, lymphocyte precursors, monocytes, B cells, T cells, and the like. In this regard, colonies of differentiated ES
cells develop into hematopoietic colonies when harvested, separated into single cells, and plated into appropriate cultures. These colonies demonstrate the development of colony-forming cells which proliferate into colony-forming units (including colony forming unit-erythroid (CFU-E), blast forming unit-eythroid (BFU-E), colony forming unit-macrophage (CFU-M), colony forming unit-granulocyte/macrophage (CFU-GM) and colony forming unit-high proliferative potential (CFU-HPP)). The identification of colony forming cells indicates the differentiation of embryonic stem cells into hematopoietic cells capable of expanding into defined hematopoietic lineages under defined conditions.
The objects of the present invention therefore include providing:
(a) methods of the above kind for obtaining hematopoietic cells;
(b) cells derived using those methods; and (c) methods for using those derived cells for transplantation, transfusion and other purposes.
These and still other objects and advantages of the present invention will be apparent from the description of the preferred embodiments that follows. However, the claims should be looked to in order to judge the full scope of the invention.
DETAILED DESCRIPTION
Embryonic Stem Cell Culture The previously described human ES cell line Hl was used for the majority of experiments, albeit some of the WO 01/34776 CA 02390281 2002-05-03 pCT/US00/23469 following studies were done with the previously described ES cell lines H9 (or H9.2) with similar results. These cells were removed from frozen (liquid nitrogen) stocks of cells derived from the original isolated and propagated cell line. The Hl ES cells were grown in 6 well culture dishes (Nunclon, Fisher).
The dish was first coated with O.lo gelatin solution (Sigma) for one or more days in a 37°C/5o CO~ incubator.
After the one or more days, the gelatin solution was removed and the wells of the plate were next coated with irradiated mouse embryonic fibroblast (MEF) cells. MEF
cells were derived from day 12-13 mouse embryos in medium consisting of DMEM (GibcoBRL) supplemented with loo fetal bovine serum (Hyclone or Harlan), 2 mM 1-glutamine (GibcoBRL), and 100 units/ml. Penicillin, 100 mg/ml streptomycin (Sigma).
The MEF cells were irradiated with 5500 cGy from a cesium source prior to plating in the wells. The MEFs were added at a density of 5 x lO4cells/ml, 2.5 ml/well.
The plate coated with MEFs was then placed in 37°C/5o CO2 incubator for one or more days until addition of ES
cells.
ES cells were passed onto new MEFs at approximately 5-8 day intervals. The time depends on cell density and morphologic appearance of differentiation. For passage, the medium in a well of ES cells was removed and 1-2 ml of medium containing 1 mg/ml collagenase IV in DMEM
(GibcoBRL) was added. The plate was then placed at 37°C/5o CO2 for 5-20 minutes until the colonies of ES
cells began to round up.
The well was then scraped with a 5 ml pipette to detach the ES cells from the plate. The contents of the harvested well were placed in a 15 ml conical tube (Fisher) and spun in a centrifuge at 1000 rpm for 5 minutes. The medium was removed and 10 ml of fresh medium was added. This ES cell medium consists of F12/DMEM
_7_ (GibcoBRL)) supplemented with 20o serum replacement medium (GibcoBRL), 8 ng/ml of bFGF (GibcoBRL), 1o non-essential amino acid solution (GibcoBRL), 1 mM 1-glutamine (GibcoBRL), and 0.1M (3-mercaptoethanol.
The cells were again spun (5 min/1000 rpm), medium removed and resuspended at a concentration of 2.5 ml of medium for each (typically 15 ml medium for plating into 6 new wells, this would be a 1:6 passage). The cells were then pipetted into the wells of a plate that had been previously coated with MEFs as described above. The cells were evenly distributed into each well and the plate was placed in an incubator at 37°C/5o CO2.
At times if there were colonies of ES cells showing morphologic appearance of differentiation prior to cell passage, these colonies were removed by gentle scraping with a pulled glass pipette. This was done with observation through a dissecting microscope. After removal of the differentiated cells, the remaining colonies were passaged as above.
After passage, each well of ES cells was "fed" with fresh medium at 24-48 hour intervals. Here, the medium of each well was removed and 2.5 ml of fresh ES medium was added. All feeding and passage of ES cells were done in a sterile environment.
Differentiation Of ES Cells To promote hematopoietic differentiation of the human ES cells, the ES cells were harvested as above. The cells were then plated in 6 well plates coated with a mammalian stromal cell. In one experiment we used C166 cells that were previously irradiated with 2500 cGy. The C166 cells were originally obtained from the yolk sac of mice at embryonic day 12 and were graciously provided by Dr. Robert Auerbach (UW-Madison).
In another experiment, S17 cells were used. They were originally obtained from mouse bone marrow, and were -g-graciously provided by Dr. Kenneth Dorshkind (then at UC-Riverside, now at UCLA).
The C166 or S17 cells were plated at a density of 1 x 10'' cells/ml, 2.5 ml/well. The ES cells plated onto either S17 of C166 cells were then allowed to grow in a medium consisting of DMEM (GibcoBRL) supplemented with 20o fetal bovine serum (Hyclone), to nonessential amino acid solution, O.1M (3-mercaptoethanol, and 1 mM 1-glutamine. This medium was replaced in each well at 24-72 hour intervals with fresh medium. In selecting an appropriate medium, one merely needs to provide conventional conditions for cell growth, albeit supplemented with the specified stromal cells.
After 3-7 days from plating onto S17 or C166 cells, the ES cells began to visually appear differentiated in that they did not have the same uniform appearance as the undifferentiated ES cells maintained on MEF feeder cells.
The colonies of ES cells began to form multiple different cell types. Some of these colonies had regions that appeared to consist of cells with a cobblestone morphology indicative of colonies of early hematopoietic progenitor cells.
Confirming Blood-Related Cells One method to determine the presence of appropriate hematopoietic cells is to assay for hematopoietic colony forming cells (CFCs) in semisolid methylcellulose-containing medium. Here, the ES cells were allowed to differentiate on either C166 or S17 cells for 2-3 weeks, maintained as described above. After this time the medium was removed. 2.5 ml of calcium and magnesium free phosphate buffered saline (PBS) was added for 2-5 minutes, removed, and 1.5 ml. of trypsin (0.125x)-EDTA
(1mM) medium was added.
The cells were then placed at 37°C/5o C02 for 10 minutes. After this time, the colonies began to disassociate. The cells were further disassociated by _g_ pipetting and scraping the wells. The cells were placed in a 15 ml. conical, spun 5 min/1000 rpm, medium removed and 10 ml fresh medium (DMEM + 10 o FBS + 1-glutamine+
pen/strep) was added, and spun again. The cells were then suspended in 5 ml medium and passaged through a 100 mM
nytex filter to remove clumps of cells.
The filter was washed with an additional 5 ml medium. The disassociated/filtered cells were then counted on a hemacytometer and 1 x 10~ (usually, but not always this many cells) cells were placed in a new 15 ml conical. These cells were then spun, medium removed and 5 ml medium consisting of IMDM (GibcoBRL) supplemented with 2o fetal bovine serum (Hyclone) was added. Cells were spun, medium removed and 250 ul medium (IMDM + 2o FBS) was added.
In accordance with the specified test conditions, these cells were then added to 2.5 ml of Methocult GF+
H4435 medium (StemCell Technologies). This medium consists of l.Oo methylcellulose, supplemented with 300 FBS, 20 ng/ml IL-3, 20 ng/ml IL-6, 50 ng/ml stem cell factor, 3 units/ml erythropoietin, 20 ng/ml GM-CSF, 20 ng/ml G-CSF, 2 mM 1-glutamine, O.lmM b-mercaptoethanol, 1% bovine serum albumin. The cells in methylcellulose were then vortexed vigorously and then 1.1 ml of the mixture was plated onto a P35 plastic dish (Stem Cell Technologies), spread evenly on the dish and placed at 37°C/5% CO,.
Duplicate plates of each sample were typically plated with 4 x 10' cells/plate. After 14-21 days, the plates were analyzed under a microscope for the presence of hematopoietic colonies. The colonies were identified by comparison to a colony atlas (StemCell Technologies) or the book: Culture of Hematopoietic Cells, RI Freshney, IB Pragnell, MG Freshney, eds., Wiley-Liss, Inc. 1994.
Colonies were identified as one of the following: colony forming unit-erythroid (CFU-E), blast forming unit-WO 01/34776 CA 02390281 2002-05-03 pCT/US00/23469 eythroid (BFU-E), colony forming unit-macrophage (CFU-M), colony forming unit-granulocyte/macrophage (CFU-GM) or colony forming unit-high proliferative potential (CFU-HPP) .
The presence of the desired hematopoietic cells can also be confirmed by flow cytometry. One can look for specified cell surface antigens by flow cytometry. Here, ES cells differentiated on S17 cells or C166 cells as described above for 14-21 days, were harvested with trypsin/EDTA as described above and passed through a 100 mM nytex filter. The filtered cells were counted on a hemacytometer, then aliquotted into 15 X 75 plastic tubes (Fisher) at approximately 1 x 105 cells/tube. The cells were then spun, medium removed and 2-3 ml of FACS medium was added. (FRCS medium is PBS with O.So BSA (Sigma), O.lo sodium azide (Sigma)).
The cells were again spun and medium removed. Next an antibody directly linked to a fluorescent marker (FITC
or PE) was added to the wells at a concentration as recommended by the supplier. Cells have been analyzed with the following antibodies: CD34-FITC (Immunotech), CD45-PE (Pharmingen). IgG1-FITC and IgGl-PE were used as isotype controls for non-specific staining of the cells.
Cells were incubated with the appropriate antibody for approximately 30 min on ice, washed 1-2 times with 2-3 ml FAGS medium and resuspended in approximately 0.5 ml FRCS
medium.
The antibody labeled cells were then analyzed using a FACScan (Becton Dickinson) as per manufacturers recommendations. The presence of dead cells was determined by addition of propidium iodide (1 mg/ml solution, 5 ul added per tube) or 7-AAD (Calbiochem) (0.2 mg/ml ,5 ul/tube). The software for analysis was eithe r PC Lysis or Cellquest.
The following experimental techniques were used to analyze antigen expression by immunohistochemistry (IHC).

WO 01/34776 CA 02390281 2002-05-03 pCT~S00/23469 Here, differentiated ES cells that have been co-cultured with either C166 or S17 as above, were harvested with trypsin/EDTA as above. The cells were resuspended in medium containing DMEM supplemented with loo FBS at a concentration of approximately 1 x 109 - 1 x 10'.
"Cytospin" preparations of these cells were then made by spinning 1 x 10=~ - 1x 104 cells onto a glass slide (Superfrost/plus, Fisher) with a Cytospin II centrifuge (Shanndon).
These slides were then fixed with cold acetone and stored frozen at -20°C. For IHC staining the slides were thawed at room temperature and the cell pellet was outlined with a wax pen (DAKO). The cells were then stained as follows using a Vectastain ABC kit (Vector Laboratories, Burlingame, CA), all incubations were at room temperature. 100-200 ul PBS was added onto the cells for 5 minutes then removed. Vectastain blocking antibody solution (horse serum) was then added onto the cells for 15 minutes. The cells were then blotted dry and 100-200 ul of primary antibody solution was added. The primary antibodies were: IgGl (1 ug/sample, Sigma), anti-CD34 (0.5 ug/sample, Immunotech), anti-CD45 (1 ug/sample, DAKO), anti-class I (1 ug/sample, gift from Dr. Paul Leibson, Mayo Clinic), anti-CD14 (1 ug/sample, Pharmingen), anti-CD31 (1 ug/sample, Pharmingen).
Primary antibody was added for 30 minutes followed by PBS for 10 minutes. Next, biotinylated anti-IgG
antibody was added (Vectastain kit, solution B) for 30 minutes followed by PBS for 10 minutes. Next Vectastain ABC solution was added for 30 minutes at room temperature followed by PBS for 10 minutes. Next DAB solution (Vectastain) was added for 5 minutes followed by washing under running tap water for 10 minutes. In some experiments, the slides were then counterstained with Gill's hematoxylline solution (Vector labs) for 3 minutes followed by washing with running tap water for 10 minutes. The slides were then air dried. Cells staining positive appear brown.
CD34+ was demonstrated within a mixed population of cells (about lo) after 2-3 weeks. Even more importantly, differentiated ES cells were shown to develop into hematopoietic colonies when harvested, separated into cells and plated into methylcellulose (semi-solid) cultures.
Transplantation Currently hematopoietic cell transplantation is conducted clinically primarily for patients who have received high dose chemotherapy for treatment of malignancies. These patients typically receive a heterogeneous mixture of hematopoietic cells either from an autologous or allogeneic source. Human ES-derived hematopoietic stem cells will at minimum provide a more homogeneous cell population for hematopoietic cell transplantation.
Further, as discussed above, the MHC characteristics of the transplantation can now be controlled, thereby enabling treatment of autoimmune diseases. For example, both hematopoietic stem cells (HSCs) and a second lineage (e. g. pancreatic islets for diabetes or oligodendrocytes for multiple sclerosis) could be derived from the same parental ES cell line. With both lineages available, a hematopoietic chimera could be first created by performing a fully allogeneic hematopoietic cell transplant (HCT). The established state of chimerism would allow the recipient's immune system to "see" the subsequent transplant of the second cell type (e. g.
pancreatic islets cell or oligodendrocyte) as "self" and should not be rejected.
Note for example that oligodendrocytes have been obtained from mouse ES cells (O. Brustle et al., 285 Science 754-6 (1999)), as have cardiac muscle cells (M.
Klug et al., 98 J. Clin. Invest. 216-224 (1996)).

This method of creating hematopoietic chimeras will also promote acceptance of tissues transplanted for reasons other than autoimmunity. In this regard, mice receiving allogeneic hematopoietic stem cells do not reject other tissues with the same genetic background as the hematopoietic cells, but will still reject third-party grafts. See K. Gandy et al., 65 Transplantation 295-304 (1998).
In addition to animal studies, there are now clinical case reports of human patients who have previously received a hematopoietic cell transplant later requiring a solid organ (kidney) transplant. In these instances, the kidney transplant from the same person who had previously supplied the bone marrow transplant is immunologically accepted without further immunosuppression. See T. Spitzer et al., 68 Transplantation 480-484 (1999).
Work in canine models and more recently in human clinical trials has shown that milder non-myeloablative conditioning regimens can be used to better prepare hosts for allogenic HCT. Here, only moderate doses of total body irradiation and a short course of immunosuppression are used to prepare the hosts prior to receiving allogeneic HCTs.
Even though the preferred embodiments have been described above, it will be appreciated by those skilled in the art that other modifications can be made within the scope of the invention. For example, while two specific stromal type cells have been selected for use, many others are also suitable. For example, one publicly available stromal cell line is the M2-lOB4 cell line having ATCC designation number CRL-1972.
Further, while the above description focuses on the creation of precursors for red blood cells and bone marrow, various other blood-related cells of interest can be obtained in quantity using the above techniques. See also U.S. patent 5,914,268. Thus, the claims should be looked to in order to judge the full scope of the invention.
Industrial Applicability The invention provides blood-related cells useful for transplantation, research and other purposes.

Claims (6)

THE EMBODIMENTS OF THE INVENTION IN WHICH AN EXCLUSIVE
PROPERTY OF PRIVILEGE IS CLAIMED ARE DEFINED AS FOLLOWS:
1. A method for obtaining human hematopoietic cells, comprising culturing human embryonic stem cells on a layer of mammalian hematopoietic stromal cells in the presence of medium, such that the human embryonic stem cells differentiate into human hematopoietic cells.
2. The method of claim 1, wherein the human hematopoietic cells that are obtained are CD34+.
3. The method of claim 1, wherein the human hematopoietic cells that are obtained form hematopoietic cell colony forming units in methylcellulose culture.
4. The method of claim 1, wherein the stromal cells are selected from the group consisting of bone marrow cells and embryonic yolk sac cells.
5. A method for obtaining human hematopoietic cells, comprising culturing human embryonic stem cells in vitro on a layer of mammalian hematopoietic stromal cells in the presence of medium, such that the human embryonic stem cells differentiate into human hematopoietic cells, wherein the human hematopoietic cells that are obtained form erythroid blast forming units in methylcellulose culture.
6. The method of claim 5, wherein the cell culture has hematopoietic cells that are CD34+.
CA002390281A 1999-11-08 2000-08-25 Hematopoietic differentiation of human embryonic stem cells Expired - Lifetime CA2390281C (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US09/435,578 1999-11-08
US09/435,578 US6280718B1 (en) 1999-11-08 1999-11-08 Hematopoietic differentiation of human pluripotent embryonic stem cells
PCT/US2000/023469 WO2001034776A1 (en) 1999-11-08 2000-08-25 Hematopoietic differentiation of human embryonic stem cells

Publications (2)

Publication Number Publication Date
CA2390281A1 CA2390281A1 (en) 2001-05-17
CA2390281C true CA2390281C (en) 2005-10-25

Family

ID=23728955

Family Applications (1)

Application Number Title Priority Date Filing Date
CA002390281A Expired - Lifetime CA2390281C (en) 1999-11-08 2000-08-25 Hematopoietic differentiation of human embryonic stem cells

Country Status (13)

Country Link
US (3) US6280718B1 (en)
EP (1) EP1228194A1 (en)
JP (3) JP2003513664A (en)
KR (2) KR20030022766A (en)
CN (1) CN1228443C (en)
AU (1) AU784928B2 (en)
BR (1) BR0015374A (en)
CA (1) CA2390281C (en)
IL (2) IL149387A0 (en)
IS (1) IS6373A (en)
NO (1) NO20022180L (en)
SE (1) SE526490C2 (en)
WO (1) WO2001034776A1 (en)

Families Citing this family (176)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7410798B2 (en) * 2001-01-10 2008-08-12 Geron Corporation Culture system for rapid expansion of human embryonic stem cells
WO2000027995A1 (en) * 1998-11-09 2000-05-18 Monash University Embryonic stem cells
US8252280B1 (en) 1999-08-05 2012-08-28 Regents Of The University Of Minnesota MAPC generation of muscle
US7015037B1 (en) * 1999-08-05 2006-03-21 Regents Of The University Of Minnesota Multiponent adult stem cells and methods for isolation
US10638734B2 (en) 2004-01-05 2020-05-05 Abt Holding Company Multipotent adult stem cells, sources thereof, methods of obtaining and maintaining same, methods of differentiation thereof, methods of use thereof and cells derived thereof
US20030129745A1 (en) 1999-10-28 2003-07-10 Robl James M. Gynogenetic or androgenetic production of pluripotent cells and cell lines, and use thereof to produce differentiated cells and tissues
US6280718B1 (en) * 1999-11-08 2001-08-28 Wisconsin Alumni Reasearch Foundation Hematopoietic differentiation of human pluripotent embryonic stem cells
US6602711B1 (en) * 2000-02-21 2003-08-05 Wisconsin Alumni Research Foundation Method of making embryoid bodies from primate embryonic stem cells
US6534052B1 (en) 2000-09-05 2003-03-18 Yong-Fu Xiao Cardiac function comprising implantation of embryonic stem cell in which differentiation has been initiated
US6607720B1 (en) 2000-09-05 2003-08-19 Yong-Fu Xiao Genetically altered mammalian embryonic stem cells, their living progeny, and their therapeutic application for improving cardiac function after myocardial infarction
IL156303A0 (en) 2000-12-06 2004-01-04 Robert J Hariri Method of collecting placental stem cells
US7311905B2 (en) 2002-02-13 2007-12-25 Anthrogenesis Corporation Embryonic-like stem cells derived from post-partum mammalian placenta, and uses and methods of treatment using said cells
EP1366148A2 (en) * 2001-01-24 2003-12-03 THE GOVERNMENT OF THE UNITED STATES OF AMERICA, represented by THE DEPARTMENT OF HEALTH & HUMAN SERVICES Differentiation of stem cells to pancreatic endocrine cells
EP1367899A4 (en) * 2001-02-14 2004-07-28 Leo T Furcht Multipotent adult stem cells, sources thereof, methods of obtaining and maintaining same, methods of differentiation thereof, methods of use thereof and cells derived thereof
EP1362095B1 (en) 2001-02-14 2015-05-27 Anthrogenesis Corporation Post-partum mammalian placenta, its use and placental stem cells therefrom
US20030211605A1 (en) * 2001-05-01 2003-11-13 Lee Sang-Hun Derivation of midbrain dopaminergic neurons from embryonic stem cells
AU2002317039B2 (en) 2001-07-24 2007-10-04 Es Cell International Pte Ltd Methods of inducing differentiation of stem cells
MXPA04001725A (en) * 2001-08-24 2005-04-11 Advanced Cell Tech Inc Screening assays for identifying differentiation-inducing agents and production of differentiated cells for cell therapy.
DE10144326B4 (en) * 2001-09-10 2005-09-22 Siemens Ag Method and system for monitoring a tire air pressure
US6759244B2 (en) * 2001-11-08 2004-07-06 Art Institute Of New York And New Jersey, Inc. Composite blastocysts (CBs) from aggregates of dissociated cells of non-viable pre-embryos
PL373957A1 (en) * 2001-11-09 2005-09-19 Artecel Sciences, Inc. Methods and compositions for the use of stromal cells to support embryonic and adult stem cells
US20040224403A1 (en) * 2001-12-07 2004-11-11 Robarts Research Institute Reconstituting hematopoietic cell function using human embryonic stem cells
JP5364224B2 (en) * 2001-12-07 2013-12-11 ジェロン・コーポレーション Hematopoietic cells derived from human embryonic stem cells
AU2012258384B2 (en) * 2001-12-07 2016-04-21 Asterias Biotherapeutics, Inc. Hematopoietic cells from human embryonic stem cells
US7799324B2 (en) * 2001-12-07 2010-09-21 Geron Corporation Using undifferentiated embryonic stem cells to control the immune system
AU2016206280B2 (en) * 2001-12-07 2018-05-17 Asterias Biotherapeutics, Inc. Hematopoietic cells from human embryonic stem cells
AU2003235652A1 (en) * 2002-01-15 2003-07-30 Advanced Cell Technology, Inc. Cloning b and t lymphocytes
US20050090004A1 (en) * 2003-01-16 2005-04-28 Sayre Chauncey B. Stem cell maturation for all tissue lines
US20030134422A1 (en) * 2002-01-16 2003-07-17 Sayre Chauncey Bigelow Stem cell maturation for all tissue lines
US20050170506A1 (en) * 2002-01-16 2005-08-04 Primegen Biotech Llc Therapeutic reprogramming, hybrid stem cells and maturation
EP1490478A4 (en) * 2002-03-18 2006-03-22 Nat Jewish Med & Res Center Method for production of neutrophils and uses therefor
US7498171B2 (en) * 2002-04-12 2009-03-03 Anthrogenesis Corporation Modulation of stem and progenitor cell differentiation, assays, and uses thereof
GB0210741D0 (en) * 2002-05-10 2002-06-19 Medical Res Council Methods of therapy
US20040091936A1 (en) * 2002-05-24 2004-05-13 Michael West Bank of stem cells for producing cells for transplantation having HLA antigens matching those of transplant recipients, and methods for making and using such a stem cell bank
EP1513928B1 (en) 2002-05-24 2019-08-21 Advanced Cell Technology, Inc. A bank of stem cells for producing cells for transplantation having hla antigens matching those of transplant recipients, and methods for making and using such a stem cell bank
CN1668733A (en) * 2002-05-30 2005-09-14 细胞基因公司 Methods of using JNK or MKK inhibitors to modulate cell differentiation and to treat myeloproliferative disorders and myelodysplastic syndromes
US20060121607A1 (en) * 2002-08-08 2006-06-08 Thomas Schulz Compositions and methods for neural differentiation of embryonic stem cells
WO2004035748A2 (en) * 2002-10-16 2004-04-29 Advanced Cell Technology, Inc. Methods using gene trapped stem cells for marking pathways of stem cell differentiation and making and isolating differentiated cells
GB0224415D0 (en) * 2002-10-21 2002-11-27 Medical Res Council Compositions
WO2004047770A2 (en) 2002-11-26 2004-06-10 Anthrogenesis Corporation Cytotherapeutics, cytotherapeutic units and methods for treatments using them
EP1567639A4 (en) * 2002-12-05 2005-12-21 Technion Res & Dev Foundation Cultured human pancreatic islets, and uses thereof
US20040110286A1 (en) * 2002-12-06 2004-06-10 The John P. Robarts Research Institute Method for making hematopoietic cells
KR20050105467A (en) * 2003-02-13 2005-11-04 안트로제네시스 코포레이션 Use of umbilical cord blood to treat individuals having a disease, disorder or condition
US20060177929A1 (en) * 2003-03-24 2006-08-10 Klug Christopher A Regulation of self-renewal in stem cells
AU2004227204B2 (en) 2003-04-08 2010-06-03 Yeda Research And Development Co. Ltd Stem cells having increased sensitivity to SDF-1 and methods of generating and using same
FR2853551B1 (en) 2003-04-09 2006-08-04 Lab Francais Du Fractionnement STABILIZING FORMULATION FOR IMMUNOGLOBULIN G COMPOSITIONS IN LIQUID FORM AND LYOPHILIZED FORM
AU2004242091C1 (en) * 2003-05-07 2009-12-24 La Jolla Institute For Molecular Medicine Administration of hyaluronic acid to enhance the function of transplanted stem cells
NZ547105A (en) * 2003-10-16 2009-02-28 Univ Edinburgh Control of ES cell self-renewal and lineage specification, and medium therefor
IL158868A0 (en) 2003-11-13 2004-05-12 Yeda Res & Dev Methods of generating and using stem cells enriched with immature primitive progenitor
US20070141703A1 (en) * 2003-11-19 2007-06-21 Stanley Edouard G Methods for producing blood products from pluripotent cells in cell culture
JP4676442B2 (en) * 2003-12-02 2011-04-27 セラヴィー バイオサイエンシズ エルエルシー Compositions and methods for growing neural progenitor cells
US20070269412A1 (en) * 2003-12-02 2007-11-22 Celavie Biosciences, Llc Pluripotent cells
WO2005080551A2 (en) * 2004-02-12 2005-09-01 University Of Newcastle Upon Tyne Stem cells
US20050232905A1 (en) * 2004-03-26 2005-10-20 Yeh Edward T Use of peripheral blood cells for cardiac regeneration
WO2005097979A2 (en) * 2004-03-31 2005-10-20 Newlink Genetics Corporation Methods and compositions for obtaining hematopoietic stem cells derived from embryonic stem cells and uses thereof
US7622108B2 (en) * 2004-04-23 2009-11-24 Bioe, Inc. Multi-lineage progenitor cells
CN101080486B (en) * 2004-04-23 2012-05-16 佰欧益股份有限公司 Multi-lineage progenitor cells
WO2005113749A2 (en) 2004-05-14 2005-12-01 Becton, Dickinson And Company Stem cell populations and methods of use
EP1809739B1 (en) 2004-07-13 2014-10-15 Asterias Biotherapeutics, Inc. Medium for growing human embryonic stem cells
CA2504451A1 (en) * 2004-08-10 2006-02-10 Geron Corporation Dendritic cell vaccines for treating cancer made from embryonic stem cells
EP1799810A2 (en) * 2004-10-12 2007-06-27 Technion Research & Development Foundation Ltd. Isolated primate embryonic cells and methods of generating and using same
WO2006050330A2 (en) * 2004-11-01 2006-05-11 Wisconsin Alumni Research Foundation Platelets from stem cells
US20070077654A1 (en) * 2004-11-01 2007-04-05 Thomson James A Platelets from stem cells
US20060263879A1 (en) * 2004-12-30 2006-11-23 Stemlifeline, Inc. Methods and systems relating to embryonic stem cell lines
US20060275899A1 (en) * 2004-12-30 2006-12-07 Stemlifeline, Inc. Methods and compositions relating to embryonic stem cell lines
WO2006086746A2 (en) * 2005-02-09 2006-08-17 Burnham Institute For Medical Research Homogeneous neural precursor cells
AU2006243800B2 (en) 2005-05-04 2012-02-02 Commonwealth Scientific And Industrial Research Organisation Selecting, culturing and creating lineage committed hematopoietic stem cells
US8034613B2 (en) 2005-06-01 2011-10-11 Wisconsin Alumni Research Foundation Multipotent lymphohematopoietic progenitor cells
AU2006252576B2 (en) * 2005-06-01 2011-01-20 Wisconsin Alumni Research Foundation Method of forming dendritic cells from embryonic stem cells
JP5560393B2 (en) 2005-06-22 2014-07-23 アステリアス バイオセラピューティクス インコーポレイテッド Differentiation of primate pluripotent stem cells into cardiomyocyte lineage cells
GB2441488C (en) * 2005-06-22 2011-10-05 Geron Corp Suspension culture of human embryonic stem cells
EP1904624B1 (en) * 2005-07-20 2011-09-21 Seoul National University Industry Foundation Method for culturing and proliferating hematopoietic stem cells and progenitor cells using human endometrial cells
EP2530145A1 (en) 2005-10-13 2012-12-05 Anthrogenesis Corporation Immunomodulation using placental stem cells
US8129187B2 (en) 2005-12-13 2012-03-06 Kyoto University Somatic cell reprogramming by retroviral vectors encoding Oct3/4. Klf4, c-Myc and Sox2
US8048999B2 (en) 2005-12-13 2011-11-01 Kyoto University Nuclear reprogramming factor
US8278104B2 (en) 2005-12-13 2012-10-02 Kyoto University Induced pluripotent stem cells produced with Oct3/4, Klf4 and Sox2
EP1976978A2 (en) 2005-12-29 2008-10-08 Anthrogenesis Corporation Co-culture of placental stem cells and stem cells from a second source
ES2549111T3 (en) 2005-12-29 2015-10-23 Anthrogenesis Corporation Placental stem cell populations
US7727763B2 (en) * 2006-04-17 2010-06-01 Bioe, Llc Differentiation of multi-lineage progenitor cells to respiratory epithelial cells
EP2064319B1 (en) 2006-08-28 2017-02-22 Yeda Research and Development Co. Ltd. Methods of generating glial and neuronal cells and use of same for the treatment of medical conditions of the cns
US20080108044A1 (en) * 2006-11-08 2008-05-08 Deepika Rajesh In vitro differentiation of hematopoietic cells from primate embryonic stem cells
CA2670497A1 (en) * 2006-11-24 2008-05-29 Regents Of The University Of Minnesota Endodermal progenitor cells
US7883698B2 (en) * 2007-01-17 2011-02-08 Maria Michejda Isolation and preservation of fetal hematopoietic and mesencymal system cells from non-controversial materials and/or tissues resulting from miscarriages and methods of therapeutic use
NZ597779A (en) 2007-02-12 2013-07-26 Anthrogenesis Corp Treatment of inflammatory diseases using placental stem cells
JP2008307007A (en) 2007-06-15 2008-12-25 Bayer Schering Pharma Ag Human pluripotent stem cell induced from human tissue-originated undifferentiated stem cell after birth
US9029147B2 (en) * 2007-06-15 2015-05-12 Massachusetts Institute Of Technology Methods and compositions for enhanced differentiation from embryonic stem cells
US9213999B2 (en) 2007-06-15 2015-12-15 Kyoto University Providing iPSCs to a customer
US20090029463A1 (en) * 2007-07-25 2009-01-29 Bioe, Inc. Differentiation of Multi-Lineage Progenitor Cells to Chondrocytes
EP3524253A1 (en) 2007-09-28 2019-08-14 Celularity, Inc. Tumor suppression using human placental perfusate and human placenta-derived intermediate natural killer cells
JP5558097B2 (en) 2007-12-10 2014-07-23 国立大学法人京都大学 Efficient nuclear initialization method
US9683232B2 (en) 2007-12-10 2017-06-20 Kyoto University Efficient method for nuclear reprogramming
AU2009241736B2 (en) * 2008-04-30 2014-05-08 Sanbio, Inc. Neural regenerating cells with alterations in DNA methylation
CN101855350B (en) 2008-05-02 2014-12-31 国立大学法人京都大学 Method of nuclear reprogramming
CA2724839A1 (en) * 2008-05-21 2009-11-26 Bioe Llc Differentiation of multi-lineage progenitor cells to pancreatic cells
KR20180108887A (en) 2008-08-20 2018-10-04 안트로제네시스 코포레이션 Treatment of stroke using isolated placental cells
EP3539380A3 (en) 2008-08-20 2019-12-18 Celularity, Inc. Improved cell composition and methods of making the same
CA2734446C (en) 2008-08-22 2017-06-20 Anthrogenesis Corporation Methods and compositions for treatment of bone defects with placental cell populations
RU2562154C2 (en) 2008-11-19 2015-09-10 Антродженезис Корпорейшн Amniotic adhesive cells
US20100209399A1 (en) * 2009-02-13 2010-08-19 Celavie Biosciences, Llc Brain-derived stem cells for repair of musculoskeletal system in vertebrate subjects
WO2010098079A1 (en) 2009-02-24 2010-09-02 学校法人金沢医科大学 Method for denucleating nucleated erythrocyte, and denucleation inducer
WO2011005326A1 (en) 2009-07-09 2011-01-13 Massachusetts Institute Of Technology Methods and compositions for increased safety of stem cell-derived populations
AU2010279572B2 (en) 2009-08-03 2017-02-02 Recombinetics, Inc. Methods and compositions for targeted gene modification
CA2777552C (en) 2009-10-19 2019-05-21 Cellular Dynamics International, Inc. Cardiomyocyte production
ES2646750T3 (en) 2010-01-26 2017-12-15 Anthrogenesis Corporation Treatment of bone-related cancers using placental stem cells
TWI578993B (en) 2010-04-07 2017-04-21 安瑟吉納西斯公司 Angiogenesis using placental stem cells
TW201138792A (en) 2010-04-08 2011-11-16 Anthrogenesis Corp Treatment of sarcoidosis using placental 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
KR20130093091A (en) 2010-07-13 2013-08-21 안트로제네시스 코포레이션 Methods of generating natural killer cells
CA2806858C (en) 2010-08-04 2021-06-15 Cellular Dynamics International, Inc. Reprogramming immortalized b cells
CA2838330C (en) 2010-08-23 2021-01-26 President And Fellows Of Harvard College Optogenetic probes for measuring membrane potential
US8895291B2 (en) 2010-10-08 2014-11-25 Terumo Bct, Inc. Methods and systems of growing and harvesting cells in a hollow fiber bioreactor system with control conditions
WO2012054896A1 (en) 2010-10-22 2012-04-26 Biotime Inc. Methods of modifying transcriptional regulatory networks in stem cells
EP2658557A1 (en) 2010-12-31 2013-11-06 Anthrogenesis Corporation Enhancement of placental stem cell potency using modulatory rna molecules
AU2012225784B2 (en) 2011-03-04 2016-03-17 The Regents Of The University Of California Locally released growth factors to mediate motor recovery after stroke
EP2691512B1 (en) 2011-03-29 2019-05-01 Asterias Biotherapeutics, Inc. Enriched populations of cardiomyocyte lineage cells from pluripotent stem cells
WO2012154344A1 (en) 2011-04-06 2012-11-15 Sanbio, Inc. Methods and compositions for modulating peripheral immune function
TWI602570B (en) 2011-06-01 2017-10-21 安瑟吉納西斯公司 Treatment of pain using placental stem cells
US10865383B2 (en) 2011-07-12 2020-12-15 Lineage Cell Therapeutics, Inc. Methods and formulations for orthopedic cell therapy
AU2012298997B2 (en) 2011-08-23 2018-10-04 Wisconsin Alumni Research Foundation Angiohematopoietic progenitor cells
WO2013055476A1 (en) 2011-09-09 2013-04-18 Anthrogenesis Corporation Treatment of amyotrophic lateral sclerosis using placental stem cells
EP2594295A1 (en) 2011-11-16 2013-05-22 Servicio Andaluz De Salud Nerve implants based on a compacted biomaterial containing cells
US9447378B2 (en) 2012-04-27 2016-09-20 Massachusetts Institute Of Technology Method for differentiating human embryonic stem cells into β-cells for the treatment of type I diabetes
JP5432322B2 (en) 2012-05-08 2014-03-05 株式会社大塚製薬工場 Mammalian cell suspension for prevention of pulmonary embolism containing trehalose
US20140178988A1 (en) 2012-10-08 2014-06-26 Biotime, Inc. Differentiated Progeny of Clonal Progenitor Cell Lines
EP3622960A1 (en) 2013-02-05 2020-03-18 Celularity, Inc. Natural killer cells from placenta
WO2014171486A1 (en) 2013-04-17 2014-10-23 日産化学工業株式会社 Medium composition and method for producing red blood cells using same
EP3003290B1 (en) 2013-06-05 2021-03-10 AgeX Therapeutics, Inc. Compositions for use in the treatment of wounds in mammalian species
WO2014201431A1 (en) 2013-06-14 2014-12-18 Massachusetts Institute Of Technology Articles and methods for stem cell differentiation
NZ714410A (en) 2013-06-28 2020-02-28 Otsuka Pharma Factory Inc Trehalose and dextran-containing solution for transplanting mammalian cells
CN105992816B (en) 2013-11-16 2018-04-17 泰尔茂比司特公司 Cell amplification in bioreactor
CN103740642A (en) * 2014-01-28 2014-04-23 娄彩玲 Method for preparing human erythrocytes
US11078462B2 (en) 2014-02-18 2021-08-03 ReCyte Therapeutics, Inc. Perivascular stromal cells from primate pluripotent stem cells
JP6783143B2 (en) 2014-03-25 2020-11-11 テルモ ビーシーティー、インコーポレーテッド Passive replenishment of medium
US10240127B2 (en) 2014-07-03 2019-03-26 ReCyte Therapeutics, Inc. Exosomes from clonal progenitor cells
WO2016049421A1 (en) 2014-09-26 2016-03-31 Terumo Bct, Inc. Scheduled feed
CN107002040B (en) 2014-10-24 2021-08-10 大日本住友制药株式会社 Method for preparing retinal tissue
US10286009B2 (en) * 2015-05-16 2019-05-14 Asterias Biotherapeutics, Inc. Pluripotent stem cell-derived oligodendrocyte progenitor cells for the treatment of spinal cord injury
WO2017004592A1 (en) 2015-07-02 2017-01-05 Terumo Bct, Inc. Cell growth with mechanical stimuli
EP3354721B1 (en) 2015-09-08 2022-06-08 Sumitomo Pharma Co., Ltd. Method for producing retinal pigment epithelial cells
CA3007733A1 (en) 2015-12-07 2017-06-15 Biotime, Inc. Methods for the re-derivation of diverse pluripotent stem cell-derived brown fat cells
CN109069870B (en) 2016-02-24 2022-04-29 洛克菲勒大学 Embryonic cell-based therapeutic candidate screening systems, models for huntington's disease and uses thereof
US11920155B2 (en) 2016-03-30 2024-03-05 Asterias Biotherapeutics, Inc. Oligodendrocyte progenitor cell compositions
US11685883B2 (en) 2016-06-07 2023-06-27 Terumo Bct, Inc. Methods and systems for coating a cell growth surface
US11104874B2 (en) 2016-06-07 2021-08-31 Terumo Bct, Inc. Coating a bioreactor
WO2018084228A1 (en) 2016-11-04 2018-05-11 国立大学法人東京大学 Solution for cryopreservation of animal cells or animal tissues, cryopreserved product, and cryopreservation method
JP7184283B2 (en) 2016-11-25 2022-12-06 国立研究開発法人理化学研究所 CELL POPULATION FOR TRANSPLANTATION AND METHOD FOR PRODUCING THE SAME
US11889829B2 (en) 2016-12-14 2024-02-06 Otsuka Pharmaceutical Factory, Inc. Mammalian cell cryopreservation liquid
JP7007686B2 (en) 2017-03-08 2022-01-25 大日本住友製薬株式会社 Manufacturing method of retinal pigment epithelial cells
US11624046B2 (en) 2017-03-31 2023-04-11 Terumo Bct, Inc. Cell expansion
EP3656842A1 (en) 2017-03-31 2020-05-27 Terumo BCT, Inc. Cell expansion
CA3070215A1 (en) 2017-07-20 2019-01-24 Riken Method for maturation of retinal tissue containing continuous epithelium
CA3070212A1 (en) 2017-07-20 2019-01-24 Riken Method for preserving neural tissue
JP7162221B2 (en) 2017-09-08 2022-10-28 国立研究開発法人理化学研究所 Cell aggregate containing retinal tissue and method for producing the same
CN111094554A (en) 2017-09-14 2020-05-01 国立研究开发法人理化学研究所 Method for preparing retinal tissue
WO2019054515A1 (en) 2017-09-14 2019-03-21 国立研究開発法人理化学研究所 Method for amplifying cone photoreceptors or rod photoreceptors using dorsalization signal transmitter or ventralization signal transmitter
SG11202004829WA (en) 2017-11-24 2020-06-29 Sumitomo Chemical Co Method for producing cell mass including pituitary tissue, and cell mass thereof
WO2019103125A1 (en) 2017-11-24 2019-05-31 住友化学株式会社 Production method for cell mass including neural cells/tissue and non-neural epithelial tissue, and cell mass from same
CA3096870A1 (en) 2018-02-19 2019-08-22 Sumitomo Dainippon Pharma Co., Ltd. Cell aggregate, mixture of cell aggregates, and method for preparing same
WO2020039732A1 (en) 2018-08-24 2020-02-27 住友化学株式会社 Cell aggregation including olfactory neuron or precursor cell thereof, and method for producing same
CA3112902A1 (en) 2018-09-28 2020-04-02 Otsuka Pharmaceutical Factory, Inc. Mammal cell preserving solution containing acarbose or stachyose
EP3882342A4 (en) 2018-11-15 2022-08-24 JSR Corporation Method for producing brain organoids
TW202035683A (en) 2018-12-20 2020-10-01 日商住友化學股份有限公司 Embryonic erythroblast-containing cell population and method for producing same, cell culture composition, and compound test method
EP3903789A4 (en) 2018-12-28 2022-10-05 Riken Therapeutic drug for disease accompanied by disorders in retinal system cells or retinal tissue
JP2022520722A (en) 2019-01-23 2022-04-01 アステリアス バイオセラピューティクス インコーポレイテッド Dorsal-derived oligodendrocyte progenitor cells from human pluripotent stem cells
JPWO2020184720A1 (en) 2019-03-13 2020-09-17
WO2020218480A1 (en) 2019-04-26 2020-10-29 国立研究開発法人理化学研究所 Composite including neural retina, retinal pigment epithelial cells, and hydrogel, and method for producing same
CN113728089A (en) 2019-04-26 2021-11-30 株式会社大塚制药工场 Trehalose-containing solution for preserving mammalian cells
JPWO2021045217A1 (en) 2019-09-06 2021-03-11
CA3162273A1 (en) 2019-11-20 2021-05-27 Sumitomo Pharma Co., Ltd. Method for freezing cell aggregates
WO2021100829A1 (en) 2019-11-20 2021-05-27 大日本住友製薬株式会社 Method for freezing neural cells
US20230310614A1 (en) 2020-09-11 2023-10-05 Sumitomo Pharma Co., Ltd. Medium for Tissue for Transplantation
CN116157512A (en) 2020-09-11 2023-05-23 国立研究开发法人理化学研究所 Composite comprising cell aggregate containing neural retina and matrix, and method for producing same
JPWO2022191216A1 (en) 2021-03-09 2022-09-15
IL309344A (en) 2021-06-17 2024-02-01 Univ Kyoto Method for producing cerebral cortical cell preparation derived from human pluripotent stem cells
AU2022343749A1 (en) 2021-09-13 2024-03-28 FUJIFILM Cellular Dynamics, Inc. Methods for the production of committed cardiac progenitor cells
CA3232099A1 (en) 2021-09-23 2023-03-30 Adam Ezra Cohen Genetically encoded voltage indicators and uses thereof

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5635386A (en) * 1989-06-15 1997-06-03 The Regents Of The University Of Michigan Methods for regulating the specific lineages of cells produced in a human hematopoietic cell culture
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
US5914268A (en) 1994-11-21 1999-06-22 National Jewish Center For Immunology & Respiratory Medicine Embryonic cell populations and methods to isolate such populations
US5843780A (en) 1995-01-20 1998-12-01 Wisconsin Alumni Research Foundation Primate embryonic stem cells
WO1998007841A1 (en) 1996-08-19 1998-02-26 University Of Massachusetts Embryonic or stem-like cell lines produced by cross species nuclear transplantation
US6280718B1 (en) * 1999-11-08 2001-08-28 Wisconsin Alumni Reasearch Foundation Hematopoietic differentiation of human pluripotent embryonic stem cells

Also Published As

Publication number Publication date
JP2003513664A (en) 2003-04-15
SE0201328D0 (en) 2002-05-03
IL149387A0 (en) 2002-11-10
IL149387A (en) 2008-08-07
AU784928B2 (en) 2006-07-27
US20020015694A1 (en) 2002-02-07
CN1387565A (en) 2002-12-25
CA2390281A1 (en) 2001-05-17
KR20070047850A (en) 2007-05-07
US6280718B1 (en) 2001-08-28
NO20022180D0 (en) 2002-05-07
BR0015374A (en) 2002-12-24
SE0201328L (en) 2002-07-05
CN1228443C (en) 2005-11-23
KR20030022766A (en) 2003-03-17
US20040043484A1 (en) 2004-03-04
US6613568B2 (en) 2003-09-02
SE526490C2 (en) 2005-09-27
AU6940400A (en) 2001-06-06
JP2010042011A (en) 2010-02-25
EP1228194A1 (en) 2002-08-07
IS6373A (en) 2002-05-06
JP2014000096A (en) 2014-01-09
WO2001034776A1 (en) 2001-05-17
NO20022180L (en) 2002-06-03

Similar Documents

Publication Publication Date Title
CA2390281C (en) Hematopoietic differentiation of human embryonic stem cells
US11261426B2 (en) Pluripotent stem cell that can be isolated from body tissue
US20030153082A1 (en) Hematopoietic cells from human embryonic stem cells
US9255249B2 (en) Isolation and purification of hematopoietic stem cells from post-liposuction lipoaspirates
US20130064801A1 (en) Cellular Compositions and Methods of Making and Using Them
JP2024026891A (en) Methods and systems for producing hematopoietic lineage cells
US20050221482A1 (en) Methods and compositions for obtaining hematopoietic stem cells derived from embryonic stem cells and uses thereof
JP2021535747A (en) Generation of hematopoietic progenitor cells from human pluripotent stem cells
JPWO2008056779A1 (en) Method for culturing and subculture of primate embryonic stem cells and method for inducing differentiation thereof
US20040110286A1 (en) Method for making hematopoietic cells
CA2330208A1 (en) Compositions and methods for use in affecting hematopoietic stem cell populations in mammals
Gordon-Keylock et al. Induction of hematopoietic differentiation of mouse embryonic stem cells by an AGM-derived stromal cell line is not further enhanced by overexpression of HOXB4
CN113557296A (en) Expansion of hematopoietic stem cells
Schubbert et al. Methods for PTEN in stem cells and cancer stem cells
Basak et al. Human embryonic stem cells hemangioblast express HLA-antigens
US20230374458A1 (en) Compositions and methods for generating human yolk sac-like hematopoietic cells
Kaufman et al. Embryonic Stem Cells: Unique Potential to Treat Autoimmune Diseases
Kelley et al. Collection and Expansion of Stem Cells
Kaufman et al. Embryonicmaintainedlines, of yet human retain stem indefinitely embryonic (ES) their cells ability in are stem culture pluripotent to cells form as provides undifferentiated any cells cell a that new type. can model The cell
EP2749640A1 (en) Processes for Producing Ectodermal, Mesodermal and Endodermal Cells as well as Pluripotent Stem Cells, Haematopoietic Stem Cells, Side Population Cells and Mesenchymal Stem Cells, and a Method of De-differentiating Peripheral Blood Cells
AU2016206280A1 (en) Hematopoietic cells from human embryonic stem cells

Legal Events

Date Code Title Description
EEER Examination request
MKEX Expiry

Effective date: 20200825