WO2014171486A1 - 培地組成物及び当該培地組成物を用いた赤血球の製造方法 - Google Patents
培地組成物及び当該培地組成物を用いた赤血球の製造方法 Download PDFInfo
- Publication number
- WO2014171486A1 WO2014171486A1 PCT/JP2014/060829 JP2014060829W WO2014171486A1 WO 2014171486 A1 WO2014171486 A1 WO 2014171486A1 JP 2014060829 W JP2014060829 W JP 2014060829W WO 2014171486 A1 WO2014171486 A1 WO 2014171486A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cells
- stem cells
- medium
- hematopoietic stem
- hematopoietic
- Prior art date
Links
Classifications
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N5/00—Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
- C12N5/06—Animal cells or tissues; Human cells or tissues
- C12N5/0602—Vertebrate cells
- C12N5/0634—Cells from the blood or the immune system
- C12N5/0641—Erythrocytes
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/10—Growth factors
- C12N2501/125—Stem cell factor [SCF], c-kit ligand [KL]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/10—Growth factors
- C12N2501/14—Erythropoietin [EPO]
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/20—Cytokines; Chemokines
- C12N2501/23—Interleukins [IL]
- C12N2501/2303—Interleukin-3 (IL-3)
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2501/00—Active agents used in cell culture processes, e.g. differentation
- C12N2501/90—Polysaccharides
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N2533/00—Supports or coatings for cell culture, characterised by material
- C12N2533/70—Polysaccharides
Definitions
- the present invention relates to a medium composition containing a polysaccharide and a method for producing erythrocytes in vitro, characterized by using the medium composition.
- Red blood cell transfusion is used as a treatment for anemia, drug injury and bleeding due to trauma and surgery.
- the supply mainly depends on voluntary blood donation, but a great deal of labor is required for collection, inspection and storage to ensure its stable supply.
- blood donation-derived red blood cell preparations cannot completely eliminate viral infections such as HIV and hepatitis virus, and unknown infectious diseases may not be detected. Under such circumstances, in order to stably supply safe red blood cells, there is an increasing need to manufacture red blood cells in vitro as an alternative to blood donation (Non-patent Document 1).
- erythrocytes In vivo, it begins with hematopoietic stem cells, erythrocyte / megakaryocytic progenitor cells, erythroid progenitor cells (BFU-E, CFU-E), proerythroblasts, basophil erythroblasts, It is known that erythrocytes are produced via dyeable erythroblasts, normal dyeable erythroblasts, and reticulocytes. In addition, erythropoietin (EPO) and stem cell factor (SCF) have been reported as main factors for promoting erythroid differentiation (Non-Patent Documents 2 and 3).
- EPO erythropoietin
- SCF stem cell factor
- Non-Patent Document 1 a method for inducing red blood cells from ES cells or iPS cells which are pluripotent stem cells (Patent Document 1, Non-Patent Documents 4 and 5), or red blood cells from CD34 positive cells derived from peripheral blood, fetal liver, bone marrow or umbilical cord blood The method (Nonpatent literature 3, 6) which guide
- induced is developed.
- Non-patent Document 7 a technique for establishing an erythrocyte progenitor cell line from pluripotent stem cells and preparing a large amount of erythrocytes from the cells has been studied (Non-patent Document 7).
- Patent Document 2 the efficiency of the enucleation process is a problem (Patent Document 2). Furthermore, the concern of canceration by transfusion of erythrocyte progenitor cells having nuclei is also a problem in in vitro amplification of erythrocytes.
- An object of the present invention is to solve the above-mentioned problems of the prior art and to provide a medium composition for producing red blood cells in a short time and with high efficiency in vitro and a method for producing red blood cells using the composition. is there.
- the present inventors have succeeded in finding a medium composition that promotes red blood cell differentiation. Furthermore, it has been found that when the medium composition is used, differentiation from erythrocyte progenitor cells to erythrocytes is induced, and erythrocytes can be efficiently produced in vitro.
- the present invention is as follows: (1) A culture medium additive containing a polysaccharide used for differentiating hematopoietic stem cells and / or hematopoietic progenitor cells into erythrocytes. (2) The additive according to (1), wherein the polysaccharide has an anionic functional group. (3) The additive according to (2), wherein the anionic functional group of the polysaccharide is at least one selected from the group consisting of a carboxyl group, a sulfo group, and a phosphate group.
- the polysaccharide is hyaluronic acid, gellan gum, deacylated gellan gum, xanthan gum, carrageenan, diyutan gum, alginic acid, fucoidan, pectin, pectinic acid, pectinic acid, heparan sulfate, heparin, heparitin sulfate, keratosulfate, chondroitin
- the additive according to (3) which is at least one selected from the group consisting of sulfuric acid, dermatan sulfate, rhamnan sulfate, or salts thereof.
- a medium composition for erythroid differentiation comprising the additive according to any one of (1) to (5).
- the medium composition according to (6) further comprising one or more factors selected from the group consisting of SCF, IL-3, IL-6, IL-11, FL, TPO and EPO.
- a method for producing erythrocytes from hematopoietic stem cells and / or hematopoietic progenitor cells, and in the presence of the additive according to any one of (1) to (5), or according to (6) or (7) A method comprising culturing hematopoietic stem cells and / or hematopoietic progenitor cells in the medium composition.
- a method for inducing differentiation of hematopoietic stem cells into red blood cells or progenitor cells thereof, in the presence of the additive according to any one of (1) to (5), or in (6) or (7) A method comprising culturing hematopoietic stem cells in the medium composition described.
- the present invention provides a medium composition containing a specific compound (hereinafter also referred to as a specific compound), particularly a polymer compound having an anionic functional group (such as a polysaccharide).
- a specific compound hereinafter also referred to as a specific compound
- a polymer compound having an anionic functional group such as a polysaccharide
- the hematopoietic stem cell in the present invention is a cell having multipotency capable of differentiating into all blood cell differentiation lineages of blood cells and capable of self-replicating while maintaining the multipotency.
- the hematopoietic progenitor cell is a cell group including both a pluripotent hematopoietic progenitor cell that can differentiate into a plurality of blood cell differentiation lineages and a unipotent hematopoietic progenitor cell that can differentiate into a single blood cell differentiation lineage.
- Erythroid progenitor cells are hematopoietic progenitor cells that can only differentiate into unidirectional blood cells of the erythroid system.
- Hematopoietic stem cells, hematopoietic progenitor cells, and erythroid progenitor cells are pluripotent such as those collected from bone marrow, umbilical cord blood, spleen, fetal liver or peripheral blood, iPS cells (induced pluripotent stem cells) and ES cells (Embryonic stem cells). Those derived from sex stem cells in vitro can also be used.
- these cells may be purchased from reagent companies such as Takara Bio Inc., Lonza Japan Inc., and Veritas Inc.
- the origin of hematopoietic stem cells, hematopoietic progenitor cells and erythroid progenitor cells is not particularly limited as long as it is derived from a mammal. Preferred examples include humans, dogs, cats, mice, rats, rabbits, pigs, cows, horses, sheep, goats and the like, with humans being more preferred.
- CD34 positive means that CD (cluster of differentiation) 34 antigen is expressed on the cell surface. This antigen is a marker for hematopoietic stem cells and hematopoietic progenitor cells, and disappears as it differentiates.
- CD34-positive cells are a cell population containing a large amount of hematopoietic stem cells and hematopoietic progenitor cells, and can be suitably used when the production of erythrocytes in the present invention is carried out. Similar cell populations also include CD133 positive cells.
- Pluripotent stem cells are cells of the endoderm (eg internal gastric wall, gastrointestinal tract or lung), mesoderm (eg muscle, bone, blood or genitourinary) or ectoderm (eg epidermal tissue or nervous system) system
- the cells have both differentiation pluripotency capable of differentiating into many types of cells constituting the living body and self-replicating ability capable of maintaining differentiation pluripotency even after undergoing divisional proliferation. Examples thereof include ES cells, iPS cells, embryonic germ stem cells (EG cells), Muse cells, and the like.
- ES cells refer to pluripotent stem cells derived from embryos at the blastocyst stage, which is the early stage of animal development.
- iPS cells are also referred to as induced pluripotent stem cells or induced pluripotent stem cells.
- somatic cells such as fibroblasts
- differentiation pluripotency and self-similarity to ES cells are obtained.
- EG cells are pluripotent stem cells derived from spermatogonia (Reference: Nature. 2008, 456, 344-349).
- the pluripotent stem cell used in the present invention may be any pluripotent stem cell that has both pluripotency and self-renewal ability and can differentiate into erythrocytes.
- pluripotent stem cells include ES cells, iPS cells, embryonic germ stem cells (EG cells), Muse cells, and the like, more preferably ES cells and iPS cells.
- transcription factor genes necessary for acquiring pluripotency include Nanog, Oct3 / 4, Sox2, Sox1, Sox3, Sox15, Sox17, Klf4, c-Myc, N -Myc, L-Myc, Lin28, ERas, etc. are known. These initialization factors may be used in any combination.
- iPS cells By introducing into somatic cells such as blasts, iPS cells can be established.
- the iPS cell used in the present invention is not limited to the method of its establishment, but other than the cells established by the above-described gene introduction method, the establishment method by introduction of a gene different from the above, proteins and low molecular compounds (histone)
- the iPS cell may be an established method using a deacetylase (HDAC) inhibitor, MEK inhibitor or the like.
- HDAC deacetylase
- Examples of hematopoietic progenitor cells derived from pluripotent stem cells in the present invention include embryoid bodies (embryoid bodies) obtained by culturing iPS cells or ES cells under conditions suitable for induction of hematopoietic cell differentiation, or nets. Examples include cells contained in like structures.
- embryoid body refers to a cell having a cystic structure obtained by removing factors or feeder cells that maintain the undifferentiated nature of iPS cells or ES cells, and subjecting the iPS cells or ES cells to suspension culture. It is a mass (reference: Blood, 2003, 102, 906-915).
- the “net-like structure” is a three-dimensional sac-like structure (with space inside) derived from iPS cells or ES cells, which is formed by an endothelial cell population and the like, and contains blood progenitor cells inside It's about things.
- TAKAYAMA et al., BLOOD 2008, 111: 5298-5306 can be referred to.
- there is a report of promoting the induction of hematopoietic progenitor cells from pluripotent stem cells by co-culture with stromal cells reference document: WO2001 / 34776).
- hematopoietic progenitor cells prepared from pluripotent stem cells can further enhance their cell proliferation ability by establishing a cell line by long-term culture or introducing an oncogene. (Reference: WO2011 / 034073, PLoS ONE 2008, 3: e1544).
- the differentiation of hematopoietic stem cells and / or hematopoietic progenitor cells means that hematopoietic stem cells have a function unique to hematopoietic progenitor cells, pluripotent hematopoietic progenitor cells have become unipotent hematopoietic progenitor cells, That is, it is converted into mature blood cells such as red blood cells, white blood cells, and megakaryocytes.
- the medium composition of the present invention acts on hematopoietic stem cells and / or hematopoietic progenitor cells and exhibits an effect of supporting differentiation into erythrocytes when hematopoietic stem cells and / or hematopoietic progenitor cells are cultured in vitro. Therefore, when hematopoietic stem cells are cultured in the medium composition of the present invention, differentiation from hematopoietic stem cells to erythrocytes or progenitor cells thereof is promoted. Specifically, erythrocytes can be prepared in large quantities in vitro by culturing hematopoietic stem cells and / or hematopoietic progenitor cells using the medium composition.
- the present invention also provides a culture preparation of hematopoietic stem cells and / or hematopoietic progenitor cells obtained by culturing hematopoietic stem cells and / or hematopoietic progenitor cells in the medium composition of the present invention.
- the culture preparation includes hematopoietic stem cells and / or hematopoietic progenitor cells and the medium composition of the present invention.
- the culture vessel used for culturing hematopoietic stem cells and / or hematopoietic progenitor cells is not particularly limited as long as it can generally cultivate animal cells.
- flasks, dishes, petri dishes, tissue culture dishes, Multidish, microplate, microwell plate, multiplate, multiwell plate, chamber slide, petri dish, tube, tray, culture bag, roller bottle and the like can be mentioned.
- the material of these culture equipment is not particularly limited, and examples thereof include glass, polyvinyl chloride, cellulosic polymer, polystyrene, polymethyl methacrylate, polycarbonate, polysulfone, polyurethane, polyester, polyamide, polystyrene, polypropylene, and the like. .
- any medium can be used as the medium used in the present invention as long as it is a medium used for culturing hematopoietic stem cells and / or hematopoietic progenitor cells.
- Examples of such a medium include Dulbecco's Modified Eagle Medium (Dulbecco's Modified Eagles's Medium; DMEM), Ham F12 Medium (Ham's Nutrient Mixture F12), DMEM / F12 Medium, McCoy's 5A Medium (McCoy's 5).
- IMDM Modified D ulbecco's Medium
- StemPro34 manufactured by Invitrogen
- X-VIVO 10 manufactured by Cambridge
- X-VIVO 15 manufactured by Cambridge
- HPGM manufactured by Cambridge
- StemSpan H3000 stem cell technology
- StemSpanSFEM manufactured by Stem Cell Technology
- Stemline II manufactured by Sigma Aldrich
- QBSF-60 manufactured by Quality Biological
- the normal culture medium used in order to maintain a pluripotent stem cell can be used for culture
- DMEM / F12 medium Iskov modified Dulbecco medium (IMDM), Dulbecco modified Eagle medium (DMEM), Ham F-12 medium, X-VIVO 10 (Lonza), X-VIVO 15 (Lonza), mTeSR (Manufactured by Stem Cell Technology), TeSR2 (manufactured by Stem Cell Technology), StemProhESC SFM (manufactured by Invitrogen) and the like.
- These media can contain cell adhesion factors, examples of which include matrigel, collagen gel, gelatin, poly-L-lysine, poly-D-lysine, laminin, and fibronectin. These cell adhesion factors can be added in combination of two or more.
- thickeners such as guar gum, tamarind gum, propylene glycol alginate, locust bean gum, gum arabic, tara gum, tamarind gum, and methylcellulose can be further mixed with the above medium.
- hematopoietic stem cells and / or hematopoietic progenitor cells those skilled in the art can add one or more other chemical components or biological components in combination according to the purpose.
- Components added to the medium of hematopoietic stem cells and / or hematopoietic progenitor cells include fetal bovine serum, human serum, horse serum, insulin, transferrin, lactoferrin, cholesterol, ethanolamine, sodium selenite, monothioglycerol, 2- Examples include mercaptoethanol, bovine serum albumin, sodium pyruvate, polyethylene glycol, various vitamins, various amino acids, agar, agarose, collagen, methylcellulose, various cytokines, various hormones, various growth factors, various extracellular matrices, and various cell adhesion molecules. It is done.
- cytokines added to the medium include interleukin-1 (IL-1), interleukin-2 (IL-2), interleukin-3 (IL-3), interleukin-4 (IL-4), Interleukin-5 (IL-5), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin-8 (IL-8), interleukin-9 (IL-9), Interleukin-10 (IL-10), interleukin-11 (IL-11), interleukin-12 (IL-12), interleukin-13 (IL-13), interleukin-14 (IL-14), Interleukin-15 (IL-15), Interleukin-18 (IL-18), Interleukin-21 (IL-21), Interferon - ⁇ (IFN- ⁇ ), interferon- ⁇ (IFN- ⁇ ), interferon- ⁇ (IFN- ⁇ ), granulocyte colony stimulating factor (G-CSF), monocyte colony stimulating factor (M-CSF), granule Sphere-macrophage colony stimulating factor (GM-CSF), stem cell factor (SCF), f
- Hormones added to the medium include melatonin, serotonin, thyroxine, triiodothyronine, epinephrine, norepinephrine, dopamine, anti-Muellerian hormone, adiponectin, corticotropin, angiotensinogen and angiotensin, antidiuretic hormone, atrium Natriuretic peptide, calcitonin, cholecystokinin, corticotropin releasing hormone, erythropoietin, follicle stimulating hormone, gastrin, ghrelin, glucagon, gonadotropin releasing hormone, growth hormone releasing hormone, human chorionic gonadotropin, human placental lactogen, growth hormone , Inhibin, insulin, insulin-like growth factor, leptin, luteinizing hormone, melanocyte stimulating hormone, oxytocin, parathyroid hormone Prolactin, secretin, somatostatin, thro
- Growth factors added to the medium include transforming growth factor- ⁇ (TGF- ⁇ ), transforming growth factor- ⁇ (TGF- ⁇ ), macrophage inflammatory protein-1 ⁇ (MIP-1 ⁇ ), epidermal growth factor ( EGF), fibroblast growth factor-1, 2, 3, 4, 5, 6, 7, 8, or 9 (FGF-1, 2, 3, 4, 5, 6, 7, 8, 9), nerve Cell growth factor (NGF), hepatocyte growth factor (HGF), leukemia inhibitory factor (LIF), protease nexin I, protease nexin II, platelet derived growth factor (PDGF), cholinergic differentiation factor (CDF), various Chemokines, Notch ligands (such as Delta1), Wnt proteins, angiopoietin-like proteins 2, 3, 5 or 7 (Angpt2, 3, 5, 7), insulin-like growth factors ( GF), insulin-like growth factor binding protein (IGFBP), but such pleiotrophin (Pleiotrophin), and the like, but is not limited to these.
- TGF- ⁇
- cytokines and growth factors obtained by artificially modifying the amino acid sequences of these cytokines and growth factors by gene recombination techniques can also be added.
- examples thereof include IL-6 / soluble IL-6 receptor complex or Hyper IL-6 (a fusion protein of IL-6 and soluble IL-6 receptor).
- Examples of various extracellular matrix and various cell adhesion molecules include collagen I to XIX, fibronectin, vitronectin, laminin-1 to 12, nitogen, tenascin, thrombospondin, von Willebrand factor, osteopontin, fibrinogen, Various elastins, various proteoglycans, various cadherins, desmocollins, desmogleins, various integrins, E-selectin, P-selectin, L-selectin, immunoglobulin superfamily, matrigel, poly-D-lysine, poly-L-lysine, chitin, Examples include chitosan, sepharose, hyaluronic acid, alginic acid gel, various hydrogels, and cut fragments thereof.
- the medium composition of the present invention is preferably known as a factor that induces differentiation into erythrocytes among the above-mentioned cytokines and growth factors.
- Stem cell factor SCF
- interleukin-3 IL-3
- interleukin-6 IL-6
- interleukin-11 IL-11
- flk2 / flt3 ligand FL
- thrombopoietin TPO
- EPO erythropoietin
- SCF stem cell factor
- flk2 / flt3 ligand FL
- interleukin-3 IL-3
- thrombopoietin Including one or more factors selected from the group consisting of TPO
- EPO erythropoietin
- Also preferably includes SCF, a factor of 1, 2 or 3 is selected from the group consisting of IL-3 and EPO.
- the concentration at the time of adding a cytokine or a growth factor can be appropriately set within a range in which differentiation induction of hematopoietic stem cells and / or hematopoietic progenitor cells into erythrocytes can be achieved, and usually 0.1 ng / mL to 1000 ng / mL, preferably 1 ng / mL to 100 ng / mL.
- the above-mentioned chemical components or biological components can be used not only by adding them to the medium, but also by immobilizing them on the substrate or carrier surface during culture. Specifically, it is achieved by dissolving the target component in an appropriate solvent, coating the substrate or the carrier surface, and then washing the excess component.
- a substrate or a carrier surface may be coated in advance with a substance that specifically binds to the target component, and the target component may be added to the substrate.
- antibiotics added to the medium include sulfa drugs, penicillin, pheneticillin, methicillin, oxacillin, cloxacillin, dicloxacillin, flucloxacillin, nafcillin, ampicillin, penicillin, amoxicillin, cyclacillin, carbenicillin, ticarcillin, piperacillin, piperacillin, Mecuzurocillin, mecillinam, andinocillin, cephalosporin and its derivatives, oxophosphoric acid, amifloxacin, temafloxacin, nalidixic acid, pyromido acid, ciprofloxane, sinoxacin, norfloxacin, perfloxacin, rosoxacin, ofloxacin, enoxacin, pipexamic acid, sulbactam acid, sulbactam acid, sulbactam acid, sulbactam acid , ⁇ -bromopenicill
- the temperature for culturing hematopoietic stem cells and / or hematopoietic progenitor cells is usually 25 to 39 ° C, preferably 33 to 39 ° C.
- the CO 2 concentration is usually 4 to 10% by volume in the culture atmosphere, and preferably 4 to 6% by volume.
- the culture period is usually 3 to 120 days, preferably 7 to 35 days, more preferably 14 to 21 days.
- this co-culture can be performed by collecting bone marrow cells and culturing them as they are.
- stromal cells, hematopoietic stem cells and / or hematopoietic progenitor cells, and other cell groups are separated, and a combination of stromal cells and hematopoietic stem cells and / or hematopoietic progenitor cells other than the individual from whom the bone marrow was collected It is also possible to carry out co-culture with It is also possible to carry out co-culture by adding hematopoietic stem cells and / or hematopoietic progenitor cells after culturing and proliferating only stromal cells. In this case, the culture conditions and medium composition described above can be used.
- stromal cell any cell that contributes to the proliferation and maintenance of hematopoietic stem cells and / or hematopoietic progenitor cells can be used.
- mouse embryo fibroblasts (MEF cells) SL10 cells, preferably C3H10T1 / 2 cell line, OP9 cell, ST2 cell, NIH3T3 cell, PA6 cell, M15 cell, human mesenchymal stem cell (MSC), human umbilical vein endothelial cell (HUVEC), human endometrial epithelial cell, etc., more preferably C3H10T1 / 2 cell line, OP9 cell, human mesenchymal stem cell (MSC) can be used.
- stromal cells When stromal cells are used, cell proliferation can be suppressed by, for example, mitomycin C treatment or radiation irradiation.
- the culture of hematopoietic stem cells and / or hematopoietic progenitor cells is performed by automatically performing cell seeding, medium exchange, cell image acquisition, and cultured cell collection in a closed environment under mechanical control, and controlling pH, temperature, oxygen concentration, etc.
- it can also be performed by a bioreactor capable of high-density culture or an automatic culture apparatus.
- feed-batch culture, continuous culture, and perfusion culture as a method of supplying a new medium in the middle of culture using these devices and supplying the required substance to cells and / or tissues without excess or deficiency. It can be used for the culture method of the present invention.
- the specific compound used in the present invention is the differentiation of hematopoietic stem cells and / or hematopoietic progenitor cells into erythrocytes (preferably, the above-described factors that induce differentiation into erythrocytes into hematopoietic stem cells and / or hematopoietic progenitor cells into erythrocytes. Promote differentiation).
- the specific compound used in the present invention promotes differentiation of hematopoietic stem cells into erythrocytes or progenitor cells (preferably differentiation of hematopoietic stem cells into erythrocytes or progenitor cells by the above-described factor that induces differentiation into erythrocytes). To do.
- Examples of the specific compound used in the present invention include, but are not particularly limited to, a polymer compound, preferably a polymer compound having an anionic functional group.
- examples of the anionic functional group include carboxylic acid, sulfonic acid, phosphoric acid, and salts thereof, and carboxylic acid or a salt thereof is preferable.
- the polymer compound used in the present invention one composed of one or more kinds from the group of the anionic functional groups can be used.
- the polymer compound used in the present invention include, but are not limited to, a polysaccharide obtained by polymerizing 10 or more monosaccharides (for example, triose, tetrose, pentose, hexose, heptose, etc.). More preferably, an acidic polysaccharide having an anionic functional group is used.
- the acidic polysaccharide here is not particularly limited as long as it has an anionic functional group in its structure.
- a polysaccharide having uronic acid for example, glucuronic acid, iduronic acid, galacturonic acid, mannuronic acid is used.
- hyaluronic acid gellan gum, deacylated gellan gum, rhamzan gum, diyutan gum, xanthan gum, carrageenan, xanthan gum, hexuronic acid, alginic acid, fucoidan, pectin, pectinic acid, pectinic acid, heparan sulfate, heparin, heparin sulfate And those composed of one or more selected from the group consisting of keratosulfuric acid, chondroitin sulfate, dermatan sulfate, rhamnan sulfate and salts thereof.
- salts of alkali metals such as lithium, sodium and potassium
- salts of alkaline earth metals such as calcium, barium and magnesium
- salts of aluminum, zinc, copper, iron, ammonium, organic bases and amino acids Is mentioned.
- polymer compounds or polysaccharides preferably have a weight average molecular weight of 10,000 to 50,000,000, more preferably 100,000 to 20,000,000, still more preferably 1,000,000 to 10,000,000.
- the molecular weight can be measured in pullulan conversion by gel permeation chromatography (GPC).
- More preferable specific examples of the specific compound used in the present invention include hyaluronic acid, deacylated gellan gum, diyutan gum, carrageenan, xanthan gum and salts thereof, and can reduce the viscosity of the medium composition and cells or tissues.
- the most preferred example is deacylated gellan gum or a salt thereof.
- the deacylated gellan gum in the present invention is a linear chain composed of 4 molecules of sugar of 1-3 linked glucose, 1-4 bonded glucuronic acid, 1-4 bonded glucose and 1-4 bonded rhamnose.
- R1 and R2 are both hydrogen atoms, and n is a polysaccharide represented by an integer of 2 or more.
- R1 may contain a glyceryl group and R2 may contain an acetyl group
- the content of acetyl group and glyceryl group is preferably 10% or less, more preferably 1% or less.
- the specific compound of the present invention takes various forms upon addition to the liquid medium.
- the metal ions For example, calcium ions are taken in, and an amorphous structure is formed via the metal ions.
- the viscosity of the medium composition of the present invention prepared from deacylated gellan gum is 8 mPa ⁇ s or less, preferably 4 mPa ⁇ s or less, and in view of the ease of cell or tissue recovery, Preferably, it is 2 mPa ⁇ s or less.
- the specific compound in the present invention can be obtained by a chemical synthesis method, but when the compound is a natural product, it can be extracted from various plants, animals, and microorganisms containing the compound using conventional techniques. It is preferable to obtain by separation and purification. In the extraction, the compound can be extracted efficiently by using water or supercritical gas. For example, as a method for producing gellan gum, the produced microorganism is cultured in a fermentation medium, and the mucosa produced outside the cells is collected by a normal purification method, and after drying, pulverization, etc., it is powdered. Good.
- deacylated gellan gum it may be recovered after subjecting the mucosa to an alkali treatment to deacylate glyceryl groups and acetyl groups bound to 1-3-bound glucose residues.
- Purification methods include, for example, liquid-liquid extraction, fractional precipitation, crystallization, various ion exchange chromatography, gel filtration chromatography using Sephadex LH-20, etc., adsorption chromatography using activated carbon, silica gel, etc. or thin layer It is possible to purify by removing impurities by using adsorption / desorption treatment of an active substance by chromatography or high-performance liquid chromatography using a reverse phase column alone or in combination in any order and repeatedly.
- gellan gum producing microorganisms examples include, but are not limited to, Sphingomonas erodea and microorganisms modified from the genes of the microorganisms.
- deacylated gellan gum commercially available products such as “KELCAOGEL (registered trademark of C. Kelco) CG-LA” manufactured by Sanki Co., Ltd., “Kelco Gel (C.P. ⁇ Registered trademark of Kelco) ”or the like.
- the concentration of the specific compound in the medium can be appropriately set within a range that promotes differentiation of hematopoietic stem cells and / or hematopoietic progenitor cells into erythrocytes, but is usually 0.0005% to 1.0% (weight) / Volume), preferably 0.001% to 0.4% (weight / volume), more preferably 0.005% to 0.1% (weight / volume), still more preferably 0.005% to 0.05. % (Weight / volume). For example, in the case of deacylated gellan gum, it is usually 0.001 to 1.0, preferably 0.003 to 0.5, more preferably 0.005 to 0.1, and most preferably 0.015 to 0.03. % (Weight / volume) medium may be added.
- the said compound can also be changed into another derivative by the chemical synthesis method, and the said derivative obtained in that way can also be used effectively in this invention.
- the hydroxyl group corresponding to R1 and / or R2 of the compound represented by the general formula (I) is a C 1-3 alkoxy group, a C 1-3 alkylsulfonyl group, glucose or Derivatives substituted with monosaccharide residues such as fructose, oligosaccharide residues such as sucrose and lactose, and amino acid residues such as glycine and arginine can also be used in the present invention.
- the compound can be cross-linked using a cross-linker such as 1-ethyl-3- (3-di-methylaminopropyl) carbodiimide (EDC).
- the specific compound or salt thereof used in the present invention can exist in any crystal form depending on the production conditions, and can exist as any hydrate. These crystal forms, hydrates, and mixtures thereof. Are also included within the scope of the present invention. Moreover, although it may exist as a solvate containing organic solvents, such as acetone, ethanol, and tetrahydrofuran, all of these forms are contained in the scope of the present invention.
- the specific compound used in the present invention exists in the form of a tautomer, geometric isomer, tautomer or mixture of geometric isomers, or a mixture thereof formed by intra-ring or exocyclic isomerization. Also good. Regardless of whether the compound of the present invention is produced by isomerization, when it has an asymmetric center, it may exist in the form of a resolved optical isomer or a mixture containing them in an arbitrary ratio.
- metal ions such as divalent metal ions (calcium ions, magnesium ions, zinc ions, iron ions, copper ions, etc.) may be present, preferably calcium ions.
- the specific compound in the present invention When the specific compound in the present invention is added to the above medium, the specific compound is first dissolved or dispersed in an appropriate solvent (this is referred to as a medium additive). Thereafter, the concentration of the specific compound in the medium is a concentration that promotes differentiation of hematopoietic stem cells and / or hematopoietic progenitor cells into erythrocytes (usually 0.0005% to 1.0% (weight / volume), preferably 0.001%. To 0.4% (weight / volume), more preferably 0.005% to 0.1% (weight / volume), and still more preferably 0.005% to 0.05% (weight / volume). In addition, the medium additive may be added to the medium.
- deacylated gellan gum In the case of deacylated gellan gum, it is usually 0.001 to 1.0, preferably 0.003 to 0.5, more preferably 0.005 to 0.1, most preferably 0.015 to 0.03% ( (Weight / volume) It may be added to the medium.
- the medium additive is used for differentiation of hematopoietic stem cells and / or hematopoietic progenitor cells into erythrocytes (preferably differentiation of hematopoietic stem cells and / or hematopoietic progenitor cells into erythrocytes by the above-described factor that induces differentiation into erythrocytes). Used to promote.
- the medium additive is used to promote differentiation of hematopoietic stem cells into red blood cells or progenitor cells (preferably differentiation of hematopoietic stem cells into red blood cells or progenitor cells by the above-described factor that induces differentiation into red blood cells). Used.
- suitable solvents used for the medium additive include aqueous solvents such as water, dimethyl sulfoxide (DMSO), methanol, ethanol, butanol, propanol, glycerin, propylene glycol, butylene glycol, and other various alcohols.
- aqueous solvents such as water, dimethyl sulfoxide (DMSO), methanol, ethanol, butanol, propanol, glycerin, propylene glycol, butylene glycol, and other various alcohols.
- DMSO dimethyl sulfoxide
- methanol ethanol
- ethanol ethanol
- butanol propanol
- glycerin propylene glycol
- butylene glycol butylene glycol
- other various alcohols such as water, dimethyl sulfoxide (DMSO), methanol, ethanol, butanol, propanol, glycerin, propylene glycol, butylene glycol, and other various alcohol
- one or more polysaccharides such as guar gum, tamarind gum, propylene glycol alginate, locust bean gum, gum arabic, tara gum, tamarind gum, and methylcellulose can be mixed.
- the specific compound can be used by immobilizing on the surface of the carrier or supporting the specific compound inside the carrier.
- the specific compound can be in any shape at the time of provision or storage.
- the specific compound is bound to formulated solids such as tablets, pills, capsules, granules, liquids such as solutions or suspensions dissolved in appropriate solvents and solubilizers, or substrates or single substances. It can be in the state.
- Additives for formulation include antiseptics such as p-hydroxybenzoates; excipients such as lactose, glucose, sucrose, and mannitol; lubricants such as magnesium stearate and talc; polyvinyl Examples include binders such as alcohol, hydroxypropyl cellulose, and gelatin; surfactants such as fatty acid esters; and plasticizers such as glycerin. These additives are not limited to those described above, and can be freely selected as long as they are available to those skilled in the art.
- the specific compound in the present invention may be sterilized as necessary.
- the sterilization method is not particularly limited, and examples thereof include radiation sterilization, ethylene oxide gas sterilization, and autoclave sterilization. In these sterilization treatments, the specific compound may be in a solid state or a solution state.
- a specific compound is added to ion exchange water or ultrapure water. And it stirs, heating at the temperature (for example, 60 degreeC or more, 80 degreeC or more, 90 degreeC or more) which can melt
- stirring for example, a homomixer
- the method for mixing the aqueous solution and the medium is not particularly limited, and examples thereof include manual mixing such as pipetting, and mixing using equipment such as a magnetic stirrer, mechanical stirrer, homomixer, and homogenizer. Moreover, you may filter the culture medium composition of this invention with a filter after mixing.
- the size of the pores of the filter used for the filtration treatment is 5 ⁇ m to 100 ⁇ m, preferably 5 ⁇ m to 70 ⁇ m, more preferably 10 ⁇ m to 70 ⁇ m.
- deacylated gellan gum when preparing deacylated gellan gum, 0.1 to 1% (weight / volume), preferably 0.2 to 0.5% (weight / volume), more preferably 0.3 to 0.4%.
- Deacylated gellan gum is added to ion-exchanged water or ultrapure water so as to be (weight / volume).
- the temperature can be any number of times as long as the deacylated gellan gum can be dissolved, but until it becomes transparent by stirring while heating to 60 ° C or higher, preferably 80 ° C or higher, more preferably 90 ° C or higher. Dissolve. After dissolution, the mixture is allowed to cool with stirring and, for example, autoclaved at 121 ° C. for 20 minutes.
- the aqueous solution is added to the medium so as to have a desired final concentration (for example, 0.3% when the final concentration is 0.015%).
- the ratio of aqueous solution: medium is 1:20) and mixed uniformly.
- the method for mixing the aqueous solution and the medium is not particularly limited, and examples thereof include manual mixing such as pipetting, and mixing using equipment such as a magnetic stirrer, mechanical stirrer, homomixer, and homogenizer.
- the size of the pores of the filter used for the filtration treatment is 5 ⁇ m to 100 ⁇ m, preferably 5 ⁇ m to 70 ⁇ m, more preferably 10 ⁇ m to 70 ⁇ m.
- the form and state of hematopoietic stem cells and / or hematopoietic progenitor cells cultured by the method of the present invention can be arbitrarily selected by those skilled in the art.
- the preferred specific examples are not particularly limited, but the cells are dispersed in the medium composition alone, a plurality of cells are aggregated to form a cell mass (sphere), or two or more types A state in which the cells gather to form a cell sphere.
- hematopoietic stem cells and / or hematopoietic progenitor cells When culturing hematopoietic stem cells and / or hematopoietic progenitor cells by the method of the present invention, separately prepared hematopoietic stem cells and / or hematopoietic progenitor cells are added to the culture composition of the present invention so that they are uniformly dispersed. To be mixed.
- the mixing method in that case is not particularly limited, and examples thereof include manual mixing such as pipetting, and mixing using equipment such as a stirrer, a vortex mixer, a microplate mixer, and a shaker.
- the culture solution may be allowed to stand, or the culture solution may be rotated, shaken or stirred as necessary.
- the number of rotations and frequency may be appropriately set according to the purpose of those skilled in the art.
- the cells and / or tissues are separated from the medium composition by centrifugation or filtration, and then the new medium composition is added to the cells. And / or may be added to the tissue.
- cells and / or tissues may be appropriately concentrated by performing centrifugation or filtration treatment, and then a new medium composition may be added to the concentrated solution.
- the gravitational acceleration (G) at the time of centrifugation is 100 to 400 G
- the pore size of the filter used for the filtration treatment is 10 ⁇ m to 100 ⁇ m, but is not limited thereto.
- cultured cells and / or tissues can be separated by magnetic force using magnetic fine particles coated on the surface with an antibody that specifically binds to the target cells.
- magnetic fine particles include Dynabead (manufactured by Veritas), MACS microbead (manufactured by Miltenyi Biotech), and BioMag (manufactured by Technochemical).
- Exchange of these medium compositions can also be performed by a bioreactor or an automatic culture apparatus that can be executed in a closed environment under mechanical control.
- the frequency of medium replacement is not particularly limited and can be appropriately selected by those skilled in the art.
- a preferred method for producing erythrocytes from hematopoietic stem cells and / or hematopoietic progenitor cells according to the present invention is exemplified below.
- hematopoietic stem cells and / or hematopoietic progenitor cells for example, umbilical cord blood, bone marrow, peripheral blood, etc. are collected, and a cell population rich in hematopoietic stem cells and / or hematopoietic progenitor cells is isolated therefrom. Is done.
- cell populations include CD34 positive cells, CD133 positive cells and the like.
- CD34-positive cells can be separated by a combination of specific gravity centrifugation and a magnetic cell sorting (MACS) system or flow cytometry.
- MCS magnetic cell sorting
- CPD solution citrate-phosphate-dextran
- nucleated cell fraction a fraction containing a large amount of mononuclear cells
- Specific gravity centrifugation methods include, for example, specific gravity centrifugation using dextran and Ficoll solution, Ficoll-paque density gradient method, Percoll discontinuous density gradient specific gravity centrifugation method, and density gradient specific gravity centrifugation method using Lymphoprep.
- CD34 antibody magnetic beads produced by Miltenyi Biotech; hereinafter referred to as CD34 antibody magnetic beads
- CD34 antibody magnetic beads immobilized with anti-human CD34 monoclonal antibody and the separated and collected nucleated cell fraction are mixed, and then incubated at about 2 to 8 ° C. (About 30 minutes) and bind CD34 positive cells in the nucleated cell fraction to the antibody magnetic beads.
- the bound antibody magnetic beads / CD34 positive cells are separated and recovered using a dedicated magnetic cell separation device such as an auto MACS system (Miltenyi Biotech).
- the purity of the isolated hematopoietic stem cells and / or hematopoietic progenitor cells (eg, CD34 positive cells) (percentage of the desired number of cells in the total number of cells) is usually 70% or more, preferably 80% or more. More preferably, it is 90% or more, more preferably 99% or more, and most preferably 100%.
- the CD34 positive cells thus obtained are cultured in the culture composition of the present invention to differentiate the CD34 positive cells into erythrocytes. Conditions for culturing CD34 positive cells, culture apparatus, type of medium, type of compound of the present invention, content of compound of the present invention, type of additive, content of additive, culture period, culture temperature, etc. However, the present invention is not limited thereto.
- the total cell count is measured by the trypan blue method, and the cultured cells are labeled with a fluorescent dye such as FITC (fluorescein isothiocyanate), PE (phycoerythrin), APC (allophycocyanin), etc.
- FITC fluorescein isothiocyanate
- PE phycoerythrin
- APC allophycocyanin
- red blood cells can be confirmed by the presence of typical biconcave cells.
- the presence of erythrocytes can also be confirmed by staining with deoxyribonucleic acid (DNA) such as Hoechst 33342, TO-PRO (registered trademark) -3, DRAG5.
- DNA deoxyribonucleic acid
- Red blood cells and reticulocytes are generally negative in these stains.
- a proportion of the erythroid hematopoietic progenitor cells can be determined by subjecting a part of the culture solution to a colony assay and measuring the number of erythrocyte colonies formed.
- Red blood cells produced by the above method can be separated from hematopoietic stem cells and / or hematopoietic progenitor cells.
- the separation can be achieved, for example, by using an antibody against CD36 and / or glycophorin A.
- Examples of the separation method include magnetic bead separation via antibodies, cell sorting, passage of cells through a membrane or column carrying antibodies against CD36 and / or glycophorin A, and the like.
- hematopoietic stem cells and / or hematopoietic progenitor cells in the culture medium can be killed by ultraviolet irradiation.
- the concentration (%) of CO 2 in the CO 2 incubator was expressed by volume% of CO 2 in the atmosphere.
- PBS phosphate buffered saline (manufactured by Sigma Aldrich Japan)
- FBS fetal bovine serum (manufactured by Biological Industries).
- (w / v) represents the weight per volume.
- Test Example 1 Amplification test of erythrocyte progenitor cells using human umbilical cord blood CD34 positive cells
- Deacylated gellan gum (KELCOGEL CG-LA, manufactured by Sanki Co., Ltd.) was suspended in ultrapure water (Milli-Q water) to a concentration of 0.3% (w / v), and then heated at 90 ° C. The aqueous solution was autoclaved at 121 ° C. for 20 minutes.
- a medium composition was prepared by adding Deacylated gellan gum at a final concentration of 0.015% or 0.030% (w / v) to StemSpanSFEM (manufactured by Pharmaceutical).
- human umbilical cord blood CD34-positive cells purchased from Lonza were seeded in a medium composition to which the above-mentioned deacylated gellan gum was added so as to be 10,000 cells / 1 mL, and then a 24-well plate (manufactured by Corning). The well was dispensed at 1 mL per well. As a negative control, a suspension of CD34 positive cells in the same medium without deacylated gellan gum was dispensed.
- the plate was cultured in a CO 2 incubator (37 ° C., 5% CO 2 ) for 7 days, and the number of viable cells was measured by the trypan blue method.
- the number of glycophorin A positive CD34 negative cells was calculated as follows. First, the cells after liquid culture were stained with an anti-glycophorin A antibody (APC, manufactured by Becton Dickinson) and an anti-CD34 antibody (PE, manufactured by Becton Dickinson).
- APC anti-glycophorin A antibody
- PE anti-CD34 antibody
- the stained cells were washed with a PBS ( ⁇ ) solution containing 2% (v / v) FBS, and then propidium iodide (manufactured by Sigma-Aldrich Japan) was added to a final concentration of 1 ⁇ g / mL and stained.
- propidium iodide manufactured by Sigma-Aldrich Japan
- the medium composition of the present invention showed excellent glycophorin A positive CD34 negative cell amplification activity, and was confirmed to have red blood cell progenitor cell amplification activity.
- Table 1 shows the amplification rate when 0.015% or 0.030% of the deacylated gellan gum was added when the number of glycophorin A positive CD34 negative cells when deacylated gellan gum was not added was 1.
- the medium composition according to the present invention is extremely useful when producing erythrocytes in vitro from hematopoietic stem cells and / or hematopoietic progenitor cells.
- Red blood cells produced by the method of the present invention are extremely useful in treatments that require red blood cell transfusion.
Landscapes
- Health & Medical Sciences (AREA)
- Engineering & Computer Science (AREA)
- Biomedical Technology (AREA)
- Life Sciences & Earth Sciences (AREA)
- Wood Science & Technology (AREA)
- Organic Chemistry (AREA)
- Chemical & Material Sciences (AREA)
- Biotechnology (AREA)
- Zoology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Genetics & Genomics (AREA)
- Microbiology (AREA)
- Cell Biology (AREA)
- Immunology (AREA)
- Biochemistry (AREA)
- General Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- Hematology (AREA)
- Micro-Organisms Or Cultivation Processes Thereof (AREA)
- Medicines Containing Material From Animals Or Micro-Organisms (AREA)
Abstract
Description
例えば、多能性幹細胞であるES細胞或いはiPS細胞から赤血球を誘導する方法(特許文献1、非特許文献4、5)、或いは末梢血、胎児肝臓、骨髄若しくは臍帯血由来のCD34陽性細胞から赤血球を誘導する方法(非特許文献3、6)が開発されている。また、多能性幹細胞から赤血球前駆細胞株を樹立し、当該細胞から赤血球を大量に調製する技術の検討も行われている(非特許文献7)。しかしながら、これらの培養法は、赤血球を短時間かつ高効率に生産させることが困難であり、特に脱核過程の効率化が課題となっている(特許文献2)。更に、核を有する赤血球前駆細胞を輸血することによる癌化の懸念も赤血球の生体外増幅において問題となっている。
(1)造血幹細胞及び/又は造血前駆細胞を赤血球に分化させる際に用いられる、多糖類を含む培地添加剤。
(2)前記多糖類が、アニオン性官能基を有する、(1)に記載の添加剤。
(3)前記多糖類のアニオン性官能基が、カルボキシル基、スルホ基及びリン酸基からなる群から選択される少なくとも1種である、(2)に記載の添加剤。
(4)前記多糖類が、ヒアルロン酸、ジェランガム、脱アシル化ジェランガム、キサンタンガム、カラギーナン、ダイユータンガム、アルギン酸、フコイダン、ペクチン、ペクチン酸、ペクチニン酸、ヘパラン硫酸、ヘパリン、ヘパリチン硫酸、ケラト硫酸、コンドロイチン硫酸、デルマタン硫酸、ラムナン硫酸またはそれらの塩からなる群から選択される少なくとも1種である、(3)に記載の添加剤。
(5)前記多糖類が、脱アシル化ジェランガムまたはその塩である、(4)に記載の添加剤。
(6)(1)乃至(5)のいずれかに記載の添加剤を含有してなる赤血球分化用の培地組成物。
(7)SCF、IL-3、IL-6、IL-11、FL、TPO及びEPOからなる群から選択される1又は2以上の因子を更に含む、(6)に記載の培地組成物。
(8)造血幹細胞及び/又は造血前駆細胞から赤血球を製造する方法であって、(1)乃至(5)のいずれかに記載の添加剤の存在下、或いは(6)又は(7)に記載の培地組成物中で、造血幹細胞及び/又は造血前駆細胞を培養することを特徴とする、方法。
(9)造血幹細胞の赤血球又はその前駆細胞への分化を誘導する方法であって、(1)乃至(5)のいずれかに記載の添加剤の存在下、或いは(6)又は(7)に記載の培地組成物中で、造血幹細胞を培養することを特徴とする、方法。
(10)造血幹細胞及び/又は造血前駆細胞、並びに(6)又は(7)に記載の培地組成物を含む、造血幹細胞及び/又は造血前駆細胞の培養調製物。
本明細書において用いる用語につき、以下の通り定義する。
培地に添加される増殖因子としては、トランスフォーミング成長因子-α(TGF-α)、トランスフォーミング成長因子-β(TGF-β)、マクロファージ炎症蛋白質-1α(MIP-1α)、上皮細胞増殖因子(EGF)、線維芽細胞増殖因子-1、2、3、4、5、6、7、8、又は9(FGF-1、2、3、4、5、6、7、8、9)、神経細胞増殖因子(NGF)、肝細胞増殖因子(HGF)、白血病阻止因子(LIF)、プロテアーゼネキシンI、プロテアーゼネキシンII、血小板由来成長因子(PDGF)、コリン作動性分化因子(CDF)、各種ケモカイン、Notchリガンド(Delta1など)、Wnt蛋白質、アンジオポエチン様蛋白質2、3、5または7(Angpt2、3、5、7)、インスリン様成長因子(IGF)、インスリン様成長因子結合蛋白質(IGFBP)、プレイオトロフィン(Pleiotrophin)などが挙げられるが、これらに限られるわけではない。
各種細胞外マトリックスや各種細胞接着分子の例としては、コラーゲンI乃至XIX、フィブロネクチン、ビトロネクチン、ラミニン-1乃至12、ニトジェン、テネイシン、トロンボスポンジン、フォンビルブランド(von Willebrand)因子、オステオポンチン、フィブリノーゲン、各種エラスチン、各種プロテオグリカン、各種カドヘリン、デスモコリン、デスモグレイン、各種インテグリン、E-セレクチン、P-セレクチン、L-セレクチン、免疫グロブリンスーパーファミリー、マトリゲル、ポリ-D-リジン、ポリ-L-リジン、キチン、キトサン、セファロース、ヒアルロン酸、アルギン酸ゲル、各種ハイドロゲル、さらにこれらの切断断片などが挙げられる。
本発明に用いる特定化合物の例としては、特に制限されるものではないが、高分子化合物が挙げられ、好ましくはアニオン性の官能基を有する高分子化合物が挙げられる。
アニオン性の官能基としては、カルボン酸、スルホン酸、リン酸及びそれらの塩が挙げられ、カルボン酸またはその塩が好ましい。
本発明に用いる高分子化合物は、前記アニオン性の官能基の群より1種又は2種以上から構成されるものを使用できる。
脱アシル化ジェランガムから調製される本発明の培地組成物の粘度は、8mPa・s以下であり、好ましくは4mPa・s以下であり、細胞または組織の回収のしやすさの点を考慮すると、より好ましくは2mPa・s以下である。
そして、脱アシル化ジェランガムの場合、市販のもの、例えば、三晶株式会社製「KELCAOGEL(シーピー・ケルコ社の登録商標)CG-LA」、三栄源エフ・エフ・アイ株式会社製「ケルコゲル(シーピー・ケルコ社の登録商標)」等を使用することができる。
なお該濃度は、以下の式で算出できる。
濃度(%)=特定化合物の重量(g)/培地組成物の容量(ml)×100
なお該濃度は、以下の式で算出できる。
濃度(%)=特定化合物の重量(g)/培地組成物の容量(ml)×100
CO2インキュベーターにおけるCO2の濃度(%)は、雰囲気中のCO2の体積%で示した。また、PBSはリン酸緩衝生理食塩水(シグマアルドリッチジャパン社製)を意味し、FBSは牛胎児血清(Biological Industries社製)を意味する。また、(w/v)は、1体積あたりの重量を表わす。
脱アシル化ジェランガム(KELCOGEL CG-LA、三晶株式会社製)を0.3%(w/v)となるように超純水(Milli-Q水)に懸濁した後、90℃にて加熱しながらの撹拌により溶解し、本水溶液を121℃で20分オートクレーブ滅菌した。本溶液を用いて最終濃度100ng/mLのSCF(和光純薬工業社製)、最終濃度20ng/mLのIL-3(和光純薬工業社製)及び最終濃度1ユニット/mLのEPO(田辺三菱製薬社製)を添加したStemSpanSFEM(ステムセルテクノロジー社製)に終濃度0.015%或いは0.030%(w/v)の脱アシル化ジェランガムを添加した培地組成物を調製した。引き続き、Lonza社より購入したヒト臍帯血のCD34陽性細胞を、10000細胞/1mLとなるように上記の脱アシル化ジェランガムを添加した培地組成物に播種した後、24ウエルプレート(コーニング社製)のウェルに1ウェル当たり1mLになるように分注した。なお、陰性対照として脱アシル化ジェランガムを含まない同上培地にCD34陽性細胞を懸濁したものを分注した。
Claims (10)
- 造血幹細胞及び/又は造血前駆細胞を赤血球に分化させる際に用いられる、多糖類を含む培地添加剤。
- 前記多糖類が、アニオン性官能基を有する、請求項1に記載の添加剤。
- 前記多糖類のアニオン性官能基が、カルボキシル基、スルホ基及びリン酸基からなる群から選択される少なくとも1種である、請求項2に記載の添加剤。
- 前記多糖類が、ヒアルロン酸、ジェランガム、脱アシル化ジェランガム、キサンタンガム、カラギーナン、ダイユータンガム、アルギン酸、フコイダン、ペクチン、ペクチン酸、ペクチニン酸、ヘパラン硫酸、ヘパリン、ヘパリチン硫酸、ケラト硫酸、コンドロイチン硫酸、デルマタン硫酸、ラムナン硫酸またはそれらの塩からなる群から選択される少なくとも1種である、請求項3に記載の添加剤。
- 前記多糖類が、脱アシル化ジェランガムまたはその塩である、請求項4に記載の添加剤。
- 請求項1乃至5のいずれか1項に記載の添加剤を含有してなる赤血球分化用の培地組成物。
- SCF、IL-3、IL-6、IL-11、FL、TPO及びEPOからなる群から選択される1又は2以上の因子を更に含む、請求項6に記載の培地組成物。
- 造血幹細胞及び/又は造血前駆細胞から赤血球を製造する方法であって、請求項1乃至5のいずれか1項に記載の添加剤の存在下、或いは請求項6又は7に記載の培地組成物中で、造血幹細胞及び/又は造血前駆細胞を培養することを特徴とする、方法。
- 造血幹細胞の赤血球又はその前駆細胞への分化を誘導する方法であって、請求項1乃至5のいずれか1項に記載の添加剤の存在下、或いは請求項6又は7に記載の培地組成物中で、造血幹細胞を培養することを特徴とする、方法。
- 造血幹細胞及び/又は造血前駆細胞、並びに請求項6又は7に記載の培地組成物を含む、造血幹細胞及び/又は造血前駆細胞の培養調製物。
Priority Applications (3)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
EP14785390.7A EP2987852A4 (en) | 2013-04-17 | 2014-04-16 | MIDDLE COMPOSITION AND PROCESS FOR PRODUCING RED GLOBLES USING THE SAME |
US14/784,843 US20160060601A1 (en) | 2013-04-17 | 2014-04-16 | Medium composition and method for producing red blood cells using same |
JP2015512506A JP6319304B2 (ja) | 2013-04-17 | 2014-04-16 | 培地組成物及び当該培地組成物を用いた赤血球の製造方法 |
Applications Claiming Priority (2)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
JP2013-086904 | 2013-04-17 | ||
JP2013086904 | 2013-04-17 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2014171486A1 true WO2014171486A1 (ja) | 2014-10-23 |
Family
ID=51731423
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/JP2014/060829 WO2014171486A1 (ja) | 2013-04-17 | 2014-04-16 | 培地組成物及び当該培地組成物を用いた赤血球の製造方法 |
Country Status (4)
Country | Link |
---|---|
US (1) | US20160060601A1 (ja) |
EP (1) | EP2987852A4 (ja) |
JP (1) | JP6319304B2 (ja) |
WO (1) | WO2014171486A1 (ja) |
Cited By (4)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2016039391A1 (ja) * | 2014-09-09 | 2016-03-17 | 日産化学工業株式会社 | 細胞回収に関する方法 |
JP2018526992A (ja) * | 2015-08-31 | 2018-09-20 | アイ ピース,インコーポレイテッド | 多能性幹細胞製造システム、幹細胞の誘導方法、幹細胞の浮遊培養方法、幹細胞の浮遊培養器、人工多能性幹細胞の作製方法、及び動物細胞から特定の体細胞を作製する方法 |
CN112771151A (zh) * | 2018-11-13 | 2021-05-07 | 韩国化学研究院 | 利用细胞培养用载体制备的类器官及利用其的药物毒性评价方法 |
CN113785049A (zh) * | 2019-06-10 | 2021-12-10 | 爱平世股份有限公司 | 红细胞除去装置、单核细胞回收器、细胞培养装置、细胞培养系统、细胞培养方法及单核细胞的回收方法 |
Families Citing this family (5)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US9664671B2 (en) | 2012-07-24 | 2017-05-30 | Nissan Chemical Industries, Ltd. | Culture medium composition and method of culturing cell or tissue using thereof |
US10017805B2 (en) | 2012-08-23 | 2018-07-10 | Nissan Chemical Industries, Ltd. | Enhancing ingredients for protein production from various cells |
US11708559B2 (en) | 2017-10-27 | 2023-07-25 | The Children's Hospital Of Philadelphia | Engineered red blood cells having rare antigen phenotypes |
JP7322707B2 (ja) | 2017-11-16 | 2023-08-08 | 日産化学株式会社 | ベージュおよび白色脂肪細胞への分化誘導および生産法 |
CN109439617A (zh) * | 2018-12-25 | 2019-03-08 | 成都赋智健康科技有限公司 | 一种干细胞无血清培养基及其制作方法 |
Citations (8)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2001034776A1 (en) | 1999-11-08 | 2001-05-17 | Wisconsin Alumni Research Foundation | Hematopoietic differentiation of human embryonic stem cells |
WO2006090886A1 (ja) * | 2005-02-23 | 2006-08-31 | Foundation For Biomedical Research And Innovation | 血管内皮前駆細胞分化動態解析方法 |
WO2009137629A2 (en) | 2008-05-06 | 2009-11-12 | Advanced Cell Technology, Inc. | Methods for producing enucleated erythroid cells derived from pluripotent stem cells |
JP2010525836A (ja) * | 2007-05-04 | 2010-07-29 | ホワイトヘッド・インスティテュート・フォー・バイオメディカル・リサーチ | ヒト造血幹細胞のexvivo増殖 |
WO2010098079A1 (ja) | 2009-02-24 | 2010-09-02 | 学校法人金沢医科大学 | 有核赤血球の脱核方法及び脱核誘導剤 |
JP2011024423A (ja) * | 2008-06-04 | 2011-02-10 | Masayuki Ishihara | 微粒子細胞担体及びそれを含む細胞導入システム及び細胞培養システム |
WO2011034073A1 (ja) | 2009-09-15 | 2011-03-24 | 国立大学法人東京大学 | 分化細胞の新規製造法 |
WO2014017513A1 (ja) * | 2012-07-24 | 2014-01-30 | 日産化学工業株式会社 | 培地組成物及び当該組成物を用いた細胞又は組織の培養方法 |
Family Cites Families (6)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
IT1271088B (it) * | 1994-11-24 | 1997-05-26 | Dompe Spa | Uso di fattore di crescita degli epatociti per indurre la proliferazione e differenziazione di cellule emopoietiche |
US9127254B2 (en) * | 2006-12-18 | 2015-09-08 | Ben-Gurion University Of The Negev Research And Development Authority | Scaffolding for tissue regeneration or repair |
EP2142642B1 (en) * | 2007-03-27 | 2017-08-09 | IPD-Therapeutics B.V. | Methods and means for stem cell proliferation and subsequent generation and expansion of progenitor cells, as well as production of effector cells as clinical therapeutics |
CA2767014C (en) * | 2009-07-02 | 2022-01-25 | Anthrogenesis Corporation | Method of producing erythrocytes without feeder cells |
US9044496B2 (en) * | 2010-10-08 | 2015-06-02 | Agency For Science, Technology And Research | Heparan sulphate |
WO2015111685A1 (ja) * | 2014-01-23 | 2015-07-30 | 日産化学工業株式会社 | 培地組成物の製造方法 |
-
2014
- 2014-04-16 EP EP14785390.7A patent/EP2987852A4/en not_active Withdrawn
- 2014-04-16 JP JP2015512506A patent/JP6319304B2/ja active Active
- 2014-04-16 US US14/784,843 patent/US20160060601A1/en not_active Abandoned
- 2014-04-16 WO PCT/JP2014/060829 patent/WO2014171486A1/ja active Application Filing
Patent Citations (8)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2001034776A1 (en) | 1999-11-08 | 2001-05-17 | Wisconsin Alumni Research Foundation | Hematopoietic differentiation of human embryonic stem cells |
WO2006090886A1 (ja) * | 2005-02-23 | 2006-08-31 | Foundation For Biomedical Research And Innovation | 血管内皮前駆細胞分化動態解析方法 |
JP2010525836A (ja) * | 2007-05-04 | 2010-07-29 | ホワイトヘッド・インスティテュート・フォー・バイオメディカル・リサーチ | ヒト造血幹細胞のexvivo増殖 |
WO2009137629A2 (en) | 2008-05-06 | 2009-11-12 | Advanced Cell Technology, Inc. | Methods for producing enucleated erythroid cells derived from pluripotent stem cells |
JP2011024423A (ja) * | 2008-06-04 | 2011-02-10 | Masayuki Ishihara | 微粒子細胞担体及びそれを含む細胞導入システム及び細胞培養システム |
WO2010098079A1 (ja) | 2009-02-24 | 2010-09-02 | 学校法人金沢医科大学 | 有核赤血球の脱核方法及び脱核誘導剤 |
WO2011034073A1 (ja) | 2009-09-15 | 2011-03-24 | 国立大学法人東京大学 | 分化細胞の新規製造法 |
WO2014017513A1 (ja) * | 2012-07-24 | 2014-01-30 | 日産化学工業株式会社 | 培地組成物及び当該組成物を用いた細胞又は組織の培養方法 |
Non-Patent Citations (14)
Title |
---|
AYAKO OTANI ET AL.: "Kobunshi Zairyo o Mochiita Shinki Sanjigen Baiyoho no Kaihatsu", DAI 36 KAI THE MOLECULAR BIOLOGY SOCIETY OF JAPAN NENKAI KOEN YOSHISHU, 20 November 2013 (2013-11-20), pages 2P-0990, XP008181249 * |
BLOOD, vol. 102, 2003, pages 906 - 915 |
DIAS ET AL., STEM CELLS AND DEVELOPMENT, vol. 20, 2011, pages 1639 - 1647 |
DOLZNIG ET AL., CURR. BIOL., vol. 12, 2002, pages 1076 - 1085 |
FUJIMI ET AL., INTERNATIONAL JOURNAL OF HEMATOLOGY, vol. 87, 2008, pages 339 - 350 |
GIARRATANA ET AL., BLOOD, vol. 118, 2011, pages 5071 - 5079 |
HIROYAMA ET AL., PLOS ONE, vol. 3, 2008, pages E1544 |
KISHIMOTO S. ET AL.: "Cytokine-immobilized microparticle-coated plates for culturing hematopoetic progenitor cells", JOURNAL OF CONTROLLED RELEASE, vol. 133, no. 3, 2009, pages 185 - 190, XP025893046 * |
MA ET AL., PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, vol. 105, 2008, pages 13087 - 13092 |
MOUNTFORD ET AL., BRITISH JOURNAL OF HAEMATOLOGY, vol. 149, 2010, pages 22 - 34 |
NATURE, vol. 456, 2008, pages 344 - 349 |
PLOS ONE, vol. 3, 2008, pages E1544 |
See also references of EP2987852A4 * |
TAKAYAMA ET AL., BLOOD, vol. 111, 2008, pages 5298 - 5306 |
Cited By (7)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
WO2016039391A1 (ja) * | 2014-09-09 | 2016-03-17 | 日産化学工業株式会社 | 細胞回収に関する方法 |
JPWO2016039391A1 (ja) * | 2014-09-09 | 2017-06-22 | 日産化学工業株式会社 | 細胞回収に関する方法 |
US10370696B2 (en) | 2014-09-09 | 2019-08-06 | Nissan Chemical Industries, Ltd. | Method for cell recovery |
JP2018526992A (ja) * | 2015-08-31 | 2018-09-20 | アイ ピース,インコーポレイテッド | 多能性幹細胞製造システム、幹細胞の誘導方法、幹細胞の浮遊培養方法、幹細胞の浮遊培養器、人工多能性幹細胞の作製方法、及び動物細胞から特定の体細胞を作製する方法 |
CN112771151A (zh) * | 2018-11-13 | 2021-05-07 | 韩国化学研究院 | 利用细胞培养用载体制备的类器官及利用其的药物毒性评价方法 |
CN112771151B (zh) * | 2018-11-13 | 2024-01-23 | 韩国化学研究院 | 利用细胞培养用载体制备的类器官及利用其的药物毒性评价方法 |
CN113785049A (zh) * | 2019-06-10 | 2021-12-10 | 爱平世股份有限公司 | 红细胞除去装置、单核细胞回收器、细胞培养装置、细胞培养系统、细胞培养方法及单核细胞的回收方法 |
Also Published As
Publication number | Publication date |
---|---|
JP6319304B2 (ja) | 2018-05-09 |
EP2987852A4 (en) | 2016-09-07 |
EP2987852A1 (en) | 2016-02-24 |
JPWO2014171486A1 (ja) | 2017-02-23 |
US20160060601A1 (en) | 2016-03-03 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
JP6319304B2 (ja) | 培地組成物及び当該培地組成物を用いた赤血球の製造方法 | |
JP6879517B2 (ja) | 培地組成物及び当該組成物を用いた細胞又は組織の培養方法 | |
JP7248166B2 (ja) | 培地組成物 | |
KR102282265B1 (ko) | 배양 배지 조성물의 제조 방법 | |
JP7283621B2 (ja) | 接着性細胞の浮遊培養用培地組成物 | |
JP7264198B2 (ja) | 細胞回収が容易な浮遊培養用培地組成物、及び細胞回収方法 | |
TWI814738B (zh) | 細胞保存材料 | |
JP6642439B2 (ja) | 細胞回収に関する方法 |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 14785390 Country of ref document: EP Kind code of ref document: A1 |
|
ENP | Entry into the national phase |
Ref document number: 2015512506 Country of ref document: JP Kind code of ref document: A |
|
WWE | Wipo information: entry into national phase |
Ref document number: 14784843 Country of ref document: US |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
WWE | Wipo information: entry into national phase |
Ref document number: 2014785390 Country of ref document: EP |