WO2019144605A1 - High performance method for differentiation of hpscs into mscs - Google Patents

High performance method for differentiation of hpscs into mscs Download PDF

Info

Publication number
WO2019144605A1
WO2019144605A1 PCT/CN2018/101076 CN2018101076W WO2019144605A1 WO 2019144605 A1 WO2019144605 A1 WO 2019144605A1 CN 2018101076 W CN2018101076 W CN 2018101076W WO 2019144605 A1 WO2019144605 A1 WO 2019144605A1
Authority
WO
WIPO (PCT)
Prior art keywords
differentiation
signaling pathway
mscs
activin
culture
Prior art date
Application number
PCT/CN2018/101076
Other languages
French (fr)
Chinese (zh)
Inventor
王淋立
李强
陈月花
莫健
Original Assignee
皓昇莱生物制药有限公司
王淋立
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 皓昇莱生物制药有限公司, 王淋立 filed Critical 皓昇莱生物制药有限公司
Publication of WO2019144605A1 publication Critical patent/WO2019144605A1/en

Links

Images

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
    • C12N5/0652Cells of skeletal and connective tissues; Mesenchyme
    • C12N5/0662Stem cells
    • C12N5/0668Mesenchymal stem cells from other natural sources
    • 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
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/115Basic fibroblast growth factor (bFGF, FGF-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/10Growth factors
    • C12N2501/15Transforming growth factor beta (TGF-β)
    • 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
    • C12N2501/155Bone morphogenic proteins [BMP]; Osteogenins; Osteogenic factor; Bone inducing factor
    • 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
    • C12N2501/40Regulators of development
    • C12N2501/415Wnt; Frizzeled
    • 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
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/58Adhesion molecules, e.g. ICAM, VCAM, CD18 (ligand), CD11 (ligand), CD49 (ligand)
    • 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
    • C12N2501/50Cell markers; Cell surface determinants
    • C12N2501/599Cell markers; Cell surface determinants with CD designations not provided for elsewhere
    • 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"
    • C12N2502/1352Mesenchymal stem 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
    • 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
    • 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
    • C12N2506/45Differentiation of animal cells from one lineage to another; Differentiation of pluripotent cells from artificially induced pluripotent stem 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
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins
    • C12N2533/52Fibronectin; Laminin
    • 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
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/50Proteins
    • C12N2533/54Collagen; Gelatin
    • 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
    • C12N2533/00Supports or coatings for cell culture, characterised by material
    • C12N2533/90Substrates of biological origin, e.g. extracellular matrix, decellularised tissue

Definitions

  • the invention relates to the field of cell culture, in particular to a method for efficiently differentiating hPSCs into MSCs.
  • the source of cells for clinical regenerative medicine transplantation mainly includes human pluripotent stem cells (hPSCs), namely human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs).
  • hPSCs human pluripotent stem cells
  • hESCs human embryonic stem cells
  • hiPSCs human induced pluripotent stem cells
  • the general term and adult stem cells are two major categories, including adult stem cells including hematopoietic stem cells and mesenchymal stem cells.
  • Mesenchymal stem cells derived from mesoderm were originally isolated and identified by fibroblast-like cells from mouse bone marrow extracts by Friedenstein et al. in 1966. In 1991, Caplan initially referred to these cells as mesenchymal stem cells ( Mesenchymal stem cells, MSCs).
  • MSCs multipotent mesenchymal stromal cells' or 'mesenchymal stem cells' (MSCs); and define MSCs as 1) to grow adherently; 2) can differentiate into mesoderm lineages such as osteoblasts, cartilage and adipocytes; 3) high-level expression of CD105, CD90, CD73 and other MSCs positive markers; basically do not express CD45, CD34, CD14 or CD11b, CD79 ⁇ or CD19 and HLA-DR Such as MSCs negative markers.
  • ISCT International Society for Cellular Therapy
  • MSCs differentiate into the muscle, bone, cartilage, fat, tendon and ligament of the mesoderm lineage, and even transdifferentiate into the multi-lineage differentiation and immune regulation of neurons and islet cells, and avoid tumors.
  • the 2016 Clinical Trial Database shows that ⁇ 500 clinical trials are related to MSCs, many of which have evaluated MSC vs.
  • MSCs are widely available, clinical applications are generally obtained from bone marrow, umbilical cord and adipose tissue, but the isolation of MSCs from tissues will result in low yield, presence of mixed cells and purification of cells, and a more mature table. The longer time required for the type (about 1 month) is likely to cause cell aging, allogeneic immune rejection (the source of the umbilical cord) and the like. Therefore, in terms of clinical needs, the shortage of donor sources is still a problem that must be solved. In contrast, MSCs derived from autologous or immunomatch-matched and immortalized differentiation of hiPSCs are better at addressing these clinical cell therapies than MSCs derived from fat, cord blood, bone marrow, and ESCs. .
  • hPSC-MSCs MSCs
  • hiPSCs and hESCs the most classical method for the derivation of MSCs (hPSC-MSCs) from hPSCs (hiPSCs and hESCs) is to block the ALK5,4,7 kinase in the transforming growth factor TGF- ⁇ (Transforming Growth Factor) signaling pathway by the small molecule compound SB431542.
  • TGF- ⁇ Transforming Growth Factor
  • the object of the present invention is a method for efficiently differentiating hPSCs into MSCs.
  • a method for efficiently inducing differentiation of hPSCs into MSCs the steps of which are:
  • the new culture plate is coated with a gelatin-based extracellular matrix for cell screening.
  • the cells obtained by cell screening are cells having a spindle-type morphological feature, positive surface markers CD90, CD73, CD105, negative for CD34, CD45, CD14 or CD11b, CD79 ⁇ or CD19, and HLA-DR.
  • the gelatin-based extracellular matrix is gelatin for cell culture or type I, III, IV, V-type collagen and elastin, and laminin.
  • culture is carried out using a differentiation medium containing BMP-SMAD1/5/8 signaling pathway activator, TGF- ⁇ 1/Activin/Nodal-SMAD2/3 signaling pathway activator, Wnt activator, PI3K inhibitor.
  • the time is 12 to 72 hours.
  • step 1) at least one of BMP-SMAD1/5/8 signaling pathway activators BMP2, BMP4, BMP7.
  • the TGF- ⁇ 1/Activin/Nodal-SMAD2/3 signaling pathway activator is at least one of ActivinA, Activin B, TGF- ⁇ 1, and Nodal.
  • the Wnt activator is at least one of CHIR99021, BIO, WNT-3a, and R-spondin-2.
  • the PI3K inhibitor is at least one of TG100713, PIK90, and PI-103.
  • the TGF- ⁇ 1/Activin/Nodal-SMAD2/3 signaling pathway inhibitor is at least one of SB431542, SB505124, A8301, and RepSox.
  • step 3 the culture is continued for 2 to 30 days.
  • step 2 culture with a differentiation medium containing BMP-SMAD1/5/8 signaling pathway activator, TGF- ⁇ 1/Activin/Nodal-SMAD2/3 signaling pathway activator, Wnt inhibitor .
  • step 2) the culture is continued for 2 to 10 days.
  • the Wnt inhibitor is at least one of Wnt-C59 and XAV-939.
  • the method of continuing the culture in step 4) is: culturing with a medium containing mesenchymal stem cells.
  • At least one medium contains basic fibroblast growth factor bFGF.
  • the obtained MSCs are obtained by inducing hPSCs to differentiate into MSCs through the middle stage or the mesoderm stage.
  • An MSCs cell prepared by the above differentiation method An MSCs cell prepared by the above differentiation method.
  • the beneficial effects of the present invention are as follows: 1) The present invention is the first method in the world to establish intact hPSCs in vitro to differentiate into MSCs through the middle stage or the lateral mesoderm stage. To provide a reliable technical resource for studying the differences between strains derived from different cells of human pluripotent stem cells and the differences between MSCs in vitro and in vivo.
  • the method for direct differentiation of hPSC-MSCs of the present invention has the advantages of simple steps, simple operation and high reproducibility, and phenotypic mature and high quality MSCs derived from mesodermal lineage can be obtained in only 12 days.
  • Figure 1 shows undifferentiated cells of hiPSCs.
  • Figure 2 shows undifferentiated cells of hESCs.
  • Figure 3 shows the differentiation of MSCs precursor cells obtained from the middle segment.
  • Figure 4 shows MSCs precursor cells obtained by differentiation through the mesoderm.
  • Figure 5 is a MSCs precursor cell obtained by adding bFGF to differentiate into a middle segment.
  • Figure 6 shows MSCs cells differentiated from hiPSCs into phenotypic and spindle-shaped forms.
  • Figure 7 shows MSCs cells differentiated from hESCs into phenotypic and spindle-shaped forms.
  • Figure 8 shows no cells found to adhere to growth.
  • hPSCs cells (Fig. 1 and Fig. 2) to matrigel (Matrigel, MG; one of extracellular matrices), coated with a 30-well adherent plate for 30 min, and differentiated with 0.5 ml of pluripotent stem cells.
  • the medium was cultured at 37 ° C, 5% CO 2 for 24 hours (Day 0), and changed to 0.5 ml of pluripotent stem cell differentiation medium containing no TGF- ⁇ 1.
  • the medium also contained 40 ng/ml BMP4 and 30 ng/ml Activin A, 6 ⁇ M CHIR99021, 100 nM PIK90.
  • BMP4 is bone morphogenetic protein 4
  • CHIR99021 is 6-[2-[4-(2,4-dichlorophenyl)-5-(4-methyl-1H-imidazol-2-yl)pyrimidine-2 -ylamino]ethylamino]pyridine-3-carbonitrile (CAS: 252917-06-9)
  • Activin A is activin A
  • PIK90 is N-(2,3-dihydro-7,8-dimethoxy Imidazo[1,2-c]quinazolin-5-yl)-3-pyridinecarboxamide (CAS: 677338-12-4). Cultivate for 24 hours.
  • SB431542 is 4-[4-(1,3-benzothiazol-5-yl)-5-(2-pyridyl)-1H-imidazol-2-yl]-benzamide hydrate (CAS: 301836-41 -9).
  • the expressions of pluripotency markers Oct3/4, Nanog, Sox2, etc. gradually decreased (Table 2), BRACHYURY decreased to no expression, and the mesodermal markers Nkx2.5, HAND1, FOXF1, etc. continued to be highly expressed. It can be concluded that the cells are in the stage of the mesoderm at this time (Table 1).
  • the cell confluence can reach ⁇ 90% by GT stage, and the cells are digested with 2ml of 0.05% Trypsin-EDTA for 3min, and then added with 4ml DMEM/F12 (containing 10% FBS). The 15ml centrifuge tube is recovered and centrifuged at 200g for 5min.
  • the cells can express MSC positive Markers such as CD105, CD90 and CD73 at high levels; they do not express MSC-negative Markers such as CD45, CD34, CD14 or CD11b, CD79 ⁇ , CD19 and HLA-DR.
  • MSC positive Markers such as CD105, CD90 and CD73 at high levels; they do not express MSC-negative Markers such as CD45, CD34, CD14 or CD11b, CD79 ⁇ , CD19 and HLA-DR.
  • the cells have a typical spindle-shaped morphology, and the cells can be identified as MSCs cells (Fig. 6 and Fig. 7).
  • BMP4 was replaced with BMP2 and BMP7;
  • Activin A was replaced with Activin B, TGF- ⁇ 1, Nodal, and CHIR99021 was used with BIO (CAS: 667463-62-9), WNT-3a (WNT signaling protein 3a), R-spondin- 2 (R-spondin signal protein 2) substitution;
  • SB431542 was replaced with SB505124 (CAS: 694433-59-5), A8301 (CAS: 909910-43-6), RepSox (CAS: 446859-33-2) PI-103;
  • a similar effect can also be produced by replacing PIK90 with TG100713 (CAS: 925705-73-3), PI-103 (CAS: 371935-74-9).
  • step 2) containing 20 ng/ml BMP4 (BMP-SMAD1/5/8 signaling pathway activator), 3 uM/ml SB505124 (TGF- ⁇ 1/Activin/Nodal-
  • the differentiation medium of SMAD2/3 signaling pathway activator and 1uM/ml Wnt-C59 (Wnt inhibitor) was further cultured for 24 hours, at which time the mesodermal markers Nkx2.5, HAND1, FOXF1 and the like were highly expressed (Table 4).
  • SB505124 is 2-[4-(1,3-benzobisazol-5-yl)-2-(1,1-dimethylethyl)-1H-imidazol-5-yl]-6-methylpyridine (CAS: 694433-59-5), Wnt-C59 is 4-(2-methyl-4-pyridyl)-N-[4-(3-pyridyl)phenyl]phenylacetamide (CAS: 1243243- 89-1).
  • step 4 typical spindle-shaped MSCs precursor cells are obtained, as shown in FIG. The remaining steps were unchanged. Finally, after FACS identification, the cells could express MSC-positive Markers such as CD105, CD90 and CD73 at high levels; MSC-negative Markers such as CD45, CD34, CD14 or CD11b, CD79 ⁇ or CD19 and HLA-DR were not expressed at all. 5 is shown.
  • MSC-positive Markers such as CD105, CD90 and CD73 at high levels
  • MSC-negative Markers such as CD45, CD34, CD14 or CD11b, CD79 ⁇ or CD19 and HLA-DR were not expressed at all. 5 is shown.
  • Step 1) Starting with the addition of basic fibroblast growth factor (bFGF) on the basis of the differentiation medium, at step 4) More and more typical spindle-shaped morphological MSCs precursor cells will be obtained, as shown in Figure 5. The remaining steps were unchanged. Finally, after FACS identification, the cells could express MSC-positive Markers such as CD105, CD90, CD73, etc.; MSC-negative Markers such as CD45, CD34, CD14, CD11b, CD79 ⁇ , CD19 and HLA-DR were not expressed at all. 6 is shown.
  • MSC-positive Markers such as CD105, CD90, CD73, etc.
  • MSC-negative Markers such as CD45, CD34, CD14, CD11b, CD79 ⁇ , CD19 and HLA-DR were not expressed at all. 6 is shown.
  • the undifferentiated hPSCs cells were transferred to a 24-well adherent culture plate coated with extracellular matrix (MG) for 30 min, and cultured with 0.5 ml of pluripotent stem cell differentiation medium at 37 ° C, 5% CO 2 for 24 hours ( Day 0), 0.5 ml of pluripotent stem cell differentiation medium containing no TGF- ⁇ 1, containing 30 ng/ml Activin A, 6 ⁇ M CHIR99021, and 100 nM PIK90 for 24 hours.
  • the expression levels of the protocolonary markers such as MIXL1, BRACHYURY, and TBX6 were detected, and it was found that high expression was obtained, and the cells were in the pre-segment phase (Table 7).

Abstract

Disclosed in the present invention is a high performance method for inducing differentiation of hPSCs into MSCs, comprising the steps of: 1) transferring undifferentiated hPSC cell lines onto a culture plate coated with an extracellular matrix for cultivation; and then cultivating same by using a differentiation culture medium containing BMP-SMAD1/5/8 signaling pathway activator, TGF-β1/Activin/Nodal-SMAD2/3 signaling pathway activator, Wnt activator and PI3K inhibitor; 2) removing the old culture medium, and continuing to cultivate by using a differentiation culture medium containing TGF-β1/Activin/Nodal-SMAD2/3 signaling pathway inhibitor; 3) digesting and transferring the cells onto a new cultivation plate, and continuing to cultivate by using a differentiation culture medium containing TGF-β1/Activin/Nodal-SMAD2/3 signaling pathway inhibitor; and 4) digesting the cells and transferring same onto an adherent culture plate for continued cultivation, so as to obtain MSCs. The present invention establishes in vitro a complete method for the directed differentiation of hPSCs into MSCs through the middle primitive streak stage or the lateral mesoderm stage. Compared with the conventional methods, the technical steps of the present method are simple, easy to operate, and highly reproducible, and MSCs having mature phenotype and high quality can be obtained within only 12 days.

Description

一种高效的hPSCs向MSCs分化的方法A method for efficiently differentiating hPSCs into MSCs 技术领域Technical field
本发明涉及细胞培养领域,特别涉及一种高效的hPSCs向MSCs分化的方法。The invention relates to the field of cell culture, in particular to a method for efficiently differentiating hPSCs into MSCs.
背景技术Background technique
作为临床再生医学移植的细胞来源主要包括人多能干细胞(human pluripotent stem cells,hPSCs)即人胚胎干细胞(human embryonic stem cells,hESCs)和人诱导多能干细胞(human induced pluripotent stem cells,hiPSCs)等的统称以及成体干细胞这两大类,其中成体干细胞包括造血干细胞,间充质干细胞等。来源于中胚层的间充质干细胞最初是由1966年Friedenstein等从老鼠骨髓抽出物中发现存在成纤维细胞样的细胞而且分离鉴定出来的,1991年Caplan最初将这些细胞称为间充质干细胞(mesenchymal stem cells,MSCs)。2006年国际细胞治疗协会(International Society for Cellular Therapy,ISCT)提议把这些细胞正式命名为‘multipotent mesenchymal stromal cells’或‘mesenchymal stem cells’(MSCs);并且把MSCs定义为1)可以贴壁生长;2)能向成骨、软骨和脂肪细胞等中胚层谱系分化;3)高水平表达CD105、CD90、CD73等MSCs阳性标志;基本不表达CD45、CD34、CD14或CD11b、CD79α或CD19以及HLA-DR等MSCs阴性标志。MSCs除了自我更新,分化成中胚层谱系的肌肉、骨骼、软骨、脂肪、肌腱和韧带,甚至还可以跨胚层转分化为神经、胰岛细胞等多谱系分化和免疫调节的能力,且避免发生肿瘤。根据美国NIH临床试验注册中心ClinicalTrials.gov,2016年临床试验数据库(拥有201个国家的约25万种试验记录)显示~500个临床试验是与MSCs有关,其中有许多临床试验评估了MSC对多种疾病的影响,包括骨关节炎,创面愈合,退行性疾病,自身免疫性疾病等。由此可见MSCs在细胞再生医疗的重要位置。虽然MSCs的来源广泛获取方法众多,临床应用一般是从骨髓、脐带及脂肪组织中分离而获得,但从组织中分离MSCs都会面临低收率、存在混杂细胞以及纯化细胞、获得一个更成熟的表型所需时间较长(约1个多月)容易造成细胞衰老,同种异体的免疫排斥(脐带来源)等问题。所以从临床需求而言,其供体来源短缺依然是必须解决的问题。相比之下,从自体来源或免疫配型匹配并且无限增殖的hiPSCs定向分化衍生而来的MSCs,比从脂肪、脐血、骨髓以及ESCs来源的MSCs能更好地解决这些临床细胞治疗的问题。The source of cells for clinical regenerative medicine transplantation mainly includes human pluripotent stem cells (hPSCs), namely human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs). The general term and adult stem cells are two major categories, including adult stem cells including hematopoietic stem cells and mesenchymal stem cells. Mesenchymal stem cells derived from mesoderm were originally isolated and identified by fibroblast-like cells from mouse bone marrow extracts by Friedenstein et al. in 1966. In 1991, Caplan initially referred to these cells as mesenchymal stem cells ( Mesenchymal stem cells, MSCs). In 2006, the International Society for Cellular Therapy (ISCT) proposed to officially name these cells as 'multipotent mesenchymal stromal cells' or 'mesenchymal stem cells' (MSCs); and define MSCs as 1) to grow adherently; 2) can differentiate into mesoderm lineages such as osteoblasts, cartilage and adipocytes; 3) high-level expression of CD105, CD90, CD73 and other MSCs positive markers; basically do not express CD45, CD34, CD14 or CD11b, CD79α or CD19 and HLA-DR Such as MSCs negative markers. In addition to self-renewal, MSCs differentiate into the muscle, bone, cartilage, fat, tendon and ligament of the mesoderm lineage, and even transdifferentiate into the multi-lineage differentiation and immune regulation of neurons and islet cells, and avoid tumors. According to the National NIH Clinical Trial Registry ClinicalTrials.gov, the 2016 Clinical Trial Database (with approximately 250,000 trial records in 201 countries) shows that ~500 clinical trials are related to MSCs, many of which have evaluated MSC vs. The effects of various diseases, including osteoarthritis, wound healing, degenerative diseases, autoimmune diseases, etc. This shows that MSCs are important in cell regenerative medicine. Although the sources of MSCs are widely available, clinical applications are generally obtained from bone marrow, umbilical cord and adipose tissue, but the isolation of MSCs from tissues will result in low yield, presence of mixed cells and purification of cells, and a more mature table. The longer time required for the type (about 1 month) is likely to cause cell aging, allogeneic immune rejection (the source of the umbilical cord) and the like. Therefore, in terms of clinical needs, the shortage of donor sources is still a problem that must be solved. In contrast, MSCs derived from autologous or immunomatch-matched and immortalized differentiation of hiPSCs are better at addressing these clinical cell therapies than MSCs derived from fat, cord blood, bone marrow, and ESCs. .
目前,由hPSCs(hiPSCs和hESCs)分化衍生出MSCs(hPSC-MSCs)最经典的方法是通过小分子化合物SB431542阻碍转化生长因子TGF-β(Transforming Growth Factor)信号通道中ALK5,4,7激酶,达到抑制SMAD2/3信号的作用。从而降解hiPSCs或hESCs的多 能性,诱发上皮向间质转化EMT(Epithelial-to-MesenchymalTransition)的发生,促进hPSC-MSCs,从而获得表型成熟、高质量的hPSC-MSCs。但难免会导致衍生出不需要的细胞谱系。然而干细胞分化到所需的细胞谱系是相当复杂而难以掌控的一个过程,尤其是人胚胎生物学(限于伦理道德),所以在体外要高度模拟、遵循人胚胎发育期间的生物学原理来研究干细胞定向分化是一份十分艰巨而又非常具有挑战性的科研工作。因此,到目前为止还没有出现过将hPSCs定向分化成高度模拟胚胎期MSCs完整的方案方法。At present, the most classical method for the derivation of MSCs (hPSC-MSCs) from hPSCs (hiPSCs and hESCs) is to block the ALK5,4,7 kinase in the transforming growth factor TGF-β (Transforming Growth Factor) signaling pathway by the small molecule compound SB431542. The effect of suppressing the SMAD2/3 signal is achieved. Thus, the pluripotency of hiPSCs or hESCs is degraded, EMT (Epithelial-to-Mesenchymal Transition) is induced, and hPSC-MSCs are promoted, thereby obtaining phenotypic mature and high-quality hPSC-MSCs. But it will inevitably lead to the derivation of unwanted cell lineages. However, the differentiation of stem cells into the desired cell lineage is a complex and difficult process to control, especially human embryo biology (limited to ethics), so it is highly simulated in vitro, following the biological principles of human embryo development to study stem cells. Directional differentiation is a very difficult and challenging research work. Therefore, there has not been a complete method for the differentiation of hPSCs into highly mimetic embryonic MSCs.
发明内容Summary of the invention
本发明的目的在于一种高效的hPSCs向MSCs分化的方法。The object of the present invention is a method for efficiently differentiating hPSCs into MSCs.
本发明所采取的技术方案是:The technical solution adopted by the present invention is:
一种高效的hPSCs向MSCs分化诱导的方法,其步骤是:A method for efficiently inducing differentiation of hPSCs into MSCs, the steps of which are:
1)将未分化hPSCs细胞株转移到细胞外基质包被的培养板上培养后;换用含有BMP-SMAD1/5/8信号通路激活剂、TGF-β1/Activin/Nodal-SMAD2/3信号通路激活剂、Wnt激活剂、PI3K抑制剂的分化培养基培养;2)去除旧培养基,换含有TGF-β1/Activin/Nodal-SMAD2/3信号通路抑制剂分化培养基继续培养;3)将细胞消化并转移到新的培养板上,换含有TGF-β1/Activin/Nodal-SMAD2/3信号通路抑制剂分化培养基继续培养;4)将细胞消化并转移到贴壁用培养板上继续培养,获得MSCs。1) Transfer undifferentiated hPSCs cell line to extracellular matrix-coated culture plates; switch to BMP-SMAD1/5/8 signaling pathway activator, TGF-β1/Activin/Nodal-SMAD2/3 signaling pathway Activator, Wnt activator, PI3K inhibitor differentiation medium culture; 2) Remove old medium, continue to culture with TGF-β1/Activin/Nodal-SMAD2/3 signaling pathway inhibitor differentiation medium; 3) Cell Digest and transfer to a new culture plate, and continue to culture with TGF-β1/Activin/Nodal-SMAD2/3 signaling pathway inhibitor differentiation medium; 4) Digest the cells and transfer them to the adherent culture plate for further cultivation. Obtain MSCs.
优选的,步骤3)中,新的培养板是用明胶类细胞外基质包被的,进行细胞筛选。Preferably, in step 3), the new culture plate is coated with a gelatin-based extracellular matrix for cell screening.
优选的,细胞筛选所得细胞为具有梭型形态特征,表面标志CD90、CD73、CD105为阳性,CD34、CD45、CD14或CD11b、CD79α或CD19以及HLA-DR为阴性的细胞。Preferably, the cells obtained by cell screening are cells having a spindle-type morphological feature, positive surface markers CD90, CD73, CD105, negative for CD34, CD45, CD14 or CD11b, CD79α or CD19, and HLA-DR.
优选的,明胶类细胞外基质为细胞培养用明胶或I型、III型、IV型、V型胶原蛋白和弹性蛋白以及层粘连蛋白。Preferably, the gelatin-based extracellular matrix is gelatin for cell culture or type I, III, IV, V-type collagen and elastin, and laminin.
优选的,步骤1)中,用含有BMP-SMAD1/5/8信号通路激活剂、TGF-β1/Activin/Nodal-SMAD2/3信号通路激活剂、Wnt激活剂、PI3K抑制剂的分化培养基培养时间为12~72h。Preferably, in step 1), culture is carried out using a differentiation medium containing BMP-SMAD1/5/8 signaling pathway activator, TGF-β1/Activin/Nodal-SMAD2/3 signaling pathway activator, Wnt activator, PI3K inhibitor. The time is 12 to 72 hours.
优选的,步骤1)中,BMP-SMAD1/5/8信号通路激活剂BMP2、BMP4、BMP7中的至少一种。Preferably, in step 1), at least one of BMP-SMAD1/5/8 signaling pathway activators BMP2, BMP4, BMP7.
优选的,步骤1)中,TGF-β1/Activin/Nodal-SMAD2/3信号通路激活剂为ActivinA、Activin B、TGF-β1、Nodal中的至少一种。Preferably, in step 1), the TGF-β1/Activin/Nodal-SMAD2/3 signaling pathway activator is at least one of ActivinA, Activin B, TGF-β1, and Nodal.
优选的,步骤1)中,Wnt激活剂为CHIR99021,BIO、WNT-3a、R-spondin-2中的至少一种。Preferably, in step 1), the Wnt activator is at least one of CHIR99021, BIO, WNT-3a, and R-spondin-2.
优选的,步骤1)中,PI3K抑制剂为TG100713、PIK90、PI-103中的至少一种。Preferably, in step 1), the PI3K inhibitor is at least one of TG100713, PIK90, and PI-103.
优选的,TGF-β1/Activin/Nodal-SMAD2/3信号通路抑制剂为SB431542、SB505124、A8301、 RepSox中的至少一种。Preferably, the TGF-β1/Activin/Nodal-SMAD2/3 signaling pathway inhibitor is at least one of SB431542, SB505124, A8301, and RepSox.
优选的,步骤3)中,继续培养的时间为2~30天。Preferably, in step 3), the culture is continued for 2 to 30 days.
优选的,在步骤2)继续培养之前,先用含有BMP-SMAD1/5/8信号通路激活剂、TGF-β1/Activin/Nodal-SMAD2/3信号通路激活剂、Wnt抑制剂的分化培养基培养。Preferably, prior to continuing the culture in step 2), culture with a differentiation medium containing BMP-SMAD1/5/8 signaling pathway activator, TGF-β1/Activin/Nodal-SMAD2/3 signaling pathway activator, Wnt inhibitor .
优选的,在步骤2)中,继续培养的时间为2~10天。Preferably, in step 2), the culture is continued for 2 to 10 days.
优选的,Wnt抑制剂为Wnt-C59、XAV-939中的至少一种。Preferably, the Wnt inhibitor is at least one of Wnt-C59 and XAV-939.
优选的,步骤4)中继续培养的方法是:用含有间充质干细胞培养基培养。Preferably, the method of continuing the culture in step 4) is: culturing with a medium containing mesenchymal stem cells.
优选的,在步骤1)至步骤4)中,至少一种培养基中含有碱性成纤维细胞生长因子bFGF。Preferably, in step 1) to step 4), at least one medium contains basic fibroblast growth factor bFGF.
优选的,获得的MSCs是使用诱导hPSCs通过中段原条阶段或侧中胚层阶段向MSCs定向分化的方法来获得的。Preferably, the obtained MSCs are obtained by inducing hPSCs to differentiate into MSCs through the middle stage or the mesoderm stage.
一种MSCs细胞,该细胞由上述分化方法制备而成。An MSCs cell prepared by the above differentiation method.
本发明的有益效果是:1)本发明是世界上首次在体外建立了完整的hPSCs通过中段原条阶段或侧中胚层阶段向MSC定向分化方案方法。为研究来源于人多能干细胞不同细胞的株间差,以及体内外MSCs的差异性提供了可靠的技术资源。The beneficial effects of the present invention are as follows: 1) The present invention is the first method in the world to establish intact hPSCs in vitro to differentiate into MSCs through the middle stage or the lateral mesoderm stage. To provide a reliable technical resource for studying the differences between strains derived from different cells of human pluripotent stem cells and the differences between MSCs in vitro and in vivo.
2)本发明hPSC-MSCs定向分化方案方法,步骤精简,操作简易,再现性高,仅仅用12天便可以获得表型成熟、高质量的来源于中胚层谱系的MSCs。2) The method for direct differentiation of hPSC-MSCs of the present invention has the advantages of simple steps, simple operation and high reproducibility, and phenotypic mature and high quality MSCs derived from mesodermal lineage can be obtained in only 12 days.
附图说明DRAWINGS
图1为hiPSCs未分化的细胞。Figure 1 shows undifferentiated cells of hiPSCs.
图2为hESCs未分化的细胞。Figure 2 shows undifferentiated cells of hESCs.
图3为分化经中段原条获得的MSCs前体细胞。Figure 3 shows the differentiation of MSCs precursor cells obtained from the middle segment.
图4为分化经侧中胚层获得的MSCs前体细胞。Figure 4 shows MSCs precursor cells obtained by differentiation through the mesoderm.
图5为添加bFGF分化经中段原条获得的MSCs前体细胞。Figure 5 is a MSCs precursor cell obtained by adding bFGF to differentiate into a middle segment.
图6为从hiPSCs分化成表型成、梭型形态的MSCs细胞。Figure 6 shows MSCs cells differentiated from hiPSCs into phenotypic and spindle-shaped forms.
图7为从hESCs分化成表型成、梭型形态的MSCs细胞。Figure 7 shows MSCs cells differentiated from hESCs into phenotypic and spindle-shaped forms.
图8为没有发现可以粘附生长的细胞。Figure 8 shows no cells found to adhere to growth.
具体实施方式Detailed ways
hPSCs向MSCs分化的培养方法:Culture method for differentiation of hPSCs into MSCs:
实施例1:Example 1:
1)将未分化hPSCs细胞(图1和图2)转移到基质胶(Matrigel,MG;细胞外基质的一种)包被30min的24孔贴壁用培养板上,用0.5ml多能干细胞分化培养基,37℃、5%CO 2培养24小时后(Day 0),改为0.5ml不含TGF-β1的多能干细胞分化培养基培养。另外,培 养基还含有40ng/ml BMP4和30ng/ml Activin A,6μM CHIR99021,100nM PIK90。其中,BMP4为骨形态发生蛋白4,CHIR99021为6-[2-[4-(2,4-二氯苯基)-5-(4-甲基-1H-咪唑-2-基)嘧啶-2-基氨基]乙基氨基]吡啶-3-甲腈(CAS:252917-06-9),Activin A为激活素A,PIK90为N-(2,3-二氢-7,8-二甲氧基咪唑并[1,2-C]喹唑啉-5-基)-3-吡啶甲酰胺(CAS:677338-12-4)。培养24小时。检测MIXL1、BRACHYURY、HAND1等中段原条标记物的表达量。发现三个中段原条标记物均获得高表达,可以断定此时细胞处于中段原条的阶段(表1)。 1) Transfer undifferentiated hPSCs cells (Fig. 1 and Fig. 2) to matrigel (Matrigel, MG; one of extracellular matrices), coated with a 30-well adherent plate for 30 min, and differentiated with 0.5 ml of pluripotent stem cells. The medium was cultured at 37 ° C, 5% CO 2 for 24 hours (Day 0), and changed to 0.5 ml of pluripotent stem cell differentiation medium containing no TGF-β1. In addition, the medium also contained 40 ng/ml BMP4 and 30 ng/ml Activin A, 6 μM CHIR99021, 100 nM PIK90. Among them, BMP4 is bone morphogenetic protein 4, and CHIR99021 is 6-[2-[4-(2,4-dichlorophenyl)-5-(4-methyl-1H-imidazol-2-yl)pyrimidine-2 -ylamino]ethylamino]pyridine-3-carbonitrile (CAS: 252917-06-9), Activin A is activin A, PIK90 is N-(2,3-dihydro-7,8-dimethoxy Imidazo[1,2-c]quinazolin-5-yl)-3-pyridinecarboxamide (CAS: 677338-12-4). Cultivate for 24 hours. The expression levels of the raw strip markers in the middle segments such as MIXL1, BRACHYURY, and HAND1 were detected. It was found that all the three middle-section original strip markers were highly expressed, and it can be concluded that the cells were in the middle stage of the original strip (Table 1).
2)去掉旧液,改为含有10uM/ml SB431542的0.5ml不含TGF-β1的多能干细胞分化培养基连续培养5天,隔天换液,期间不传代。SB431542是4-[4-(1,3-苯并二唑-5-基)-5-(2-吡啶基)-1H-咪唑-2-基]-苯酰胺水合物(CAS:301836-41-9)。在经SB431542处理后,多能性标记Oct3/4、Nanog、Sox2等表达逐渐衰减(表2),BRACHYURY急减至基本不表达,侧中胚层标记物Nkx2.5、HAND1、FOXF1等持续高表达,可以断定此时细胞处于侧中胚层的阶段(表1)。2) The old solution was removed, and 0.5 ml of the pluripotent stem cell differentiation medium containing no TGF-β1 containing 10 uM/ml SB431542 was continuously cultured for 5 days, and the solution was changed every other day without passage. SB431542 is 4-[4-(1,3-benzothiazol-5-yl)-5-(2-pyridyl)-1H-imidazol-2-yl]-benzamide hydrate (CAS: 301836-41 -9). After treatment with SB431542, the expressions of pluripotency markers Oct3/4, Nanog, Sox2, etc. gradually decreased (Table 2), BRACHYURY decreased to no expression, and the mesodermal markers Nkx2.5, HAND1, FOXF1, etc. continued to be highly expressed. It can be concluded that the cells are in the stage of the mesoderm at this time (Table 1).
表1Table 1
Figure PCTCN2018101076-appb-000001
Figure PCTCN2018101076-appb-000001
Figure PCTCN2018101076-appb-000002
Figure PCTCN2018101076-appb-000002
表2Table 2
Figure PCTCN2018101076-appb-000003
Figure PCTCN2018101076-appb-000003
Figure PCTCN2018101076-appb-000004
Figure PCTCN2018101076-appb-000004
3)去掉旧液,加2ml 0.05%Trypsin-EDTA将SB处理5天后的细胞消化5min,加4ml DMEM/F12(含10%FBS)终止,回收到15ml离心管,200g、5min离心。3) Remove the old solution, add 2 ml of 0.05% Trypsin-EDTA, digest the cells after 5 days of SB treatment for 5 min, add 4 ml of DMEM/F12 (containing 10% FBS) to terminate, collect 15 ml centrifuge tubes, and centrifuge at 200 g for 5 min.
4)除掉上清,加含有10uM/ml SB431542的1ml不含TGF-β1的多能干细胞分化培养基轻轻将细胞悬浮,移到经明胶(Gelatin,GT)包被30min、加含有10uM/ml SB431542的9ml不含TGF-β1的多能干细胞分化培养基的P100贴壁用培养板上,培养48h,得到具有贴壁性能、梭型形态特征的细胞(图3)。37℃、5%CO 2继续培养3天,隔日换液,此时即为MSCs前体细胞阶段。明胶为细胞培养用明胶或I型、III型、IV型、V型胶原蛋白和弹性蛋白以及层粘连蛋白。 4) Remove the supernatant, add 1 ml of pluripotent cell differentiation medium containing 10 uM/ml SB431542 without TGF-β1, gently suspend the cells, transfer to gelatin (GT) for 30 min, add 10 uM/ MLC SB431542 was cultured on a P100 adherent plate of 9 ml of pluripotent stem cell differentiation medium containing no TGF-β1 for 48 hours to obtain cells having adherent properties and spindle-shaped morphology (Fig. 3). The culture was continued for 3 days at 37 ° C, 5% CO 2 , and the solution was changed every other day, which is the precursor cell stage of MSCs. Gelatin is gelatin for cell culture or type I, III, IV, V collagen and elastin, and laminin.
5)经GT阶段细胞汇合度可以到达~90%,加2ml 0.05%Trypsin-EDTA将细胞消化3min,加4ml DMEM/F12(含10%FBS)终止,回收到15ml离心管,200g、5min离心。5) The cell confluence can reach ~90% by GT stage, and the cells are digested with 2ml of 0.05% Trypsin-EDTA for 3min, and then added with 4ml DMEM/F12 (containing 10% FBS). The 15ml centrifuge tube is recovered and centrifuged at 200g for 5min.
6)除掉上清,加1ml含有10uM/ml SB431542且不含TGF-β1抑制剂的多能干细胞分化培养基轻轻将细胞悬浮,移到(1∶5传)没有任何包被的加有9ml含有10uM/ml SB431542且不含TGF-β1抑制剂的多能干细胞分化培养基的P100贴壁用培养板上培养24小时后换成10%FBS的α-MEM,37℃、5%CO 2培养3天,流式鉴定结果如表3所示。经过FACS鉴定,细胞能高水平表达CD105、CD90、CD73等MSC阳性Markers;基本不表达CD45、CD34、CD14 or CD11b、CD79α、CD19以及HLA-DR等MSC阴性Markers。另外,细胞是具有典型的梭型形态,可以认定细胞为MSCs细胞(图6和图7)。 6) Remove the supernatant, add 1 ml of pluripotent stem cell differentiation medium containing 10 uM/ml SB431542 and no TGF-β1 inhibitor, gently suspend the cells, and transfer to (1:5) without any coating. 9 ml of P100 adherent medium containing 10 uM/ml SB431542 and no TGF-β1 inhibitor was cultured on a culture plate for 24 hours and then replaced with 10% FBS α-MEM, 37 ° C, 5% CO 2 The culture was carried out for 3 days, and the results of flow identification were shown in Table 3. After FACS identification, the cells can express MSC positive Markers such as CD105, CD90 and CD73 at high levels; they do not express MSC-negative Markers such as CD45, CD34, CD14 or CD11b, CD79α, CD19 and HLA-DR. In addition, the cells have a typical spindle-shaped morphology, and the cells can be identified as MSCs cells (Fig. 6 and Fig. 7).
表3table 3
Figure PCTCN2018101076-appb-000005
Figure PCTCN2018101076-appb-000005
另外,将BMP4用BMP2、BMP7替换;Activin A用Activin B、TGF-β1、Nodal替换,CHIR99021用BIO(CAS:667463-62-9)、WNT-3a(WNT信号蛋白3a)、R-spondin-2(R-spondin信号蛋白2)替换;SB431542用SB505124(CAS:694433-59-5)、A8301(CAS:909910-43-6)、RepSox(CAS:446859-33-2)PI-103替换;PIK90用TG100713(CAS:925705-73-3)、PI-103(CAS:371935-74-9)替换也可以产生类似的效果。In addition, BMP4 was replaced with BMP2 and BMP7; Activin A was replaced with Activin B, TGF-β1, Nodal, and CHIR99021 was used with BIO (CAS: 667463-62-9), WNT-3a (WNT signaling protein 3a), R-spondin- 2 (R-spondin signal protein 2) substitution; SB431542 was replaced with SB505124 (CAS: 694433-59-5), A8301 (CAS: 909910-43-6), RepSox (CAS: 446859-33-2) PI-103; A similar effect can also be produced by replacing PIK90 with TG100713 (CAS: 925705-73-3), PI-103 (CAS: 371935-74-9).
实施例2:Example 2:
按照实施例1的方法进行分化培养,做如下改进:步骤2)换含有20ng/ml BMP4(BMP-SMAD1/5/8信号通路激活剂)、3uM/ml SB505124(TGF-β1/Activin/Nodal-SMAD2/3信号通路激活剂)、1uM/ml Wnt-C59(Wnt抑制剂)的分化培养基继续培养24小时,此时侧中胚层标记物Nkx2.5、HAND1、FOXF1等高表达(表4),可以断定此时细胞处于侧中胚层的阶段,然后换含有10uM/ml SB431542的0.5ml不含TGF-β1的多能干细胞分化培养基继续培养,隔天换液,期间不传代。SB505124是2-[4-(1,3-苯并二唑-5-基)-2-(1,1-二甲基乙基)-1H-咪唑-5-基]-6-甲基吡啶(CAS:694433-59-5),Wnt-C59是4-(2-甲基-4-吡啶基)-N-[4-(3-吡啶基)苯基]苯乙酰胺(CAS:1243243-89-1)。在步骤4)的时候会获得典型的梭型形态特征的MSCs前体细胞,如图4所示。其余步骤不变,最后经过FACS鉴定,细胞能高水平表达CD105、CD90、CD73等MSC阳性Markers;基本不表达CD45、CD34、CD14 or CD11b、CD79α or CD19以及HLA-DR等MSC阴性Markers,如表5所示。The differentiation culture was carried out in accordance with the method of Example 1, and the following improvements were made: step 2) containing 20 ng/ml BMP4 (BMP-SMAD1/5/8 signaling pathway activator), 3 uM/ml SB505124 (TGF-β1/Activin/Nodal- The differentiation medium of SMAD2/3 signaling pathway activator and 1uM/ml Wnt-C59 (Wnt inhibitor) was further cultured for 24 hours, at which time the mesodermal markers Nkx2.5, HAND1, FOXF1 and the like were highly expressed (Table 4). It can be concluded that the cells are in the stage of mesoderm at this time, and then cultured in 0.5 ml of pluripotent stem cell differentiation medium containing no TGF-β1 containing 10 uM/ml SB431542, and the cells were changed every other day without passage. SB505124 is 2-[4-(1,3-benzobisazol-5-yl)-2-(1,1-dimethylethyl)-1H-imidazol-5-yl]-6-methylpyridine (CAS: 694433-59-5), Wnt-C59 is 4-(2-methyl-4-pyridyl)-N-[4-(3-pyridyl)phenyl]phenylacetamide (CAS: 1243243- 89-1). At the time of step 4), typical spindle-shaped MSCs precursor cells are obtained, as shown in FIG. The remaining steps were unchanged. Finally, after FACS identification, the cells could express MSC-positive Markers such as CD105, CD90 and CD73 at high levels; MSC-negative Markers such as CD45, CD34, CD14 or CD11b, CD79α or CD19 and HLA-DR were not expressed at all. 5 is shown.
表4Table 4
Figure PCTCN2018101076-appb-000006
Figure PCTCN2018101076-appb-000006
表5table 5
Figure PCTCN2018101076-appb-000007
Figure PCTCN2018101076-appb-000007
实施例3:Example 3:
按照实施例1的方法进行分化培养,做如下改进:步骤1)开始在分化培养基基础上额外加上碱性成纤维细胞生长因子(Basic fibroblast growth factor,bFGF)后,在步骤4)的时候会获得更多、更典型的梭型形态特征的MSCs前体细胞,如图5所示。其余步骤不变,最后经过FACS鉴定,细胞能高水平表达CD105、CD90、CD73等MSC阳性Markers;基本不表达CD45、CD34、CD14、CD11b、CD79α、CD19以及HLA-DR等MSC阴性Markers,如表6所示。The differentiation culture was carried out in accordance with the method of Example 1, and the following improvements were made: Step 1) Starting with the addition of basic fibroblast growth factor (bFGF) on the basis of the differentiation medium, at step 4) More and more typical spindle-shaped morphological MSCs precursor cells will be obtained, as shown in Figure 5. The remaining steps were unchanged. Finally, after FACS identification, the cells could express MSC-positive Markers such as CD105, CD90, CD73, etc.; MSC-negative Markers such as CD45, CD34, CD14, CD11b, CD79α, CD19 and HLA-DR were not expressed at all. 6 is shown.
表6Table 6
Figure PCTCN2018101076-appb-000008
Figure PCTCN2018101076-appb-000008
对比例Comparative example
1)将未分化hPSCs细胞转移到细胞外基质(MG)包被30min的24孔贴壁用培养板上,用0.5ml多能干细胞分化培养基,37℃、5%CO 2培养24小时后(Day 0),换0.5ml不含TGF-β1的多能干细胞分化培养基,培养基中含有30ng/ml Activin A,6μM CHIR99021,100nM PIK90培养24小时。检测MIXL1、BRACHYURY、TBX6等前段原条标记物的表达量,发现均获得高表达,此时细胞处于前段原条阶段(表7)。 1) The undifferentiated hPSCs cells were transferred to a 24-well adherent culture plate coated with extracellular matrix (MG) for 30 min, and cultured with 0.5 ml of pluripotent stem cell differentiation medium at 37 ° C, 5% CO 2 for 24 hours ( Day 0), 0.5 ml of pluripotent stem cell differentiation medium containing no TGF-β1, containing 30 ng/ml Activin A, 6 μM CHIR99021, and 100 nM PIK90 for 24 hours. The expression levels of the protocolonary markers such as MIXL1, BRACHYURY, and TBX6 were detected, and it was found that high expression was obtained, and the cells were in the pre-segment phase (Table 7).
表7Table 7
Figure PCTCN2018101076-appb-000009
Figure PCTCN2018101076-appb-000009
Figure PCTCN2018101076-appb-000010
Figure PCTCN2018101076-appb-000010
2)去掉旧液,改为含有10uM/ml SB431542的0.5ml不含TGF-β1的多能干细胞分化培养基连续培养5天,隔天换液,期间不传代。2) The old solution was removed, and 0.5 ml of the pluripotent stem cell differentiation medium containing no TGF-β1 containing 10 uM/ml SB431542 was continuously cultured for 5 days, and the solution was changed every other day without passage.
3)去掉旧液,加2ml 0.05%Trypsin-EDTA将SB处理5天后的细胞消化5min,加4ml DMEM/F12(含10%FBS)终止,回收到15ml离心管,200g、5min离心。3) Remove the old solution, add 2 ml of 0.05% Trypsin-EDTA, digest the cells after 5 days of SB treatment for 5 min, add 4 ml of DMEM/F12 (containing 10% FBS) to terminate, collect 15 ml centrifuge tubes, and centrifuge at 200 g for 5 min.
4)除掉上清,加含有10uM/ml SB431542的1ml不含TGF-β1的多能干细胞分化培养基轻轻将细胞悬浮,移到经明胶(GT)包被30min、加含有10uM/ml SB431542的9ml不含TGF-β1的多能干细胞分化培养基的P100贴壁用培养板上,培养48h,没有发现可以粘附生长的细胞(图8)。4) Remove the supernatant, add 1 ml of pluripotent cell differentiation medium containing 10 uM/ml SB431542 without TGF-β1, gently suspend the cells, transfer to gelatin (GT) for 30 min, and add 10 uM/ml SB431542. 9 ml of the P100 adherent culture medium containing no TGF-β1 pluripotent stem cell differentiation medium was cultured for 48 hours, and no cells capable of adhering to growth were found (Fig. 8).

Claims (18)

  1. 一种高效的hPSCs向MSCs分化诱导的方法,其步骤是:A method for efficiently inducing differentiation of hPSCs into MSCs, the steps of which are:
    1)将未分化hPSCs细胞株转移到细胞外基质包被的培养板上培养后;换用含有BMP-SMAD1/5/8信号通路激活剂、TGF-β1/Activin/Nodal-SMAD2/3信号通路激活剂、Wnt激活剂、PI3K抑制剂的分化培养基培养;1) Transfer undifferentiated hPSCs cell line to extracellular matrix-coated culture plates; switch to BMP-SMAD1/5/8 signaling pathway activator, TGF-β1/Activin/Nodal-SMAD2/3 signaling pathway Differentiation medium culture of activator, Wnt activator, PI3K inhibitor;
    2)去除旧培养基,换含有TGF-β1/Activin/Nodal-SMAD2/3信号通路抑制剂分化培养基继续培养;2) Remove the old medium and continue to culture with the differentiation medium containing TGF-β1/Activin/Nodal-SMAD2/3 signaling pathway inhibitor;
    3)将细胞消化并转移到新的培养板上,换含有TGF-β1/Activin/Nodal-SMAD2/3信号通路抑制剂分化培养基继续培养;3) Digest and transfer the cells to a new culture plate, and continue to culture with TGF-β1/Activin/Nodal-SMAD2/3 signaling pathway inhibitor differentiation medium;
    4)将细胞消化并转移到贴壁用培养板上继续培养,获得MSCs。4) The cells were digested and transferred to an adherent culture plate for further culture to obtain MSCs.
  2. 根据权利要求1所述的分化诱导的方法,其特征在于,步骤3)中,新的培养板是用明胶类细胞外基质包被的,进行细胞筛选。The method of differentiation induction according to claim 1, wherein in the step 3), the new culture plate is coated with a gelatin-based extracellular matrix for cell selection.
  3. 根据权利要求2所述的分化方法,其特征在于,细胞筛选所得细胞为具有梭型形态特征,表面标志CD90、CD73、CD105为阳性,CD34、CD45、CD14或CD11b、CD79α或CD19以及HLA-DR为阴性的细胞。The differentiation method according to claim 2, wherein the cells obtained by the cell screening have a spindle-shaped morphological characteristic, and the surface markers CD90, CD73, and CD105 are positive, CD34, CD45, CD14 or CD11b, CD79α or CD19, and HLA-DR. Negative cells.
  4. 根据权利要求2所述的分化方法,其特征在于,明胶类细胞外基质为细胞培养用明胶或I型、III型、IV型、V型胶原蛋白和弹性蛋白以及层粘连蛋白。The differentiation method according to claim 2, wherein the gelatin-based extracellular matrix is gelatin for cell culture or type I, III, IV, V-type collagen and elastin, and laminin.
  5. 根据权利要求1所述的分化方法,其特征在于,步骤1)中,用含有BMP-SMAD1/5/8信号通路激活剂、TGF-β1/Activin/Nodal-SMAD2/3信号通路激活剂、Wnt激活剂、PI3K抑制剂的分化培养基培养时间为12~72h。The differentiation method according to claim 1, wherein in step 1), a BMP-SMAD1/5/8 signaling pathway activator, a TGF-β1/Activin/Nodal-SMAD2/3 signaling pathway activator, Wnt is used. The differentiation medium culture time of the activator and the PI3K inhibitor is 12 to 72 hours.
  6. 根据权利要求1所述的分化方法,其特征在于,步骤1)中,BMP-SMAD1/5/8信号通路激活剂BMP2、BMP4、BMP7中的至少一种。The differentiation method according to claim 1, wherein in step 1), at least one of BMP-SMAD1/5/8 signaling pathway activators BMP2, BMP4, BMP7.
  7. 根据权利要求1所述的分化方法,其特征在于,步骤1)中,TGF-β1/Activin/Nodal-SMAD2/3信号通路激活剂为Activin A、Activin B、TGF-β1、Nodal中的至少一种。The differentiation method according to claim 1, wherein in step 1), the TGF-β1/Activin/Nodal-SMAD2/3 signaling pathway activator is at least one of Activin A, Activin B, TGF-β1, and Nodal. Kind.
  8. 根据权利要求1所述的分化方法,其特征在于,步骤1)中,Wnt激活剂为CHIR99021,BIO、WNT-3a、R-spondin-2中的至少一种。The differentiation method according to claim 1, wherein in the step 1), the Wnt activator is at least one of CHIR99021, BIO, WNT-3a, and R-spondin-2.
  9. 根据权利要求1所述的分化方法,其特征在于,步骤1)中,PI3K抑制剂为TG100713、PIK90、PI-103中的至少一种。The differentiation method according to claim 1, wherein in the step 1), the PI3K inhibitor is at least one of TG100713, PIK90, and PI-103.
  10. 根据权利要求1所述的分化方法,其特征在于,TGF-β1/Activin/Nodal-SMAD2/3信号通路抑制剂为SB431542、SB505124、A8301、RepSox中的至少一种。The differentiation method according to claim 1, wherein the TGF-β1/Activin/Nodal-SMAD2/3 signaling pathway inhibitor is at least one of SB431542, SB505124, A8301, and RepSox.
  11. 根据权利要求1所述的分化方法,其特征在于,步骤3)中,继续培养的时间为2~30天。The differentiation method according to claim 1, wherein in the step 3), the culture is continued for 2 to 30 days.
  12. 根据权利要求1所述的分化方法,其特征在于,在步骤2)继续培养之前,先用含有 BMP-SMAD1/5/8信号通路激活剂、TGF-β1/Activin/Nodal-SMAD2/3信号通路激活剂、Wnt抑制剂的分化培养基培养。The differentiation method according to claim 1, characterized in that before the step 2), the BMP-SMAD1/5/8 signaling pathway activator, TGF-β1/Activin/Nodal-SMAD2/3 signaling pathway is used. The differentiation medium of the activator and the Wnt inhibitor is cultured.
  13. 根据权利要求1所述的分化方法,其特征在于,在步骤2)中,继续培养的时间为2~10天。The differentiation method according to claim 1, wherein in the step 2), the culture is continued for 2 to 10 days.
  14. 根据权利要求12所述的分化方法,其特征在于,Wnt抑制剂为Wnt-C59、XAV-939中的至少一种。The differentiation method according to claim 12, wherein the Wnt inhibitor is at least one of Wnt-C59 and XAV-939.
  15. 根据权利要求1所述的分化方法,其特征在于,步骤4)中继续培养的方法是:用含有间充质干细胞培养基培养。The differentiation method according to claim 1, wherein the step of continuing the culture in the step 4) is: culturing with a medium containing mesenchymal stem cells.
  16. 根据权利要求1所述的分化方法,其特征在于,在步骤1)至步骤4)中,至少一种培养基中含有碱性成纤维细胞生长因子bFGF。The differentiation method according to claim 1, wherein in the step 1) to the step 4), the at least one medium contains basic fibroblast growth factor bFGF.
  17. 根据权利要求1所述的分化方法,其特征在于,获得的MSCs是使用诱导hPSCs通过中段原条阶段或侧中胚层阶段向MSCs定向分化的方法来获得的。The differentiation method according to claim 1, wherein the obtained MSCs are obtained by inducing differentiation of hPSCs into MSCs by a middle stage or a mesoderm stage.
  18. 一种MSCs细胞,由权利要求1~17任意一项所述的分化方法制备而成。An MSCs cell prepared by the differentiation method according to any one of claims 1 to 17.
PCT/CN2018/101076 2018-01-26 2018-08-17 High performance method for differentiation of hpscs into mscs WO2019144605A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201810075968.9A CN108570448B (en) 2018-01-26 2018-01-26 A kind of method that efficient hPSCs breaks up to MSCs
CN201810075968.9 2018-01-26

Publications (1)

Publication Number Publication Date
WO2019144605A1 true WO2019144605A1 (en) 2019-08-01

Family

ID=63576718

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/101076 WO2019144605A1 (en) 2018-01-26 2018-08-17 High performance method for differentiation of hpscs into mscs

Country Status (2)

Country Link
CN (1) CN108570448B (en)
WO (1) WO2019144605A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022025598A1 (en) * 2020-07-28 2022-02-03 삼성전자 주식회사 Composition for inducing differentiation from pluripotent stem cells into mesenchymal stem cells comprising smad1/5/8 activator and use thereof

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110592007B (en) * 2019-09-19 2020-08-21 安徽中盛溯源生物科技有限公司 Mesenchymal stem cell and preparation method and application thereof
CN111876380B (en) * 2020-08-04 2022-06-28 浙江大学 Induction medium for differentiating human embryonic stem cells into mesenchymal stem cells
WO2023011114A1 (en) * 2021-08-06 2023-02-09 中国科学院动物研究所 Ric cell and preparation method therefor and use thereof
CN113462642A (en) * 2021-08-12 2021-10-01 呈诺再生医学科技(珠海横琴新区)有限公司 Rapid induced differentiation method and kit of mesenchymal stem cells and application of kit

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1636054A (en) * 2001-07-06 2005-07-06 杰龙公司 Mesenchymal cells and osteoblasts from human embryonic stem cell
CN101712947A (en) * 2009-11-12 2010-05-26 浙江大学 Preparation method and application of mesenchymal stem cells deriving from embryonic stem cells
CN101984048A (en) * 2010-11-24 2011-03-09 中国人民解放军军事医学科学院放射与辐射医学研究所 Culture medium for culturing mesenchymal stem cells
CN102686724A (en) * 2009-10-27 2012-09-19 财团法人首尔大学校产学协力团 Method for producing mesenchymal stem cells from human pluripotent stem cells, and mesenchymal stem cells produced by same
CN104487568A (en) * 2012-07-11 2015-04-01 爱姆斯坦生物技术公司 Mesenchymal-like stem cells derived from human embryonic stem cells, methods and uses thereof
CN105154393A (en) * 2014-12-22 2015-12-16 浙江大学 Method for differentiation of embryonic stem cells (ESC) into mesenchymal stem cells (MSC)
TW201819625A (en) * 2016-08-23 2018-06-01 中央研究院 Method for preparing induced mesenchymal stem cells and improving mesenchymal stem cell's characters and its applications

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1636054A (en) * 2001-07-06 2005-07-06 杰龙公司 Mesenchymal cells and osteoblasts from human embryonic stem cell
CN102686724A (en) * 2009-10-27 2012-09-19 财团法人首尔大学校产学协力团 Method for producing mesenchymal stem cells from human pluripotent stem cells, and mesenchymal stem cells produced by same
CN101712947A (en) * 2009-11-12 2010-05-26 浙江大学 Preparation method and application of mesenchymal stem cells deriving from embryonic stem cells
CN101984048A (en) * 2010-11-24 2011-03-09 中国人民解放军军事医学科学院放射与辐射医学研究所 Culture medium for culturing mesenchymal stem cells
CN104487568A (en) * 2012-07-11 2015-04-01 爱姆斯坦生物技术公司 Mesenchymal-like stem cells derived from human embryonic stem cells, methods and uses thereof
CN105154393A (en) * 2014-12-22 2015-12-16 浙江大学 Method for differentiation of embryonic stem cells (ESC) into mesenchymal stem cells (MSC)
TW201819625A (en) * 2016-08-23 2018-06-01 中央研究院 Method for preparing induced mesenchymal stem cells and improving mesenchymal stem cell's characters and its applications

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
HU JIABO ET AL.: "Preliminary investigation on the differantiation of human embryonic stem cells into mesenchymal stem cells", CHINSES JOURNAL OF CLINICAL LABORATORY SCIENCE, vol. 31, no. 4, 30 April 2012 (2012-04-30), pages 276 - 278 *
SHI LUNGANG ET AL.: "Induction and Identification of Mesenchymal stem lioke cells Huamn embryonic stem cells", JOURNAL OF SHANGHAI JIAOTONG UNIVERSITY- MEDICAL SCIENCE, vol. 29, no. 3, 31 December 2009 (2009-12-31) - 311, pages 307 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022025598A1 (en) * 2020-07-28 2022-02-03 삼성전자 주식회사 Composition for inducing differentiation from pluripotent stem cells into mesenchymal stem cells comprising smad1/5/8 activator and use thereof

Also Published As

Publication number Publication date
CN108570448B (en) 2019-04-02
CN108570448A (en) 2018-09-25

Similar Documents

Publication Publication Date Title
WO2019144605A1 (en) High performance method for differentiation of hpscs into mscs
Chen et al. Small molecule mesengenic induction of human induced pluripotent stem cells to generate mesenchymal stem/stromal cells
Salero et al. Adult human RPE can be activated into a multipotent stem cell that produces mesenchymal derivatives
Liu et al. Neural crest stem cells and their potential therapeutic applications
JP6678107B2 (en) Method of expanding pancreatic progenitor cells
Huang et al. Plasticity of stem cells derived from adult periodontal ligament
Techawattanawisal et al. Isolation of multipotent stem cells from adult rat periodontal ligament by neurosphere-forming culture system
Boyd et al. Human embryonic stem cell–derived mesoderm-like epithelium transitions to mesenchymal progenitor cells
Lee et al. Enhancement of osteogenic and chondrogenic differentiation of human embryonic stem cells by mesodermal lineage induction with BMP-4 and FGF2 treatment
JP7287948B2 (en) Pluripotent stem cell differentiation promoter
Świerczek et al. From pluripotency to myogenesis: a multistep process in the dish
WO2023106122A1 (en) Method for producing neural crest cells specialized for differentiation into mesenchymal lineage
Kumar et al. Generation of an expandable intermediate mesoderm restricted progenitor cell line from human pluripotent stem cells
Sauerzweig et al. A population of serumdeprivation-induced bone marrow stem cells (SD-BMSC) expresses marker typical for embryonic and neural stem cells
WO2023127824A1 (en) Neural crest cell culturing method and production method
EP2882848B1 (en) Method, combination and/or composition for inducing cardiomyocyte differentation
Mu et al. Enhanced differentiation of human amniotic fluid‐derived stem cells into insulin‐producing cells in vitro
Honda et al. N-cadherin is a useful marker for the progenitor of cardiomyocytes differentiated from mouse ES cells in serum-free condition
US20140314725A1 (en) Method for preparing mesenchymal stem cell-like cells and cardiomyocyte-like cells
Yamasaki et al. Long-term serial cultivation of mouse induced pluripotent stem cells in serum-free and feeder-free defined medium.
KR101896803B1 (en) Method for increasing the rate of inducing differentiation of human pluripotent stem cells to mesenchymal stem cells, and mesenchymal stem cells produced by thereof
JPWO2019107485A1 (en) Cell culture method
KR20110095218A (en) Cd49f promoting proliferation, multipotency and reprogramming of adult stem cells via pi3k/akt/gsk3 pathway
Vasyliev et al. Large-scale expansion and characterization of human adult neural crest-derived multipotent stem cells from hair follicle for regenerative medicine applications
JP7306667B2 (en) Differentiation-promoting pluripotent stem cell and use thereof

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: 18902917

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18902917

Country of ref document: EP

Kind code of ref document: A1