WO2012075636A1 - 预测干细胞分化潜能的表观遗传修饰标签 - Google Patents

预测干细胞分化潜能的表观遗传修饰标签 Download PDF

Info

Publication number
WO2012075636A1
WO2012075636A1 PCT/CN2010/079608 CN2010079608W WO2012075636A1 WO 2012075636 A1 WO2012075636 A1 WO 2012075636A1 CN 2010079608 W CN2010079608 W CN 2010079608W WO 2012075636 A1 WO2012075636 A1 WO 2012075636A1
Authority
WO
WIPO (PCT)
Prior art keywords
differentiation
histone
modification
gene
stem cells
Prior art date
Application number
PCT/CN2010/079608
Other languages
English (en)
French (fr)
Inventor
李红凌
赵春华
Original Assignee
中国医学科学院基础医学研究所
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 中国医学科学院基础医学研究所 filed Critical 中国医学科学院基础医学研究所
Priority to PCT/CN2010/079608 priority Critical patent/WO2012075636A1/zh
Priority to US13/992,619 priority patent/US9523074B2/en
Priority to CN201180057318.8A priority patent/CN103459592B/zh
Priority to EP11847131.7A priority patent/EP2636732B1/en
Priority to AU2011341213A priority patent/AU2011341213B2/en
Priority to CA2820395A priority patent/CA2820395C/en
Priority to PCT/CN2011/083380 priority patent/WO2012075912A1/zh
Priority to IN1005MUN2013 priority patent/IN2013MN01005A/en
Publication of WO2012075636A1 publication Critical patent/WO2012075636A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0607Non-embryonic pluripotent stem cells, e.g. MASC
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6881Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for tissue or cell typing, e.g. human leukocyte antigen [HLA] probes
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56966Animal cells
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6875Nucleoproteins

Definitions

  • the present invention relates to a method for predicting the differentiation potential of stem cells and a histone modification state of a pluripotency gene and/or a differentiation-related gene as an epigenetic modified label for predicting the differentiation potential of stem cells.
  • Epigenetic modifications usually include DNA methylation, histone modification, and RA modification; while histone modifications include histone methylation, acetylation, phosphorylation, and ubiquitination; the modified sites are mostly located at the N-terminus of histones. Modification can affect the state of chromatin by affecting the affinity of histones and DNA, and can also affect the binding of transcription factors to DNA sequences. It has a DNA-like genetic code for gene expression regulation, so it is called "histone.” Password”. Histone methylation refers to methylation that occurs at the N-terminal arginine or lysine residues of H3 and H4 histones and is mediated by histone methyltransferase.
  • Histone H3 4th lysine trimethylation (H3K4me3) is generally associated with promoter activation [6]
  • histone H3 27th lysine trimethylation (H3K27me3) is expressed with promoter Silence related [7-8].
  • the simultaneous presence of the two promoter histone modifications, H3K4me3 and H3K27me3, is called bivalent modification. This bivalent modification keeps the gene at a relatively low level of expression and maintains a "pre-transcriptional" state. The state allows the gene to respond rapidly to transcriptional activation or inhibition according to appropriate stimuli [9-12].
  • H3K4me3 modification is ubiquitous in the promoter pair, while H3K27me3 is only found in 10% of the gene promoter region, and that the region in which H3K27me3 is modified is also modified by H3K4me3, which is subject to bivalent
  • the modified gene is preferentially activated during ESC differentiation, suggesting that the presence of bivalent modification may be to maintain development-related genes in an equilibrium state, ready for later activation [10]; and those without any modification are inhibited. The state is completely silent.
  • Stem cell transplantation can be used to treat Parkinson's disease, cardiomyopathy, liver disease, and the treatment of bone defects in the treatment of bone defects, large-area burns, and much-needed skin materials.
  • the adult stem cells are obtained from the autologous tissue, and the tissue induced by the differentiation has no advantages such as immunological rejection during the transplantation, and has a wide variety of tissue types for differentiation, and has broad application value. It is expected to become the main force for future stem cell transplantation to treat end-stage diseases of various organs. However, there are still many safety problems in stem cell transplantation. If it is reported that the application of embryonic stem cells to the heart for coronary heart disease may lead to teratoma, the application of skeletal muscle stem cells may cause malignant arrhythmia.
  • stem cell transplantation therapy depends on two important factors: (1) acquisition, purification and amplification of seed stem cells in vitro; (2) stem cells undergo specific and functional differentiation in vivo according to therapeutic purposes. How to control proliferation, avoid tumors, and initiate the required pathways for differentiation at the appropriate time is essential for stem cell transplantation.
  • obtaining a stem cell with appropriate differentiation ability, identifying it according to the purpose of treatment, and then transplanting it into the body to cause specific differentiation in a timely manner without occurrence of teratoma, etc. requires a set of stem cell differentiation. Potential identification, differentiation stages, and whether stem cells have an accurate identification and evaluation index for controllable specific differentiation in vivo.
  • pluripotent stem cells depends on their differentiation potential, and this differentiation potential carries an epigenetic code, which is likely to be regulated by epigenetic modifications. Therefore, the role of histone methylation modification in stem cell differentiation has important clinical application value.
  • Chromatin Immunoprecipitation is the most important method for detecting histone modifications.
  • Chromatin immunoprecipitation also known as binding site analysis, is a powerful tool for studying the interaction of proteins and DNA in vivo. It is commonly used in the study of transcription factor binding sites or histone-specific modification sites.
  • Chromatin immunoprecipitation analysis is a method developed based on in vivo analysis. Its basic principle is to immobilize a protein-DNA complex in a living cell state and randomly sever it into one.
  • a small fragment of chromatin in a fixed length range and then immunologically precipitated the complex, specifically enriching the DNA fragment bound by the target protein, and obtaining information on the interaction between the protein and the DNA by purifying and detecting the target fragment.
  • the target fragment can be detected by tiling array or high-throughput sequencing, the former being called ChlP-on-chip and the latter being called ChIP-Seq.
  • the ChlP-Seq technology which combines ChIP with second-generation sequencing technology, efficiently detects DNA segments that interact with histones, transcription factors, etc., across the genome.
  • ChIP chromatin immunoprecipitation
  • ChlP-Seq Since the data of ChlP-Seq is the result of DNA sequencing, researchers can provide resources for further exploration of biological information. researchers can conduct research in the following aspects:
  • Fig. 7 shows a schematic diagram of the technology of the present invention. Summary of the invention
  • the object of the present invention is to find a relationship between histone methylation modification and stem cell differentiation ability, thereby providing a powerful tool for rapidly and accurately determining the differentiation ability of stem cells.
  • a first aspect of the invention provides a method of predicting the differentiation potential of stem cells, comprising the steps of:
  • the target gene belongs to histone H3 4th lysine trimethylation modification or histone H3 4th Coexistence of a lysine trimethylation modification and a histone H3 27th lysine trimethylation modification indicates that the target stem cell has the ability to differentiate to a particular cell type indicated by the gene of interest.
  • the gene of interest is selected from a lineage of a totipotent gene, a neural differentiation-related gene, a lipid-forming gene, an osteogenic gene, a hematopoietic-related gene, or a hepatic epithelial differentiation-related gene, a plurality of lineages, or all differentiation including other lineages.
  • transcription factors including OCT4, NANOG, c-MYC, SALL4, SOX2, KLF4, and neural differentiation related genes including TUBB3, NKX2-2, SOX1, NEUROG 1, ASCL1, BR 2, MYT1L, ZIC1, NEUROG2, HES1 , DLX1, PAX6, TLX2, MSI1, GFRA1, GFRA3, MAPT, NES, OLIG2, NEUROD1, NEUROD2, lipid-forming genes including C/EBP ⁇ , PPAR ⁇ , ERK5, GSK3 ⁇ , GSK3 ⁇ , C/EBP ⁇ , C/ EBP ⁇ , osteogenic genes include RUNX2 BMP4, Smad5, TAZ, MSX2, DLX5, BMPR2, Wnt5a, hematopoietic-related genes including c-Myb, EGR1, FOG1, SCL, E47, Ikaros, GATA1, BCL-6, hepatic epithelial differentiation Related genes include M
  • the second aspect of the present invention provides pluripotency genes and/or differentiation.
  • the stage of differentiation in which the cell is located is identified by detecting the histone methylation modification status of a particular lineage differentiation stage transcription factor and marker gene.
  • analysis of a histone modification state change that initiates a non-target lineage differentiation-related gene identifies the specificity of cell differentiation to a target lineage.
  • the histone methylation modification is a histone H3 4th lysine trimethylation modification and a histone H3 4th lysine trimethylation modification and histone H3 27th lysine
  • the acid trimethylation modification coexists.
  • the pluripotency gene and/or the differentiation-related gene is selected from the group consisting of a totipotent gene, a neural differentiation-related gene, a fatogenic gene, an osteogenic gene, a hematopoietic-related gene, or a hepatic epithelial differentiation-related gene, a plurality of lineages, or includes All differentiation-related transcription factors of other lineages, including tonal genes including OCT4, NANOG, c-MYC, SALL4, SOX2, KLF4, and neural differentiation-related genes including TUBB3, NKX2-2, SOXl, NEUROGK ASCL1, BRN2, MYT1L, ZIC1, NEUROG2 , HES1, DLX1, PAX6, TLX2, MSI 1, GFRA 1, GFRA3, MAPT, NES, OLIG2, NEUROD 1, NEUROD2, lipidogenic genes including C/EBP ⁇ , PPAR ⁇ , ER 5, GSK3 ⁇ , GSK3 ⁇ , C /
  • ChlP-seq or ChIP-PCR is used to detect the histone methylation modification status of the pluripotency gene and/or differentiation-related gene.
  • different histone methylation status of the pluripotency gene and/or differentiation-related gene indicates different differentiation potential of stem cells
  • a lineage differentiation-related genomic protein methylation modification is histone H3 position 4 lysine Trimethylation modification and histone H3 position 4 lysine trimethylation modification and histone H3 position 27 lysine trimethylation modification coexist, indicating that this stem cell has differentiation to this lineage Potential
  • two or more stem cells are compared
  • the lineage-related genes are generally modified by histone H3 lysine trimethylation and histone H3 lysine trimethylation modification and group Stem cells with a high proportion of genes coexisting with the lysine trimethylation modification at the 27th position of the protein H3 are more likely to differentiate into the lineage.
  • the present invention utilizes chromatin immunoprecipitation (ChIP) to obtain the target lysine H3 at position 4 of the specific anti-histone H3 and the lysine trimethylation at position 27 of the anti-histone H3.
  • ChIP chromatin immunoprecipitation
  • the antibody binds to all the DNA, and then obtains the sequence information of the DNA by high-throughput sequencing technology.
  • the primers use PCR technology to obtain the histone modification state of a specific gene.
  • the target gene belongs to histone H3 4th lysine trimethylation modification (H3K4me3) or histone H3 4th lysine trimethylation modification (H3K4me3) and histone H3 27th lysine
  • H3K27me3 The coexistence of the trimethylation modification indicates that the target stem cells have the ability to differentiate to the specific cell type indicated by the gene of interest.
  • the methylation modification of a lineage-associated genomic protein is the lysine trimethylation of histidine H3 and the lysine trimethylation of histidine H3 and the 27th lysine of histone H3.
  • the coexistence of trimethylation modification indicates that this stem cell has the potential to differentiate into the lineage.
  • the lineage-related gene is generally affected by histone H3 lysine 4
  • the grouping modification and the stem cell of the histone H3 4th lysine trimethylation modification and the gene of the histone H3 27th lysine trimethylation modification are more likely to differentiate into the lineage.
  • Any gene having the meaning of pluripotency or lineage differentiation in pluripotent stem cells or specific differentiation potential stem cells can be used in the methods of the present invention, including but not limited to the pluripotency genes OCT4, NANOG, c-MYC, SALL4, SOX2, KLF4.
  • the differentiation-related genomic protein modification state will be re-arranged, making the progenitor modification state of the target lineage-related genes more activated, and the histone modification that initiates other lineage differentiation-related genes becomes further suppressed or Silencing, therefore, dynamic changes in the methylation status of genomic proteins associated with this lineage and other non-target lineage differentiation during stem cell differentiation into a lineage can be used to identify the stage and specificity of stem cell stem cell differentiation.
  • the method of the invention can accurately determine the differentiation state and differentiation potential of a certain stem cell, thereby providing crucial guiding information for its correct clinical use.
  • Figure 1 shows the differentiation ability of aMSC.
  • A Ammonia-derived aMSCs obtained by limiting dilution method
  • aMSCs differentiate into adipogenic and osteogenic lineages
  • C aMSCs differentiate into hematopoietic differentiation and identification
  • OC Osteocalcin
  • BFU-E erythroid blast colony forming unit
  • CFU-G macrophage colony forming unit
  • CFU-MK megakaryocyte colony forming unit
  • HPP-CFC high proliferative potential cell colony forming unit
  • Figure 2 Differential histone methylation modification showing genes at different levels of stem cell pluripotency and neural differentiation.
  • A Whole-genome histone H3K4me3 and H3K27me3 modification panoramas in aMSC,
  • B histone methylation modification of pluripotency-related genes, and
  • C histone methylation modification of neural differentiation-related genes.
  • Figure 3 shows the differential repair profile of methylation (A) and osteogenic (B) differentiation-related genes in different stem cells.
  • Figure 4 shows the difference in methylation differences between liver epithelial (A) and hematopoietic (B) differentiation genes in different stem cells.
  • Figure 5 Comparison of methylation modification and differentiation ability of histoproteins related to adipogenic differentiation in aMSC and bMSC.
  • A ChlP-PCR analysis of osteogenic genomic protein methylation modification status in aMSC and bMSC
  • B ChlP-PCR analysis of adipose genomic protein methylation status in aMSC and bMSC
  • FIG. 6 shows dynamic changes in related genomic protein methylation modifications in neural, adipogenic and osteogenic differentiation.
  • A real-time PCR to determine the expression of related genes before and after neural differentiation
  • B ChlP-PCR to detect histone methylation status of related genes before and after neural differentiation
  • C real-time PCR and gene chip analysis of adipogenic differentiation Pre- and post-related gene expression and histone methylation status
  • D real-time PCR and gene chip analysis of related gene expression and histone methylation status before and after osteogenic differentiation
  • E real-time PCR and gene chip The expression of other lineage-related genes and histone methylation status in adipogenic differentiation or osteogenic differentiation were analyzed.
  • Figure 7 Schematic diagram of histone methylation status predicting stem cell differentiation potential. detailed description
  • aMSCs adult adipose-derived mesenchymal stem cells
  • adipose tissue was obtained from patients who had liposuction surgery (plastic surgery hospital of Chinese Academy of Medical Sciences) and signed informed consent with the donor. The donors were healthy women aged 25 to 35 years old.
  • the method for isolating aMSCs from adipose tissue is described with reference to the method of Zuk et al. [20] with minor modifications. Briefly described as follows: Adipose tissue collected by liposuction was washed with D-Hanks to remove blood cells and anesthetics, digested with 0.2% sputum collagenase for 1 hour, and then washed twice with D-Hanks to remove collagenase.
  • the cells were collected by centrifugation, and the cells were seeded at a density of 2 ⁇ 10 6 /ml in 58% DMEM/F12 + 40% MCDB-20K 2% fetal bovine serum (FCS), 10 ng/ml EGF, 10 ng/ml PDGF, lx insulin-transfer.
  • FCS fetal bovine serum
  • ITS Insulin-Transferrin-Selenium
  • LA-BSA lx linoleic acid-bovine serum albumin
  • 50 ⁇ ⁇ -mercaptoethanol 2 mM L-glutamine
  • 10 ⁇ g/ml penicillin and 100 U/ml streptomycin sulfate culture medium, incubate in 37 ° C, 5% CO 2, 95% humidity incubator.
  • bMSCs bone marrow-derived mesenchymal stem cells
  • the above mononuclear cells were seeded at a density of 1 ⁇ 10 6 /cm 2 in a 25 cm 2 culture flask, and the cell culture system contained 58% DMEM/F12 + 40% MCDB-201, 2% fetal bovine serum.
  • FCS 10 ng/ml EGF, 10 ng/ml PDGF, l Insulin-Transferrin-Selenium (ITS), lx linoleic acid-bovine serum albumin (linoleic acid-bovine serum albumin, LA-BSA), 50 ⁇ ⁇ -mercaptoethanol, 2mM L-glutamine, 10 ( ⁇ g/ml penicillin and 100U/ml streptomycin sulfate, 37°C, 5% CO 2, 95% humidity incubator to cultivate.
  • ITS Insulin-Transferrin-Selenium
  • aMSCs By limiting dilution, we inoculated aMSCs into 96-well plates at a density of 1 cell/well. After 3 weeks, approximately 24.55% ⁇ 0.66% of the wells were observed to grow monoclonal. After further amplification of these monoclonals, each monoclonal-derived cell was divided into 6 equal portions and induced to differentiate into hepatic epithelium, nerve, hematopoietic, adipose and osteogenic lineages, and another cell was further expanded for each A control of lineage differentiation (Fig. 1A). After 14 days of induction, the cytoplasm of the adipogenic induction group was filled with fat droplets under light microscope, and the positive rate of oil red 0 staining was 80%.
  • the cells were cultured, fixed with 1% formaldehyde, and the protein and DNA were cross-linked at room temperature for 10 minutes.
  • anti-R A Polymerase II a combination of all promoter regions that activate transcription.
  • Control primer promoter region of the GAPDH gene.
  • Each IP requires 900 ul Dilution Buffer plus 4.5 ul PI Cocktail.
  • the negative control IgG is recommended to be consistent with the species from which the protein of interest is derived.
  • the product can be placed in a large tube (EP tube capable of holding 1.1 ml of solution).
  • Each chromatin lOOul required product contained approximately 2 X 10 6 cells derived.
  • Protein G Agarose is a 50% slurry that is gently mixed before use.
  • This step is a "preclear" chromatin, which removes the protein that is non-specifically bound to Protein G Agarose and adds a corresponding amount of Protein G Agarose when combined. (10) Incubate for 1 hour at 4 °C.
  • each sample takes 1% of chromatin as Input.
  • Positive control tube force B l.Oug anti-R A polymerase antibody.
  • Negative control tube plus l.Oug normal same species IgG.
  • IP Incubate overnight at 4 °C.
  • the incubation time of IP can be shortened, depending on factors such as antibody, target gene and cell type.
  • the amount of Bind Reagent A added is 5 times the volume of the sample.
  • step 2 If precipitation is observed in step 2, this step collects sediment at the bottom of the tube, but does not affect the experiment.
  • control primers including IP for positive and negative control antibodies, and Input and DNA free tubes as control tubes with or without DNA contamination.
  • control primers are directed against the specific human GAPDH gene. For other substances, it is recommended that the user design specific primers based on experience.
  • Hot start Taq enzymes are recommended. If the hot start Taq enzyme is not used, it is recommended to add the Taq enzyme after the initial denaturation step.
  • a primer specific for the gene of interest was designed, and all of the DNA samples obtained by the above ChiP binding to the H3K4me3 and /H3K27me3 antibodies were used as substrates to carry out a PCR reaction, which is a ChlP-PCR technique.
  • stem cells with different differentiation potentials such as ESC (embryonic stem cells), aMSC (fat-derived mesenchymal stem cells), HSC (hematopoietic stem cells) and HPC (hematopoietic progenitor cells) as the research object [21, 22].
  • ESC epionic stem cells
  • aMSC fat-derived mesenchymal stem cells
  • HSC hematopoietic stem cells
  • HPC hematopoietic progenitor cells
  • ubiquitin genomic protein methylation showed that the pluripotency genes OCT4, NANOG, c-MYC, SALL4 and SOX2 in ESC were all modified by H3K4me3, and KLF4 was a bivalent modification of H3K4me3 and H3K27me3 coexistence; -MYC and KLF4 are H3K4me3 activation modifications, SALL4 and SOX2 are bivalent modifications, OCT4 and NANOG are essentially no modification signals; in addition to c-MYC, which is considered to be closely related to cell cycle, HSC and HPC are activation modifications, and other pluripotency genes are Modification or no modification for H3K27me3 (Fig. 2B).
  • the genes related to neural differentiation in the literature mainly include 22 transcription factors such as BRN2, MYT1L, ZIC1, NEUROG2, HES1, DLX1, PAX6, TLX2, MSI1, GFRA1, GFRA3, MAPT, NES and OLIG2 [23-25].
  • ChlP-seq data analysis showed that 17 genes in ESCs showed H3K4me3 modification or bivalent modification status; the results in aMSC were similar to those in ESC; and the analysis showed that the three genes related to the initiation of neural differentiation NES , MSI1 and HES1, their histone modification status in ESC and aMSC is H3K4me3 activation state; however, in HSC and HPC, some of these genes are expressed as H3K27me3 inhibitory modification, and no other modification signals are detected (Fig. 2C). ).
  • the osteogenesis-critical transcription factor RUNX2 was modified by H3K4me3 activation in aMSC, and bivalently modified in ESC; BMP2 and BMP4 upstream of RUNX2
  • the histone methylation modifications of Smad5, TAZ, MSX2, DLX5 and Wnt5a were similar in both cells (Fig. 3B).
  • PROX1 HNF1B, HNF6, E-cadherin Foxa-1 and Foxa-2 are all activated or bivalently modified in ESC; among them, Mxill, GSC, Soxl7, HNF6, PROX1 and Foxa-1 are also activated or double in aMSC.
  • Valence modification, Foxa-2, which is similar to Foxa-1, is H3K27me3 inhibitory modification, and SNAI1, an upstream regulator of the epithelial marker molecule E-cadherin, is an activation signal; in addition to Mxill, a weaker activating modification signal is present in HSC.
  • Other hepatic epithelial differentiation-associated genomic proteins were H3K27me3 inhibitory signals or unmodified; whereas in HPC, all hepatic genes were H3K27me3 inhibitory or unmodified (Fig. 4B).
  • H3K27me3 inhibitory or no modification signals Other lineage-related genes in HPCs were H3K27me3 inhibitory or no modification signals, and hematopoietic differentiation-related genes were all activated by H3K4me3, and their activation signals were stronger than HSC; the hematopoietic lineage-directed differentiation factor GATA1 was shown in ESCs, aMSCs and HSCs. For inhibition or no modification, but in the hematopoietic progenitor cells for H3K4me3 activation modification. It is speculated that it is activated and modified in the stage of pluripotent stem cells to hematopoietic differentiation to hematopoietic progenitor cells, in order to facilitate further differentiation of the hematopoietic lineage.
  • the methylation status of hematopoietic-related genomic protein is basically a process in which H3K27me3 inhibitory modification disappears and H3K4me3 activation modification signal gradually increases, while other non-hematopoietic related lineages It is shown that the activating modification is attenuated and the inhibitory modification signal (including H3K27me3 and no modification, both of which lead to gene silencing) is enhanced.
  • HSCs only have the ability to differentiate into hematopoietic lineages.
  • HPCs are cells that are more oriented and differentiated than HSCs, and our experiments have shown that aMSCs have multiple processes such as hepatic epithelium, nerve, hematopoiesis, vascular endothelium, adipogenesis and osteogenesis. The sub-totality of germline multi-lineage differentiation.
  • the differentiation of aMSC and bMSC into osteogenic and adipogenic directions showed that the differentiation ratio of aMSC and bMSC was 50% and 65%, respectively, on the 8th day of osteogenic induction, and the expression of ALP and OPN were statistically significant. Differences; on day 8 of adipogenic induction, the aMSC and bMSC differentiation ratios were 80% and 27%, respectively, and the expression of the marker genes LPL and AP2 were significantly different (Fig. 5C).
  • adipogenic and osteogenic related genes of MSCs from two different sources are H3K4me3 or bivalent modification
  • the proportion of adipogenic related gene H3K4me3 in aMSC is significantly higher than that of bMSC, which is more difficult than bMSC than aMSC.
  • the results of differentiation into the adipogenic lineage were consistent; the methylation-activated modification of osteogenesis-associated genomic protein in bMSC was not significantly different from that of aMSC, which was consistent with the observation that bMSC and aMSC differentiated into osteogenic lineages.
  • genomic protein modification analysis and differentiation ability of different sources of MSC differentiation further confirmed the feasibility of analysis of the genomic protein H3K4me3 and H3K27me3 modification status of lineage differentiation as an epigenetic modified label for predicting the differentiation potential of stem cells.
  • Example 4 Dynamic analysis of the genomic proteins associated with differentiation stages H3K4me3 and H3K27me3 can be used to predict the degree of cell differentiation.
  • TLX2 and MSI1 changed from bivalent K27 modification to K4 predominance, GFRA1 changed from unmodified state to divalent state; NEUROG2, PAX6, TLX2, NEUROD2 and MSI1 expression were significantly up-regulated.
  • aMSC into the adipogenic lineage, the expression of early lipid-producing transcription factors C/ ⁇ and C/ ⁇ was transiently up-regulated, and their histone modification status changed from H3K4me3 activated state to bivalent state, and the expression of GSK3P was increased.
  • histone modification also maintains the H3K4me3 sustained activation state; the downstream effector molecule PPARY of these genes remains in a sustained state, while C/ ⁇ changes from a bivalent state.
  • the activated state as the expression of PPAR Y and C/EBPct continued to increase, the adipogenic differentiation progressed smoothly, and the expression of the marker genes LPL and AP2 increased significantly.
  • BMP2, TAZ, MSX2, Smad5 and BMPR2 also experienced up-regulation from expression up to induction 4 to 6 days, then The dynamic process of down-regulation is expressed.
  • This dynamic expression change ensures the initiation of osteogenic differentiation and further maturation of osteoblast function.
  • the osteogenesis-critical gene RUNX2 maintains the H3K4me3 activation state, and the expression level continues to increase, further promoting Expression of its downstream target gene OSX and osteogenic marker genes ALP and OPN.
  • the differentiation status of differentiation-related genomic proteins H3K4me3 and H3K27me3 is closely related to the differentiation ability and differentiation stage of stem cells, and can be used as an epigenetic label for predicting the differentiation potential and cell differentiation stage and maturity of different levels of stem cells from different sources.
  • This finding provides a new standard for clinically better screening and identification of seed cells required for the regenerative repair of various tissues and organs.
  • This histone methylation tag is relatively easy to obtain: First, the whole genome histone methylation modification spectrum of unknown differentiation potential stem cells is obtained by using the ChlP-seq technique, and then a targeted selection analysis is performed according to the purpose of the stem cell application.
  • the histone H3K4me3 and B H3K27me3 modification status of the genes related to the lineage differentiation, and further verification of the ChlP-seq results by the ChlP-PCR technique, can predict whether the stem cells have the ability to differentiate into the lineage.
  • the stem cells are located on a stem cell grade pyramid with apical embryonic stem cells as the apex.
  • ChlP-seq technology can quickly and easily predict whether stem cells have the potential to differentiate into the lineage of interest under suitable external conditions or in vivo microenvironment after obtaining the genome-wide histone methylation modification profile.
  • ChlP-seq or ChlP-PCR was used to analyze the histone methylation status of stem cells in a lineage differentiation process and related transcription factors and real-time quantitation of these genes. PCR can be used to identify the specific differentiation stage of stem cells. (Note: Changes in histone methylation are first and gene expression changes, when the histone methylation status of a gene becomes more activated.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Cell Biology (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Zoology (AREA)
  • Organic Chemistry (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • General Health & Medical Sciences (AREA)
  • Analytical Chemistry (AREA)
  • Microbiology (AREA)
  • Wood Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Genetics & Genomics (AREA)
  • General Physics & Mathematics (AREA)
  • Food Science & Technology (AREA)
  • Pathology (AREA)
  • Medicinal Chemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Biophysics (AREA)
  • Developmental Biology & Embryology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Virology (AREA)
  • Measuring Or Testing Involving Enzymes Or Micro-Organisms (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Immobilizing And Processing Of Enzymes And Microorganisms (AREA)

Description

预测干细胞分化潜能的表观遗传修饰标签
技术领域
本发明涉及预测干细胞分化潜能的方法及全能性基因和 /或分化相关基因的组 蛋白修饰状态作为预测干细胞分化潜能的表观遗传修饰标签的用途。 背景技术
表观遗传修饰通常包括 DNA甲基化、 组蛋白修饰及 R A修饰; 而组蛋白修饰 又包括组蛋白甲基化、 乙酰化、 磷酸化和泛素化; 修饰部位大多位于组蛋白 N端, 这些修饰可通过影响组蛋白与 DNA的亲和性, 从而改变染色质的状态, 也可以影 响转录因子与 DNA序列的结合, 对基因表达调控具有类似 DNA遗传密码的作用, 故被称作 "组蛋白密码" 。 组蛋白甲基化是指发生在 H3和 H4组蛋白 N端精氨酸或 者赖氨酸残基上的甲基化, 由组蛋白甲基转移酶介导。 组蛋白赖氨酸的甲基化已 成为转录的重要调控机制, 在异染色质的形成、 X染色体的失活、 基因组印迹、 DNA损伤修复及基因转录调控中担任重要角色 [1-5]。组蛋白 H3第 4位赖氨酸三甲基 化 ( H3K4me3 ) —般与启动子活化有关 [6], 而组蛋白 H3第 27位赖氨酸三甲基化 ( H3K27me3 ) 则与启动子的表达沉默相关 [7-8]。 基因启动子区 H3K4me3和 H3K27me3两种组蛋白修饰状态同时存在则被称为双价修饰,这种双价修饰使基因 保持在相对低的表达水平, 维持在一种 "预备转录" 的状态, 这种状态可以使基 因根据适当的刺激做出迅速的转录活化或抑制等反应 [9-12]。
最近越来越多的研究开始关注组蛋白赖氨酸甲基化在胚胎发育中的作用。 对 斑马鱼的研究发现, 受精后基因组失活, 直到母源性向合子转换(maternal- zygotic transition) 完成才重新起始转录 [13- 15], 基因组组蛋白 H3赖氨酸三甲基化的修饰 分析结果显示,转换前组蛋白 H3K27me3抑制性修锦及 H3K4me3活化性修饰均未检 测到; 转换完成, 基因组激活后, 80%的基因出现了 H3K4me3修饰, 其中未活化 的一些发育调控相关基因同时还存在: H3K27me3修饰。 上述结果提示, 这种在母源 性向合子转换过程中建立起来的染色质组蛋白 H3双价或单价修饰谱很可能与全能 性的建立有关 [16]。先前的研究发现,在小鼠胚胎干细胞中, H3K4me3和 H3K27me3 共定位存在与由约 2.5%基因组组成的高度保守的区域, 提示这种双价修饰状态在 全能性细胞保持这种 "预备"活化的状态中担任重要角色 [9]。 对人胚胎干细胞组 蛋白修饰的研究发现, 在启动子對近, H3K4me3修饰普遍存在, 而 H3K27me3仅见 于 10%的基因启动子区, 而且, H3K27me3修饰存在的区域同时受 H3K4me3修饰, 这些受双价修饰的基因在 ESC分化时优先被激活,提示双价修饰的存在可能是维持 发育相关基因在一种平衡状态, 为以后的活化做准备 [10]; 而那些没有任何修饰的 基因则处在抑制状态, 完全被沉默。利用 ChlP-seq检测结果建立的数据库对多能性 神经祖细胞(multipotent neural progenitor cells , NPCs)、鼠胚胎纤维母细胞(murine embryonic fibroblasts , MEFs)及原代人 T细胞中 H3K4me3 和 H3K27me3修饰分析 发现, 在 NPCs或 MEFs中, 受双价修饰的基因数量下降 [5, 12]。 由此推测受双价 修饰的区域大部分是胚胎干细胞特异性的 [9]。 但最近全基因组组蛋白甲基化修饰 谱的分析显示, 在分化的细胞 (如 T细胞和 MEFs) 中也发现有这种受双价修饰的 区域, 因此双价修饰并不是 ESC特有的 [5, 12, 17]。 尽管与 ESC相比, 人 T细胞 中受双价修饰基因的数量下降, 但之前一些没有任何修饰的基因的组蛋白却重新 被甲基化修饰, 推测组蛋白修饰的这种变化可能与 T细胞的特化及其它谱系的抑制 有关 [10]。
尽管研究提示, 组蛋白甲基化在异染色质的形成、 X染色体的失活、 基因组 印迹、 DNA损伤修复及基因转录调控中有重要作用, 而且发现组蛋白甲基化位点 在不同物种中是高度保守的, 在分化能力不同的细胞中存在不同的组蛋白甲基化 修饰谱。 但迄今为止, 组蛋白甲基化修饰在细胞分化中的作用及意义还知之甚少。 表观遗传调节是一个动态变化的过程, 这使得表观遗传的研究变得复杂。 近年来 随着测序技术的迅速发展及其成本的降低, 染色质免疫共沉淀与测序相结合的技 术 (ChlP-seq) 得到到了广泛应用 [18, 19]。 干细胞移植可以用于治疗帕金森症、 心肌病、 肝脏疾病、 以及诱导成骨治疗骨缺损、 大面积烧伤治疗急需的皮肤材料 等。 成体具有干细胞取自自体、 由它诱导分化而来的组织在进行移植时不存在免 疫排斥问题, 诱导分化的组织类型广泛等优点, 具有广阔的应用价值。 有望成为 未来干细胞移植治疗各种器官终末期疾病的主力军。 但是, 目前干细胞移植还存 在众多的安全性问题, 如有报道指出, 应用胚胎干细胞植入心脏治疗冠心病时, 可能会导致畸胎瘤, 应用骨骼肌干细胞则有产生恶性心律失常的危险, 还有骨髓 细胞移植后出现严重心肌钙化的报告。 因此, 上述干细胞移植治疗的成功取决于 以下两大重要因素: (1 ) 种子干细胞在体外的获取、 纯化和扩增; (2) 干细胞 在体内按照治疗目的发生专一性有功能的分化。 如何使既控制增殖, 避免发生肿 瘤, 又能在适当的时候启动所需要的途径进行分化, 对干细胞移植治疗至关重要。 但是, 获取具有适当分化能力的干细胞, 根据治疗目的对其进行鉴定, 并进而移 植到体内使之适时的发生专一性分化而不发生畸胎瘤等等一系列问题均需要有一 套对干细胞分化潜能、 分化阶段及干细胞是否在体内发生可控的专一性分化等准 确鉴定和评价指标。 因此, 多能性干细胞在临床上的应用前景依赖于它们的分化 潜能, 而这种分化潜能携带有表观遗传密码, 很可能受表观遗传修饰的调节。 因 此探讨组蛋白甲基化修饰在干细胞分化中的作用有着及其重要的临床应用价值。
染色质免疫沉淀分析 (Chromatin Immunoprecipitation, ChIP) 是检测组蛋白 修饰的最重要的方法。 染色质免疫共沉淀技术 (ChIP) 也称结合位点分析法, 是 研究体内蛋白质与 DNA相互作用的有力工具,通常用于转录因子结合位点或组蛋 白特异性修饰位点的研究。 染色质免疫沉淀分析是基于体内分析发展起来的方法, 它的基本原理是在活细胞状态下固定蛋白质一 DNA复合物, 并将其随机切断为一 定长度范围内的染色质小片段, 然后通过免疫学方法沉淀此复合体, 特异性地富 集目的蛋白结合的 DNA片段, 通过对目的片断的纯化与检测, 从而获得蛋白质与 DNA相互作用的信息。目的片断可以通过 tiling array或高通量测序的方法来检测, 前者称为 ChlP-on-chip而后者称为 ChIP-Seq。将 ChIP与第二代测序技术相结合的 ChlP-Seq技术, 能够高效地在全基因组范围内检测与组蛋白、 转录因子等相互作 用的 DNA区段。
ChlP-Seq 的原理是, 首先通过染色质免疫共沉淀技术 (ChIP) 特异性地富集 目的蛋白结合的 DNA片段,并对其进行纯化与文库构建;然后对富集得到的 DNA 片段进行高通量测序。 研究人员通过将获得的数百万条序列标签精确定位到基因 组上,从而获得全基因组范围内与组蛋白、转录因子等相互作用的 DNA区段信息。
由于 ChlP-Seq的数据是 DNA测序的结果, 为研究者提供了进一步深入挖掘 生物信息的资源, 研究者可以在以下几方面展开研究:
( 1 ) 判断 DNA链的某一特定位置会出现何种组蛋白修饰;
(2) 检测 R A polymerase II及其它反式因子在基因组上结合位点的精确定 位;
(3 ) 研究组蛋白共价修饰与基因表达的关系;
(4) CTCF转录因子研究。
为了进一步揭示 "组蛋白甲基化密码"在细胞分化中的神秘角色, 我们利用 ChIP检测与生物信息学分析相结合的方法(ChlP-seq)研究了 ESC (胚胎干细胞)、 aMSC (脂肪来源间充质干细胞) 、 HSC (造血干细胞)及 HPC (造血祖细胞) 等 多种级别干细胞的全基因组组蛋白甲基化修饰谱, 试图找到组蛋白甲基化修饰与 干细胞分化能力间的关系。 图 7显示了本发明的技术示意图。 发明内容
因此, 本发明的目的在于寻找组蛋白甲基化修饰和干细胞分化能力间的关系, 从而为快速、 准确地确定干细胞的分化能力提供一种有力的工具。
因此, 本发明的第一方面提供了一种预测干细胞分化潜能的方法, 其包括下 述步骤:
1 ) 获取目标干细胞;
2) 使用特异性抗组蛋白 H3第 4位赖氨酸三甲基化和抗组蛋白 H3第 27位赖 氨酸三甲基化的抗体以 ChIP技术获得目标干细胞中所有与所述抗体结合的 DNA 样品;
3 )将 ChIP获得的 DNA样品进行高通量测序以获取目标干细胞的全基因组组 蛋白甲基化修饰谱和 /或设计特异于目的基因的引物, 以上述 DNA样品作为底物, 进行 PCR反应以获取目的基因的组蛋白甲基化修饰结果,
其中, 目标基因属于组蛋白 H3第 4位赖氨酸三甲基化修饰或组蛋白 H3第 4 位赖氨酸三甲基化修饰和组蛋白 H3第 27位赖氨酸三甲基化修饰共存则指示目标 干细胞具有分化到目的基因所指示的特定细胞类型的能力。
优选地, 所述目的基因选自全能基因、 神经分化相关基因、 成脂性基因、 成 骨性基因、 造血相关基因或肝上皮分化相关基因的一种谱系、 多种谱系或包括其 他谱系的全部分化相关转录因子, 其中全能基因包括 OCT4、 NANOG、 c-MYC、 SALL4、 SOX2、 KLF4,神经分化相关基因包括 TUBB3、 NKX2-2、 SOX1、NEUROG 1、 ASCL1、 BR 2、 MYT1L、 ZIC1、 NEUROG2、 HES1、 DLX1、 PAX6、 TLX2、 MSI1、 GFRA1、 GFRA3、 MAPT、 NES、 OLIG2、 NEURODl、 NEUROD2, 成脂性基因 包括 C/EBP α、 PPAR γ、 ERK5、 GSK3 α、 GSK3 β、 C/EBP δ、 C/EBP β, 成骨 性基因包括 RUNX2 BMP4、 Smad5、 TAZ、 MSX2、 DLX5、 BMPR2、 Wnt5a, 造血相关基因包括 c-Myb、 EGR1、 FOGl、 SCL、 E47、 Ikaros、 GATA1、 BCL-6, 肝上皮分化相关基因包括 Mxill、 GSC、 Soxl7、 PROX1、 HNF1B、丽 F6、 E-cadherin、 Foxa-l、 Foxa-2、 SNAI1、 NEUROD2、 GFRA2。 (注: 终末分化的标志基因如 AP2、 LPL、 c-Kit、 ALP、 OPN、 CK8、 CK18等不宜作为分化潜能预测的候选基因) 本发明的第二方面提供了全能性基因和 /或分化相关基因组蛋白修饰状态作为 预测干细胞分化潜能的表观遗传修饰标签的用途, 其中通过检测所述全能性基因 和 /或分化相关基因的组蛋白甲基化修饰状态预测干细胞的分化潜能。 优选地, 通 过检测特定谱系分化阶段性转录因子及标志基因的组蛋白甲基化修饰状态鉴定该 细胞所处的分化阶段。 优选地, 分析启动非目标谱系分化相关基因的组蛋白修饰 状态变化鉴定细胞向目标谱系分化的专一性。
优选地,所述组蛋白甲基化修饰为组蛋白 H3第 4位赖氨酸三甲基化修饰和组 蛋白 H3第 4位赖氨酸三甲基化修饰与组蛋白 H3第 27位赖氨酸三甲基化修饰并 存。
优选地, 全能性基因和 /或分化相关基因选自全能基因、 神经分化相关基因、 成脂性基因、 成骨性基因、 造血相关基因或肝上皮分化相关基因的一种谱系、 多 种谱系或包括其他谱系的全部分化相关转录因子, 其中全能基因包括 OCT4、 NANOG、 c-MYC、 SALL4、 SOX2、 KLF4,神经分化相关基因包括 TUBB3、NKX2-2、 SOXl、 NEUROGK ASCL1、 BRN2、 MYT1L、 ZIC1、 NEUROG2、 HES1、 DLX1、 PAX6、 TLX2、 MSI 1、 GFRA 1、 GFRA3、 MAPT、 NES、 OLIG2、 NEUROD 1、 NEUROD2, 成脂性基因包括 C/EBP α、 PPAR γ、 ER 5、 GSK3 α、 GSK3 β、 C/EBP δ、 C/EBP β, 成骨性基因包括 RUNX2 BMP4、 Smad5、 TAZ、 MSX2、 DLX5、 BMPR2、 Wnt5a, 造血相关基因包括 c-Myb、 EGR1、 FOGl、 SCL、 E47、 Ikaros、 GATA1、 BCL-6, 肝上皮分化相关基因包括 Mxill、 GSC、 Soxl7、 PROX1 HNF1B、 HNF6、 E-cadherin Foxa-l、 Foxa-2、 SNAI1、 NEUROD2、 GFRA2。 (注: 终末分化的标 志基因如 AP2、 LPL、 c-Kit、 ALP、 OPN、 CK8、 CK18等不宜作为分化潜能预测 的候选基因) 优选地, 检测所述全能性基因和 /或分化相关基因的组蛋白甲基化修饰状态时 使用 ChlP-seq或 ChIP-PCR。
优选地, 所述全能性基因和 /或分化相关基因的不同组蛋白甲基化状态指示了 干细胞的不同分化潜能, 某谱系分化相关基因组蛋白甲基化修饰是组蛋白 H3第 4 位赖氨酸三甲基化修饰和组蛋白 H3第 4位赖氨酸三甲基化修饰与组蛋白 H3第 27 位赖氨酸三甲基化修饰并存为主, 则指示这种干细胞具有向该谱系分化的潜能, 两种或多种干细胞进行比较,则该谱系相关基因总体上受组蛋白 H3第 4位赖氨酸 三甲基化修饰和组蛋白 H3第 4位赖氨酸三甲基化修饰与组蛋白 H3第 27位赖氨 酸三甲基化修饰并存的基因所占比例高的干细胞更易于向该谱系分化。
换言之, 本发明利用染色质免疫沉淀技术 (ChIP) 获得了目标干细胞的与特 异性抗组蛋白 H3第 4位赖氨酸三甲基化和抗组蛋白 H3第 27位赖氨酸三甲基化 的抗体结合的所有 DNA, 然后通过高通量测序技术获得了所述 DNA的序列信息, 通过与基因组信息相比对, 获得了目标干细胞的全基因组的组蛋白修饰谱, 或者 通过设计针对特定基因的引物利用 PCR技术获得特定基因的组蛋白修饰状态。 其 中, 目标基因属于组蛋白 H3第 4位赖氨酸三甲基化修饰 (H3K4me3 ) 或组蛋白 H3第 4位赖氨酸三甲基化修饰 (H3K4me3)和组蛋白 H3第 27位赖氨酸三甲基化修 饰 (H3K27me3)共存则指示目标干细胞具有分化到目的基因所指示的特定细胞类型 的能力。某谱系分化相关基因组蛋白甲基化修饰是组蛋白 H3第 4位赖氨酸三甲基 化修饰和组蛋白 H3第 4位赖氨酸三甲基化修饰与组蛋白 H3第 27位赖氨酸三甲 基化修饰并存为主, 则指示这种干细胞具有向该谱系分化的潜能, 两种或多种干 细胞进行比较,则该谱系相关基因总体上受组蛋白 H3第 4位赖氨酸三甲基化修饰 和组蛋白 H3第 4位赖氨酸三甲基化修饰与组蛋白 H3第 27位赖氨酸三甲基化修 饰并存的基因所占比例高的干细胞更易于向该谱系分化。 任何在全能性干细胞或 特定分化潜能干细胞内具有全能性或谱系分化标识意义的基因均可用于本发明所 述的方法, 包括但不限于全能基因 OCT4、 NANOG、 c-MYC、 SALL4、 SOX2、 KLF4, 神经分化相关基因 TUBB3、 NKX2-2、 S0X1、 NEUR0G1、 ASCL1、 BR 2、 MYT1L、 ZIC1、 NEUROG2、 HES1、 DLX1、 PAX6、 TLX2、 MSI1、 GFRA1、 GFRA3、 MAPT、 NES、 OLIG2、 NEUR0D1、 NEUROD2, 成脂性基因 C/EBP a、 PPAR y ERK5、 GSK3 a、 GSK3 β、 C/EBP δ、 C/EBP β, 成骨性基因 RUNX2、 BMP4、 Smad5、 TAZ、 MSX2、 DLX5、 BMPR2、 Wnt5a, 造血相关基因 c-Myb、 EGR1、 F0G1、 SCL、 E47、 Ikaros、 GATA1、 BCL-6, 肝上皮分化相关基因 Mxill、 GSC、 Soxl7、 PROX1 HNF1B、 HNF6、 E-cadherin Foxa-l、 Foxa-2、 SNAI1、 NEUROD2、 GFRA2等的一种谱系、 多种谱系或包括其他谱系的全部分化相关转录因子。 (注: 终末分化的标志基因如 AP2、 LPL、 c-Kit、 ALP、 OPN、 CK8、 CK18等不宜作为 分化潜能预测的候选基因)
也即, 通过对具有不同分化能力的干细胞的组蛋白修饰状态的分析之后发现: 干细胞中全能及各系分化相关基因不同组蛋白 H3K4及 H3K27三甲基化的修饰状 态与干细胞的分化能力密切相关, 可以用来预测干细胞的分化潜能; 不仅如此, 干细胞向特定谱系分化启动时, 先于基因表达的改变, 分化相关基因组蛋白修饰 状态将发生重新布局, 使得目标谱系相关基因的祖丹修饰状态变得更加活化, 而 启动其它谱系分化相关基因的组蛋白修饰则变得进一步抑制或沉默, 因此, 对干 细胞向某一谱系分化过程中该谱系及其它非目标谱系分化相关基因组蛋白甲基化 修饰状态的动态变化可用来鉴定干细胞干细胞分化的阶段及专一性。 一旦向某一 谱系分化相关基因的表达已经被激活, 这时的细胞已经是发生部分分化了的细胞, 因此, 如果用谱系分化相关基因的表达量来衡量干细胞的分化潜能会漏掉很多更 原始、 分化潜能更广泛的干细胞来源, 而组蛋白修饰的变化先于基因表达的改变, 干细胞中组蛋白 H3上的 K4或 K4和 K27同时被三甲基化后能够维持这些基因在 非常低的表达水平, 处在一种 "预备"活化或失活的状态, 这种组蛋白修饰状态 有利于干细胞根据根据微环境或外界条件的改变向不同谱系方向分化, 因此分化 相关基因组蛋白修饰状态的差异可作为预测和评价不同来源干细胞分化潜能的有 力指标。
利用本发明的方法可准确地确定某种干细胞的分化状态及分化潜能, 从而为 其正确的用于临床提供了至关重要的指导信息。 附图说明
图 1 : 显示 aMSC的分化能力。 (A) 极限稀释法获得单克隆来源的 aMSCs,
( B ) aMSCs 向成脂和成骨谱系分化, (C ) aMSCs 向造血分化及鉴定 (OC : Osteocalcin, BFU-E: 红系爆式集落形成单位, CFU-G: 巨嗜细胞集落形成单位, CFU-MK: 巨核细胞集落形成单位, HPP-CFC: 高增殖潜能细胞集落形成单位),
(D) aMSCs向肝上皮诱导分化及鉴定, (E) aMSCs向神经方向诱导分化及鉴定。
图 2:显示不同级别干细胞全能性及神经分化相关基因的组蛋白甲基化修饰差 异谱。 (A) aMSC中全基因组组蛋白 H3K4me3和 H3K27me3修饰全景图, (B) 全能性相关基因的组蛋白甲基化修饰, (C) 神经分化相关基因的组蛋白甲基化修 饰。
图 3: 显示不同干细胞中成脂 (A) 及成骨 (B) 分化相关基因甲基化差异修 饰谱。
图 4: 显示不同干细胞中肝上皮 (A) 及造血 (B) 分化相关基因甲基化差异 修饰谱。
图 5:显示 aMSC和 bMSC中成脂成骨分化相关基因组蛋白甲基化修饰比较及 分化能力比较。 (A)ChlP-PCR分析 aMSC和 bMSC中成骨性基因组蛋白甲基化修 饰状态, (B)ChlP-PCR分析 aMSC和 bMSC中成脂性基因组蛋白甲基化修饰状态,
(C) aMSC和 bMSC向成骨谱系和成脂谱系分化能力的比较。 图 6:显示神经、成脂和成骨分化中的相关基因组蛋白甲基化修饰的动态变化。 (A) real-time PCR测定神经分化前后相关基因的表达, (B) ChlP-PCR检测神经 分化前后相关基因的组蛋白甲基化状态, (C) real-time PCR及基因芯片分析成脂 分化前后相关基因的表达及组蛋白甲基化状态, (D) real-time PCR及基因芯片分 析成骨分化前后相关基因的表达及组蛋白甲基化状态, (E) real-time PCR及基因 芯片分析成脂分化或成骨分化中其它谱系相关基因的表达及组蛋白甲基化状态。
图 7: 组蛋白甲基化修饰状态预测干细胞分化潜能示意图。 具体实施方式
下面将通过下述非限制性实施例进一步说明本发明, 本领域技术人员公知, 在不背离本发明精神的情况下, 可以对本发明做出许多修改, 这样的修改也落入 本发明的范围。
下述实验方法如无特别说明, 均为常规方法, 所使用的实验材料如无特别说 明, 均可容易地从商业公司获取。 实施例
实施例 1验证单细胞来源 aMSC的分化能力
为了探讨组蛋白甲基化在干细胞分化中的意义, 我们首先对单细胞来源 aMSC 的分化能力进行了验证。
成人脂肪样品取自医科院整形医院, 成人骨髓样品取自解放军 307 医院。 所 有样品均签订知情同意书。
成人脂肪源间充质干细胞 (aMSCs) 的分离:
成人脂肪组织取自吸脂手术患者 (中国医学科学院整形医院), 与供者签订知 情同意书, 供者均为 25〜35 岁的健康女性。 从脂肪组织中分离 aMSCs的方法参照 Zuk等 [20]方法,并稍加改动。简述如下:采用吸脂术采集出来的脂肪组织用 D-Hanks 洗去血细胞和麻醉药, 0.2%Π 型胶原酶消化 1小时, 之后用 D-Hanks洗涤 2 遍以除 去胶原酶。离心收集细胞,细胞以 2x l06/ml 的密度接种于含 58% DMEM/F12 + 40% MCDB-20K 2%胎牛血清 (FCS)、 10ng/ml EGF、 10ng/ml PDGF, l x胰岛素 -转铁 蛋白-亚硒酸(Insulin-Transferrin- Selenium, ITS)、 l x亚油酸-牛血清白蛋白(linoleic acid-bovine serum albumin, LA-BSA) , 50μΜ β巯基乙醇, 2mM L-谷氨酰胺, 10(^g/ml 青霉素和 100U/ml硫酸链霉素的培养液, 37°C、 5%C02、 95%湿度的培 养箱培养。 2d 后换液, 弃去未贴壁的细胞, 以后每 3 天半量换液。 当细胞达 70%〜80%汇合时, 0.25%胰酶(Gibco 公司)常规消化, 细胞按照 1 : 3 进行传代。
成人骨髓源间充质干细胞 (bMSCs) 的分离:
( 1 ) 无菌采集健康志愿者的骨髓 5-10 ml 于无菌的肝素管中。
(2)取一无菌的离心管, 用 D-Hanks 液适当稀释骨髓后计数, 并调整骨髓细 胞浓度至 l X 107/ml。
(3 )取一新的离心管, 分别加入恢复至室温的淋巴细胞分离液和上述骨髓细 胞悬液, 加入时仔细操作勿破坏界面, 二者的比例为 1 : 1。
(4)将上述离心管配平后放入常温台式离心机中, 20°C以 1800 转 /分钟的速 度离心 20 分钟。 取出离心管后, 无菌操作仔细吸取白膜层即获得了单个核细胞, 将单个核细胞用 D-Hanks 液洗涤两次并计数。
(5 )将上述单个核细胞以 l X 106/cm2 的密度接种于 25cm2 的培养瓶中, 细胞 培养体系均为含 58% DMEM/F12 + 40% MCDB-201、 2%胎牛血清 (FCS )、 10ng/mlEGF 、 10ng/ml PDGF , l 胰岛素 - 转铁蛋 白 - 亚硒酸 ( Insulin-Transferrin-Selenium, ITS)、 l x亚油酸-牛血清白蛋白( linoleic acid-bovine serum albumin, LA-BSA), 50μΜ β巯基乙醇, 2mM L-谷氨酰胺, 10(^g/ml青霉 素和 100U/ml硫酸链霉素的培养液, 37°C、 5%C02、 95%湿度的培养箱培养。
( 6) 24 小时后, 去除悬浮细胞, 补充培养基, 细胞每隔三天换液一次, 待 细胞长至 70-80%汇合时, 用 0.05%胰蛋白酶 -0.01%EDTA消化传代。 1-2代的间充 质干细胞冻存于液氮罐备用。
通过极限稀释法, 我们以 1个细胞 /孔的密度将 aMSC接种与 96孔板中, 三周后 观察约有 24.55%±0.66%的孔长出了单克隆。 将这些单克隆进一步扩增后, 每个单 克隆来源的细胞分成 6等份, 分别向肝上皮、 神经、 造血、 成脂肪及成骨谱系诱导 分化, 另一份细胞继续扩增后用于各谱系分化的对照 (图 1A) 。 诱导 14天后, 光 镜下可见成脂诱导组细胞胞浆内充满了脂肪滴, 油红 0染色阳性率达 80%, 实时定 量 PCR检测显示成脂标志基因 AP2及 LPL高表达 (图 1B) ; 成骨诱导组 ALP及茜素 红染色阳性率达 65%, 实时定量 PCR检测显示, 成骨标志基因 ALP和 OPN表达较诱 导前明显上调 (图 IB ) 。 aMSC向造血方向诱导第 3天, 造血相关的标志分子 Osteocalcin (OC) 、 c-Kit和 CD34染色阳性, 诱导 14天时可见 BFU-E (红系爆式集 落形成单位) 、 CFU-G (巨嗜细胞集落形成单位) 、 CFU-MK (巨核细胞集落形成 单位) 和 HPP-CFC (高增殖潜能细胞集落形成单位) 等各系造血集落的形成 (图
IC)。诱导第 21天, 肝上皮诱导组细胞 CK8、 CK18和 CK19免疫组化检测阳性(图
ID)。 诱导第 12天, 神经诱导组细胞 Nestin和 Musashi免疫组化检测阳性 (图 1E)。 上述结果表明, aMSC在一定的诱导条件下能够向肝上皮、 神经、 造血及成脂和成 骨等不同胚层来源的多谱系方向分化。 实施例 2分化相关基因组蛋白 H3K4me3和 H3K27me3的不同修饰状态与干 细胞的分化潜能密切相关
验证了 aMSC具有向肝上皮、 神经、 造血、 成脂及成骨等多胚层谱系分化能力 后, 我们进一步利用 ChIP检测技术与生物信息学分析相结合的方法(ChlP-seq)获 得了 aMSC全基因组组蛋白甲基化修饰谱。 H3K4me3 (Abeam 8580)和 H3K27me3 (Upstate 07-449)的 ChlP实验按照 EZ ChlP™ kit (Millipore)的标准操作方法进行
基本过程如下:
1. 染色质样本准备和免疫选择
( 1 ) 培养细胞, 1%甲醛固定, 使蛋白和 DNA交联, 室温 10分钟,
(2)裂解和超声(Branson 250D 超声仪)处理细胞,染色质片段在 200-1000bp,
(3 )免疫选择; 用特异抗体, H3K4me3 (Abeam 8580) 禾 B H3K27me3 (Upstate 07-449)。
2. DNA纯化和检测
(4) 分离纯化 DNA, 去除蛋白质, 65摄氏度孵育去除蛋白 -DNA交联,
(5 )针对目的基因设计特异性引物, 利用 PCR或 real-time PCR对 DNA序列 进行鉴定。
阳性对照;
anti-R A Polymerase II, 所有活化转录的基因启动子区都有结合。
阴性对照;
抗同一种属来源的正常 IgG。
对照引物; GAPDH基因的启动子区。
3. 免疫沉淀 (IP) 交联物
开始前准备: 将蛋白酶抑制剂 Cocktail II放置室温溶解, 该试剂含 DMSO, 在 18.4°C以下时仍处于冰冻状态。
( 6 ) 制备含蛋白酶抑制剂的 Dilution buffer, 放置在冰上。
每个 IP 需要 900 ul Dilution Buffer加 4.5ul PI Cocktail。
样品包括阳性对照(抗 R A polymerase Π)、阴性对照(正常的相同物种 IgG) 和目的蛋白。 阴性对照 IgG 建议与目的蛋白抗体来源的物种一致。
(7) 将制备的含 lOOul产物的 EP管放冰上, 进行染色质免疫沉淀 (IP)。 冻 存的样品需提前冰上溶解。
如果同一份染色质产物要进行多个 IP, 可将产物放在一个大管内 (能容纳 1.1ml溶液的 EP管)。
每 lOOul产物需含有大约 2 X 106细胞来源的染色质。
( 8) 每 lOOul染色质产物中加入 900ul含 PI cocktail的 Dilution Buffer。
如果有多分 IP, 加入相应数量的 Dilution Buffer。
(9) 每个 IP加入 60ul Protein G Agarose。
Protein G Agarose是 50%浆液, 使用前颠倒轻轻混匀。
这一步是 "preclear"染色质, 去除非特异结合 Protein G Agarose的蛋白和 多分合并处理时加相应数量的 Protein G Agarose。 (10) 4°C旋转孵育 1小时。
(11) 3000-5000g离心 1分钟, 沉淀 agarose。
不要高速离心 Protein GAgarose。 离心力太大, 可能造成 beads粉碎或变形。
(12) 取 lOul (1%) 上清作为 Input, 保存在 4°C, 待第 5步进行处理。
多个样本同时处理时, 各取 1%的染色质样本做 Input.
(13) 收集上清液 lml转移至新的 EP管。
(14) 在上清液中加入免疫沉淀的抗体。
阳性对照管, 力 B l.Oug抗 -R A polymerase抗体。
阴性对照管, 加 l.Oug正常的相同物种 IgG。
检测管加 1-lOug抗体。 加抗体的量需要根据以往经验确定。
(15) 4°C旋转孵育过夜。 IP的孵育时间可以縮短, 主要取决于抗体、 靶基因 和细胞类型等多个因素。
(16) 加 60ul Protein G Agarose, 4°C旋转孵育 1小时。
(17) 3000-5000g离心 1分钟, 沉淀 agarose, 去上清。
( 18) 用以下溶液每个 lml依次洗 Protein G Agarose/染色质复合物, 重悬后, 旋转孵育 3-5分钟, 低速离心 3000-5000g离心 1分钟, 小心去除上清。
a. Low salt immune Complex Wash Buffer, 一次;
b. High Salt Immune Complex Wash Buffer, —次;
c. LiCl Immune Complex Wash Buffer, 一次;
d. TE Buffer, 二次。
4. 蛋白 /DNA复合物洗脱
开始前准备:
将 IM NaHC03 放置至室温。 可能会有少量沉淀, 等恢复到室温后, 沉淀可 溶解。 1 M NaHC03可漩涡混匀。
准备 65 °C水浴。
(1) 为所有 IP管准备 Elution buffer, 包括 Input管 (见节 3, 步骤 7)。 每管 需 200ul Elution buffer,配制方法: lOul 20%SDS, 20ul IM NaHC03,加 170ul dH20。
(2) 或者可以在大管中一起配制, 如有 10个 IP 管, 可将 105ul 20%SDS, 210ul IM NaHC03, 力 B 1.785ul dH20混合.
(3) Input管加入 200 ul Elution buffer后, 放置室温第 5步继续处理。
( 4 ) 每管抗体 /agarose复合物中加 100ul Elution buffer, 轻弹混匀。
(5) 室温孵育 15分钟。
(6) 3000-5000g离心 1分钟以沉淀 agarose, 将上清液收集到新的 EP管中。
(7) 重复步骤 (4) 到 (6), 将洗脱液合并, 总体积 200ul。
5. 蛋白 /DNA去交联, 获得游离 DNA
(1) 所有管 (包括 IPs和 Inputs)加入 8ul 5MNaCl, 65°C孵育 4到 5小时或 过夜, 以去除 DNA-蛋白交联。 完成后样本可储存在 -20°C, 隔天继续进行后续实 验。
(2) 所有管中加 lulR aseA, 37°C孵育 30分钟。
(3) 力 B 4ul 0.5M EDTA, 8ul 1M Tris-HCl和 lul 蛋白酶 K, 45°C孵育 1-2小 时。
6. 用 Spin Columns纯化 DNA
( 1 )每个样品准备一个收集管和一个分离管。将 Spin Column放入收集管中。
(2) 每 200 ul DNA样品加 1ml Bind Reagent A, 混匀。
所加 Bind Reagent A的量为样品体积的 5倍。
可见到沉淀, 但不会影响本步骤。
( 3 ) 将 600ul样品 / Bind Reagent A混合物加到收集管的 Spin滤器上。
(4) >10,000g离心 30秒。
(5) 移开 Spin滤器, 弃去收集管中的液体, 保留收集管。
如果步骤 2中见沉淀, 本步骤收集管底可见沉淀, 但不会影响实验。
(6) 将 Spin滤器重新放入收集管。
(7)将步骤 2中的样品 / Bind Reagent A混合物 600ul加入 Spin滤器, 重复步 骤 (4) 到 (6)。
(8) 在收集管的 Spin滤器中加入 500ul Wash Reagent B。
(9) >10,000g离心 30秒。
(10) 从收集管中移出 Spin滤器, 弃去收集管中的液体, 保留收集管。
(11) 将 Spin滤器重新放入原收集管。
(12) >10,000g离心 30秒。
(13) 弃去收集管和液体。
(14) 将 Spin滤器放入收集管中。
(15) 直接在白色 Spin滤膜的中央, 加入 50ul Elution Buffer C。
(16) >10,000g离心 30秒。
(17) 弃去 Spin滤器。 洗出液为纯化的 DNA。 可立即分析或 -20°C冻存。
7. 对照 PCR
注意: 本部分所有枪和枪尖要尽可能避免污染。
(1) 在 0.2ml PCR管上做好标记, 放冰上。
使用本试剂盒, 至少有 4个 DNA样品要用对照引物进行 PCR分析, 包括阳 性和阴性对照抗体的 IP物, Input和无 DNA得空管作为有无 DNA污染的对照管。
对照引物是针对特异性人 GAPDH基因。 针对其它物质, 建议使用者根据经 验设计特异的引物。
(2) 每管加 2ul样品, 放回冰上。
(3) 每个反应管中加入合适数量的试剂, 根据表格 1, 依次加入水, Taq酶 等。
推荐使用热启动 Taq酶。 如果没有使用热启动 Taq酶, 建议起始变性步骤后 再加入 Taq酶。
8. 将上述 ChiP获得的所有与 H3K4me3和 /H3K27me3抗体结合的 DNA样 品进行高通量测序即为 ChlP-Seq技术。
9. 设计针对目的基因特异的引物, 以上述 ChiP获得的所有与 H3K4me3和 /H3K27me3抗体结合的 DNA样品作为底物, 进行 PCR反应, 此为 ChlP-PCR技 术。
根据人类基因组数据库(Hgl8)进行校正后得到的 aMSC全基因组组蛋白甲基 化 K4和 K27位点修饰全景图 (图 2A)。 我们选取了 ESC (胚胎干细胞)、 aMSC (脂 肪来源间充质干细胞)、 HSC (造血干细胞) 和 HPC (造血祖细胞) 等具有不同分 化潜能的干细胞作为研究对象 [21, 22], 分析比较了这些干细胞中全能性基因和肝 上皮、 神经、 造血、 成脂及成骨等谱系分化相关基因的组蛋白甲基化修饰状态。
对全能性基因组蛋白甲基化修饰的分析发现, ESC中全能性基因 OCT4、 NANOG、 c-MYC、 SALL4禾卩 SOX2均为 H3K4me3活化修饰, KLF4为 H3K4me3 和 H3K27me3共存的双价修饰; MSC中 c-MYC和 KLF4为 H3K4me3活化修饰, SALL4 和 SOX2为双价修饰, OCT4和 NANOG基本无修饰信号; HSC和 HPC中除了被认为 与细胞周期关系密切的 c-MYC为活化修饰外,其它全能基因均为 H3K27me3抑制修 饰或无修饰(图 2B)。 目前文献报道与神经分化相关的基因主要有 BRN2、 MYT1L、 ZIC1、 NEUROG2、 HES1、 DLX1、 PAX6、 TLX2、 MSI1、 GFRA1、 GFRA3、 MAPT、 NES和 OLIG2等 22个转录因子 [23-25]。 ChlP-seq数据分析显示, 在 ESCs 中有 17个基因表现为 H3K4me3修饰或双价修饰状态; 在 aMSC中的分析结果与 ESC 中相似; 而且分析结果显示,与神经分化启动相关的三个基因 NES、 MSI1和 HES1 , 它们在 ESC及 aMSC中的组蛋白修饰状态均为 H3K4me3活化状态; 但在 HSC和 HPC 中,这些基因一部分表现为 H3K27me3抑制性修饰,其它的均未检测到修饰信号(图 2C) 。
接下来, 我们又比较了上述干细胞中中胚层相关谱系成脂、 成骨及造血分化 相关基因的组蛋白修饰状态。 结果显示, 成脂性关键转录因子 C/ΕΒΡα和 ΡΡΑΙ γ在 aMSC中是 H3K4me3活化修饰, 而在 ESC中是双价修饰; 它们上游的调节因子 ERK5、 BMP2、 GSK3a GSK3p C/ΕΒΡδ 禾口 C/ΕΒΡβ在 ESC及 aMSC中的组蛋白修 饰相似; 同样, 在 HSC和 HPC中这些基因的组蛋白修饰亦为 H3K27me3抑制状态或 无修饰 (图 3A) 。 对成骨相关基因的组蛋白甲基化分析得到了类似的结果, 即成 骨关键转录因子 RUNX2在 aMSC中为 H3K4me3活化修饰, 而在 ESC中为双价修饰; RUNX2上游的调控因子 BMP2、 BMP4、 Smad5、 TAZ、 MSX2、 DLX5禾卩 Wnt5a 等在两种细胞中的组蛋白甲基化修饰相似 (图 3B) 。 造血分化相关基因 c-Myb、 EGR1、 FOG1 (ZFPM1 )、 SCL (TALI )、 E47 (TCF3 )、 Ikaros (IKZF1 )、 GATA1 和 BCL-6等组蛋白甲基化修饰分析显示, c-Myb、 EGR1、 E47和 BCL-6在四种干 / 祖细胞中均为 H3K4me3活化修饰状态; F0G1、 SCL和 Ikaros在 ESC和 MSC中为双 价修饰, 而在 HSC和 HPC中为 H3K4me3活化性修饰; GATA1在 ESC和 aMSC中显示 为抑制性修饰或无修饰, 在 HSC和 HPC中则为 H3K4me3活化性修饰 (图 4A) 。
进一步对内胚层肝上皮谱系相关基因的分析显示, Mxill、 GSC、 Soxl7
PROX1 HNF1B、 HNF6、 E-cadherin Foxa-1和 Foxa-2等在 ESC中均为活化或双 价修饰; 其中 Mxill、 GSC、 Soxl7、 HNF6、 PROX1 禾口 Foxa-1在 aMSC中亦为活化 或双价修饰, 与 Foxa-1作用相似的 Foxa-2为 H3K27me3抑制修饰, 上皮性标志分子 E-cadherin 的上游调控因子 SNAI1则为活化信号; 在 HSC中除 Mxill有较弱的活化 性修饰信号外, 其它肝上皮分化相关基因组蛋白均为 H3K27me3抑制信号或无修 饰; 而在 HPC中, 所有肝性基因均为 H3K27me3抑制信号或无修饰 (图 4B) 。
全基因组组蛋白修饰的分析比较显示, ESCs中六个全能基因有五个是活化修 饰, 一个为双价修饰, 向各胚层分化的关键转录因子的组蛋白甲基化状态总体上 以 H3K4me3活化或双价修饰为主; aMSCs中六个全能基因有两个是活化修饰, 四 个是双价修饰。 HSCs中几乎所有造血分化相关基因均以活化修饰为主外, 其它谱 系方向相关基因则以 H3K27me3或无修饰信号为主。 HPCs中其它谱系相关基因均 为 H3K27me3抑制修饰或无修饰信号, 而造血分化相关基因均为 H3K4me3活化修 饰,而且其活化信号较 HSC强;造血谱系定向分化相关因子 GATA1在 ESCs、 aMSCs 和 HSCs中显示为抑制或无修饰, 但在造血祖细胞中为 H3K4me3活化修饰。 推测它 是在多能干细胞向造血分化到造血祖细胞阶段被活化修饰, 以利于造血谱系进一 步的定向分化。 整体上看, 从 ESC、 aMSC、 HSC到 HPC, 造血分化相关基因组蛋 白甲基化修饰状态基本是一个 H3K27me3抑制性修饰逐渐消失、 H3K4me3活化性修 饰信号逐渐增强的过程, 而其它非造血相关谱系则表现为活化性修饰减弱而抑制 性修饰信号 (包括 H3K27me3及无修饰, 这两种情况都导致基因沉默) 增强。
先前已有大量的研究证明, ESCs 具有向所有胚层所有谱系分化的全能性,
HSCs只有向造血相关谱系分化的能力, HPCs是较 HSCs更进一步定向与造血谱 系分化的细胞, 而我们的实验证明, aMSCs 具有向肝上皮、 神经、 造血、 血管内 皮、 成脂和成骨等多胚层多谱系分化的亚全能性。 结合上述 ESC、 aMSC, HSC和 HPC 中各谱系分化相关基因不同组蛋白修饰状态的分析结果及这些干细胞不同的 分化潜能提示我们: 随着多能性等级的下降, 组蛋白甲基化修饰状态的改变, 干 细胞逐渐丢失其分化的全能性 (ESC) 转变成具有多胚层分化能力 (MSC) 或仅 具有单一胚层 (HSC) 甚至单一谱系分化的能力 (祖细胞) 的细胞。 分化相关基 因组蛋白 H3K4me3 和 H3K27me3修饰状态与干细胞的分化潜能密切相关, 可作 为预测干细胞分化潜能的表观遗传修饰标签。 实施例 3谱系分化相关基因组蛋白 H3K4me3和 H3K27me3修饰状态的分 析可用来预测干细胞分化潜能
为了进一步验证分化相关基因组蛋白 H3K4me3 和 H3K27me3修饰状态与干 细胞的分化潜能的相关性, 我们利用 ChlP-PCR进一步对脂肪来源 MSC (aMSC) 和骨髓来源 MSC (bMSC) 的成脂和成骨谱系分化相关基因的组蛋白修饰进行了 分析, 并与它们向上述两种谱系分化的能力进行了比较。 组蛋白甲基化分析的结 果显示, aMSC中成骨性基因 RUNX2、 BMP2、 Smad5、 TAZ、 Wnt5a禾 B BMPR2 为活化修饰, MSX2和 BMP4为双价修饰; bMSC中除了 MSX2为 H3K4me3占优 势的双价修饰外, 其它均为活化修饰(图 5A)。 aMSC中成脂性基因除 C/ΕΒΡ α为 双价修饰外, ERK5、 GSK3a GSK3p C/EBPS、 PPARy禾 B C/ΕΒΡβ均为活化性 修饰;而 bMSC中则以双价修饰为主(图 5B)。同样诱导条件下,对 aMSC和 bMSC 向成骨和成脂方向分化的比较显示,成骨诱导第 8天, aMSC和 bMSC分化比率分 别为 50%和 65%, 标志基因 ALP和 OPN表达有统计学差异; 成脂诱导第 8天, aMSC和 bMSC分化比率分别为 80%和 27%,标志基因 LPL和 AP2表达有显著差 异 (图 5C)。
由此可见, 尽管两种不同来源的 MSC中成脂和成骨相关基因均为 H3K4me3 或双价修饰, 但 aMSC中成脂相关基因 H3K4me3修饰所占比率明显高于 bMSC, 这与 bMSC比 aMSC难于向成脂谱系分化的结果相吻合; bMSC中成骨相关基因 组蛋白甲基化活化修饰与 aMSC差别不大, 这与我们观察到 bMSC和 aMSC向成 骨谱系分化能力相似的结果一致。对不同来源 MSC分化相关基因组蛋白修饰分析 及分化能力的比较结果进一步验证了谱系分化相关基因组蛋白 H3K4me3 和 H3K27me3 修饰状态的分析作为预测干细胞分化潜能的表观遗传修饰标签的可行 性。 实施例 4分化阶段相关基因组蛋白 H3K4me3和 H3K27me3修饰状态的动 态分析可用来预测细胞的分化程度
验证了组蛋白甲基化分析可作为预测干细胞分化潜能的表观遗传修饰标签之 后,我们又利用 ChlP-PCR对 aMSC分化前后相关转录因子组蛋白甲基化修饰的动 态变化进行了分析,结果显示, aMSC向神经谱系分化过程中,关键转录因子 PAX6 和 NEUROG2 的组蛋白修饰状态从 H3K27me3 抑制态转变为双价修饰, NEUROD2从 H3K27me3抑制态变为活化态, GFRA2从双价态变为活化态, TLX2 和 MSI1则从双价的 K27修饰占优势变为 K4占优势状态, GFRA1则从无修饰状 态变为双价态; NEUROG2、 PAX6、 TLX2、 NEUROD2 禾卩 MSI1 的表达明显上 调。 aMSC向成脂谱系分化过程中, 伴随早期成脂性转录因子 C/ΕΒΡβ和 C/ΕΒΡδ 表达短暂上调, 它们的组蛋白修饰状态从 H3K4me3活化态变成双价态, 调节因子 GSK3P表达量升高后维持在较高水平, 其组蛋白修饰亦维持 H3K4me3 持续活化 态; 这些基因的下游效应分子 PPARY保持持续活化态, 而 C/ΕΒΡα则从双价态变 为激活态, 随着 PPARY和 C/EBPct表达持续升高, 成脂分化顺利进行, 标志基因 LPL和 AP2表达明显增加。 aMSC向成骨谱系分化过程中, 伴随早期调控基因组 蛋白修饰从 H3K4me3到双价修饰的变化, BMP2、 TAZ、 MSX2、 Smad5和 BMPR2 亦经历了从表达上调到诱导 4到 6天表达达峰值, 然后表达下调的动态过程, 这 种动态表达变化即确保了成骨分化的启动又有利用成骨细胞功能的进一步成熟; 成骨关键性基因 RUNX2维持 H3K4me3活化态, 表达量持续升高 , 进一步促进 了其下游靶基因 OSX及成骨标志基因 ALP 和 OPN的表达。 有趣的是, 我们发现 aMSC走向成脂分化时,成骨分化相关转录因子 RUNX2、 TAZ、 MSX2、 Smad5 和 BMPR2的组蛋白修饰从 H3K4me3活化态变为双价修饰, MSX2则从双价修饰变 为抑制态, 这些基因的表达均下调。 而当 aMSC走向成骨分化时, 成脂分化的关 键转录因子 σΕΒΡβ、 ΟΕΒΡδ、 GSK3P和 ΡΡΑΙ γ等的组蛋白修饰从 H3K4me3活 化态变成双价态, C/EBPct则从双价态变为 H3K27me3抑制态, 这些基因的表达亦 降低。而在 aMSC向成指或成骨谱系分化时,神经分化相关的转录因子 MSI1、TLX2 和 NES等的组蛋白修饰则进一步走向抑制。终末分化的成脂或成骨细胞中该谱系 分化相关的转录因子及标志基因的组蛋白修饰以 H3K4me3为主,而其它谱系则表 现为 H3K27me3。 这些结果提示, MSC向特定谱系分化开始前, 某些未知机制使 相关基因的组蛋白修饰状态发生改变, 以利于该谱系分化所需基因的活化表达, 同时抑制或关闭其他谱系相关因子的表达, 从而使特定谱系分化得以顺利进行。
由上述组蛋白修饰的动态分析可见, aMSC向神经、 成脂及成骨谱系分化后, 该谱系相关转录因子组蛋白修饰表现为进一步活化 (即从抑制或无修饰到双价、 从 K27 占优势的双价态到 K4 占优势的双价态、 从双价到活化态或保持持续活化 等方式), 各种方式的进一步活化的组蛋白修饰变化为特定谱系分化相关基因的活 化或表达上调提供了可能性。 不仅如此, 当 aMSC 向特定谱系分化启动时, 该谱 系分化相关基因组蛋白修饰进一步转向活化的同时, 其它谱系分化相关转录因子 的组蛋白则进一步转向抑制性修饰为主, 这样就很好的确保了干细胞向特定谱系 分化的专一性及分化效率。 总之, 在细胞向特定谱系分化过程中, 组蛋白甲基化 修饰亦发生着动态变化, 以满足开启不同分化阶段性事件相关基因活化的需要。 对未知分化程度的细胞进行分化阶段相关基因的组蛋白甲基化修饰状态进行分析 就可评估此细胞所处的分化阶段。 因此, 组蛋白甲基化修饰状态的分析可作为鉴 定细胞分化阶段或成熟度的辅助指标。 讨论:
上述的研究结果表明, 不同级别干 /祖细胞细胞中各谱系分化关键基因的不同 组蛋白修饰状态与这些细胞所具有的分化潜能密切相关。 对未知分化潜能的干 /祖 细胞中组蛋白甲基化的分析可作为预测此类细胞分化潜能。 而且, 在特定谱系分 化启动前, 在某些未知机制的调控下, 组蛋白修饰状态会发生重新布局, 以利用 特定谱系分化相关基因的活化及保持其它谱系分化相关基因不被激活, 从而实现 了定向分化的特异性。 因此, 对分化阶段性相关基因的组蛋白甲基化修饰的分析 还可以用来鉴定细胞所处的分化阶段及成熟度。 因此我们提出, 分化相关基因组 蛋白 H3K4me3 和 H3K27me3修饰状态与干细胞的分化能力及分化阶段密切相关, 可以作为预测不同来源不同级别干细胞分化潜能及细胞分化阶段和成熟度的表观 遗传学标签。 这一发现为临床上更好的筛选和鉴定各种组织器官再生修复治疗所 需种子细胞提供了新的标准。 这种组蛋白甲基化标签比较容易得到: 首先, 利用 ChlP-seq 技术获得未知分化潜能干细胞的全基因组组蛋白甲基化修饰谱, 再根据 此干细胞应用的目的有针对性的选择分析某一 /些谱系分化相关基因的组蛋白 H3K4me3 禾 B H3K27me3修饰状态,进一步应用 ChlP-PCR技术对 ChlP-seq结果进 行验证, 即可对此干细胞是否具有向该谱系分化的能力作出预测。 另外还可通过 目前已经获得人类胚胎干细胞、 脂肪来源间充质干细胞、 造血干细胞、 造血祖细 胞及成熟 T细胞等不断丰富的网络数据库资源进行比对分析, 按照分化能力的差 别, 将未知分化能力的干细胞在以具有分化全能性的胚胎干细胞为塔尖的干细胞 等级金字塔上进行定位。 ChlP-seq技术获得全基因组组蛋白甲基化修饰谱后就可 简单快捷的预测此干细胞在适合的外界条件下或体内微环境下是否具有向感兴趣 谱系分化潜能。 同样, 利用 ChlP-seq或 ChlP-PCR技术对干细胞在某一谱系分化 过程中相关转录因子及阶段性分化标志基因的组蛋白甲基化修饰状态进行分析, 结合这些转录因子及标志基因的实时定量 PCR, 就可很好的对干细胞所处的具体 分化阶段进行鉴定 (注: 因组蛋白甲基化修饰的变化先与基因表达改变, 当某一 基因的组蛋白甲基化状态变为更加活化,包括双价修饰变成 H3K4me3活化修饰或 H3K27me3抑制修饰变成双价修饰,预示着这个基因将有进一步被活化的可能,而 该基因最终表达是否上调或上调程度受控于其上游的活化因子、 细胞因子或 miRNA等的调控)。 同时, 当干细胞向目的谱系分化时, 对其它谱系相关基因的组 蛋白修饰状态的变化分析还可鉴定干细胞是否专一性的向目的基因分化。
因此,利用全基因组 ChlP-seq技术和 ChlP-seq对相关基因组蛋白 H3K4me3 和 H3K27me3修饰状态进行分析,联合相应的基因芯片结果,针对不同谱系关键转录 因子制定组合性表观遗传学检测标签, 结合基因及非编码 R A等标志物的联合应 用, 有望成为临床上各种组织器官再生修复治疗所需种子细胞的筛选及分化阶段 和分化专一性鉴定的金标准。 参考文献
[1] Boggs, B.A., Cheung, P., Heard, E., Spector, D丄., Chinault, A.C., and Allis, CD. Differentially methylated forms of histone H3 show unique association patterns with inactive human X chromosomes. Nat. Genet. 2002, 30: 73-76.
[2] Peters, A.H., Mermoud, J.E., O' Carroll, D., Pagani, M., Schweizer, D., Brockdorff, N., and Jenuwein, T. Histone H3 lysine 9 methylation is an epigenetic imprint of facultative heterochromatin. Nat. Genet. 2002, 30: 77-80.
[3] Plath, K., Fang, J., Mlynarczyk-Evans, S.K., Cao, R., Worringer, K.A., Wang, H., de la Cruz, C.C., Otte, A.P., Panning, B., and Zhang, Y. Role of histone H3 lysine 27 methylation in X inactivation. Science. 2003, 300: 131-135.
[4] Silva, J., Mak, W., Zvetkova, I., Appanah, R., Nesterova, T.B., Webster, Z.,
Peters, A.H., Jenuwein, T., Otte, A.P., and Brockdorff, N. Establishment of histone h3 methylation on the inactive X chromosome requires transient recruitment of Eed-Enxl polycomb group complexes. Dev. Cell. 2003, 4:481-495.
[5] Barski, A., et al., High-resolution profiling of histone methylations in the human genome. Cell, 2007. 129(4): 823-37
[6] Martin, C., and Zhang, Y. . The diverse functions of histone lysine methylation. Nat. Rev. Mol. Cell Biol. 2005, 6: 838-849.
[7] Schneider, R., Bannister, A.J., Myers, F.A., Thome, A.W., Crane-Robinson, C., and Kouzarides, T. Histone H3 lysine 4 methylation patterns in higher eukaryotic genes. Nat. Cell Biol.2003, 6: 73-77.
[8] Schubeler, D., MacAlpine, D.M., Scalzo, D., Wirbelauer, C., Kooperberg, C., van Leeuwen, F., Gottschling, D.E., O'Neill, L.P., Turner, B.M., Delrow, J., et al. (2004). The histone modification pattern of active genes revealed through genome-wide chromatin analysis of a higher eukaryote. 2004, Genes Dev. 18: 1263-1271.
[9] Bernstein, B.E., et al., A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell, 2006. 125(2): 315-26.
[10] Xiao Dong Zhao, Xu Han, Joon Lin Chew, Jun Liu, Kuo Ping Chiu, Andre Choo, Yuriy L. Orlov, Wing-Kin Sung, Atif Shahab, Vladimir A. Kuznetsov, Guillaume Bourque, Steve Oh, Yijun Ruan, Huck-Hui Ng, and Chia-Lin Wei. Whole-Genome Mapping of Histone H3Lys4and27 Trimethylations RevealsDistinctGenomic Compartments in Human Embryonic Stem Cells. Cell stem cell. 2007, 1 : 286-298.
[11] Pan, G., et al., Whole-genome analysis of histone H3 lysine 4 and lysine 27 methylation in human embryonic stem cells. Cell Stem Cell, 2007. 1(3): 299-312.
[12] Mikkelsen, T.S., et al., Genome-wide maps of chromatin state in pluripotent and lineage-committed cells. Nature, 2007. 448(7153): 553-560.
[13] "Newport, J. & Kirschner, M. A major developmental transition in early Xenopus embryos: IL Control of the onset of transcription. Cell. 1982, 30: 687-696.
[14] Schier, A. F The mateni al-zygoti c trail si tion: death and birth of RNAs, Scieiice. 2007, 316; 406-407,
[15] Tadros, W, & Lipshitz, H, D. The HI atemal ~lo~zygotic transition; a pl y in two acts, DeveSopment. 2009, 136: 3033-3042.
[16] Nadine L Vastenhomv, Yong Zhang, Ian G, Woods, Farhad lmam; Aviv Regev, X, Shirley Liu; John Rixin& Alexander F. Schier. Chromatin signature of embiyonic pluripoiency is established during genome activation. 2010, 464; 922 -925.
[17] Roh, Ί\Υ., Cuddapah, S,, C i, K,; and Zhao, K. The genomic landscape of histoiie modifications in human T cells. Proc, Nail Acad Sci. USA. 2006, 103, 15782 15787,
[18] Barski, A, and . Zhao, Genomic location analysis by ChIP Seq. J Cell Blochem, 2009. 107(1): i i 8.
[19] Park, P. J., ChIP- seq: advantages and challenges of a maturing technology. Nat
Rev Genet., 2009 10(10): 669-680'
[20] Patricia A. Zuk, Min Zhu. Peter Ashjian, Daniel A , De Ugarte, Jerry I. Huang, Fliroslii Mizuno, Zeni C, Alfonso, John 【 Frasex Prosper Benhaim, and Marc H. Hedrick, Human Adipose Tissue Is a Source of iiltipotent Stem Cells. Mol Biol Cell, 2002, .13(12); 4279-4295.
[21] Ku, M5 ei al., Genomewide anaSysis of PRC1 and P C2 occupancy identifies two classes of bivalent doniams. PLoS Genet, 2008, 4(10): el 000242„
[22] Cui, K" ei al, Chromatin signatures in删 Itipotent human he atopoietic stem ceils indicate the fate of bivalent genes during differed tiatioii, CeSI Stem Ce L 2009, 4(1): 80 93,
[23] Thomas Vierbuchen, AustinOstermeier, Zhiping P, Pang, Yuko Kokubu, Thomas C, Su'dhof &Marius Wernig. Direct conversion of fibroblasts to functional neurons by defined factors. Nature. 2010, 463; 1035 -1041.
[24] Jung Hee Shim, Sun A C o; Min Ji Seo, Jung Hee Kim, Na Kyung Ryu, Kyung Hyun Yoo, Moon Hee Yang, Seyoon Kim, Young Yil Balik, and Jong Hoon Park. Proteomic Analysis of Time -Dependant Difference of Protein Expression Profile Changes during Neuronal Differentiation of Mouse Embryonic Stem Cells. MoL Cells. 2010, 29: 239 244,
[25] Chin-Hsing Lin, Stacey Hansen, Zhenshan Wang, Daniel R. Storm, Stephen J. Tapscott, and James M. Olson. The dosage of the neuroD2 transcription factor regulates amygdala development and emotional learning. PNAS. 2005, 102(41): 14877-14882.

Claims

权利要求书
1、 一种预测干细胞分化潜能的方法, 其包括下述步骤:
1 ) 获取目标干细胞;
2 ) 使用特异性抗组蛋白 H3第 4位赖氨酸三甲基化和抗组蛋白 H3第 27位赖 氨酸三甲基化的抗体以 CMP技术获得目标干细胞中所有与所述抗体结合的 DNA 样品;
3 )将 CMP获得的 DNA样品进行高通量测序以获取目标干细胞的全基因组组 蛋白甲基化修饰谱和 /或设计特异于目的基因的引物, 以上述 DNA样品作为底物, 进行 PCR反应以获取目的基因的组蛋白甲基化修饰状态,
其中, 目标基因属于组蛋白 H3第 4位赖氨酸三甲基化修饰或组蛋白 H3第 4 位赖氨酸三甲基化修饰和组蛋白 H3第 27位赖氨酸三甲基化修饰共存则指示目标 干细胞具有分化到目的基因所指示的特定细胞类型的能力。 2、 根据权利要求 1所述的预测干细胞分化潜能的方法, 其特征在于所述目的 基因选自全能基因、 神经分化相关基因、 成脂性基因、 成骨性基因、 造血相关基 因或肝上皮分化相关基因的一种谱系、 多种谱系或包括其他谱系的全部分化相关 转录因子, 其中全能基因包括 OCT4、 NANOG、 c-MYC、 SALL4、 SOX2、 KLF4 , 神经分化相关基因包括 TUBB3、 NKX2-2、 S0X1、 NEUR0G1、 ASCL1、 BRN2、 MYT1L、 ZIC1、 NEUROG2、 HES1、 DLX1、 PAX6、 TLX2、 MSI1、 GFRA1、 GFRA3、 MAPT、 NES、 OLIG2 NEUR0D1、 NEUROD2, 成脂性基因包括 C/EBP a、 PPAR Y ERK5、 GSK3 a、 GSK3 β、 C/EBP δ、 C/EBP β , 成骨性基因包括 RUNX2、 BMP4、 Smad5、 TAZ、 MSX2、 DLX5、 BMPR2、 Wnt5a,造血相关基因包括 c-Myb、 EGR1、 F0G1、 SCL、 E47、 Ikaros、 GATA1、 BCL-6 , 肝上皮分化相关基因包括 Mxill、 GSC、 Soxl7、 PROX1、 HNF1B、 HNF6、 E-cadherin、 Foxa- 1、 Foxa-2 SNAI1、 NEUROD2、 GFRA2。
3、全能性基因和 /或分化相关基因的组蛋白修饰状态作为预测干细胞分化潜能 的表观遗传修饰标签的用途, 其中通过检测所述全能性基因和 /或分化相关基因的 组蛋白甲基化修饰状态预测干细胞的分化潜能。
4、根据权利要求 3所述的全能性基因和 /或分化相关基因的组蛋白修饰状态作 为预测干细胞分化潜能的表观遗传修饰标签的用途, 其特征在于通过检测特定谱 系分化阶段性转录因子及标志基因的组蛋白甲基化修饰状态鉴定该细胞所处的分 化阶段。
5、根据权利要求 3所述的全能性基因和 /或分化相关基因的组蛋白修饰状态作 为预测干细胞分化潜能的表观遗传修饰标签的用途, 其特征在于分析启动其它非 目标谱系分化相关基因的组蛋白修饰状态变化鉴定细胞向目标谱系分化的专一 性。
6、根据权利要求 3至 5任一项所述的全能性基因和 /或分化相关基因的组蛋白 修饰状态作为预测干细胞分化潜能的表观遗传修饰标签的用途, 其特征在于所述 组蛋白甲基化修饰为组蛋白 H3第 4位赖氨酸三甲基化修饰或组蛋白 H3第 4位赖 氨酸三甲基化修饰与组蛋白 H3第 27位赖氨酸三甲基化修饰并存。
7、根据权利要求 3或 6任一项所述的全能性基因和 /或分化相关基因的组蛋白 修饰状态作为预测干细胞分化潜能的表观遗传修饰标签的用途, 其特征在于全能 性基因和 /或分化相关基因选自全能基因、 神经分化相关基因、 成脂性基因、 成骨 性基因、 造血相关基因或肝上皮分化相关基因的一种谱系、 多种谱系或包括其他 谱系的全部分化相关转录因子, 其中全能基因包括 OCT4、 NANOG、 c-MYC、 SALL4、 SOX2、 KLF4,神经分化相关基因包括 TUBB3、 NKX2-2、 SOX1、 NEUROG1、 ASCL1 BRN2、 MYT1L、 ZIC1、 NEUROG2、 HES1、 DLX1、 PAX6、 TLX2、 MSI1、 GFRA1 GFRA3、 MAPT、 NES、 OLIG2、 NEURODl、 NEUROD2, 成脂性基因 包括 C/EBP α、 PPAR γ、 ERK5、 GSK3 α、 GSK3 β、 C/EBP δ、 C/EBP β, 成骨 性基因包括 RUNX2、 BMP4、 Smad5、 TAZ、 MSX2、 DLX5、 BMPR2、 Wnt5a, 造血相关基因包括 c-Myb、 EGR1、 FOGl、 SCL、 E47、 Ikaros、 GATA1、 BCL-6, 肝上皮分化相关基因包括 Mxill、 GSC、 Soxl7、 PROX1、 HNF1B、 HNF6、 E-cadherin、 Foxa-l Foxa-2 SNAI1、 NEUROD2、 GFRA2。
8、根据权利要求 3至 Ί任一项所述的全能性基因和 /或分化相关基因的组蛋白 修饰状态作为预测干细胞分化潜能的表观遗传修饰标签的用途, 其特征在于检测 所述全能性基因和 /或分化相关基因的组蛋白甲基化修饰状态时使用 ChlP-seq 或
9、根据权利要求 3至 8任一项所述的全能性基因和 /或分化相关基因的组蛋白 修饰状态作为预测干细胞分化潜能的表观遗传修饰标签的用途, 其特征在于所述 全能性基因和 /或分化相关基因的不同组蛋白甲基化状态指示了干细胞的不同分化 潜能,某谱系分化相关基因组蛋白甲基化修饰是组蛋白 H3第 4位赖氨酸三甲基化 修饰和组蛋白 H3第 4位赖氨酸三甲基化修饰与组蛋白 H3第 27位赖氨酸三甲基 化修饰并存为主, 则指示这种干细胞具有向该谱系分化的潜能, 两种或多种干细 胞进行比较,则该谱系相关基因总体上受组蛋白 H3第 4位赖氨酸三甲基化修饰和 组蛋白 H3第 4位赖氨酸三甲基化修饰与组蛋白 H3第 27位赖氨酸三甲基化修饰 并存的基因所占比例高的干细胞更易于向该谱系分化。
PCT/CN2010/079608 2010-12-09 2010-12-09 预测干细胞分化潜能的表观遗传修饰标签 WO2012075636A1 (zh)

Priority Applications (8)

Application Number Priority Date Filing Date Title
PCT/CN2010/079608 WO2012075636A1 (zh) 2010-12-09 2010-12-09 预测干细胞分化潜能的表观遗传修饰标签
US13/992,619 US9523074B2 (en) 2010-12-09 2011-12-02 Sub-totipotent stem cell product and apparent hereditary modifying label thereof
CN201180057318.8A CN103459592B (zh) 2010-12-09 2011-12-02 亚全能干细胞产品及其表观遗传修饰标签
EP11847131.7A EP2636732B1 (en) 2010-12-09 2011-12-02 Sub-totipotent stem cell product and apparent hereditary modifying label thereof
AU2011341213A AU2011341213B2 (en) 2010-12-09 2011-12-02 Sub-totipotent stem cell product and apparent hereditary modifying label thereof
CA2820395A CA2820395C (en) 2010-12-09 2011-12-02 Sub-totipotent stem cell product and epigenetic modification label thereof
PCT/CN2011/083380 WO2012075912A1 (zh) 2010-12-09 2011-12-02 亚全能干细胞产品及其表观遗传修饰标签
IN1005MUN2013 IN2013MN01005A (zh) 2010-12-09 2011-12-02

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2010/079608 WO2012075636A1 (zh) 2010-12-09 2010-12-09 预测干细胞分化潜能的表观遗传修饰标签

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US13/488,695 Continuation US9806850B2 (en) 2009-12-07 2012-06-05 Method and apparatus for transmitting uplink control information

Publications (1)

Publication Number Publication Date
WO2012075636A1 true WO2012075636A1 (zh) 2012-06-14

Family

ID=46206539

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/CN2010/079608 WO2012075636A1 (zh) 2010-12-09 2010-12-09 预测干细胞分化潜能的表观遗传修饰标签
PCT/CN2011/083380 WO2012075912A1 (zh) 2010-12-09 2011-12-02 亚全能干细胞产品及其表观遗传修饰标签

Family Applications After (1)

Application Number Title Priority Date Filing Date
PCT/CN2011/083380 WO2012075912A1 (zh) 2010-12-09 2011-12-02 亚全能干细胞产品及其表观遗传修饰标签

Country Status (6)

Country Link
US (1) US9523074B2 (zh)
EP (1) EP2636732B1 (zh)
AU (1) AU2011341213B2 (zh)
CA (1) CA2820395C (zh)
IN (1) IN2013MN01005A (zh)
WO (2) WO2012075636A1 (zh)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103667190A (zh) * 2012-09-07 2014-03-26 上海吉凯基因化学技术有限公司 一种诱导形成神经元的方法和组合物
CN110499283A (zh) * 2018-05-17 2019-11-26 西安组织工程与再生医学研究所 Wnt信号通路激活剂在制备改善低碱性磷酸酯酶症干细胞成骨分化能力异常产品中的应用

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103865873B (zh) * 2012-12-12 2017-04-05 中国医学科学院基础医学研究所 亚全能干细胞分泌的外来体及其应用

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007085827A2 (en) * 2006-01-26 2007-08-02 The University Of Birmingham Epigenetic analyses

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7056738B2 (en) * 2001-03-23 2006-06-06 Tulane University Early stage multipotential stem cells in colonies of bone marrow stromal cells
JP2004248505A (ja) * 2001-09-21 2004-09-09 Norio Nakatsuji 移植抗原の一部または全てを欠除したes細胞由来の未分化な体細胞融合細胞およびその製造
CN1778905B (zh) * 2004-11-22 2010-05-05 赵春华 一种脂肪间充质干细胞的分离培养方法及其应用
CN101821383B (zh) * 2007-09-05 2013-12-18 中国医学科学院基础医学研究所 一种用于人成体原始间充质干细胞体外规模化培养的培养基、方法及获得的原始间充质干细胞及其应用
US8524498B2 (en) * 2009-05-29 2013-09-03 The General Hospital Corporation Methods and compositions for homologous recombination in human cells

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007085827A2 (en) * 2006-01-26 2007-08-02 The University Of Birmingham Epigenetic analyses

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BRADLEY E. BERNSTEIN ET AL.: "A Bivalent Chromatin Structure Marks Key Developmental Genes in Embryonic Stem Cells", CELL, vol. 125, 2006, pages 315 - 326 *
TARJEI S. MIKKELSEN ET AL.: "Genome-wide maps of chromatin state in pluripotent and lineage-committed cells", NATURE, vol. 448, 2007, pages 553 - 562 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103667190A (zh) * 2012-09-07 2014-03-26 上海吉凯基因化学技术有限公司 一种诱导形成神经元的方法和组合物
CN110499283A (zh) * 2018-05-17 2019-11-26 西安组织工程与再生医学研究所 Wnt信号通路激活剂在制备改善低碱性磷酸酯酶症干细胞成骨分化能力异常产品中的应用

Also Published As

Publication number Publication date
WO2012075912A9 (zh) 2013-09-06
AU2011341213B2 (en) 2016-08-18
CA2820395C (en) 2017-03-07
US20140287930A1 (en) 2014-09-25
US9523074B2 (en) 2016-12-20
EP2636732A4 (en) 2014-05-07
EP2636732B1 (en) 2018-05-30
IN2013MN01005A (zh) 2015-09-25
WO2012075912A1 (zh) 2012-06-14
EP2636732A1 (en) 2013-09-11
AU2011341213A1 (en) 2013-07-11
CA2820395A1 (en) 2012-06-14

Similar Documents

Publication Publication Date Title
Okae et al. Derivation of human trophoblast stem cells
Sandberg et al. Comparison of glioma stem cells to neural stem cells from the adult human brain identifies dysregulated Wnt-signaling and a fingerprint associated with clinical outcome
Hammoud et al. Chromatin and transcription transitions of mammalian adult germline stem cells and spermatogenesis
Mahmud et al. Differential effects of epigenetic modifiers on the expansion and maintenance of human cord blood stem/progenitor cells
Raynaud et al. Endothelial cells provide a niche for placental hematopoietic stem/progenitor cell expansion through broad transcriptomic modification
Chiriac et al. SDF-1-enhanced cardiogenesis requires CXCR4 induction in pluripotent stem cells
Jiang et al. Maintenance of human haematopoietic stem and progenitor cells in vitro using a chemical cocktail
CN103459592A (zh) 亚全能干细胞产品及其表观遗传修饰标签
WO2012075636A1 (zh) 预测干细胞分化潜能的表观遗传修饰标签
Suthapot et al. The RNA helicases DDX5 and DDX17 facilitate neural differentiation of human pluripotent stem cells NTERA2
US20130309209A1 (en) Formation of hematopoietic progenitor cells from mesenchymal stem cells
Han et al. Global transcriptome profiling of genes that are differentially regulated during differentiation of mouse embryonic neural stem cells into astrocytes
Liedtke et al. Neonatal mesenchymal-like cells adapt to surrounding cells
WO2018037091A1 (en) Methods for the identification and isolation of hematopoietic stem and progenitor cells
JP2023550036A (ja) 富化された生体活性腎細胞集団、その特徴及び使用
Mazzella et al. Regulation of self-renewal and senescence in primitive mesenchymal stem cells by Wnt and TGFβ signaling
Ma et al. Genome-wide screening of differentially expressed genes and their potential associations with aging dental pulp stem cells
Iftakhar-e-Khuda et al. Vascular adhesion protein-1 defines a unique subpopulation of human hematopoietic stem cells and regulates their proliferation
US20120045758A1 (en) Imprinting in very small embryonic-like (vsel) stem cells
Lee et al. The Transcription Factor CP2-like 1 Is Expressed in Very Small Embryonic-like Stem Cells and Other Adult Stem Cells: Implications for Cancer Stem Cells
Chamling et al. Single-Cell Transcriptomic Analysis Reveals Molecular Diversity of Human Oligodendrocyte Progenitor Cells
Creamer The role of CDX4 during patterning of definitive hemogenic mesoderm
Mazurier et al. A new signaling cascade linking BMP4, BMPR1A, ΔNp73 and NANOG impacts on stem-like human cell properties and patient outcome
EP1961810A1 (en) Method for obtaining intestinal stem-precursor cell
Haque Innate Immune Protein C1q Modulation of Endothelial Cell Responses in Atherosclerosis

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: 10860558

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: 10860558

Country of ref document: EP

Kind code of ref document: A1